Androgen receptor as potential therapeutic target in metastatic endometrial cancer

Ingvild Løberg Tangen, Therese Bredholt Onyango, Reidun Kopperud, Anna Berg, Mari K Halle, Anne M Øyan, Henrica M J Werner, Jone Trovik, Karl Henning Kalland, Helga B Salvesen, Camilla Krakstad, Ingvild Løberg Tangen, Therese Bredholt Onyango, Reidun Kopperud, Anna Berg, Mari K Halle, Anne M Øyan, Henrica M J Werner, Jone Trovik, Karl Henning Kalland, Helga B Salvesen, Camilla Krakstad

Abstract

Purpose: The expression and involvement of estrogen (ER) and progesterone receptor (PR) is extensively studied in endometrial cancer. Androgen receptor (AR) is a hormone receptor less studied in female cancers, and we here aim to investigate the expression level of AR in endometrial cancer precursor lesions, primary tumors and metastases, and its potential as therapeutic target.

Results: Expression of AR was observed in 93% of hyperplasias, but only in 41% of non-endometrioid tumors. Compared to estrogen and progesterone receptor AR is more commonly expressed in metastatic lesions, and AR status is discordant in primary and metastatic lesions in a large proportion of cases. AR protein level was significantly associated with survival (P < 0.001), and a calculated AR to ERα ratio identified a subgroup of patients with particular poor outcome. The anti-androgen enzalutamide may have a growth inhibitory effect in endometrial cancer cells based on experiments with primary endometrial tumor cells.

Materials and methods: 718 primary endometrial cancers and 298 metastatic lesions (from 142 patients) were investigated for expression of AR in relation to survival, clinical and histopathological data. Protein levels were investigated by immunohistochemistry and reverse phase protein array; mRNA levels by DNA oligonucleotide microarray. The effect of androgen stimulation and inhibition was tested on primary endometrial tumor cells.

Conclusions: A large proportion of metastatic endometrial cancer lesions express AR, which may be a potential target in these patients. Treatment targeting AR may be of particular benefit in patients with high AR levels compared to ERα levels.

Keywords: androgen receptor; biomarker; endometrial cancer; survival.

Conflict of interest statement

No potential conflicts of interest was disclosed.

Figures

Figure 1. AR expression decreases with dedifferentiation
Figure 1. AR expression decreases with dedifferentiation
AR expression in glandular cells was scored, with strong AR immunohistochemical staining shown in (A) and AR loss in (B). Both AR protein level (C) and mRNA (D) level decreased with dedifferentiation. Abbreviations: AR: androgen receptor, Gr: grade, NE: non-endometrioid, PT: primary tumor, Met: metastases, *Information on grade missing for 10 patients included in the study (7 AR positive and 3 AR negative)
Figure 2
Figure 2
Expression of the individual hormone receptors AR, PR and ERα in metastatic lesions based on its expression in primary tumor. 48% of the primary tumors with corresponding metastatic lesions were AR positive (IHC score 1–9) (A). Graphs show distribution of AR expression in the metastatic lesions from AR positive primary tumors (B) and AR negative primary tumors (C). 61% of the primary tumors with corresponding metastatic lesions were PR positive (IHC score 1–9) (D). Graphs show distribution of PR expression in the metastatic lesions from the PR positive primary tumors (E) and PR negative primary tumors (F). 52% of the primary tumors with corresponding metastatic lesions were ER positive (IHC score 4–9) (G). Graphs show distribution of ER expression in the ER positive primary tumors (H) and ER negative primary tumors (I). Expression patterns in individual metastases from AR positive patients (J) and AR negative patients (K) are shown as AR (purple), PR (green) and ERα (orange) in J and K. One line of circles represents one metastasis, and shows the difference in hormone receptor expression in that specific metastasis. Only patients with one or more AR positive metastases are shown. *Only primary tumors with corresponding metastases included.
Figure 3. AR status predicts prognosis in…
Figure 3. AR status predicts prognosis in endometrial cancer
Endometrial cancer patients with AR expression (scoring index 1–9) have a significantly better survival than patient with AR loss (scoring index 0) both in the whole population (A) and in the subgroup of patients with FIGO stage I and II (B).
Figure 4. High AR to ER ratio…
Figure 4. High AR to ER ratio identifies a subgroup with particularly poor survival
Patients with a high AR to ERα ratio assessed by Reverse Phase Protein Array demonstrate a particularly poor disease specific survival compared to patients with a low AR to ERα ratio (A). The group with high AR to ERα ratio had significantly higher CCP score (B) and PCNA expression (C) compared to the group with low AR to ERα ratio.

