Cytotoxic Effect of Progesterone, Tamoxifen and Their Combination in Experimental Cell Models of Human Adrenocortical Cancer

Elisa Rossini, Mariangela Tamburello, Andrea Abate, Silvia Beretta, Martina Fragni, Manuela Cominelli, Deborah Cosentini, Constanze Hantel, Federica Bono, Salvatore Grisanti, Pietro Luigi Poliani, Guido A M Tiberio, Maurizio Memo, Sandra Sigala, Alfredo Berruti, Elisa Rossini, Mariangela Tamburello, Andrea Abate, Silvia Beretta, Martina Fragni, Manuela Cominelli, Deborah Cosentini, Constanze Hantel, Federica Bono, Salvatore Grisanti, Pietro Luigi Poliani, Guido A M Tiberio, Maurizio Memo, Sandra Sigala, Alfredo Berruti

Abstract

Progesterone (Pg) and estrogen (E) receptors (PgRs and ERs) are expressed in normal and neoplastic adrenal cortex, but their role is not fully understood. In literature, Pg demonstrated cytotoxic activity on AdrenoCortical Carcinoma (ACC) cells, while tamoxifen is cytotoxic in NCI-H295R cells. Here, we demonstrated that in ACC cell models, ERs were expressed in NCI-H295R cells with a prevalence of ER-β over the ER-α.Metastasis-derived MUC-1 and ACC115m cells displayed a very weak ER-α/β signal, while PgR cells were expressed, although at low level. Accordingly, these latter were resistant to the SERM tamoxifen and scarcely sensitive to Pg, as we observed a lower potency compared to NCI-H295R cells in cytotoxicity (IC50: MUC-1 cells: 67.58 µM (95%CI: 63.22-73.04), ACC115m cells: 51.76 µM (95%CI: 46.45-57.67) and cell proliferation rate. Exposure of NCI-H295R cells to tamoxifen induced cytotoxicity (IC50: 5.43 µM (95%CI: 5.18-5.69 µM) mainly involving ER-β, as their nuclear localization increased after tamoxifen: Δ A.U. treated vs untreated: 12 h: +27.04% (p < 0.01); 24 h: +36.46% (p < 0.0001). This effect involved the SF-1 protein reduction: Pg: -36.34 ± 9.26%; tamoxifen: -46.25 ± 15.68% (p < 0.01). Finally, in a cohort of 36 ACC samples, immunohistochemistry showed undetectable/low level of ERs, while PgR demonstrated a higher expression. In conclusion, ACC experimental cell models expressed PgR and low levels of ER in line with data obtained in patient tissues, thus limiting the possibility of a clinical approach targeting ER. Interestingly, Pg exerted cytotoxicity also in metastatic ACC cells, although with low potency.

Keywords: ACC cell lines; ACC primary cells; adrenocortical carcinoma; estrogen receptors; progesterone receptors; tamoxifen.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Rossini, Tamburello, Abate, Beretta, Fragni, Cominelli, Cosentini, Hantel, Bono, Grisanti, Poliani, Tiberio, Memo, Sigala and Berruti.

Figures

Figure 1
Figure 1
ER expression in NCI-H295R, MUC-1 cell lines and ACC115m primary culture. Cells were seeded on poly-L-lysine pre-treated coverslips following by incubation with DAPI for nuclear staining. Panel (A) DAPI; panel (B) phalloidin; panel (C) ER (red signal: ER-α; green signal: ER-β); panel (D) merge. The scale bar of 20 µm is automatically inserted by the software ZEN Black.
Figure 2
Figure 2
Effect of tamoxifen on NCI-H295R cell viability and proliferation. (A) NCI-H295R were treated with increasing concentration of tamoxifen (0.1–20 uM) and cell viability was then evaluated by MTT assay. Results are expressed as percent of viable cells vs ctrl ± SEM of three independent experiments run in triplicate. (B) NCI-H295R were treated with low, intermediate, and high dose of tamoxifen and then cell proliferation was evaluated by directing counting with trypan blue discrimination. *P < 0.0001 vs untreated cells; §P < 0.001 vs untreated cells.
