Aberrant corticosteroid metabolism in tumor cells enables GR takeover in enzalutamide resistant prostate cancer

Jianneng Li, Mohammad Alyamani, Ao Zhang, Kai-Hsiung Chang, Michael Berk, Zhenfei Li, Ziqi Zhu, Marianne Petro, Cristina Magi-Galluzzi, Mary-Ellen Taplin, Jorge A Garcia, Kevin Courtney, Eric A Klein, Nima Sharifi, Jianneng Li, Mohammad Alyamani, Ao Zhang, Kai-Hsiung Chang, Michael Berk, Zhenfei Li, Ziqi Zhu, Marianne Petro, Cristina Magi-Galluzzi, Mary-Ellen Taplin, Jorge A Garcia, Kevin Courtney, Eric A Klein, Nima Sharifi

Abstract

Prostate cancer is driven by androgen stimulation of the androgen receptor (AR). The next-generation AR antagonist, enzalutamide, prolongs survival, but resistance and lethal disease eventually prevail. Emerging data suggest that the glucocorticoid receptor (GR) is upregulated in this context, stimulating expression of AR-target genes that permit continued growth despite AR blockade. However, countering this mechanism by administration of GR antagonists is problematic because GR is essential for life. We show that enzalutamide treatment in human models of prostate cancer and patient tissues is accompanied by a ubiquitin E3-ligase, AMFR, mediating loss of 11β-hydroxysteroid dehydrogenase-2 (11β-HSD2), which otherwise inactivates cortisol, sustaining tumor cortisol concentrations to stimulate GR and enzalutamide resistance. Remarkably, reinstatement of 11β-HSD2 expression, or AMFR loss, reverses enzalutamide resistance in mouse xenograft tumors. Together, these findings reveal a surprising metabolic mechanism of enzalutamide resistance that may be targeted with a strategy that circumvents a requirement for systemic GR ablation.

Keywords: androgens; biochemistry; cancer biology; glucocorticoids; human; nuclear receptors; prostate cancer; treatment resistance.

Conflict of interest statement

The authors declare that no competing interests exist.

