Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide

Dawn R Cochrane, Sebastián Bernales, Britta M Jacobsen, Diana M Cittelly, Erin N Howe, Nicholas C D'Amato, Nicole S Spoelstra, Susan M Edgerton, Annie Jean, Javier Guerrero, Francisco Gómez, Satyanarayana Medicherla, Iván E Alfaro, Emma McCullagh, Paul Jedlicka, Kathleen C Torkko, Ann D Thor, Anthony D Elias, Andrew A Protter, Jennifer K Richer, Dawn R Cochrane, Sebastián Bernales, Britta M Jacobsen, Diana M Cittelly, Erin N Howe, Nicholas C D'Amato, Nicole S Spoelstra, Susan M Edgerton, Annie Jean, Javier Guerrero, Francisco Gómez, Satyanarayana Medicherla, Iván E Alfaro, Emma McCullagh, Paul Jedlicka, Kathleen C Torkko, Ann D Thor, Anthony D Elias, Andrew A Protter, Jennifer K Richer

Abstract

Introduction: The androgen receptor (AR) is widely expressed in breast cancers and has been proposed as a therapeutic target in estrogen receptor alpha (ER) negative breast cancers that retain AR. However, controversy exists regarding the role of AR, particularly in ER + tumors. Enzalutamide, an AR inhibitor that impairs nuclear localization of AR, was used to elucidate the role of AR in preclinical models of ER positive and negative breast cancer.

Methods: We examined nuclear AR to ER protein ratios in primary breast cancers in relation to response to endocrine therapy. The effects of AR inhibition with enzalutamide were examined in vitro and in preclinical models of ER positive and negative breast cancer that express AR.

Results: In a cohort of 192 women with ER + breast cancers, a high ratio of AR:ER (≥2.0) indicated an over four fold increased risk for failure while on tamoxifen (HR = 4.43). The AR:ER ratio had an independent effect on risk for failure above ER % staining alone. AR:ER ratio is also an independent predictor of disease-free survival (HR = 4.04, 95% CI: 1.68, 9.69; p = 0.002) and disease specific survival (HR = 2.75, 95% CI: 1.11, 6.86; p = 0.03). Both enzalutamide and bicalutamide inhibited 5-alpha-dihydrotestosterone (DHT)-mediated proliferation of breast cancer lines in vitro; however, enzalutamide uniquely inhibited estradiol (E2)-mediated proliferation of ER+/AR + breast cancer cells. In MCF7 xenografts (ER+/AR+) enzalutamide inhibited E2-driven tumor growth as effectively as tamoxifen by decreasing proliferation. Enzalutamide also inhibited DHT- driven tumor growth in both ER positive (MCF7) and negative (MDA-MB-453) xenografts, but did so by increasing apoptosis.

Conclusions: AR to ER ratio may influence breast cancer response to traditional endocrine therapy. Enzalutamide elicits different effects on E2-mediated breast cancer cell proliferation than bicalutamide. This preclinical study supports the initiation of clinical studies evaluating enzalutamide for treatment of AR+ tumors regardless of ER status, since it blocks both androgen- and estrogen- mediated tumor growth.

Figures

Figure 1
Figure 1
Women with tumors having a higher AR:ER ratio have a shorter disease-free and disease-specific overall survival as compared with patients with lower AR:ER ratio. Immunohistochemistry for androgen receptor (AR) and estrogen receptor (ER) were performed on formalin-fixed paraffin-embedded sections of primary breast cancers. Slides were scored for the percent of positive nuclear staining for AR and ER. Ratios were calculated to determine the best cutoff point for analysis. For (A) to (D) women are divided into two groups: those with AR:ER ratios <2.0 (blue squares) and those with AR:ER ratios ≥2.0 (red circles). The number of patients at risk at each time point is reflective of the number of patients censored due to no further follow-up data at each time point (underneath). Kaplan–Meier survival curve for: (A) disease-free survival (DFS) for all patients; (B) disease-specific survival (DSS) overall for all patients; (C) DFS for patients who failed while on tamoxifen therapy; (D) DSS overall for patients who failed while on tamoxifen therapy; and (E) representative images of AR and ER staining from the two groups (400× magnification).