Figure 3
Figure 3
Tamoxifen exposure selectively modified the ER intracellular localization in NCI-H295R cells. (A) Cells were treated for different times with tamoxifen IC50 value. Slides were observed by a LSM 880 Zeiss confocal laser microscope or by a LSM 510 Zeiss confocal laser microscope (Carl Zeiss with 40× magnification. Images were then reconstructed using Zeiss ZEN 2.3 Imaging Software (Carl Zeiss). On the left the ER-β staining, on the right ER-β + DAPI staining. (B) The specific mean fluorescence intensity of the pixels of acquired images was quantified using ZEN Black software (Carl Zeiss). Several fields, randomly chosen, were acquired and then analyzed for each experimental condition. Quantified analysis was conducted by GraphPad Prism 5.02 software. *P < 0.0001 vs ctrl; #P < 0.01 vs ctrl.
Figure 4
Figure 4
PgR expression in NCI-H295R, MUC-1 cell lines and ACC115m primary culture. Cells were seeded on poly-L-lysine pre-treated coverslips following by incubation with DAPI for nuclear staining. Panel (A) DAPI; panel (B) phalloidin; panel (C): PgR; panel (D): merge. The scale bar of 20 µm is automatically inserted by the software ZEN Black.
Figure 5
Figure 5
Cytotoxic effect of Pg in ACC cell models. (A) MUC-1 cell line and ACC115m primary culture were treated with increasing concentrations of progesterone (0.1–160 uM), then cell viability was analyzed by MTT assay, (B) NCI-H295R, MUC-1 cell lines and ACC115m primary culture were treated with low, intermediate, and high dose of Pg, and cell proliferation was analyzed by directing counting with trypan blue discrimination. Results are expressed as percent of viable cells vs ctrl ± SEM; *P < 0.0001 vs untreated cells; #P < 0.01 vs untreated cells.
Figure 6
Figure 6
Combined treatment tamoxifen plus Pg in NCI-H295R. (A) Concentration–response curve of tamoxifen, Pg, and drug combination in NCI-H295R. Cells were exposed to increasing concentrations of tamoxifen and Pg alone or in combination as described in Materials and Methods. Data are expressed as percent of viable cells vs ctrl. Data are the mean ± SEM of three independent experiments; *P < 0.0001 vs untreated cells; §P < 0.001 vs untreated cells. (B) Combination index plot. Cell viability data of panel A were converted to Fa values and analyzed with CompuSyn software.
Figure 7
Figure 7
Tamoxifen and Pg reduced the SF-1 expression in NCI-H295R cell line. (1A) Representative western blot of SF-1 expression after NCI-H295R tamoxifen IC50 and Pg IC50 4 days treatment. (1B) Densitometric analysis of SF-1 expression after NCI-H295R drug treatment. Data are expressed as normalized values SF-1/GAPDH and are the mean of three independent experiments. *P < 0.0001 vs ctrl; §P < 0.001 vs ctrl. (1C) NCI-H295R were treated with tamoxifen IC50 or Pg IC50 for 4 days and then miRNA23a/b expression was investigated. Data are expressed as normalized values on internal control U6 and are the mean of three independent experiments. *P < 0.0001 vs ctrl; §P < 0.001 vs ctrl; #P < 0.01 vs ctrl. (2A) Representative western blot of SF-1 expression after MUC-1 tamoxifen IC50 and Pg IC50 5 days treatment. (2B) Densitometric analysis of SF-1 expression after MUC-1 drug treatment. Data are expressed as normalized values SF-1/GAPDH and are the mean of three independent experiments.
Figure 8
Figure 8
Immunohistochemistry for PgR and ER expression in ACC samples. Left panels show representative H&E-stained section from ACC tumor samples, middle panels PgR and right panels ER immunostainings. All images are from ×40 original magnification.