Figures

Figure 1.. GR stimulation with enzalutamide resistance…
Figure 1.. GR stimulation with enzalutamide resistance in prostate cancer is tightly regulated by glucocorticoid metabolism in target tissues.
(A) Glucocorticoid metabolism in target tissues. Stimulation of GR by cortisol in humans is limited by 11β-HSD2, which oxidizes and converts cortisol to inactive cortisone. In mice, 11β-HSD2 converts active corticosterone to inactive 11-dehydrocorticosterone. (B) Enzalutamide (Enz) sustains cortisol levels by retarding inactivation in the LAPC4 and VCaP human prostate cancer cell lines. Cells were treated with the indicated concentrations of Enz or vehicle for 36 days (LAPC4) or 40 days (VCaP), and subsequently treated with [3H]-cortisol (100 nM) for the indicated times, followed by steroid extraction from media (above) and cells (below), steroid separation and quantitation with HPLC. The experiment was done in duplicate and repeated at least three times. (C) Cortisol inactivation is impaired in xenograft tumors treated with Enz. Fresh tumor tissues were harvested from LAPC4 or VCaP xenografts grown in orchiectomized mice and treated with Enz or chow alone (n = 5 tumors per treatment group). Tumors were treated with [3H]-cortisol (100 nM) for the indicated times and steroids were extracted from media and analyzed by HPLC. Error bars represent the SD. (D) Enz suppresses LAPC4 cell line proliferation. LAPC4 cells were treated with vehicle (Ctrl) or the indicated concentration of Enz for the designated number of days and cell viability was assessed using CellTiter-Glo. Cell viability was normalized to day 0, experiments were performed in triplicate and error bars represent the SD. DOI:http://dx.doi.org/10.7554/eLife.20183.002
Figure 1—figure supplement 1.. Effects of Enz…
Figure 1—figure supplement 1.. Effects of Enz on LAPC4 cells.
(A) Short term Enz treatment does not affect cortisol metabolism. Previously untreated cells were treated with the indicated concentration of Enz or Vehicle and concomitantly with [3H]-cortisol (100 nM) for the indicated times and steroids were separated and quantitated by HPLC. (B) Enz suppresses expression of AR-regulated transcripts and has no acute effect on expression of GR, HSD11B2 or HSD11B1. LAPC4 cells were treated with the indicated concentration of Enz or Vehicle for 24 hr and the indicated transcripts were assessed by qPCR. Expression is normalized to Vehicle control and RPLP0. (C) The viability of LAPC4 cells recovers with long-term Enz treatment. The cells were treated with long-term (Enz D56), short-term (Ctrl + Enz) Enz (10 µM), or no treatment (Ctrl) for the indicated number of days and cell viability was assessed relative to day 0. Experiments were performed in triplicate and error bars represent the SD. DOI:http://dx.doi.org/10.7554/eLife.20183.003
Figure 2.. Enzalutamide promotes 11β-HSD2 protein loss…
Figure 2.. Enzalutamide promotes 11β-HSD2 protein loss in cell line models and tissues from patients with prostate cancer.
(A) Enzalutamide (Enz) treatment results in the loss of 11β-HSD2 protein that occurs concurrently with an increase in GR protein in the LAPC4 model of CRPC as assessed by Western blot. (B) 11β-HSD2 protein expression in Enz treated LAPC4 cells as assessed by immunocytochemistry. (C,D) Loss of 11β-HSD2 and increase in GR protein similarly occur with Enz treatment in the VCaP and MDA-PCa-2b models. (E) Enz induces loss of 11β-HSD2 protein in tissue from patients with prostate cancer. Local prostate biopsies were obtained from Patients #1 and #2 with image guidance in a neoadjuvant study before (Pre) and after (Post) two months of treatment with Enz and medical castration. Patients #3 and #4 had biopsies of metastatic CRPC from lymph nodes before and after three months (Patient #3) and 11 months (Patient #4) of treatment with Enz. Fresh tissues from Patients #5-#11 were obtained from surgical prostatectomy specimens and incubated with vehicle or Enz (10 µM) for 7–8 days prior to protein extraction and Western blot. DOI:http://dx.doi.org/10.7554/eLife.20183.004
Figure 2—figure supplement 1.. Response to Enz…
Figure 2—figure supplement 1.. Response to Enz in prostate cancer cell lines and human tissues.
