Phase II Study of Dehydroepiandrosterone in Androgen Receptor-Positive Metastatic Breast Cancer

Elisabetta Pietri, Ilaria Massa, Sara Bravaccini, Sara Ravaioli, Maria Maddalena Tumedei, Elisabetta Petracci, Caterina Donati, Alessio Schirone, Federico Piacentini, Lorenzo Gianni, Mario Nicolini, Enrico Campadelli, Alessandra Gennari, Alessandro Saba, Beatrice Campi, Linda Valmorri, Daniele Andreis, Francesco Fabbri, Dino Amadori, Andrea Rocca, Elisabetta Pietri, Ilaria Massa, Sara Bravaccini, Sara Ravaioli, Maria Maddalena Tumedei, Elisabetta Petracci, Caterina Donati, Alessio Schirone, Federico Piacentini, Lorenzo Gianni, Mario Nicolini, Enrico Campadelli, Alessandra Gennari, Alessandro Saba, Beatrice Campi, Linda Valmorri, Daniele Andreis, Francesco Fabbri, Dino Amadori, Andrea Rocca

Abstract

Lessons learned: The androgen receptor (AR) is present in most breast cancers (BC), but its exploitation as a therapeutic target has been limited.This study explored the activity of dehydroepiandrosterone (DHEA), a precursor being transformed into androgens within BC cells, in combination with an aromatase inhibitor (to block DHEA conversion into estrogens), in a two-stage phase II study in patients with AR-positive/estrogen receptor-positive/human epidermal growth receptor 2-negative metastatic BC.Although well tolerated, only 1 of 12 patients obtained a prolonged clinical benefit, and the study was closed after its first stage for poor activity.

Background: Androgen receptors (AR) are expressed in most breast cancers, and AR-agonists have some activity in these neoplasms. We investigated the safety and activity of the androgen precursor dehydroepiandrosterone (DHEA) in combination with an aromatase inhibitor (AI) in patients with AR-positive metastatic breast cancer (MBC).

Methods: A two-stage phase II study was conducted in two patient cohorts, one with estrogen receptor (ER)-positive (resistant to AIs) and the other with triple-negative MBC. DHEA 100 mg/day was administered orally. The combination with an AI aimed to prevent the conversion of DHEA into estrogens. The main endpoint was the clinical benefit rate. The triple-negative cohort was closed early.

Results: Twelve patients with ER-positive MBC were enrolled. DHEA-related adverse events, reported in four patients, included grade 2 fatigue, erythema, and transaminitis, and grade 1 drowsiness and musculoskeletal pain. Clinical benefit was observed in one patient with ER-positive disease whose tumor had AR gene amplification. There was wide inter- and intra-patient variation in serum levels of DHEA and its metabolites.

Conclusion: DHEA showed excellent safety but poor activity in MBC. Although dose and patient selection could be improved, high serum level variability may hamper further DHEA development in this setting.

Trial registration: ClinicalTrials.gov NCT02000375.

© AlphaMed Press; the data published online to support this summary are the property of the authors.

Figures

Figure 1.
Figure 1.
(A) Time to progression and (B) overall survival of the estrogen receptor‐positive cohort. Abbreviations: OS, overall survival; TTP, time to progression.
Figure 2.
Figure 2.
Individual serum concentrations of dehydroepiandrosterone (DHEA) and metabolites in 10 patients from the estrogen receptor‐positive cohort. The following are reported for each patient: Left panels: serum concentrations of DHEA, androstane‐3alpha,17beta‐diol‐3‐glucuronide (3α‐diol‐3G), and androstane‐3alpha,17beta‐diol‐17glucuronide (3α‐diol‐17G) at different time points during treatment. Right panels: serum concentrations of androsterone glucuronide (ADT‐G) at different time points during treatment. Solid line: DHEA levels; dotted line: 3α‐diol‐3G levels; dashed line: 3α‐diol‐17G levels; dash‐dotted line: ADT‐G levels. Abbreviations: Baseline, before starting treatment; C1D14, cycle 1 day 14; C2D1, cycle 2 day 1; EOT, end of treatment.
Figure 3.
Figure 3.
Boxplots of serum concentrations of DHEA and metabolites. Box and whisker plots, showing the median, interquartile range, and the highest and lowest values for each analyte at three time points (baseline, cycle 2 day 1, and end of treatment). Abbreviations: C2, cycle 2 day 1; DHEA, dehydroepiandrosterone; EOT, end of treatment.