References

    1. Terzolo M, Daffara F, Ardito A, Zaggia B, Basile V, Ferrari L, et al. . Management of Adrenal Cancer: A 2013 Update. J Endocrinol Invest (2014) 37(3):207–17. 10.1007/s40618-013-0049-2
    1. Terzolo M, Angeli A, Fassnacht M, Daffara F, Tauchmanova L, Conton PA, et al. . Adjuvant Mitotane Treatment for Adrenocortical Carcinoma. N Engl J Med (2007) 356(23):2372–80. 10.1056/NEJMoa063360
    1. Berruti A, Grisanti S, Pulzer A, Claps M, Daffara F, Loli P, et al. . Long-Term Outcomes of Adjuvant Mitotane Therapy in Patients With Radically Resected Adrenocortical Carcinoma. J Clin Endocrinol Metab (2017) 102(4):1358–65. 10.1210/jc.2016-2894
    1. Fassnacht M, Assie G, Baudin E, Eisenhofer G, de la Fouchardiere C, Haak HR, et al. . Esmo Guidelines Committee. Electronic Address: Clinicalguidelines@Esmo.Org. Adrenocortical Carcinomas and Malignant Phaeochromocytomas: ESMO-EURACAN Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann Oncol (2020) 31(11):1476–90. 10.1016/j.annonc.2020.08.2099
    1. Fassnacht M, Terzolo M, Allolio B, Baudin E, Haak H, Berruti A, et al. . Firm-Act Study Group. Combination Chemotherapy in Advanced Adrenocortical Carcinoma. N Engl J Med (2012) 66(23):2189–97. 10.1056/NEJMoa1200966
    1. Laganà M, Grisanti S, Cosentini D, Ferrari VD, Lazzari B, Ambrosini R, et al. . Efficacy of the EDP-M Scheme Plus Adjunctive Surgery in the Management of Patients With Advanced Adrenocortical Carcinoma: The Brescia Experience. Cancers (Basel) (2020) 12(4):941. 10.3390/cancers12040941
    1. Cosentini D, Badalamenti G, Grisanti S, Basile V, Rapa I, Cerri S, et al. . Activity and Safety of Temozolomide in Advanced Adrenocortical Carcinoma Patients. Eur J Endocrinol (2019) 181(6):681–9. 10.1530/EJE-19-0570
    1. Sperone P, Ferrero A, Daffara F, Priola A, Zaggia B, Volante M, et al. . Gemcitabine Plus Metronomic 5-Fluorouracil or Capecitabine as a Second-/Third-Line Chemotherapy in Advanced Adrenocortical Carcinoma: A Multicenter Phase II Study. Endocr Relat Cancer (2010) 17(2):445–53. 10.1677/ERC-09-0281
    1. Fiorentini C, Grisanti S, Cosentini D, Abate A, Rossini E, Berruti A, et al. . Molecular Drivers of Potential Immunotherapy Failure in Adrenocortical Carcinoma. J Oncol (2019) 1:6072863. 10.1155/2019/6072863
    1. Grisanti S, Cosentini D, Laganà M, Volta AD, Palumbo C, Massimo Tiberio GA, et al. . The Long and Winding Road to Effective Immunotherapy in Patients With Adrenocortical Carcinoma. Future Oncol (2020) 16(36):3017–20. 10.2217/fon-2020-0686
    1. de Cremoux P, Rosenberg D, Goussard J, Brémont-Weil C, Tissier F, Tran-Perennou C, et al. . Expression of Progesterone and Estradiol Receptors in Normal Adrenal Cortex, Adrenocortical Tumors, and Primary Pigmented Nodular Adrenocortical Disease. Endocr Relat Cancer (2008) 15(2):465–74. 10.1677/ERC-07-0081
