Androgen receptors beyond prostate cancer: an old marker as a new target

Javier Munoz, Jennifer J Wheler, Razelle Kurzrock, Javier Munoz, Jennifer J Wheler, Razelle Kurzrock

Abstract

Androgen receptors (ARs) play a critical role in the development of prostate cancer. Targeting ARs results in important salutary effects in this malignancy. Despite mounting evidence that ARs also participate in the pathogenesis and/or progression of diverse tumors, exploring the impact of hormonal manipulation of these receptors has not been widely pursued beyond prostate cancer. This review describes patterns of AR expression in a spectrum of cancers, and the potential to exploit this knowledge in the clinical therapeutic setting.

Conflict of interest statement

Conflicts of Interest

The authors have no conflicts of interests to disclose.

Figures

Figure 1. The road to (androgen) independence
Figure 1. The road to (androgen) independence
AR-negative prostate adult stem cells generate AR-negative transit amplifying cells, AR-negative intermediate cells and AR-positive luminal secretory cells. Once therapeutic circulating androgen blockade ensues, AR-positive luminal cells die, giving origin to the adaptive (selected AR-positive luminal cells will continue to thrive despite lack of androgen exposure) and clonal theory of resistance (pre-existent AR-negative cells develop into a malignant clone). Castration-resistant prostate cancer might develop via ligand-dependent (tissue steroidogenesis, AR mutations, AR amplification) and ligand-independent pathways (heightened AR nuclear translocation, AR cross-talk with additional pathways, disturbing the balance between co-activators and co-repressors).
Figure 2. The fate of testosterone in…
Figure 2. The fate of testosterone in prostatic tissues
Testosterone circulates in the blood and is bound to albumin, whereas free testosterone is introduced into prostate cells and is subsequently converted to DHT by 5-alpha-reductase. Binding of DHT to the AR induces dissociation from HSPs and receptor phosphorylation. The AR dimerizes and can bind to androgen-response elements in the promoter regions of target genes, leading to growth, survival and production of PSA. Enzalutamide, formerly called MDV3100, exerts its mechanism of action during several steps in the AR signaling pathway including inhibition of AR binding to androgens, inhibition of nuclear translocation of AR, inhibition of AR association to DNA, and AR amplification. As some of those aberrations may occur late in the disease, it is unknown at this point if enzalutamide will have a role upfront in the management of prostate cancer. Abbreviations: AR, androgen receptor; DHT, dihydrotestosterone; GTA, general transcription activation; HSP, heat-shock protein; SHBG, sex-hormone-binding globulin; AKT, akt serine/threonine kinase; DHEA, dihydroepiandrosterone; ERK, extracellular signal-regulated kinase; P, phosphorylated residues; PI3K, phosphoinositide 3-kinase; PTEN, phoshatase and tensin homolog.
Figure 3. Androgen blockade plus mTOR inhibitors:…
Figure 3. Androgen blockade plus mTOR inhibitors: A prime candidate for combination hormonal treatment
The mTORC2/AKT/AR pathway (red arrows) leads to tumor cell proliferation. Rapamycin combined with bicalutamide has an apoptosis-inducing effect in prostate cancer. Rapamycin inhibits both mTOR complexes, mTORC1 with raptor and mTORC2 with rictor; nevertheless abrogation of mTORC2, a kinase for AKT phosphorylation, further inhibits the AR transcription cascade in an AKT-dependent manner. As a counterpoint, abrogation of mTORC1 produces AKT/AR-independent apoptosis, though it continues to stimulate the AR transcriptional cascade and AKT phosphorylation. According to the suggested cross-talk, it would take a combination of androgen blockade plus mTOR inhibitors to fully abrogate the mTORC2/AKT/AR pathway. Barnett et al. [44] found that, out of 47 tumors evaluable by immunohistochemistry, 36% had PTEN loss which was associated with an increased relapse in high risk prostate cancer treated with chemotherapy followed by surgery. PTEN loss activates the AKT/mTOR pathway [45] thus supporting the use of mTOR inhibitors in this condition. Abbreviations: AR, androgen receptor; AKT, aKT serine/threonine kinase.