References

    1. Collins LC, Cole KS, Marotti JD et al. Androgen receptor expression in breast cancer in relation to molecular phenotype: Results from the Nurses' Health Study. Mod Pathol 2011;24:924–931.
    1. Vera‐Badillo FE, Templeton AJ, de Gouveia P et al. Androgen receptor expression and outcomes in early breast cancer: A systematic review and meta‐analysis. J Natl Cancer Inst 2014;106:djt319.
    1. Labrie F, Simard J, de Launoit Y et al. Androgens and breast cancer. Cancer Detect Prev 1992;16:31–38.
    1. Fioretti FM, Sita‐Lumsden A, Bevan CL et al. Revising the role of the androgen receptor in breast cancer. J Mol Endocrinol 2014;52:R257–265.
    1. Chia K, O'Brien M, Brown M et al. Targeting the androgen receptor in breast cancer. Curr Oncol Rep 2015;17:4.
    1. Ortmann J, Prifti S, Bohlmann MK et al. Testosterone and 5 alpha‐dihydrotestosterone inhibit in vitro growth of human breast cancer cell lines. Gynecol Endocrinol 2002;16:113–120.
    1. Greeve MA, Allan RK, Harvey JM et al. Inhibition of MCF‐7 breast cancer cell proliferation by 5alpha‐dihydrotestosterone; a role for p21(Cip1/Waf1). J Mol Endocrinol 2004;32:793–810.
    1. Macedo LF, Guo Z, Tilghman SL et al. Role of androgens on MCF‐7 breast cancer cell growth and on the inhibitory effect of letrozole. Cancer Res 2006;66:7775–7782.
    1. Cops EJ, Bianco‐Miotto T, Moore NL et al. 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.
    1. Birrell SN, Bentel JM, Hickey TE et al. Androgens induce divergent proliferative responses in human breast cancer cell lines. J Steroid Biochem Mol Biol 1995;52:459–467.
    1. Welsch CW. Host factors affecting the growth of carcinogen‐induced rat mammary carcinomas: A review and tribute to Charles Brenton Huggins. Cancer Res 1985;45:3415–3443.
    1. Poulin R, Simard J, Labrie C et al. Down‐regulation of estrogen receptors by androgens in the ZR‐75‐1 human breast cancer cell line. Endocrinology 1989;125:392–399.
    1. Nahleh Z. Androgen receptor as a target for the treatment of hormone receptor‐negative breast cancer: An unchartered territory. Future Oncol 2008;4:15–21.
    1. Najid A, Habrioux G. Biological effects of adrenal androgens on MCF‐7 and BT‐20 human breast cancer cells. Oncology 1990;47:269–274.
    1. Boccuzzi G, Brignardello E, di Monaco M et al. Influence of dehydroepiandrosterone and 5‐en‐androstene‐3 beta, 17 beta‐diol on the growth of MCF‐7 human breast cancer cells induced by 17 beta‐estradiol. Anticancer Res 1992;12:799–803.
    1. Maggiolini M, Donzé O, Jeannin et al. Adrenal androgens stimulate the proliferation of breast cancer cells as direct activators of estrogen receptor alpha. Cancer Res 1999;59:4864–4869.
    1. Cochrane DR, Bernales S, Jacobsen BM et al. Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide. Breast Cancer Res 2014;16:R7.
    1. Barton VN, D'Amato NC, Gordon MA et al. Multiple molecular subtypes of triple‐negative breast cancer critically rely on androgen receptor and respond to enzalutamide in vivo. Mol Cancer Ther 2015;14:769–778.
    1. Gordan GS, Halden A, Horn Y et al. Calusterone (7beta, 17alpha‐dimethyltestosterone) as primary and secondary therapy of advanced breast cancer. Oncology 1973;28:138–146.
    1. Goldenberg IS, Waters N, Ravdin RS et al. Androgenic therapy for advanced breast cancer in women. A report of the cooperative breast cancer group. JAMA 1973;223:1267–1268.
    1. Boni C, Pagano M, Panebianco M et al. Therapeutic activity of testosterone in metastatic breast cancer. Anticancer Res 2014;34:1287–1290.