    1. Baquedano MS, Saraco N, Berensztein E, Pepe C, Bianchini M, Levy E, et al. . Identification and Developmental Changes of Aromatase and Estrogen Receptor Expression in Prepubertal and Pubertal Human Adrenal Tissues. J Clin Endocrinol Metab (2007) 92(6):2215–22. 10.1210/jc.2006-2329
    1. Barzon L, Masi G, Pacenti M, Trevisan M, Fallo F, Remo A, et al. . Expression of Aromatase and Estrogen Receptors in Human Adrenocortical Tumors. Virchows Arch (2008) 452(2):181–91. 10.1007/s00428-007-0542-0
    1. Medwid S, Guan H, Yang K. Bisphenol A Stimulates Adrenal Cortical Cell Proliferation Via Erβ-Mediated Activation of the Sonic Hedgehog Signalling Pathway. J Steroid Biochem Mol Biol (2018) 178:254–62. 10.1016/j.jsbmb.2018.01.004
    1. Shen XC, Gu XC, Qiu YQ, Du CJ Fu YB, Wu JJ. Estrogen Receptor Expression in Adrenocortical Carcinoma. J Zhejiang Univ Sci B (2009) 10(1):1–6. 10.1631/jzus.B0820072
    1. Montanaro D, Maggiolini M, Recchia AG, Sirianni R, Aquila S, Barzon L, et al. . Antiestrogens Upregulate Estrogen Receptor Beta Expression and Inhibit Adrenocortical H295R Cell Proliferation. J Mol Endocrinol (2005) 35(2):245–56. 10.1677/jme.1.01806
    1. Zhao C, Dahlman-Wright K, Gustafsson J-Å. Estrogen Receptor ß: An Overview and Update. Nucl Recept Signal (2008) 6:e003. 10.1621/nrs.06003
    1. Fragni M, Fiorentini C, Rossini E, Fisogni S, Vezzoli S, Bonini SA, et al. . In Vitro Antitumor Activity of Progesterone in Human Adrenocortical Carcinoma. Endocrine (2019. a) 63(3):592–601. 10.1007/s12020-018-1795-x
    1. Katzenellenbogen BS, Norman MJ. Multihormonal Regulation of the Progesterone Receptor in MCF-7 Human Breast Cancer Cells: Interrelationships Among Insulin/Insulin-Like Growth factor-I, Serum, and Estrogen. Endocrinology (1990) 126(2):891–8. 10.1210/endo-126-2-891
    1. Carlson JA, Jr, Allegra JC, Day TG, Jr, Wittliff JL. Tamoxifen and Endometrial Carcinoma: Alterations in Estrogen and Progesterone Receptors in Untreated Patients and Combination Hormonal Therapy in Advanced Neoplasia. Am J Obstet Gynecol (1984) 149(2):149–53. 10.1016/0002-9378(84)90187-x
    1. Abiven-Lepage G, Coste J, Tissier F, Groussin L, Billaud L, Dousset B, et al. . Adrenocortical Carcinoma and Pregnancy: Clinical and Biological Features and Prognosis. Eur J Endocrinol (2010) 163(5):793–800. 10.1530/EJE-10-0412
    1. de Corbière P, Ritzel K, Cazabat L, Ropers J, Schott M, Libé R, et al. . Pregnancy in Women Previously Treated for an Adrenocortical Carcinoma. J Clin Endocrinol Metab (2015) 100(12):4604–11. 10.1210/jc.2015-2341
    1. Altinoz MA, Bilir A, Gedikoglu G, Ozcan E, Oktem G, Muslumanoglu M. Medroxyprogesterone and Tamoxifen Augment Anti-Proliferative Efficacy and Reduce Mitochondria-Toxicity of Epirubicin in FM3A Tumor Cells In Vitro. Cell Biol Int (2007) 31(5):473–81. 10.1016/j.cellbi.2006.11.013
    1. Zaino RJ, Satyaswaroop PG, Mortel R. Hormonal Therapy of Human Endometrial Adenocarcinoma in a Nude Mouse Model. Cancer Res (1985) 45(2):539–41.