Figure 2
Figure 2
Enzalutamide abrogates androgen mediated proliferation in estrogen receptor-positive breast cancer cells. (A) Baseline levels of androgen receptor (AR) and estrogen receptor (ER) alpha protein in whole cell lysates from ER-positive (MCF7, BCK4, T47D and ZR-75-1) and ER-negative (MDA-MB-453) breast cancer and prostate (LNCaP) cancer cell lines. (B) AR protein levels in MCF7 cells plated in charcoal-stripped serum-containing media for 48 hours prior to treatment with vehicle control, 10 nM 5-alpha-dihydrotestosterone (DHT), 10 μM enzalutamide (Enza) or a combination of DHT and Enza for 48 hours. (C) MCF7 and (D) BCK4 breast cancer cells, both ER + AR+, were treated with vehicle control, 10 nM DHT, 10 μM Enza or a combination of DHT and Enza, and MTS proliferation assays were performed. Error bars represent standard error of the mean. *P < 0.05, ***P < 0.001 for DHT versus DHT + Enza, analysis of variance with Bonferroni’s multiple comparison test correction. (E) AR levels in cytosolic and nuclear fractions of MCF7 cells treated with vehicle, 10 nM DHT, 10 μM enzalutamide or DHT + Enza for 3 hours.
Figure 3
Figure 3
Enzalutamide inhibits androgen-stimulated growth of MCF7 tumors in vivo. MCF7-TGL cells stably expressing luciferase were implanted orthotopically in the mammary gland of NOD/SCID ovariectomized female mice with a 5-alpha-dihydrotestosterone (DHT) pellet implanted subcutaneously. Mice were matched into two groups based on tumor volume (day -2) and treatment with either control chow (DHT) or chow containing 50 mg/kg enzalutamide (DHT + Enza) begun (day 0, indicated by arrow), and the tumor burden was measured by whole-body luminescence. (A) Mean total flux of all mice in each of the treatment groups. Error bar represents standard error of the mean. *P < 0.05, Wilcoxon rank sum. (B) Total luminescent flux is shown for all individual mice at the day of matching (day -2) and at the final imaging day (day 19). *P < 0.05, Wilcoxon rank sum. (C) Mice were injected with bromodeoxyuridine (BrdU) 2 hours prior to sacrifice and BrdU immunohistochemistry was performed on formalin-fixed paraffin-embedded tumor sections and quantified. *P < 0.05, Student’s t test. Representative images of BrdU staining (400× magnification) and quantification. (D) Quantification of apoptotic cells as measured by terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) staining with representative images below (400× magnification). *P < 0.05, Student’s t test. (E) Quantification of nuclear AR staining and representative images (400× magnification). ***P < 0.001, Wilcoxon rank sum.
Figure 4
Figure 4
Enzalutamide inhibits androgen-stimulated growth of MDA-MB-453 tumors. MDA-MB-453 cells were injected orthotopically in the mammary gland of female NOD-SCID-IL2Rgc-/- mice. Three groups had a 5-alpha-dihydrotestosterone (DHT) pellet implanted subcutaneously and one group had no pellet (Vehicle). Once tumors reached an average size of 100 mm3 (green arrow), mice were given either enzalutamide (Enza, 10 mg/kg) or vehicle (Vehicle and DHT groups) by daily oral gavage. Another group was given a higher dose of Enza (25 mg/kg) by oral gavage when tumors reached an average size of 400 mm3 (blue arrow). (A) Tumor volume was measured weekly by caliper. Error bars represent standard error of the mean. *P < 0.05, **P < 0.01 for DHT versus DHT + Enza (10 mg/kg), Wilcoxon rank sum. (B) Tumors were excised and weighed at the end of the experiment. (C) Tumor sections stained for cleaved caspase 3 were quantified (left) and representative images shown (right) (200× magnification). *P < 0.05, **P < 0.01, ***P < 0.001, analysis of variance with Bonferroni’s multiple comparison test correction. (D) Nuclear androgen receptor staining was quantified (left) and representative images (400× magnification) are shown (right). *P < 0.05, ***P < 0.001, Kruskal–Wallis with Dunn’s multiple comparison test correction.
Figure 5
Figure 5
Enzalutamide inhibits estradiol-mediated proliferation of breast cancer cells, while bicalutamide does not. MTS proliferation assays were performed on (A) MCF7s cells and (B) BCK4 cells treated with vehicle control, 10 nM estradiol (E2), 10 μM enzalutamide (Enza) or a combination of E2 and Enza. Error bars represent standard error of the mean (SEM). **P < 0.01, ***P < 0.001 for E2 versus E2 + Enza, analysis of variance (ANOVA) with Bonferroni’s multiple comparison test correction. (C) MCF7 cells were treated for 48 hours with treatments as above and real-time polymerase chain reaction (PCR) was performed for estrogen-responsive genes, progesterone receptor (PR) and stromal cell-derived factor 1 (SDF-1, also known as CXCL12). Each gene is normalized to 18S and shown relative to vehicle. *P < 0.05, ***P < 0.001, Student’s t test. MCF7 cells were treated with vehicle control, (D) 1 μM bicalutamide, 10 nM 5-alpha-dihydrotestosterone (DHT) and DHT + bicalutamide or (E) with 10 nM E2 and E2 + bicalutamide. **P < 0.01, ***P < 0.001 for DHT versus DHT + bicalutamide, or E2 versus E2 + bicalutamide, ANOVA with Bonferroni’s multiple comparison test correction. (F) MCF7 cells treated for 48 hours with vehicle, 1 μM bicalutamide, 10 nM E2 and E2 + bicalutamide and real-time PCR performed for PR and SDF-1. *P < 0.05, ***P < 0.001, Student’s t test. Error bars represent SEM, Student’s t test (all analyses).
Figure 6
Figure 6
Enzalutamide inhibits estrogen-stimulated growth of MCF7 tumors as effectively as tamoxifen. MCF7-TGL cells stably expressing luciferase were implanted orthotopically in the mammary gland of ovariectomized female nude mice. All mice had an estradiol (E2) pellet implanted subcutaneously and were given either control chow (E2), control chow plus a tamoxifen pellet implanted subcutaneously (E2 + Tam) or chow containing 50 mg/kg enzalutamide (E2 + Enza). The tumor burden was measured by whole-body luminescence. (A) Mean total flux. Mice were matched on day -3 and treatment began on day 0 (arrow). *P < 0.05, analysis of variance (ANOVA) with Bonferroni’s multiple comparison test correction. (B) The total luminescent flux is shown for individual mice on the day of matching (day -3) and of final imaging (day 11). *P < 0.05, ANOVA with Bonferroni’s multiple comparison test correction. (C) Images of luminescent signal in the two treatment groups at time of matching (day -3) and the final day of imaging (day 11). (D) Mice were injected with bromodeoxyuridine (BrdU) 2 hours prior to sacrifice and immunohistochemistry for BrdU was performed on tumor sections and quantified using imageJ (National Institutes of Health, Bethesda, MD, USA). Representative images of BrdU staining (left, 400× magnification) and quantification (right). **P < 0.01 for E2 versus E2 + Tam, ***P < 0.001 for E2 versus E2 + Enza, ANOVA with Bonferroni’s multiple comparison test correction.

References

    1. Guedj M, Marisa L, de Reynies A, Orsetti B, Schiappa R, Bibeau F, MacGrogan G, Lerebours F, Finetti P, Longy M, Bertheau P, Bertrand F, Bonnet F, Martin AL, Feugeas JP, Bieche I, Lehmann-Che J, Lidereau R, Birnbaum D, Bertucci F, de The H, Theillet C. A refined molecular taxonomy of breast cancer. Oncogene. 2012;31:1196–1206. doi: 10.1038/onc.2011.301.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750–2767. doi: 10.1172/JCI45014.
    1. Collins LC, Cole KS, Marotti JD, Hu R, Schnitt SJ, Tamimi RM. Androgen receptor expression in breast cancer in relation to molecular phenotype: results from the Nurses' Health Study. Mod Pathol. 2011;24:924–931. doi: 10.1038/modpathol.2011.54.