References

    1. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108.
    1. Fleming GF. Second-Line Therapy for Endometrial Cancer: The Need for Better Options. J Clin Oncol. 2015;33:3535–3540.
    1. Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012;13:e353–361.
    1. Dinkelspiel HE, Wright JD, Lewin SN, Herzog TJ. Contemporary clinical management of endometrial cancer. Obstet Gynecol Int. 2013;2013:583891.
    1. Bender D BT, Leslie KK. Hormones and Receptors in Endometrial Cancer. Proc Obstet Gynecol. 2011;2:25.
    1. Tangen IL, Werner HM, Berg A, Halle MK, Kusonmano K, Trovik J, Hoivik EA, Mills GB, Krakstad C, Salvesen HB. Loss of progesterone receptor links to high proliferation and increases from primary to metastatic endometrial cancer lesions. Eur J Cancer. 2014;50:3003–3010.
    1. Wik E, Raeder MB, Krakstad C, Trovik J, Birkeland E, Hoivik EA, Mjos S, Werner HM, Mannelqvist M, Stefansson IM, Oyan AM, Kalland KH, Akslen LA, et al. Lack of estrogen receptor-alpha is associated with epithelial-mesenchymal transition and PI3K alterations in endometrial carcinoma. Clin Cancer Res. 2013;19:1094–1105.
    1. Kokka F, Brockbank E, Oram D, Gallagher C, Bryant A. Hormonal therapy in advanced or recurrent endometrial cancer. Cochrane Database Syst Rev. 2010:CD007926.
    1. Harris KA, Small EJ. Hormonal treatment for prostate cancer. Expert Opin Investig Drugs. 2001;10:493–510.
    1. Fioretti FM, Sita-Lumsden A, Bevan CL, Brooke GN. Revising the role of the androgen receptor in breast cancer. J Mol Endocrinol. 2014;52:R257–265.
    1. Gibson DA, Simitsidellis I, Collins F, Saunders PT. Evidence of androgen action in endometrial and ovarian cancers. Endocr Relat Cancer. 2014;21:T203–218.
    1. Gelmann EP. Molecular biology of the androgen receptor. J Clin Oncol. 2002;20:3001–3015.
    1. Heinlein CA, Chang C. Androgen receptor in prostate cancer. Endocr Rev. 2004;25:276–308.
    1. Vanaja DK, Mitchell SH, Toft DO, Young CY. Effect of geldanamycin on androgen receptor function and stability. Cell Stress Chaperones. 2002;7:55–64.
    1. Harada N, Atarashi K, Murata Y, Yamaji R, Nakano Y, Inui H. Inhibitory mechanisms of the transcriptional activity of androgen receptor by resveratrol: Implication of DNA binding and acetylation of the receptor. J Steroid Biochem Mol Biol. 2011;123:65–70.
    1. Harada N, Murata Y, Yamaji R, Miura T, Inui H, Nakano Y. Resveratrol down-regulates the androgen receptor at the post-translational level in prostate cancer cells. J Nutr Sci Vitaminol (Tokyo) 2007;53:556–560.
    1. Khan SA, Faisal A, Mpindi JP, Parkkinen JA, Kalliokoski T, Poso A, Kallioniemi OP, Wennerberg K, Kaski S. Comprehensive data-driven analysis of the impact of chemoinformatic structure on the genome-wide biological response profiles of cancer cells to 1159 drugs. BMC Bioinformatics. 2012;13:112.
    1. Watson H, Franks S, Bonney RC. Regulation of epidermal growth factor receptor by androgens in human endometrial cells in culture. Hum Reprod. 1998;13:2585–2591.