    1. Wen L, Hong D, Yanyin W, Mingyue Z, Baohua L. Effect of Tamoxifen, Methoxyprogesterone Acetate and Combined Treatment on Cellular Proliferation and Apoptosis in SKOV3/DDP Cells Via the Regulation of Vascular Endothelial Growth Factor. Arch Gynecol Obstet (2013) 287(5):997–1004. 10.1007/s00404-012-2664-0
    1. Herzog TJ. What is the Clinical Value of Adding Tamoxifen to Progestins in the Treatment [Correction for Treament] of Advanced or Recurrent Endometrial Cancer? Gynecol Oncol (2004) 92(1):1–3. 10.1016/j.ygyno.2003.11.014
    1. Rainey WE, Saner K, Schimmer BP. Adrenocortical Cell Lines. Mol Cell Endocrinol (2004) 228(1-2):23–38. 10.1016/j.mce.2003.12.020
    1. Hantel, Shapiro I, Poli G, Chiapponi C, Bidlingmaier M, Reincke M, et al. . Targeting Heterogeneity of Adrenocortical Carcinoma: Evaluation and Extension of Preclinical Tumor Models to Improve Clinical Translation. Oncotarget (2016) 7(48):79292–304. 10.18632/oncotarget.12685
    1. Fragni M, Palma Lopez LP, Rossini E, Abate A, Cosentini D, Salvi V, et al. . In Vitro Cytotoxicity of Cabazitaxel in Adrenocortical Carcinoma Cell Lines and Human Adrenocortical Carcinoma Primary Cell Cultures. Mol Cell Endocrinol (2019. b) 498:110585. 10.1016/j.mce.2019.110585
    1. Lloyd RV, Osamura RY, Klöppel G, Rosai J. Who Classification of Tumours of Endocrine Organs. Lyon: IARC: WHO; (2017).
    1. Fiorentini C, Fragni M, Perego P, Vezzoli S, Bonini SA, Tortoreto M, et al. . Antisecretive and Antitumor Activity of Abiraterone Acetate in Human Adrenocortical Cancer: A Preclinical Study. J Clin Endocrinol Metab (2016) 101(12):4594–602. 10.1210/jc.2016-2414
    1. Chou TC, Talalay P. Quantitative Analysis of Dose-Effect Relationships: The Combined Effects of Multiple Drugs or Enzyme Inhibitors. Adv Enzyme Regul (1984) 22:27–55. 10.1016/0065-2571(84)90007-4
    1. Chou TC. Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies. Pharmacol Rev (2006) 58(3):621–81. 10.1124/pr.58.3.10
    1. Fragni M, Bonini SA, Stabile A, Bodei S, Cristinelli L, Simeone C, et al. . Inhibition of Survivin is Associated With Zoledronic Acid-Induced Apoptosis of Prostate Cancer Cells. Anticancer Res (2016. a) 36:913–20.
    1. Fragni M, Bonini SA, Bettinsoli P, Bodei S, Generali D, Bottini A, et al. . The miR-21/PTEN/Akt Signaling Pathway is Involved in the Anti-Tumoral Effects of Zoledronic Acid in Human Breast Cancer Cell Lines. Naunyn Schmiedebergs Arch Pharmacol (2016. b) 389(5):529–38. 10.1007/s00210-016-1224-8
    1. Lalli E, Doghman M, Latre de Late P, El Wakil A, Mus-Veteau I. Beyond Steroidogenesis: Novel Target Genes for SF-1 Discovered by Genomics. Mol Cell Endocrinol (2013) 371(1-2):154–9. 10.1016/j.mce.2012.11.005
    1. Shen L, Yang S, Huang W, Xu W, Wang Q, Song Y, et al. . MicroRNA23a and microRNA23b Deregulation Derepresses SF-1 and Upregulates Estrogen Signaling in Ovarian Endometriosis. J Clin Endocrinol Metab (2013) 98(4):1575–82. 10.1210/jc.2012-3010
    1. Gruvberger-Saal SK, Bendahl PO, Saal LH, Laakso M, Hegardt C, Edén P, et al. . Estrogen Receptor Beta Expression is Associated With Tamoxifen Response in ERalpha-negative Breast Carcinoma. Clin Cancer Res (2007) 13(7):1987–94. 10.1158/1078-0432.CCR-06-1823
    1. Tamoxifen . Ibm Micromedex® DRUGDEX® (Electronic Version) . Greenwood Village, Colorado, USA: IBM Watson Health. Available at: (Accessed January, 04, 2021).
