Skip to main content

Advertisement

Log in

Androgen metabolite-dependent growth of hormone receptor-positive breast cancer as a possible aromatase inhibitor-resistance mechanism

  • Preclinical Study
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Aromatase inhibitors (AIs) have been reported to exert their antiproliferative effects in postmenopausal women with hormone receptor-positive breast cancer not only by reducing estrogen production but also by unmasking the inhibitory effects of androgens such as testosterone (TS) and dihydrotestosterone (DHT). However, the role of androgens in AI-resistance mechanisms is not sufficiently understood. 5α-Androstane-3β,17β-diol (3β-diol) generated from DHT by 3β-hydroxysteroid dehydrogenase type 1 (HSD3B1) shows androgenic and substantial estrogenic activities, representing a potential mechanism of AI resistance. Estrogen response element (ERE)-green fluorescent protein (GFP)-transfected MCF-7 breast cancer cells (E10 cells) were cultured for 3 months under steroid-depleted, TS-supplemented conditions. Among the surviving cells, two stable variants showing androgen metabolite-dependent ER activity were selected by monitoring GFP expression. We investigated the process of adaptation to androgen-abundant conditions and the role of androgens in AI-resistance mechanisms in these variant cell lines. The variant cell lines showed increased growth and induction of estrogen-responsive genes rather than androgen-responsive genes after stimulation with androgens or 3β-diol. Further analysis suggested that increased expression of HSD3B1 and reduced expression of androgen receptor (AR) promoted adaptation to androgen-abundant conditions, as indicated by the increased conversion of DHT into 3β-diol by HSD3B1 and AR signal reduction. Furthermore, in parental E10 cells, ectopic expression of HSD3B1 or inhibition of AR resulted in adaptation to androgen-abundant conditions. Coculture with stromal cells to mimic local estrogen production from androgens reduced cell sensitivity to AIs compared with parental E10 cells. These results suggest that increased expression of HSD3B1 and reduced expression of AR might reduce the sensitivity to AIs as demonstrated by enhanced androgen metabolite-induced ER activation and growth mechanisms. Androgen metabolite-dependent growth of breast cancer cells may therefore play a role in AI-resistance.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

AI:

Aromatase inhibitor

TS:

Testosterone

DHT:

Dihydrotestosterone

3β-Diol:

5α-Androstane-3β,17β-diol

HSD3B1:

3β-Hydroxysteroid dehydrogenase type 1

AKR1C3:

Aldo–keto reductase 1C3

AR:

Androgen receptor

ERα:

Estrogen receptor α

E2:

Estradiol

OHT:

4-Hydroxytamoxifen

GFP:

Green fluorescent protein

SERM:

Selective estrogen receptor modulator

References

  1. Andò S, De Amicis F, Rago V, Carpino A, Maggiolini M, Panno ML, Lanzino M (2002) Breast cancer: from estrogen to androgen receptor. Mol Cell Endocrinol 193:121–128

    Article  PubMed  Google Scholar 

  2. Chia S, Gradishar W, Mauriac L, Bines J, Amant F, Federico M, Fein L, Romieu G, Buzdar A, Robertson JF et al (2008) Double-blind, randomized placebo controlled trial of fulvestrant compared with exemestane after prior nonsteroidal aromatase inhibitor therapy in postmenopausal women with hormone receptor-positive, advanced breast cancer: results from EFECT. J Clin Oncol 26:1664–1670

    Article  PubMed  CAS  Google Scholar 

  3. Chlebowski R, Cuzick J, Amakye D, Bauerfeind I, Buzdar A, Chia S, Cutuli B, Linforth R, Maass N, Noguchi S et al (2000) Clinical perspectives on the utility of aromatase inhibitors for the adjuvant treatment of breast cancer. The Breast 18(Suppl 2):S1–S11

    Google Scholar 

  4. Fang H, Tong W, Branham WS, Moland CL, Dial SL, Hong H, Xie Q, Perkins R, Owens W, Sheehan DM (2003) Study of 202 natural, synthetic, and environmental chemicals for binding to the androgen receptor. Chem Res Toxicol 16:1338–1358

    Article  PubMed  CAS  Google Scholar 

  5. Hayashi S, Niwa T, Yamaguchi Y (2009) Estrogen signaling pathway and its imaging in human breast cancer. Cancer Sci 100:1773–1778

    Article  PubMed  CAS  Google Scholar 

  6. Heneweer M, Muusse M, Dingemans M, de Jong PC, van den Berg M, Sanderson JT (2005) Co-culture of primary human mammary fibroblasts and MCF-7 cells as an in vitro breast cancer model. Toxicol Sci 83:257–263