    1. Nantermet PV, Masarachia P, Gentile MA, Pennypacker B, Xu J, Holder D, Gerhold D, Towler D, Schmidt A, Kimmel DB, Freedman LP, Harada S, Ray WJ. Androgenic induction of growth and differentiation in the rodent uterus involves the modulation of estrogen-regulated genetic pathways. Endocrinology. 2005;146:564–578.
    1. Qiu M, Bao W, Wang J, Yang T, He X, Liao Y, Wan X. FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer. BMC Cancer. 2014;14:78.
    1. Rose GL, Dowsett M, Mudge JE, White JO, Jeffcoate SL. The inhibitory effects of danazol, danazol metabolites, gestrinone, and testosterone on the growth of human endometrial cells in vitro. Fertil Steril. 1988;49:224–228.
    1. Neulen J, Wagner B, Runge M, Breckwoldt M. Effect of progestins, androgens, estrogens and antiestrogens on 3H-thymidine uptake by human endometrial and endosalpinx cells in vitro. Arch Gynecol. 1987;240:225–232.
    1. Hackenberg R, Beck S, Filmer A, Hushmand Nia A, Kunzmann R, Koch M, Slater EP, Schulz KD. Androgen responsiveness of the new human endometrial cancer cell line MFE-296. Int J Cancer. 1994;57:117–122.
    1. Hackenberg R, Schulz KD. Androgen receptor mediated growth control of breast cancer and endometrial cancer modulated by antiandrogen- and androgen-like steroids. J Steroid Biochem Mol Biol. 1996;56:113–117.
    1. Ito K, Suzuki T, Akahira J, Moriya T, Kaneko C, Utsunomiya H, Yaegashi N, Okamura K, Sasano H. Expression of androgen receptor and 5alpha-reductases in the human normal endometrium and its disorders. Int J Cancer. 2002;99:652–657.
    1. Sasaki M, Oh BR, Dharia A, Fujimoto S, Dahiya R. Inactivation of the human androgen receptor gene is associated with CpG hypermethylation in uterine endometrial cancer. Mol Carcinog. 2000;29:59–66.
    1. Kato J, Seto T. Correlation of androgen receptors with histological differentiation in human endometrial carcinomas. Acta obstetricia et gynecologica Scandinavica. 1985;64:209–212.
    1. Kadar N, Malfetano JH, Homesley HD. Steroid receptor concentrations in endometrial carcinoma: effect on survival in surgically staged patients. Gynecol Oncol. 1993;50:281–286.
    1. Fukuda K, Mori M, Uchiyama M, Iwai K, Iwasaka T, Sugimori H. Prognostic significance of progesterone receptor immunohistochemistry in endometrial carcinoma. Gynecol Oncol. 1998;69:220–225.
    1. Kleine W, Maier T, Geyer H, Pfleiderer A. Estrogen and progesterone receptors in endometrial cancer and their prognostic relevance. Gynecol Oncol. 1990;38:59–65.
    1. Culig Z. Targeting the androgen receptor in prostate cancer. Expert Opin Pharmacother. 2014;15:1427–1437.
    1. Elebro K, Borgquist S, Simonsson M, Markkula A, Jirstrom K, Ingvar C, Rose C, Jernstrom H. Combined Androgen and Estrogen Receptor Status in Breast Cancer: Treatment Prediction and Prognosis in a Population-Based Prospective Cohort. Clin Cancer Res. 2015;21:3640–3650.
    1. Cochrane DR, Bernales S, Jacobsen BM, Cittelly DM, Howe EN, D'Amato NC, Spoelstra NS, Edgerton SM, Jean A, Guerrero J, Gomez F, Medicherla S, Alfaro IE, et al. Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide. Breast Cancer Res. 2014;16:R7.