    1. Boonyaratanakornkit V, McGowan E, Sherman L, Mancini MA, Cheskis BJ, Edwards DP. The Role of Extranuclear Signaling Actions of Progesterone Receptor in Mediating Progesterone Regulation of Gene Expression and the Cell Cycle. Mol Endocrinol (2007) 21:359–75. 10.1210/me.2006-0337
    1. Altinoz MA, Ozpinar A, Elmaci I. Reproductive Epidemiology of Glial Tumors may Reveal Novel Treatments: High-Dose Progestins or Progesterone Antagonists as Endocrino-Immune Modifiers Against Glioma. Neurosurg Rev (2019) 42(2):351–69. 10.1007/s10143-018-0953-1
    1. Motamed HR, Shariati M, Ahmadi R, Khatamsaz S, Mokhtari M. The Apoptotic Effects of Progesterone on Breast Cancer (MCF-7) and Human Osteosarcoma (MG-636) Cells. Physiol Int (2020) 107(3):406–18. 10.1556/2060.2020.00034
    1. Truong TH, Lange CA. Deciphering Steroid Receptor Crosstalk in Hormone-Driven Cancers. Endocrinology (2018) 159(12):3897–907. 10.1210/en.2018-00831
    1. Lalli E. Adrenocortical Development and Cancer: Focus on SF-1. J Mol Endocrinol (2010) 44(6):301–7. 10.1677/JME-09-0143
    1. Doghman M, Karpova T, Rodrigues GA, Arhatte M, De Moura J, Cavalli LR, et al. . Increased Steroidogenic Factor-1 Dosage Triggers Adrenocortical Cell Proliferation and Cancer. Mol Endocrinol (2007) 12:2968–87. 10.1210/me.2007-0120
    1. Jonas S, Izaurralde E. Towards a Molecular Understanding of MicroRNA-Mediated Gene Silencing. Nat Rev Genet (2015) 16(7):421–33. 10.1038/nrg3965
    1. Bhat-Nakshatri P, Wang G, Collins NR, Thomson MJ, Geistlinger TR, Carroll JS, et al. . Estradiol-Regulated microRNAs Control Estradiol Response in Breast Cancer Cells. Nucleic Acids Res (2009) 37(14):4850–61. 10.1093/nar/gkp500
    1. Huang FY, Wong DK, Seto WK, Lai CL, Yuen MF. Estradiol Induces Apoptosis Via Activation of miRNA-23a and p53: Implication for Gender Difference in Liver Cancer Development. Oncotarget (2015) 6(33):34941–52. 10.18632/oncotarget.5472
    1. Paris O, Ferraro L, Grober OM, Ravo M, De Filippo MR, Giurato G, et al. . Direct Regulation of microRNA Biogenesis and Expression by Estrogen Receptor Beta in Hormone-Responsive Breast Cancer. Oncogene (2012) 31(38):4196–206. 10.1038/onc.2011.583
    1. McFall T, McKnight B, Rosati R, Kim S, Huang Y, Viola-Villegas N, et al. . Progesterone Receptor A Promotes Invasiveness and Metastasis of Luminal Breast Cancer by Suppressing Regulation of Critical microRNAs by Estrogen. J Biol Chem (2018) 293(4):1163–77. 10.1074/jbc.M117.812438

Source: PubMed

3
Předplatit