    Article  PubMed  CAS  Google Scholar 

  7. Inoue A, Omoto Y, Yamaguchi Y, Kiyama R, Hayashi S (2004) Transcription factor EGR3 is involved in the estrogen-signaling pathway in breast cancer cell. J Mol Endocrinol 32:649–661

    Article  PubMed  CAS  Google Scholar 

  8. Jin Y, Duan L, Lee SH, Kloosterboer HJ, Blair IA, Penning TM (2009) Human cytosolic hydroxysteroid dehydrogenases of the aldo-ketoreductase superfamily catalyze reduction of conjugated steroids. J Biol Chem 284:10013–10022

    Article  PubMed  CAS  Google Scholar 

  9. Koyama H, Iesato A, Fukushima Y, Okada T, Watanabe T, Harada M, Ito T, Maeno K, Mochizuki Y, Ito K et al (2011) A retrospective study of high-dose toremifene treatment for patients with aromatase inhibitor refractory advanced or metastatic hormone receptor-positive breast cancer. Gan To Kagaku Ryoho 38:1123–1126

    PubMed  Google Scholar 

  10. Kuiper GG, Carlsson B, Grandien K, Enmark E, Häggblad J, Nilsson S, Gustafsson JA (1997) Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 138:863–870

    Article  PubMed  CAS  Google Scholar 

  11. Labrie F, Luu-The V, Labrie C, Bélanger A, Simard J, Lin SX, Pelletier G (2003) Endocrine and intracrine sources of androgens in women: inhibition of breast cancer and other roles of androgens and their precursor dehydroepiandrosterone. Endocr Rev 24:152–182

    Article  PubMed  CAS  Google Scholar 

  12. Lawrence MG, Lai J, Clements JA (2010) Kallikreins on steroids: structure, function, and hormonal regulation of prostate-specific antigen and the extended kallikrein locus. Endocr Rev 31:407–446

    Article  PubMed  CAS  Google Scholar 

  13. Lorence MC, Murry BA, Trant JM, Mason JI (1990) Human 3 beta-hydroxysteroid dehydrogenase/delta 5 → 4 isomerase from placenta: expression in nonsteroidogenic cells of a protein that catalyzes the dehydrogenation/isomerization of C21 and C19 steroids. Endocrinology 126:2493–2498

    Article  PubMed  CAS  Google Scholar 

  14. Macedo LF, Guo Z, Tilghman SL, Sabnis GJ, Qiu Y, Brodie A (2006) Role of androgens on MCF-7 breast cancer cell growth and on the inhibitory effect of letrozole. Cancer Res 66:7775–7782

    Article  PubMed  CAS  Google Scholar 

  15. Martin LA, Farmer I, Johnston SR, Ali S, Dowsett M (2005) Elevated ERK1/ERK2/estrogen receptor cross-talk enhances estrogen-mediated signaling during long-term estrogen deprivation. Endocr Relat Cancer 12(Suppl 1):S75–S84

    Article  PubMed  CAS  Google Scholar 

  16. Matsumoto M, Yamaguchi Y, Seino Y, Hatakeyama A, Takei H, Niikura H, Ito K, Suzuki T, Sasano H, Yaegashi N et al (2008) Estrogen signaling ability in human endometrial cancer through the cancer-stromal interaction. Endocr Relat Cancer 15:451–463

    Article  PubMed  CAS  Google Scholar 

  17. Miller WR, Anderson TJ, Jack WJ (1990) Relationship between tumour aromatase activity, tumour characteristics and response to therapy. J Steroid Biochem Mol Biol 37:1055–1059

    Article  PubMed  CAS  Google Scholar 

  18. O’Neill JS, Miller WR (1987) Aromatase activity in breast adipose tissue from women with benign and malignant breast disease. Br J Cancer 56:601–604

    Article  PubMed  Google Scholar 

  19. Omoto Y, Kobayashi Y, Nishida K, Tsuchiya E, Eguchi H, Nakagawa K, Ishikawa Y, Yamori T, Iwase H, Fujii Y et al (2001) Expression, function, and clinical implications of the estrogen receptor β in human lung cancers. Biochem Biophys Res Commun 285:340–347

    Article  PubMed  CAS  Google Scholar 

  20. Ortmann J, Prifti S, Bohlmann MK, Rehberger-Schneider S, Strowitzki T, Rabe T (2002) Testosterone and 5 alpha-dihydrotestosterone inhibit in vitro growth of human breast cancer cell lines. Gynecol Endocrinol 16:113–120

    PubMed  CAS  Google Scholar 

  21. Panet-Raymond V, Gottlieb B, Beitel LK, Pinsky L, Trifiro MA (2000) Interactions between androgen and estrogen receptors and the effects on their transactivational properties. Mol Cell Endocrinol 167:139–150

    Article  PubMed  CAS  Google Scholar 

  22. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL et al (2009) Androgen receptor inhibits estrogen receptor-alpha activity and is prognostic in breast cancer. Cancer Res 69:6131–6140

    Article  PubMed  CAS  Google Scholar 

  23. Sabnis G, Brodie A (2010) Adaptive changes results in activation of alternate signaling pathways and resistance to aromatase inhibitor resistance. Mol Cell Endocrinol 340(2):142–147

    Article  PubMed  Google Scholar 

  24. Santen RJ, Santner SJ, Pauley RJ, Tait L, Kaseta J, Demers LM, Hamilton C, Yue W, Wang JP (1997) Estrogen production via the aromatase enzyme in breast carcinoma: which cell type is responsible? J Steroid Biochem Mol Biol 61:267–271

    Article  PubMed  CAS  Google Scholar 

  25. Santen RJ, Song RX, Masamura S, Yue W, Fan P, Sogon T, Hayashi S, Nakachi K, Eguchi H (2008) Adaptation to estradiol deprivation causes up-regulation of growth factor pathways and hypersensitivity to estradiol in breast cancer cells. Adv Exp Med Biol 630:19–34

    Article  PubMed  CAS  Google Scholar 

  26. Santner SJ, Chen S, Zhou D, Korsunsky Z, Martel J, Santen RJ (1993) Effect of androstenedione on growth of untransfected and aromatase-transfected MCF-7 cells in culture. J Steroid Biochem Mol Biol 44:611–616

    Article  PubMed  CAS  Google Scholar 

  27. Sasano H, Ozaki M (1997) Aromatase expression and its localization in human breast cancer. J Steroid Biochem Mol Biol 61:293–298

    Article  PubMed  CAS  Google Scholar 

  28. Sasano H, Miki Y, Nagasaki S, Suzuki T (2009) In situ estrogen production and its regulation in human breast carcinoma: from endocrinology to intracrinology. Pathol Int 59:777–789

    Article  PubMed  CAS  Google Scholar 

  29. Sikora MJ, Cordero KE, Larios JM, Johnson MD, Lippman ME, Rae JM (2009) The androgen metabolite 5alpha-androstane-3beta,17beta-diol (3betaAdiol) induces breast cancer growth via estrogen receptor: implications for aromatase inhibitor resistance. Breast Cancer Res Treat 115:289–296

    Article  PubMed  CAS  Google Scholar 

  30. Steckelbroeck S, Jin Y, Gopishetty S, Oyesanmi B, Penning TM (2004) Human cytosolic 3 alpha-hydroxysteroid dehydrogenases of the aldo-keto reductase superfamily display significant 3 beta-hydroxysteroid dehydrogenase activity. J Biol Chem 279:10784–10795

    Article  PubMed  CAS  Google Scholar 

  31. Takagi K, Miki Y, Nagasaki S, Hirakawa H, Onodera Y, Akahira J, Ishida T, Watanabe M, Kimijima I, Hayashi S et al (2010) Increased intratumoral androgens in human breast carcinoma following aromatase inhibitor exemestane treatment. Endocr Relat Cancer 17:415–430

    Article  PubMed  CAS  Google Scholar 

  32. Thürlimann B, Robertson JF, Nabholtz JM, Buzdar A, Bonneterre J, Arimidex Study Group (2003) Efficacy of tamoxifen following anastrozole (‘Arimidex’) compared with anastrozole following tamoxifen as first-line treatment for advanced breast cancer in postmenopausal women. Eur J Cancer 39:2310–2317

    Article  PubMed  Google Scholar 

  33. Wang P, Wen Y, Han G-Z, Sidhu PK, Zhu BT (2009) Characterization of the oestrogenic activity of non-aromatic steroids: are there male-specific endogenous oestrogen receptor modulators? Br J Pharmacol 158:1796–1807

    Article  PubMed  CAS  Google Scholar 

  34. Yamaguchi Y, Takei H, Suemasu K, Kobayashi Y, Kurosumi M, Harada N, Hayashi S (2005) Tumor-stromal interaction through the estrogen-signaling pathway in human breast cancer. Cancer Res 65:4653–4662

    Article  PubMed  CAS  Google Scholar 

  35. Yamamoto Y, Masuda N, Ohtake T, Yamashita H, Saji S, Kimijima I, Kasahara Y, Ishikawa T, Sawaki M, Hozumi Y et al (2010) Clinical usefulness of high-dose toremifene in patients relapsed on treatment with an aromatase inhibitor. Breast Cancer 17:254–260

    Article  PubMed  Google Scholar 

  36. Yue W, Fan P, Wang J, Li Y, Santen RJ (2007) Mechanisms of acquired resistance to endocrine therapy in hormone-dependent breast cancer cells. J Steroid Biochem Mol Biol 106:102–110

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We would like to thank Takashi Suzuki (Tohoku University Department of Pathology and Histotechnology) for discussions and helpful suggestions. This study was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan; a Grant-in-Aid for Cancer Research from the Ministry of Health, Labour and Welfare, Japan; the Program for Promotion of Fundamental Studies in Health Science of the National Institute of Biomedical Innovation (NIBIO); and a Grant from the Smoking Research Foundation.

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical standard

All experiments complied with the current laws of Japan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Toru Hanamura.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10549_2013_2595_MOESM1_ESM.ppt

Fig. S1 Establishment of E10 cells and screening of variant cell lines. a E10 cells report ER activity via GFP expression. ERE-tk-GFP-MCF-7 cells (E10 cells) were established from the human breast cancer cell line, MCF-7, by introduction of a plasmid carrying the ERE fused with ERE-GFP gene. These cells show luminescence via GFP, corresponding to ER transcription activity. b Screening of variant cell lines that show ER activity depending on androgen metabolites. We used E10 cells to establish variant cell lines. After 3 months of culture in TS-supplemented steroid-depleted medium, five clones expressing GFP (representing ER activity) were obtained. ER activities at each step of treatment (a, b, c, d, e) were assessed in a single experiment, as described in Materials and methods. E estrogen; Let letrozole; Ful fulvestrant (PPT 449 kb)

10549_2013_2595_MOESM2_ESM.ppt

Fig. S2 Expression of HSD3B1 in E10-HSD3B1 cells. Cells transfected with HSD3B1 expression vector (E10-HSD3B1) or control vector (E10-Control) were generated as described in Materials and methods. The expression level of HSD3B1 under regular growth medium was analyzed using real-time PCR. Values relative to RPL13A are shown (PPT 134 kb)

10549_2013_2595_MOESM3_ESM.ppt

Fig. S3 Interaction coculture system using insert layers. To assess the inhibitory effect of AIs in E10 and variant cell lines, we used interaction cultures, defined as cocultures where cancer cells and stromal cells are grown separately by a membrane, but are allowed to interact via soluble factors, and have the same microenvironment, such as local estrogen synthesis from androgen by aromatase in stromal cells (PPT 136 kb)

10549_2013_2595_MOESM4_ESM.ppt

Fig. S4 Induction of estrogen- or androgen-responsive genes by androgen or 3β-diol. After 3 days of incubation in steroid-depleted medium, 100 nM TS, DHT, 3β-diol or vehicle control (EtOH) was added to each cell line for 4 days before total RNA extraction. Total RNA was extracted from each cell line cultured in the indicated culture conditions, and real-time PCRs for expression of the indicated mRNA were carried out as described in Materials and methods. All PCRs were performed in duplicate, and the expression of the target gene relative to vehicle control is shown (mean ± SD) (PPT 198 kb)

10549_2013_2595_MOESM5_ESM.ppt

Fig. S5 Estrogen synthesis and androgen metabolism. In postmenopausal women, estrogens are mainly supplied by aromatase from androgens (TS and androstenedione) biosynthesized in the adrenal gland. In contrast, 5α-androstane-3β,17β-diol (3β-diol) is generated from DHT by HSD3B1 type 1 and AKR1C3. It was suggested that the metabolizing pathway from DHT to 3β-diol is up-regulated in variant cell lines (PPT 149 kb)

10549_2013_2595_MOESM6_ESM.ppt

Fig. S6 Blockage of 3β-diol-induced growth in variant cell lines by SERMs. After 3 days of culture in steroid-depleted medium, each cell line was plated in 24-well culture plates at a density of 10,000 cells/well in steroid-depleted medium. A volume of 100 nM 3β-diol or vehicle control (EtOH), with or without 1 μM OHT, 3 μM toremifene (TOR), 1 μM fulvestrant (Ful), 100 nM letrozole (Let), 100 nM anastrozole (Ana) or 100 nM exemestane (Exe), was added to each well for 4 days. Cells from each well were then harvested and counted. Values relative to the vehicle control are shown. All data are shown as mean ± SD of three independent experiments (PPT 161 kb)

Supplementary material 7 (DOC 48 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hanamura, T., Niwa, T., Nishikawa, S. et al. Androgen metabolite-dependent growth of hormone receptor-positive breast cancer as a possible aromatase inhibitor-resistance mechanism. Breast Cancer Res Treat 139, 731–740 (2013). https://doi.org/10.1007/s10549-013-2595-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-013-2595-x

Keywords

Navigation