References

    1. Loblaw DA, Virgo KS, Nam R, et al. Initial hormonal management of androgen-sensitive metastatic, recurrent, or progressive prostate cancer: 2006 update of an American Society of Clinical Oncology practice guideline. J Clin Oncol. 2007;25(12):1596–605.
    1. Askew EB, Gampe RT, Jr, Stanley TB, et al. Modulation of androgen receptor activation function 2 by testosterone and dihydrotestosterone. J Biol Chem. 2007;282(35):25801–16.
    1. Sandow J, von Rechenberg W, Engelbart K. Pharmacological studies on androgen suppression in therapy of prostate carcinoma. Am J Clin Oncol. 1988;11 Suppl 1:S6–10.
    1. Davison SL, Bell R. Androgen physiology. Semin Reprod Med. 2006;24(2):71–7.
    1. Clark AJ, Metherell LA. Mechanisms of disease: the adrenocorticotropin receptor and disease. Nat Clin Pract Endocrinol Metab. 2006;2(5):282–90.
    1. Rittmaster RS, Zwicker H, Thompson DL, et al. Androstanediol glucuronide production in human liver, prostate, and skin. Evidence for the importance of the liver in 5 alpha-reduced androgen metabolism. J Clin Endocrinol Metab. 1993;76(4):977–82.
    1. Valle LD, Toffolo V, Nardi A, et al. Tissue-specific transcriptional initiation and activity of steroid sulfatase complementing dehydroepiandrosterone sulfate uptake and intracrine steroid activations in human adipose tissue. J Endocrinol. 2006;190(1):129–39.
    1. Thomas BC, Neal DE. Androgen deprivation treatment in prostate cancer. BMJ. 2013;346:e8555.
    1. Lepor H. Comparison of single-agent androgen suppression for advanced prostate cancer. Rev Urol. 2005;7 Suppl 5:S3–S12.
    1. Lepor H, Shore ND. LHRH Agonists for the Treatment of Prostate Cancer: 2012. Rev Urol. 2012;14(1-2):1–12.
    1. Rick FG, Block NL, Schally AV. An update on the use of degarelix in the treatment of advanced hormone-dependent prostate cancer. Onco Targets Ther. 2013;6:391–402.
    1. Gauthier S, Martel C, Labrie F. Steroid derivatives as pure antagonists of the androgen receptor. J Steroid Biochem Mol Biol. 2012;132(1-2):93–104.
    1. Freedland SJ, Eastham J, Shore N. Androgen deprivation therapy and estrogen deficiency induced adverse effects in the treatment of prostate cancer. Prostate Cancer Prostatic Dis. 2009;12(4):333–8.
    1. Carruba G. Estrogen and prostate cancer: an eclipsed truth in an androgen-dominated scenario. J Cell Biochem. 2007;102(4):899–911.
    1. Langley RE, Cafferty FH, Alhasso AA, et al. Cardiovascular outcomes in patients with locally advanced and metastatic prostate cancer treated with luteinising-hormone-releasing-hormone agonists or transdermal oestrogen: the randomised, phase 2 MRC PATCH trial (PR09) Lancet Oncol. 2013;14(4):306–16.
    1. Peer A, Gottfried M, Sinibaldi V, et al. Comparison of abiraterone acetate versus ketoconazole in patients with metastatic castration resistant prostate cancer refractory to docetaxel. Prostate. 2013
    1. Salem M, Garcia JA. Abiraterone acetate, a novel adrenal inhibitor in metastatic castration-resistant prostate cancer. Curr Oncol Rep. 2011;13(2):92–6.
    1. Gardner JR, Livingston PM, Fraser SF. Effects of Exercise on Treatment-Related Adverse Effects for Patients With Prostate Cancer Receiving Androgen-Deprivation Therapy: A Systematic Review. J Clin Oncol. 2013
    1. Sountoulides P, Rountos T. Adverse effects of androgen deprivation therapy for prostate cancer: prevention and management. ISRN Urol. 2013;2013:240108.
    1. Cannata DH, Kirschenbaum A, Levine AC. Androgen deprivation therapy as primary treatment for prostate cancer. J Clin Endocrinol Metab. 2012;97(2):360–5.
    1. Yap TA, Zivi A, Omlin A, et al. The changing therapeutic landscape of castration-resistant prostate cancer. Nat Rev Clin Oncol. 2011;8(10):597–610.
    1. Hammes SR, Levin ER. Extranuclear steroid receptors: nature and actions. Endocr Rev. 2007;28(7):726–41.
    1. Tombal B. What is the pathophysiology of a hormone-resistant prostate tumour? Eur J Cancer. 2011;47 Suppl 3:S179–88.
    1. Saraon P, Jarvi K, Diamandis EP. Molecular alterations during progression of prostate cancer to androgen independence. Clin Chem. 2011;57(10):1366–75.
    1. Geller J, Albert JD. Comparison of various hormonal therapies for prostatic carcinoma. Semin Oncol. 1983;10(4 Suppl 4):34–41.
    1. Jacobi GH, Wenderoth UK. Gonadotropin-releasing hormone analogues for prostate cancer: untoward side effects of high-dose regimens acquire a therapeutical dimension. Eur Urol. 1982;8(3):129–34.
    1. Ricke WA, Wang Y, Cunha GR. Steroid hormones and carcinogenesis of the prostate: the role of estrogens. Differentiation. 2007;75(9):871–82.
    1. Shah S, Ryan C. Abiraterone acetate for castration resistant prostate cancer. Expert Opin Investig Drugs. 2010;19(4):563–70.
    1. Harris WP, Mostaghel EA, Nelson PS, et al. Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat Clin Pract Urol. 2009;6(2):76–85.
    1. Hussain M, Tangen CM, Higano CS, et al. Intermittent (IAD) versus continuous androgen deprivation (CAD) in hormone sensitive metastatic prostate cancer (HSM1PC) patients (pts): Results of S9346 (INT-0162), an international phase III trial. J Clin Oncol. 2012;30(18_suppl)
    1. Crook JM, O'Callaghan CJ, Duncan G, et al. Intermittent androgen suppression for rising PSA level after radiotherapy. N Engl J Med. 2012;367(10):895–903.
    1. Scher HI, Beer TM, Higano CS, et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study. Lancet. 2010;375(9724):1437–46.
    1. Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–97.
    1. DeBono JS, Fizazi K, Saad F, et al. Primary, secondary, and quality-of-life endpoint results from the phase III AFFIRM study of MDV3100, an androgen receptor signaling inhibitor. J Clin Oncol. 2012;30(15_suppl)
    1. Ryan CJ, Tindall DJ. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically. J Clin Oncol. 2011;29(27):3651–8.
    1. Wassersug RJ. Mastering emasculation. J Clin Oncol. 2009;27(4):634–6.
    1. Kobayashi T, Shimizu Y, Terada N, et al. Regulation of androgen receptor transactivity and mTOR-S6 kinase pathway by Rheb in prostate cancer cell proliferation. Prostate. 2010;70(8):866–74.
    1. Wang Y, Mikhailova M, Bose S, et al. Regulation of androgen receptor transcriptional activity by rapamycin in prostate cancer cell proliferation and survival. Oncogene. 2008;27(56):7106–17.
    1. McCall P, Gemmell LK, Mukherjee R, et al. Phosphorylation of the androgen receptor is associated with reduced survival in hormone-refractory prostate cancer patients. Br J Cancer. 2008;98(6):1094–101.
    1. Roychowdhury S, Chinnaiyan AM. Advancing precision medicine for prostate cancer through genomics. J Clin Oncol. 2013;31(15):1866–73.
    1. Recchia AG, Musti AM, Lanzino M, et al. A cross-talk between the androgen receptor and the epidermal growth factor receptor leads to p38MAPK-dependent activation of mTOR and cyclinD1 expression in prostate and lung cancer cells. Int J Biochem Cell Biol. 2009;41(3):603–14.
    1. Gonzalez-Angulo AM, Stemke-Hale K, Palla SL, et al. Androgen receptor levels and association with PIK3CA mutations and prognosis in breast cancer. Clin Cancer Res. 2009;15(7):2472–8.
    1. Wang Y, Romigh T, He X, et al. Differential regulation of PTEN expression by androgen receptor in prostate and breast cancers. Oncogene. 2011;30(42):4327–38.
    1. Barnett CM, Heinrich MC, Lim J, et al. Genetic Profiling to Determine Risk of Relapse Free Survival in High-risk Localized Prostate Cancer. Clin Cancer Res. 2013
    1. Zhang W, Haines BB, Efferson C, et al. Evidence of mTOR Activation by an AKT-Independent Mechanism Provides Support for the Combined Treatment of PTEN-Deficient Prostate Tumors with mTOR and AKT Inhibitors. Transl Oncol. 2012;5(6):422–9.
    1. Mulder E, van Loon D, de Boer W, et al. Mechanism of androgen action: recent observations on the domain structure of androgen receptors and the induction of EGF-receptors by androgens in prostate tumor cells. J Steroid Biochem. 1989;32(1B):151–6.
    1. Fiorelli A, Ricciardi C, Pannone G, et al. Interplay between steroid receptors and neoplastic progression in sarcoma tumors. J Cell Physiol. 2011;226(11):2997–3003.
    1. Chia KM, Liu J, Francis GD, et al. A feedback loop between androgen receptor and ERK signaling in estrogen receptor-negative breast cancer. Neoplasia. 2011;13(2):154–66.
    1. Naderi A, Hughes-Davies L. A functionally significant cross-talk between androgen receptor and ErbB2 pathways in estrogen receptor negative breast cancer. Neoplasia. 2008;10(6):542–8.
    1. Naderi A, Chia KM, Liu J. Synergy between inhibitors of androgen receptor and MEK has therapeutic implications in estrogen receptor-negative breast cancer. Breast Cancer Res. 2011;13(2):R36.
    1. Wang Y, Yang J, Gao Y, et al. Reciprocal regulation of 5alpha-dihydrotestosterone, interleukin-6 and interleukin-8 during proliferation of epithelial ovarian carcinoma. Cancer Biol Ther. 2007;6(6):864–71.
    1. Darshan MS, Loftus MS, Thadani-Mulero M, et al. Taxane-induced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer. Cancer Res. 2011;71(18):6019–29.
    1. Ware KE, Garcia-Blanco MA, Armstrong AJ, Dehm Sm. Biologic and clincial significance of androgen receptor varianrts n castration resistant prostate cancer. Endocr Relat Cancer. 2014;21(4):T87–T103. Aug.
    1. McNamara KM1, Moore NL1, Hickey TE1, Sasano H1. Tilley WDComplexities of androgen receptor signalling in breast cancer. Endocr Relat Cancer. 2014;21(4):T161–81. Aug. doi: 10.1530/ERC-14-0243. Epub 2014 Jun 2.
    1. Mechlin CW, Frankel J, McCullough A. Coadministration of anastrozole sustains therapeutic testosterone levels in hypogonadal men undergoing testosterone Pellet insertion. J Sex Med. 2013

Source: PubMed

3
Abonnere