(A) Enz treatment does not change 11β-HSD2 protein expression in the AR-negative DU145 prostate cancer cell line. (B) Enz treatment does not change 11β-HSD1 protein expression in AR-expressing prostate cancer cell lines. Cells were treated with the indicated concentrations of Enz, whole cell protein lysates were obtained, separated and assessed with anti-11β-HSD1 and anti-β-actin antibodies. (C) GR transcript increases and HSD11B2 is unchanged with long-term Enz treatment of LAPC4 and VCaP cells. Expression is normalized to vehicle treated cells and RPLP0. (D) Enz does not directly antagonize 11β-HSD2. LAPC4 cells were transfected with a vector encoding 11β-HSD2, in the presence of the indicated concentration of Enz or Vehicle, and conversion from [3H]-cortisol (100 nM) to cortisone was assessed by HPLC. Experiments performed in biological duplicate. (E) 11β-HSD2 loss is not attributable to GR stimulation. LAPC4 cells were treated with dexamethasone (DEX; 100 nM) for the indicated durations, whole cell protein lysates were obtained and assessed with anti-11β-HSD2 and anti-β-actin antibodies. (F) GR protein expression is induced in a subset of the patient tissues from Figure 2E. All six tissues that have induction of GR expression exhibit loss of 11β-HSD2 in Figure 2. DOI:http://dx.doi.org/10.7554/eLife.20183.005
Figure 3.. 11β-HSD2 expression reverses enzalutamide-sustained cortisol…
Figure 3.. 11β-HSD2 expression reverses enzalutamide-sustained cortisol levels and GR-responsive gene expression.
(A) Impeded conversion from cortisol to cortisone with Enz treatment is reversible with transient and (B) stable 11β-HSD2 expression. Cells expressing 11β-HSD2 or empty vector (control) were treated with the indicated concentration of Enz for 40 days, followed by treatment with [3H]-cortisol and analysis of steroids in media by HPLC. (C) With Enz treatment, only cortisol-induced GR signaling is specifically reversible with forced stable 11β-HSD2 expression. LAPC4 cells were treated with Enz for 36 days, starved with phenol-red-free medium containing 5% Charcoal:Dextran-stripped FBS for 48 hr and transfected with a plasmid expressing 11β-HSD2 and treated with the indicated conditions for 24 hr. Only cortisol induction of PSA expression, which is GR- and metabolism-dependent, is reversible by 11β-HSD2. Expression of KLF9, which is regulated only by GR, is induced by cortisol and dexamethasone, but only cortisol induction is reversible by 11β-HSD2. Expression of PMEPA1, which is regulated only by AR, is induced with DHT only and not reversible by 11β-HSD2. Expression is normalized to vehicle-treated cells (not shown) and RPLP0 expression. The experiment was performed four times. Error bars represent the SD of a representative experiment performed in triplicate. DOI:http://dx.doi.org/10.7554/eLife.20183.006
Figure 3—figure supplement 1.. 11β-HSD2 overexpression (OE)…
Figure 3—figure supplement 1.. 11β-HSD2 overexpression (OE) in the long-term Enz-treated LAPC4 cells is comparable to endogenous expression in the human placental derived JEG-3 cell line.
DOI:http://dx.doi.org/10.7554/eLife.20183.007
Figure 4.. Reinstatement of 11β-HSD2 expression restores…
Figure 4.. Reinstatement of 11β-HSD2 expression restores sensitivity to enzalutamide therapy by specifically suppressing tumor corticosterone.
(A) Expression of 11β-HSD2 reverses enzalutamide (Enz) resistant LAPC4 CRPC xenograft tumor growth. (B) Progression-free survival is prolonged by 11β-HSD2 expression in Enz-treated LAPC4 xenografts. N.S. = not significant. (C) 11β-HSD2 expression reverses Enz resistance in the VCaP xenograft model of CRPC as assessed by decreased tumor volume and (D) prolongation of progression-free survival. For both xenograft studies, cells expressing 11β-HSD2 or vector (control) were grown in orchiectomized mice supplemented with DHEA and arbitrarily assigned to Enz or chow (Ctrl). For the comparisons in tumor volume, the significance of the difference between 11β-HSD2/Enz and Vector/Enz was calculated with an unpaired and two-tailed t-test on day 24 (LAPC4) or day 23 (VCaP). For the comparisons in progression-free survival, the significance of the difference between 11β-HSD2/Enz and Vector/Enz was calculated with a log-rank test. (E) The absolute concentration of corticosterone is reduced in xenograft tumors expressing 11β-HSD2. (F) The percentage of corticosterone relative to 11-dehydrocorticosterone is reduced in tumors expressing 11β-HSD2. (G) The absolute concentration of corticosterone and (H) percentage of corticosterone relative to 11-dehydrocorticosterone in serum are unaffected in mice harboring tumors with restored 11β-HSD2 expression. P values in E-H were calculated with an unpaired and two-tailed t-test. DOI:http://dx.doi.org/10.7554/eLife.20183.008