    1. Glaser R, Dimitrakakis C. Testosterone and breast cancer prevention. Maturitas 2015;82:291–295.
    1. D'Amato NC, Gordon MA, Babbs B et al. Cooperative dynamics of AR and ER activity in breast cancer. Mol Cancer Res 2016;14:1054–1067.
    1. Krop I, Cortes J, Miller K et al. A single‐arm phase 2 study to assess clinical activity, efficacy and safety of enzalutamide with trastuzumab in HER2+ AR+ metastatic or locally advanced breast cancer. Cancer Res 2017;77(suppl 4):P4‐22‐08a.
    1. Schwartzberg LS, Yardley DA, Elias AD et al. A phase I/Ib study of enzalutamide alone and in combination with endocrine therapies in women with advanced breast cancer. Clin Cancer Res 2017;23:4046–4054.
    1. Krop I, Abramson V, Colleoni M et al. Results from a randomized placebo‐controlled phase 2 trial evaluating exemestane ± enzalutamide in patients with hormone receptor‐positive breast cancer. Cancer Res 2018;78(suppl 4):GS4‐07a.
    1. Traina TA, Miller K, Yardley DA et al. Enzalutamide for the treatment of androgen receptor‐expressing triple‐negative breast cancer. J Clin Oncol 2018;36:884–890.
    1. Labrie F, Luu‐The V, Bélanger A et al. Is dehydroepiandrosterone a hormone? J Endocrinol 2005;187:169–196.
    1. Labrie F, Luu‐The V, Labrie C et al. Endocrine and intracrine sources of androgens in women: inhibition of breast cancer and other roles of androgens and their precursor dehydroepiandrosterone. Endocr Rev 2003;24:152–182.
    1. Traish AM, Kang HP, Saad F et al. Dehydroepiandrosterone (DHEA)‐‐A precursor steroid or an active hormone in human physiology. J Sex Med 2011;8:2960–2982.
    1. Elraiyah T, Sonbol MB, Wang Z et al. Clinical review: The benefits and harms of systemic dehydroepiandrosterone (DHEA) in postmenopausal women with normal adrenal function: A systematic review and meta‐analysis. J Clin Endocrinol Metab 2014;99:3536–3542.
    1. Rutkowski K, Sowa P, Rutkowska‐Talipska J et al. Dehydroepiandrosterone (DHEA): Hypes and hopes. Drugs 2014;74:1195–1207.
    1. Prough R, Clark BJ, Klinge CM. Novel mechanisms for DHEA action. J Mol Endocrinol 2016;56:R139–155.
    1. Shilkaitis A, Green A, Punj V et al. Dehydroepiandrosterone inhibits the progression phase of mammary carcinogenesis by inducing cellular senescence via a p16‐dependent but p53‐independent mechanism. Breast Cancer Res 2005;7:R1132–1140.
    1. Hakkak R, Shaaf S, Jo CH et al. Dehydroepiandrosterone intake protects against 7,12‐dimethylbenz(a)anthracene‐induced mammary tumor development in the obese Zucker rat model. Oncol Rep 2010;24:357–362.
    1. Schwartz AG, Pashko L, Whitcomb JM. Inhibition of tumor development by dehydroepiandrosterone and related steroids. Toxicol Pathol 1986;14:357–362.
    1. Gordon GB, Shantz LM, Talalay P. Modulation of growth, differentiation and carcinogenesis by dehydroepiandrosterone. Adv Enzyme Regul 1987;26:355–382.
    1. Li S, Yan X, Bélanger A et al. Prevention by dehydroepiandrosterone of the development of mammary carcinoma induced by 7,12‐dimethylbenz(a)anthracene (DMBA) in the rat. Breast Cancer Res Treat 1994;29:203–217.
    1. Sandra N, Ester P, Marie‐Agnès P et al. The DHEA metabolite 7β‐hydroxy‐epiandrosterone exerts anti‐estrogenic effects on breast cancer cell lines. Steroids 2012;77:542–551.
    1. Boccuzzi G, Di Monaco M, Brignardello E et al. Dehydroepiandrosterone antiestrogenic action through androgen receptor in MCF‐7 human breast cancer cell line. Anticancer Res 1993;13:2267–2272.
    1. Labrie F. DHEA, important source of sex steroids in men and even more in women. Prog Brain Res 2010;182:97–148.
    1. Labrie F, Bélanger A, Bélanger P et al. Metabolism of DHEA in postmenopausal women following percutaneous administration. J Steroid Biochem Mol Biol 2007;103:178–188.
    1. Morales AJ, Haubrich RH, Hwang JY et al. The effect of six months treatment with a 100 mg daily dose of dehydroepiandrosterone (DHEA) on circulating sex steroids, body composition and muscle strength in age‐advanced men and women. Clin Endocrinol (Oxf) 1998;49:421–432.
    1. Campi B, Frascarelli S, Pietri E et al. Quantification of DHEA in human serum on a routine basis: Development and validation of a tandem mass spectrometry based method. Anal Bioanal Chem 2018;410:407–416.
    1. Parker LN. Control of adrenal androgen secretion. Endocrinol Metab Clin North Am 1991;20:401–421.
    1. Labrie F, Bélanger A, Labrie C et al. Bioavailability and metabolism of oral and percutaneous dehydroepiandrosterone in postmenopausal women. J Steroid Biochem Mol Biol 2007;107:57–69.
    1. Morales AJ, Nolan JJ, Nelson JC et al. Effects of replacement dose of dehydroepiandrosterone in men and women of advancing age. J Clin Endocrinol Metab 1994;78:1360–1367.
    1. Gebre‐Medhin G, Husebye ES, Mallmin H et al. Oral dehydroepiandrosterone (DHEA) replacement therapy in women with Addison's disease. Clin Endocrinol (Oxf) 2000;52:775–780.
    1. Petri MA, Lahita RG, Van Vollenhoven RF et al. Effects of prasterone on corticosteroid requirements of women with systemic lupus erythematosus: A double‐blind, randomized, placebo‐controlled trial. Arthritis Rheum 2002;46:1820–1829.
    1. van Vollenhoven RF, Engleman EG, McGuire JL. An open study of dehydroepiandrosterone in systemic lupus erythematosus. Arthritis Rheum 1994;37:1305–1310.
    1. van Vollenhoven RF, Morabito LM, Engleman EG et al. Treatment of systemic lupus erythematosus with dehydroepiandrosterone: 50 patients treated up to 12 months. J Rheumatol 1998;25:285–289.
    1. Yen SS, Morales AJ, Khorram O. Replacement of DHEA in aging men and women. Potential remedial effects. Ann N Y Acad Sci 1995;774:128–142.
    1. Wolkowitz OM, Reus VI, Roberts E et al. Dehydroepiandrosterone (DHEA) treatment of depression. Biol Psychiatry 1997;41:311–318.
    1. Bloch M, Meiboom H, Zaig I et al. The use of dehydroepiandrosterone in the treatment of hypoactive sexual desire disorder: A report of gender differences. Eur Neuropsychopharmacol 2013;23:910–918.
    1. Christiansen JJ, Bruun JM, Christiansen JS et al. Long‐term DHEA substitution in female adrenocortical failure, body composition, muscle function, and bone metabolism: A randomized trial. Eur J Endocrinol 2011;165:293–300.
    1. Genazzani AR, Stomati M, Valentino V et al. Effect of 1‐year, low‐dose DHEA therapy on climacteric symptoms and female sexuality. Climacteric 2011;14:661–668.
    1. Barry NN, McGuire JL, van Vollenhoven RF. Dehydroepiandrosterone in systemic lupus erythematosus: Relationship between dosage, serum levels, and clinical response. J Rheumatol 1998;25:2352–2356.
    1. Labrie F, Cusan L, Gomez JL et al. Changes in serum DHEA and eleven of its metabolites during 12‐month percutaneous administration of DHEA. J Steroid Biochem Mol Biol 2008;110:1–9.
    1. Labrie F, Bélanger A, Bélanger P et al. Androgen glucuronides, instead of testosterone, as the new markers of androgenic activity in women. J Steroid Biochem Mol Biol 2006;99:182–188.
    1. Africander D, Storbeck KH. Steroid metabolism in breast cancer: Where are we and what are we missing? Mol Cell Endocrinol 2018;466:86–97.

Source: PubMed

3
订阅