    1. Park S, Koo J, Park HS, Kim JH, Choi SY, Lee JH, Park BW, Lee KS. Expression of androgen receptors in primary breast cancer. Ann Oncol. 2010;21:488–492. doi: 10.1093/annonc/mdp510.
    1. Hu R, Dawood S, Holmes MD, Collins LC, Schnitt SJ, Cole K, Marotti JD, Hankinson SE, Colditz GA, Tamimi RM. Androgen receptor expression and breast cancer survival in postmenopausal women. Clin Cancer Res. 2011;17:1867–1874. doi: 10.1158/1078-0432.CCR-10-2021.
    1. Bergh J, Jonsson PE, Lidbrink EK, Trudeau M, Eiermann W, Brattstrom D, Lindemann JP, Wiklund F, Henriksson R. FACT: an open-label randomized phase III study of fulvestrant and anastrozole in combination compared with anastrozole alone as first-line therapy for patients with receptor-positive postmenopausal breast cancer. J Clin Oncol. 2012;30:1919–1925. doi: 10.1200/JCO.2011.38.1095.
    1. Mouridsen H, Gershanovich M, Sun Y, Perez-Carrion R, Boni C, Monnier A, Apffelstaedt J, Smith R, Sleeboom HP, Jaenicke F, Pluzanska A, Dank M, Becquart D, Bapsy PP, Salminen E, Snyder R, Chaudri-Ross H, Lang R, Wyld P, Bhatnagar A. Phase III study of letrozole versus tamoxifen as first-line therapy of advanced breast cancer in postmenopausal women: analysis of survival and update of efficacy from the International Letrozole Breast Cancer Group. J Clin Oncol. 2003;21:2101–2109. doi: 10.1200/JCO.2003.04.194.
    1. Harvell DM, Richer JK, Singh M, Spoelstra N, Finlayson C, Borges VF, Elias AD, Horwitz KB. Estrogen regulated gene expression in response to neoadjuvant endocrine therapy of breast cancers: tamoxifen agonist effects dominate in the presence of an aromatase inhibitor. Breast Cancer Res Treat. 2008;112:489–501. doi: 10.1007/s10549-008-9923-6.
    1. Harvell DM, Spoelstra NS, Singh M, McManaman JL, Finlayson C, Phang T, Trapp S, Hunter L, Dye WW, Borges VF, Elias A, Horwitz KB, Richer JK. Molecular signatures of neoadjuvant endocrine therapy for breast cancer: characteristics of response or intrinsic resistance. Breast Cancer Res Treat. 2008;112:475–488. doi: 10.1007/s10549-008-9897-4.
    1. De Amicis F, Thirugnansampanthan J, Cui Y, Selever J, Beyer A, Parra I, Weigel NL, Herynk MH, Tsimelzon A, Lewis MT, Chamness GC, Hilsenbeck SG, Ando S, Fuqua SA. Androgen receptor overexpression induces tamoxifen resistance in human breast cancer cells. Breast Cancer Res Treat. 2010;121:1–11. doi: 10.1007/s10549-009-0436-8.
    1. Spinola PG, Marchetti B, Merand Y, Belanger A, Labrie F. Effects of the aromatase inhibitor 4-hydroxyandrostenedione and the antiandrogen flutamide on growth and steroid levels in DMBA-induced rat mammary tumors. Breast Cancer Res Treat. 1988;12:287–296. doi: 10.1007/BF01811241.
    1. Dimitrakakis C, Bondy C. Androgens and the breast. Breast Cancer Res. 2009;11:212. doi: 10.1186/bcr2413.
    1. Gallicchio L, Macdonald R, Wood B, Rushovich E, Helzlsouer KJ. Androgens and musculoskeletal symptoms among breast cancer patients on aromatase inhibitor therapy. Breast Cancer Res Treat. 2011;130:569–577. doi: 10.1007/s10549-011-1611-2.
    1. Morris KT, Toth-Fejel S, Schmidt J, Fletcher WS, Pommier RF. High dehydroepiandrosterone-sulfate predicts breast cancer progression during new aromatase inhibitor therapy and stimulates breast cancer cell growth in tissue culture: a renewed role for adrenalectomy. Surgery. 2001;130:947–953. doi: 10.1067/msy.2001.118378.
    1. Niemeier LA, Dabbs DJ, Beriwal S, Striebel JM, Bhargava R. Androgen receptor in breast cancer: expression in estrogen receptor-positive tumors and in estrogen receptor-negative tumors with apocrine differentiation. Mod Pathol. 2010;23:205–212. doi: 10.1038/modpathol.2009.159.
    1. Tsutsumi Y. Apocrine Carcinoma as Triple-negative Breast Cancer: Novel Definition of Apocrine-type Carcinoma as Estrogen/Progesterone Receptor-negative and Androgen Receptor-positive Invasive Ductal Carcinoma. Jpn J Clin Oncol. 2012;42:375–386. doi: 10.1093/jjco/hys034.
    1. Doane AS, Danso M, Lal P, Donaton M, Zhang L, Hudis C, Gerald WL. An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen. Oncogene. 2006;25:3994–4008. doi: 10.1038/sj.onc.1209415.
    1. Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D, Macgrogan G, Bergh J, Cameron D, Goldstein D, Duss S, Nicoulaz AL, Brisken C, Fiche M, Delorenzi M, Iggo R. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene. 2005;24:4660–4671. doi: 10.1038/sj.onc.1208561.
    1. Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG, Carroll JS. Androgen receptor driven transcription in molecular apocrine breast cancer is mediated by FoxA1. Embo J. 2011;30:3019–3027. doi: 10.1038/emboj.2011.216.
    1. Ni M, Chen Y, Lim E, Wimberly H, Bailey ST, Imai Y, Rimm DL, Liu XS, Brown M. Targeting androgen receptor in estrogen receptor-negative breast cancer. Cancer Cell. 2011;20:119–131. doi: 10.1016/j.ccr.2011.05.026.
    1. Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, Blackwell K, Rugo H, Nabell L, Forero A, Stearns V, Doane AS, Danso M, Moynahan ME, Momen LF, Gonzalez JM, Akhtar A, Giri DD, Patil S, Feigin KN, Hudis CA, Traina TA. Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptor-negative metastatic Breast Cancer. Clin Cancer Res. 2013;19:5505–5512. doi: 10.1158/1078-0432.CCR-12-3327.
    1. Masiello D, Cheng S, Bubley GJ, Lu ML, Balk SP. Bicalutamide functions as an androgen receptor antagonist by assembly of a transcriptionally inactive receptor. J Biol Chem. 2002;277:26321–26326. doi: 10.1074/jbc.M203310200.
    1. Small EJ, Carroll PR. Prostate-specific antigen decline after casodex withdrawal: evidence for an antiandrogen withdrawal syndrome. Urology. 1994;43:408–410. doi: 10.1016/0090-4295(94)90092-2.
    1. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, Armstrong AJ, Flaig TW, Flechon A, Mainwaring P, Fleming M, Hainsworth JD, Hirmand M, Selby B, Seely L, de Bono JS. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367:1187–1197. doi: 10.1056/NEJMoa1207506.
    1. Jambal P, Badtke MM, Harrell JC, Borges VF, Post MD, Sollender GE, Spillman MA, Horwitz KB, Jacobsen BM. Estrogen switches pure mucinous breast cancer to invasive lobular carcinoma with mucinous features. Breast Cancer Res Treat. 2013;137:431–448. doi: 10.1007/s10549-012-2377-x.
    1. Magklara A, Brown TJ, Diamandis EP. Characterization of androgen receptor and nuclear receptor co-regulator expression in human breast cancer cell lines exhibiting differential regulation of kallikreins 2 and 3. Int J Cancer. 2002;100:507–514. doi: 10.1002/ijc.10520.
    1. Chamberlain NL, Driver ED, Miesfeld RL. The length and location of CAG trinucleotide repeats in the androgen receptor N-terminal domain affect transactivation function. Nucleic Acids Res. 1994;22:3181–3186. doi: 10.1093/nar/22.15.3181.
    1. Birrell SN, Bentel JM, Hickey TE, Ricciardelli C, Weger MA, Horsfall DJ, Tilley WD. Androgens induce divergent proliferative responses in human breast cancer cell lines. J Steroid Biochem Mol Biol. 1995;52:459–467. doi: 10.1016/0960-0760(95)00005-K.
    1. Hall RE, Birrell SN, Tilley WD, Sutherland RL. MDA-MB-453, an androgen-responsive human breast carcinoma cell line with high level androgen receptor expression. Eur J Cancer. 1994;30A:484–490.
    1. Beyer C, McDonald P, Vidal N. Failure of 5-alpha-dihydrotestosterone to elicit estrous behavior in the ovariectomized rabbit. Endocrinology. 1970;86:939–941. doi: 10.1210/endo-86-4-939.
    1. McDonald P, Beyer C, Newton F, Brien B, Baker R, Tan HS, Sampson C, Kitching P, Greenhill R, Pritchard D. Failure of 5alpha-dihydrotestosterone to initiate sexual behaviour in the castrated male rat. Nature. 1970;227:964–965. doi: 10.1038/227964a0.
    1. Ryan KJ. Biological aromatization of steroids. J Biol Chem. 1959;234:268–272.
    1. Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, Wasielewska T, Welsbie D, Chen CD, Higano CS, Beer TM, Hung DT, Scher HI, Jung ME, Sawyers CL. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 2009;324:787–790. doi: 10.1126/science.1168175.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121
    1. Moore NL, Buchanan G, Harris JM, Selth LA, Bianco-Miotto T, Hanson AR, Birrell SN, Butler LM, Hickey TE, Tilley WD. An androgen receptor mutation in the MDA-MB-453 cell line model of molecular apocrine breast cancer compromises receptor activity. Endocr Relat Cancer. 2012;19:599–613. doi: 10.1530/ERC-12-0065.
    1. Wilson VS, Bobseine K, Lambright CR, Gray LE Jr. A novel cell line, MDA-kb2, that stably expresses an androgen- and glucocorticoid-responsive reporter for the detection of hormone receptor agonists and antagonists. Toxicol Sci. 2002;66:69–81. doi: 10.1093/toxsci/66.1.69.
    1. Kuenen-Boumeester V, Van der Kwast TH, Claassen CC, Look MP, Liem GS, Klijn JG, Henzen-Logmans SC. The clinical significance of androgen receptors in breast cancer and their relation to histological and cell biological parameters. Eur J Cancer. 1996;32A:1560–1565.
    1. Soreide JA, Lea OA, Varhaug JE, Skarstein A, Kvinnsland S. Androgen receptors in operable breast cancer: relation to other steroid hormone receptors, correlations to prognostic factors and predictive value for effect of adjuvant tamoxifen treatment. Eur J Surg Oncol. 1992;18:112–118.
    1. Garay JP, Park BH. Androgen receptor as a targeted therapy for breast cancer. Am J Cancer Res. 2012;2:434–445.
    1. Hickey TE, Robinson JL, Carroll JS, Tilley WD. Minireview: the androgen receptor in breast tissues: growth inhibitor, tumor suppressor, oncogene? Mol Endocrinol. 2012;26:1252–1267. doi: 10.1210/me.2012-1107.
    1. Moe RE, Anderson BO. Androgens and androgen receptors: a clinically neglected sector in breast cancer biology. J Surg Oncol. 2007;95:437–439. doi: 10.1002/jso.20722.
    1. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL, Henshall SM, Birrell SN, Coetzee GA, Sutherland RL, Butler LM, Tilley WD. Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer Res. 2009;69:6131–6140.
    1. Poulin R, Baker D, Labrie F. Androgens inhibit basal and estrogen-induced cell proliferation in the ZR-75-1 human breast cancer cell line. Breast Cancer Res Treat. 1988;12:213–225. doi: 10.1007/BF01805942.
    1. Macedo LF, Guo Z, Tilghman SL, Sabnis GJ, Qiu Y, Brodie A. Role of androgens on MCF-7 breast cancer cell growth and on the inhibitory effect of letrozole. Cancer Res. 2006;66:7775–7782. doi: 10.1158/0008-5472.CAN-05-3984.
    1. Cops EJ, Bianco-Miotto T, Moore NL, Clarke CL, Birrell SN, Butler LM, Tilley WD. Antiproliferative actions of the synthetic androgen, mibolerone, in breast cancer cells are mediated by both androgen and progesterone receptors. J Steroid Biochem Mol Biol. 2008;110:236–243. doi: 10.1016/j.jsbmb.2007.10.014.
    1. Garay JP, Karakas B, Abukhdeir AM, Cosgrove DP, Gustin JP, Higgins MJ, Konishi H, Konishi Y, Lauring J, Mohseni M, Wang GM, Jelovac D, Weeraratna A, Sherman Baust CA, Morin PJ, Toubaji A, Meeker A, De Marzo AM, Lewis G, Subhawong A, Argani P, Park BH. The growth response to androgen receptor signaling in ERalpha-negative human breast cells is dependent on p21 and mediated by MAPK activation. Breast Cancer Res. 2012;14:R27. doi: 10.1186/bcr3112.
    1. Simon WE, Albrecht M, Trams G, Dietel M, Holzel F. In vitro growth promotion of human mammary carcinoma cells by steroid hormones, tamoxifen, and prolactin. J Natl Cancer Inst. 1984;73:313–321.
    1. Recchione C, Venturelli E, Manzari A, Cavalleri A, Martinetti A, Secreto G. Testosterone, dihydrotestosterone and oestradiol levels in postmenopausal breast cancer tissues. J Steroid Biochem Mol Biol. 1995;52:541–546. doi: 10.1016/0960-0760(95)00017-T.
    1. Swerdloff RS, Wang C. Dihydrotestosterone: a rationale for its use as a non-aromatizable androgen replacement therapeutic agent. Baillieres Clin Endocrinol Metab. 1998;12:501–506. doi: 10.1016/S0950-351X(98)80267-X.
    1. Hall JM, Korach KS. Stromal cell-derived factor 1, a novel target of estrogen receptor action, mediates the mitogenic effects of estradiol in ovarian and breast cancer cells. Mol Endocrinol. 2003;17:792–803. doi: 10.1210/me.2002-0438.
    1. Sauve K, Lepage J, Sanchez M, Heveker N, Tremblay A. Positive feedback activation of estrogen receptors by the CXCL12-CXCR4 pathway. Cancer Res. 2009;69:5793–5800. doi: 10.1158/0008-5472.CAN-08-4924.
    1. Kasina S, Macoska JA. The CXCL12/CXCR4 axis promotes ligand-independent activation of the androgen receptor. Mol Cell Endocrinol. 2012;351:249–263. doi: 10.1016/j.mce.2011.12.015.
    1. Kang Z, Janne OA, Palvimo JJ. Coregulator recruitment and histone modifications in transcriptional regulation by the androgen receptor. Mol Endocrinol. 2004;18:2633–2648. doi: 10.1210/me.2004-0245.
    1. Panet-Raymond V, Gottlieb B, Beitel LK, Pinsky L, Trifiro MA. Interactions between androgen and estrogen receptors and the effects on their transactivational properties. Mol Cell Endocrinol. 2000;167:139–150. doi: 10.1016/S0303-7207(00)00279-3.
    1. Migliaccio A, Di Domenico M, Castoria G, Nanayakkara M, Lombardi M, de Falco A, Bilancio A, Varricchio L, Ciociola A, Auricchio F. Steroid receptor regulation of epidermal growth factor signaling through Src in breast and prostate cancer cells: steroid antagonist action. Cancer Res. 2005;65:10585–10593. doi: 10.1158/0008-5472.CAN-05-0912.
    1. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Maneval EC, Chen I, Darimont B, Hager JH. A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discov. 2013;3:1020–1029. doi: 10.1158/-13-0226.
    1. Korpal M, Korn JM, Gao X, Rakiec DP, Ruddy DA, Doshi S, Yuan J, Kovats SG, Kim S, Cooke VG, Monahan JE, Stegmeier F, Roberts TM, Sellers WR, Zhou W, Zhu P. An F876L Mutation in Androgen Receptor Confers Genetic and Phenotypic Resistance to MDV3100 (Enzalutamide) Cancer Discov. 2013;3:1030–1043. doi: 10.1158/-13-0142.

Source: PubMed

3
Abonnieren