Figure 4—figure supplement 1.. Forced 11β-HSD2 expression…
Figure 4—figure supplement 1.. Forced 11β-HSD2 expression in Enz-treated LAPC4 xenografts is comparable to endogenous expression in the MDA-PCa-2b prostate cancer cell line and the human placental derived JEG-3 cell line.
DOI:http://dx.doi.org/10.7554/eLife.20183.009
Figure 5.. AMFR is required for 11β-HSD2…
Figure 5.. AMFR is required for 11β-HSD2 ubiquitination and the enzalutamide-induced metabolic phenotype that sustains local cortisol concentrations and enzalutamide-resistance.
(A) 11β-HSD2 and AMFR co-immunoprecipitate. Immunoprecipitation (IP) and immunoblot (IB) from endogenously expressed proteins in whole cell protein lysate from LAPC4 cells were performed with the indicated antibodies. The experiment was performed twice. (B) AMFR promotes 11β-HSD2 ubiquitination. Proteins were expressed in 293 cells, proteins tagged with ubiquitin-His, were pulled-down with Ni-agarose beads, and immunoblot was performed with the indicated antibodies. The experiment was performed twice. (C) Silencing AMFR expression with two independent shRNAs increases 11β-HSD2 protein. LAPC4 cells stably expressed shRNAs against AMFR (shAMFR) or non-silencing control expression vector. The experiment was performed three times. (D) Blockade of Enzalutamide (Enz)-mediated 11β-HSD2 loss by silencing AMFR reverses the metabolic phenotype that confers sustained cortisol concentrations. Cells were treated with the indicated concentration of Enz and subsequently were treated with [3H]-cortisol (100 nM) for the indicated times, followed by steroid extraction from media and cells, and steroid analysis by HPLC. Error bars represent the SD of biological triplicates. The experiment was performed three times. Enz treatment in panel D was for 38–42 days. (E) AMFR is required for tumor growth through enzalutamide therapy. Xenografts from LAPC4 cells expressing shAMFR or non-silencing control vector (shCtrl) were grown in surgically orchiectomized mice supplemented with DHEA and treated with Enz when tumors reached 100 mm3. The significance of the difference between shCtrl and shAMFR groups was calculated with an unpaired and two-tailed t-test on day 20. (F) Progression-free survival is increased in tumors lacking AMFR. The significance of the difference between shCtrl and shAMFR groups was determined with a log-rank test. (G) Xenograft tumors with genetic ablation of AMFR retain 11β-HSD2 protein expression. Xenograft tissues were collected at the end of the xenograft study and immunoblot was performed with the indicated antibodies. DOI:http://dx.doi.org/10.7554/eLife.20183.010
Figure 5—figure supplement 1.. AMFR and Erlin-2…
Figure 5—figure supplement 1.. AMFR and Erlin-2 regulation and cortisol metabolism with Enz treatment.
(A) Erlin-2 but not AMFR is consistently up-regulated with Enz treatment of LAPC4 cells. (B) Erlin-2 is up-regulated in 8 of 11 human prostate tissues. Immunoblots were performed as described previously. (C) Erlin-2 overexpression (OE) suppresses expression of 11β-HSD2 protein in LAPC4 cells and Erlin-2 knockdown by siRNA increases 11β-HSD2 expression and activity in the long-term Enz-treated LAPC4 cells. (D) AMFR silencing does not regulate HSD11B2 transcript. qPCR was performed in triplicate and expression is normalized to shControl-expressing cells and RPLP0. (E) Reversal of the metabolic phenotype that sustains cortisol with Enz treatment by AMFR knockdown is reversed again by 11β-HSD2 knockdown (compare cortisol at 24 hr in shAMFR groups between siCTRL and siHSD11B2). The specificity of siHSD11B2 is shown by qPCR and immunoblot. LAPC4 cells stably expressing stably shCTRL or an shAMFR construct were treated with Enz as described for Figure 5, transiently transfected with siHSD11B2 or siCTRL and treated with [3H]-cortisol (100 nM). Experiments were performed in duplicate. DOI:http://dx.doi.org/10.7554/eLife.20183.011

References

    1. Arora VK, Schenkein E, Murali R, Subudhi SK, Wongvipat J, Balbas MD, Shah N, Cai L, Efstathiou E, Logothetis C, Zheng D, Sawyers CL. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell. 2013;155:1309–1322. doi: 10.1016/j.cell.2013.11.012.
    1. Attard G, Parker C, Eeles RA, Schröder F, Tomlins SA, Tannock I, Drake CG, de Bono JS. Prostate cancer. The Lancet. 2016;387:70–82. doi: 10.1016/S0140-6736(14)61947-4.
    1. Attard G, Reid AH, Auchus RJ, Hughes BA, Cassidy AM, Thompson E, Oommen NB, Folkerd E, Dowsett M, Arlt W, de Bono JS. Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. The Journal of Clinical Endocrinology & Metabolism. 2012;97:507–516. doi: 10.1210/jc.2011-2189.
    1. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, Davis ID, de Bono JS, Evans CP, Fizazi K, Joshua AM, Kim CS, Kimura G, Mainwaring P, Mansbach H, Miller K, Noonberg SB, Perabo F, Phung D, Saad F, Scher HI, Taplin ME, Venner PM, Tombal B, PREVAIL Investigators Enzalutamide in metastatic prostate cancer before chemotherapy. New England Journal of Medicine. 2014;371:424–433. doi: 10.1056/NEJMoa1405095.
    1. Browman DT, Resek ME, Zajchowski LD, Robbins SM. Erlin-1 and erlin-2 are novel members of the prohibitin family of proteins that define lipid-raft-like domains of the ER. Journal of Cell Science. 2006;119:3149–3160. doi: 10.1242/jcs.03060.
    1. Chang K-H, Li R, Papari-Zareei M, Watumull L, Zhao YD, Auchus RJ, Sharifi N. Dihydrotestosterone synthesis bypasses testosterone to drive castration-resistant prostate cancer. PNAS. 2011;108:13728–13733. doi: 10.1073/pnas.1107898108.
    1. Chang KH, Li R, Kuri B, Lotan Y, Roehrborn CG, Liu J, Vessella R, Nelson PS, Kapur P, Guo X, Mirzaei H, Auchus RJ, Sharifi N. A gain-of-function mutation in DHT synthesis in castration-resistant prostate cancer. Cell. 2013;154:1074–1084. doi: 10.1016/j.cell.2013.07.029.
    1. Chapman K, Holmes M, Seckl J. 11β-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. Physiological Reviews. 2013;93:1139–1206. doi: 10.1152/physrev.00020.2012.
    1. Efstathiou E, Li W, Gormley M. Biological heterogeneity in localized high-risk prostate cancer (LHRPC) from a study of neoadjuvant abiraterone acetate plus leuprolide acetate (LHRHa) versus LHRHa. 2015 ASCO Annual Meeting.2015.
    1. Hearn JW, AbuAli G, Reichard CA, Reddy CA, Magi-Galluzzi C, Chang KH, Carlson R, Rangel L, Reagan K, Davis BJ, Karnes RJ, Kohli M, Tindall D, Klein EA, Sharifi N. HSD3B1 and resistance to androgen-deprivation therapy in prostate cancer: a retrospective, multicohort study. The Lancet Oncology. 2016;17:1435–1444. doi: 10.1016/S1470-2045(16)30227-3.
    1. Huber MD, Vesely PW, Datta K, Gerace L. Erlins restrict SREBP activation in the ER and regulate cellular cholesterol homeostasis. The Journal of Cell Biology. 2013;203:427–436. doi: 10.1083/jcb.201305076.
    1. Isikbay M, Otto K, Kregel S, Kach J, Cai Y, Vander Griend DJ, Conzen SD, Szmulewitz RZ. Glucocorticoid receptor activity contributes to resistance to androgen-targeted therapy in prostate cancer. Hormones and Cancer. 2014;5:72–89. doi: 10.1007/s12672-014-0173-2.
    1. Jeanneteau F, Garabedian MJ, Chao MV. Activation of Trk neurotrophin receptors by glucocorticoids provides a neuroprotective effect. PNAS. 2008;105:4862–4867. doi: 10.1073/pnas.0709102105.
    1. Kach J, Conzen SD, Szmulewitz RZ. Targeting the glucocorticoid receptor in breast and prostate cancers. Science Translational Medicine. 2015;7:305ps19. doi: 10.1126/scitranslmed.aac7531.
    1. Knudsen KE, Penning TM. Partners in crime: deregulation of AR activity and androgen synthesis in prostate cancer. Trends in Endocrinology & Metabolism. 2010;21:315–324. doi: 10.1016/j.tem.2010.01.002.
    1. Li J, Wang W, Liu C, Wang W, Li W, Shu Q, Chen ZJ, Sun K. Critical role of histone acetylation by p300 in human placental 11β-HSD2 expression. The Journal of Clinical Endocrinology & Metabolism. 2013;98:E1189–E1197. doi: 10.1210/jc.2012-4291.
    1. Li Z, Bishop AC, Alyamani M, Garcia JA, Dreicer R, Bunch D, Liu J, Upadhyay SK, Auchus RJ, Sharifi N. Conversion of abiraterone to D4A drives anti-tumour activity in prostate cancer. Nature. 2015;523:347–351. doi: 10.1038/nature14406.
    1. Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocrine Reviews. 2011;32:81–151. doi: 10.1210/er.2010-0013.
    1. Montgomery B, Cheng HH, Drechsler J, Mostaghel EA. Glucocorticoids and prostate cancer treatment: friend or foe? Asian Journal of Andrology. 2014;16:354–358. doi: 10.4103/1008-682X.125392.
    1. Mostaghel EA, Plymate SR, Montgomery B. Molecular pathways: targeting resistance in the androgen receptor for therapeutic benefit. Clinical Cancer Research. 2014;20:791–798. doi: 10.1158/1078-0432.CCR-12-3601.
    1. Ni M, Chen Y, Lim E, Wimberly H, Bailey ST, Imai Y, Rimm DL, Liu XS, 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. Pearce MM, Wang Y, Kelley GG, Wojcikiewicz RJ. SPFH2 mediates the endoplasmic reticulum-associated degradation of inositol 1,4,5-trisphosphate receptors and other substrates in mammalian cells. Journal of Biological Chemistry. 2007;282:20104–20115. doi: 10.1074/jbc.M701862200.
    1. Sartor O, Parker CC, de Bono J. Reappraisal of glucocorticoids in castrate-resistant prostate cancer. Asian Journal of Andrology. 2014;16:666. doi: 10.4103/1008-682X.133314.
    1. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, Armstrong AJ, Flaig TW, Fléchon A, Mainwaring P, Fleming M, Hainsworth JD, