Phospho-kinase profile of triple negative breast cancer and androgen receptor signaling

María D Cuenca-López, Juan C Montero, Jorge C Morales, Aleix Prat, Atanasio Pandiella, Alberto Ocana, María D Cuenca-López, Juan C Montero, Jorge C Morales, Aleix Prat, Atanasio Pandiella, Alberto Ocana

Abstract

Background: The androgen receptor (AR) plays a central role in the oncogenesis of different tumors, as is the case in prostate cancer. In triple negative breast cancer (TNBC) a gene expression classification has described different subgroups including a luminal androgen subtype. The AR can be controlled by several mechanisms like the activation of membrane tyrosine kinases and downstream signaling pathways. However little is known in TNBC about how the AR is modulated by these mechanisms and the potential therapeutic strategists to inhibit its expression.

Methods: We used human samples to evaluate the expression of AR by western-blot and phospho-proteomic kinase arrays that recognize membrane tyrosine kinase receptors and downstream mediators. Western-blots in human cell lines were carried out to analyze the expression and activation of individual proteins. Drugs against these kinases in different conditions were used to measure the expression of the androgen receptor. PCR experiments were performed to assess changes in the AR gene after therapeutic modulation of these pathways.

Results: AR is present in a subset of TNBC and its expression correlates with activated membrane receptor kinases-EGFR and PDGFRβ in human samples and cell lines. Inhibition of the PI3K/mTOR pathway in TNBC cell lines decreased notably the expression of the AR. Concomitant administration of the anti-androgen bicalutamide with the EGFR, PDGFRβ and Erk1/2 inhibitors, decreased the amount of AR compared to each agent given alone, and had an additive anti-proliferative effect. Administration of dihydrotestosterone augmented the expression of AR that was not modified by the inhibition of the PI3K/mTOR or Erk1/2 pathways. AR expression was posttranscriptionally regulated by PI3K or Erk1/2 inhibition.

Conclusion: Our results describe the expression of the AR in TNBC as a druggable target and further suggest the combination of bicalutamide with inhibitors of EGFR, PDGFRβ or Erk1/2 for future development.

Figures

Figure 1
Figure 1
Expression of AR in a panel of TNBC cell lines and in human samples. A) Eight cell lines, Hs578T, MDA-MB231, MDA-MB435, HCC 1187, HBL 100, BT 549, HCC 70 and HCC 3153 were cultured and cell lysates were analyzed by western blot for AR expression. α-tubuline was used as a loading control. Expression of the AR gene from public gene expression profile data bases. B) Human samples of triple negative breast cancer patients were processed for protein extraction and lysates were analyzed by western blot for AR expression. α-tubuline was used as a loading control.
Figure 2
Figure 2
Activation of TKRs and their downstream pathways in human samples and in TNBC cell lines. A) Human samples of triple negative breast cancer patients were processed for protein extraction. Lysates containing 1 mg/ml protein were analyzed for the level of tyrosine phosphorylation in a panel of 42 receptor tyrosine kinases (RTKs) and citosolic kinases using antibody array kits. The levels of phosphorylated EGFR, HER2, HER3, PDGFRβ, Akt (S473), Akt (T308) and Erk1/2 are shown (indicated by arrows). B) Relationship between pPDGFRβ and pEGFR and AR expression in human samples. Activated PDGFRβ and EGFR obtained from the antibody array kit were measured using a gray level scale in arbitrary units. AR expression measured by western blot was classified in two groups: negative or positive. Statistical difference (p < 0.05) was analyzed using a Mann–Whitney test. C) Eight cell lines Hs578T, MDA-MB231, MDA-MB435, HCC 1187, HBL 100, BT 549, HCC 70 and HCC 3153 were cultured and cell lysates were analyzed by western blot for the levels of different proteins as well as their phosphorylated forms.
Figure 3
Figure 3
Effect of PI3K/mTOR, Erk1/2 and EGFR/PDGFRβ inhibitors alone or in combination with bicalutamide on the AR expression and cell proliferation in Hs578T and BT549. A) Effect of drugs on AR expression in Hs578T and BT549. Cells were cultured and treated with drugs for 24 h. Cell lysates were analyzed by western blot for AR expression. α-tubuline was used as a loading control. B) Effect of drugs on cell proliferation in Hs578T and BT549. MTT metabolization was performed after 4 days to evaluate cell proliferation. Control cells were untreated. Statistical difference (* = p < 0.05, Bic versus control or drug combination versus drug alone.) was analyzed using a Test T. C) Effect of drugs on cell proliferation in Hs578T and BT549. Cell counting was performed after 4 days to evaluate cell proliferation. Control cells were untreated. Statistical difference (* = p < 0.05, Bic versus control or drug combination versus drug alone.) was analyzed using a Test T.
Figure 4
Figure 4
Effect of DHT and bicalutamide alone and in combination with PI3K/mTOR, Erk1/2 and EGFR/PDGFRβ inhibitors in AR expression and cell proliferation. A) Effect of DHT and bicalutamide, BEZ and PD in alone and combined on AR expression in Hs578T and BT549. Cells were cultured and treated with drugs for 24 h. Cell lysates were analyzed by western blot for AR expression. α-tubuline was used as loading control. B) In parallel, total RNA was extracted and subjected to RT-PCR. Subsequently, amplification products were resolved on agarose gel to determine AR mRNA levels. Actin was used as protein loading control and mRNA as gene control.

References

    1. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lønning PE, Børresen-Dale AL, Brown PO, Botstein D. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093.
    1. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, Thorsen T, Quist H, Matese JC, Brown PO, Botstein D, Lønning PE, Børresen-Dale AL. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001;98:10869–10874. doi: 10.1073/pnas.191367098.
    1. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13:4429–4434. doi: 10.1158/1078-0432.CCR-06-3045.
    1. Gruber CJ, Tschugguel W, Schneeberger C, Huber JC. Production and actions of estrogens. N Engl J Med. 2002;346:340–352. doi: 10.1056/NEJMra000471.
    1. Smith IE, Dowsett M. Aromatase inhibitors in breast cancer. N Engl J Med. 2003;348:2431–2442. doi: 10.1056/NEJMra023246.
    1. Agoff SN, Swanson PE, Linden H, Hawes SE, Lawton TJ. Androgen receptor expression in estrogen receptor-negative breast cancer. Immunohistochemical, clinical, and prognostic associations. Am J Clin Pathol. 2003;120:725–731. doi: 10.1309/42F00D0DJD0J5EDT.
    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. 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. 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. 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. Kotsopoulos J, Narod SA. Androgens and breast cancer. Steroids. 2012;77:1–9. doi: 10.1016/j.steroids.2011.10.002.
    1. Mellinghoff IK, Vivanco I, Kwon A, Tran C, Wongvipat J, Sawyers CL. HER2/neu kinase-dependent modulation of androgen receptor function through effects on DNA binding and stability. Cancer Cell. 2004;6:517–527. doi: 10.1016/j.ccr.2004.09.031.
    1. Ni M, Chen Y, Lim E, Wimberly H, Bailey ST, Imai Y, Rimm DL, Shirley Liu X, 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. Carver BS, Chapinski C, Wongvipat J, Hieronymus H, Chen Y, Chandarlapaty S, Arora VK, Le C, Koutcher J, Scher H, Scardino PT, Rosen N, Sawyers CL. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19:575–586. doi: 10.1016/j.ccr.2011.04.008.
    1. Mehrian-Shai R, Chen CD, Shi T, Horvath S, Nelson SF, Reichardt JK, Sawyers CL. Insulin growth factor-binding protein 2 is a candidate biomarker for PTEN status and PI3K/Akt pathway activation in glioblastoma and prostate cancer. Proc Natl Acad Sci U S A. 2007;104:5563–5568. doi: 10.1073/pnas.0609139104.
    1. Gonzalez-Angulo AM, Stemke-Hale K, Palla SL, Carey M, Agarwal R, Meric-Berstam F, Traina TA, Hudis C, Hortobagyi GN, Gerald WL, Mills GB, Hennessy BT. Androgen receptor levels and association with PIK3CA mutations and prognosis in breast cancer. Clin Cancer Res. 2009;15:2472–2478. doi: 10.1158/1078-0432.CCR-08-1763.
    1. Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004;10:5367–5374. doi: 10.1158/1078-0432.CCR-04-0220.
    1. Hochgrafe F, Zhang L, O’Toole SA, Browne BC, Pinese M, Porta Cubas A, Lehrbach GM, Croucher DR, Rickwood D, Boulghourjian A, Shearer R, Nair R, Swarbrick A, Faratian D, Mullen P, Harrison DJ, Biankin AV, Sutherland RL, Raftery MJ, Daly RJ. Tyrosine phosphorylation profiling reveals the signaling network characteristics of Basal breast cancer cells. Cancer Res. 2010;70:9391–9401. doi: 10.1158/0008-5472.CAN-10-0911.
    1. Sun T, Aceto N, Meerbrey KL, Kessler JD, Zhou C, Migliaccio I, Nguyen DX, Pavlova NN, Botero M, Huang J, Bernardi RJ, Schmitt E, Hu G, Li MZ, Dephoure N, Gygi SP, Rao M, Creighton CJ, Hilsenbeck SG, Shaw CA, Muzny D, Gibbs RA, Wheeler DA, Osborne CK, Schiff R, Bentires-Alj M, Elledge SJ, Westbrook TF. Activation of multiple proto-oncogenic tyrosine kinases in breast cancer via loss of the PTPN12 phosphatase. Cell. 2011;144:703–718. doi: 10.1016/j.cell.2011.02.003.
    1. Montero JC, Seoane S, Ocana A, Pandiella A. P-Rex1 participates in Neuregulin-ErbB signal transduction and its expression correlates with patient outcome in breast cancer. Oncogene. 2011;30:1059–1071. doi: 10.1038/onc.2010.489.
    1. Diaz-Rodriguez E, Cabrera N, Esparis-Ogando A, Montero JC, Pandiella A. Cleavage of the TrkA neurotrophin receptor by multiple metalloproteases generates signalling-competent truncated forms. Eur J Neurosci. 1999;11:1421–1430. doi: 10.1046/j.1460-9568.1999.00552.x.
    1. Yuste L, Montero JC, Esparis-Ogando A, Pandiella A. Activation of ErbB2 by overexpression or by transmembrane neuregulin results in differential signaling and sensitivity to herceptin. Cancer Res. 2005;65:6801–6810. doi: 10.1158/0008-5472.CAN-04-4023.
    1. Gregory CW, Whang YE, McCall W, Fei X, Liu Y, Ponguta LA, French FS, Wilson EM, Earp HS 3rd. Heregulin-induced activation of HER2 and HER3 increases androgen receptor transactivation and CWR-R1 human recurrent prostate cancer cell growth. Clin Cancer Res. 2005;11:1704–1712. doi: 10.1158/1078-0432.CCR-04-1158.
    1. Culig Z, Hobisch A, Cronauer MV, Radmayr C, Trapman J, Hittmair A, Bartsch G, Klocker H. Androgen receptor activation in prostatic tumor cell lines by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth factor. Cancer Res. 1994;54:5474–5478.
    1. Chen L, Mooso BA, Jathal MK, Madhav A, Johnson SD, van Spyk E, Mikhailova M, Zierenberg-Ripoll A, Xue L, Vinall RL, DeVere White RW, Ghosh PM. Dual EGFR/HER2 inhibition sensitizes prostate cancer cells to androgen withdrawal by suppressing ErbB3. Clin Cancer Res. 2011;17:6218–6228. doi: 10.1158/1078-0432.CCR-11-1548.
    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. Montero JC, Esparis-Ogando A, Re-Louhau MF, Seoane S, Abad M, Calero R, Ocana A, Pandiella A. Active kinase profiling, genetic and pharmacological data define mTOR as an important common target in triple-negative breast cancer. Oncogene. 2014;33:148–156. doi: 10.1038/onc.2012.572.
    1. Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, Stegh AH, Bradner JE, Ligon KL, Brennan C, Chin L, DePinho RA. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 2007;318:287–290. doi: 10.1126/science.1142946.
    1. Naderi A, Chia K, Liu J. Synergy between inhibitors of androgen receptor and MEK has therapeutic implications in estrogen receptor-negative breast cancer. Breast Cancer Res. 2011;13:R36. doi: 10.1186/bcr2858.
    1. Abiraterone Acetate in Treating Postmenopausal Women With Advanced or Metastatic Breast Cancer. [ ]

Source: PubMed

3
Subscribe