    1. Cochrane DR, Bernales S, Jacobsen BM, Cittelly DM, Howe EN, D'Amato NC, Spoelstra NS, Edgerton SM, Jean A, Guerrero J, Gómez F, Medicherla S, Alfaro IE, et al. Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide. Breast Cancer Res. 2014;16:R7.
    1. Carlson MJ, Thiel KW, Leslie KK. Past, present, and future of hormonal therapy in recurrent endometrial cancer. Int J Womens Health. 2014;6:429–435.
    1. Krakstad C, Trovik J, Wik E, Engelsen IB, Werner HM, Birkeland E, Raeder MB, Oyan AM, Stefansson IM, Kalland KH, Akslen LA, Salvesen HB. Loss of GPER identifies new targets for therapy among a subgroup of ERalpha-positive endometrial cancer patients with poor outcome. Br J Cancer. 2012;106:1682–1688.
    1. Lindstrom LS, Karlsson E, Wilking UM, Johansson U, Hartman J, Lidbrink EK, Hatschek T, Skoog L, Bergh J. Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol. 2012;30:2601–2608.
    1. Werner HM, Berg A, Wik E, Birkeland E, Krakstad C, Kusonmano K, Petersen K, Kalland KH, Oyan AM, Akslen LA, Trovik J, Salvesen HB. ARID1A loss is prevalent in endometrial hyperplasia with atypia and low-grade endometrioid carcinomas. Mod Pathol. 2013;26:428–434.
    1. Trovik J, Wik E, Stefansson IM, Marcickiewicz J, Tingulstad S, Staff AC, Njolstad TS, MoMaTec Study G, Vandenput I, Amant F, Akslen LA, Salvesen HB. Stathmin overexpression identifies high-risk patients and lymph node metastasis in endometrial cancer. Clin Cancer Res. 2011;17:3368–3377.
    1. Krakstad C, Tangen IL, Hoivik EA, Halle MK, Berg A, Werner HM, Raeder MB, Kusonmano K, Zou JX, Oyan AM, Stefansson I, Trovik J, Kalland KH, et al. ATAD2 overexpression links to enrichment of B-MYB-translational signatures and development of aggressive endometrial carcinoma. Oncotarget. 2015;6:28440–28452. doi: 10.18632/oncotarget.4955.
    1. Engelsen IB, Stefansson I, Akslen LA, Salvesen HB. Pathologic expression of p53 or p16 in preoperative curettage specimens identifies high-risk endometrial carcinomas. American journal of obstetrics and gynecology. 2006;195:979–986.
    1. Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998;4:844–847.
    1. Cuzick J, Swanson GP, Fisher G, Brothman AR, Berney DM, Reid JE, Mesher D, Speights VO, Stankiewicz E, Foster CS, Moller H, Scardino P, Warren JD, et al. Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol. 2011;12:245–255.
    1. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–1935.
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499:43–49.
    1. Hennessy BT, Lu Y, Gonzalez-Angulo AM, Carey MS, Myhre S, Ju Z, Davies MA, Liu W, Coombes K, Meric-Bernstam F, Bedrosian I, McGahren M, Agarwal R, et al. A Technical Assessment of the Utility of Reverse Phase Protein Arrays for the Study of the Functional Proteome in Non-microdissected Human Breast Cancers. Clin Proteomics. 2010;6:129–151.
    1. Akbani R, Ng PK, Werner HM, Shahmoradgoli M, Zhang F, Ju Z, Liu W, Yang JY, Yoshihara K, Li J, Ling S, Seviour EG, Ram PT, et al. A pan-cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun. 2014;5:3887.

Source: PubMed

3
Subscribe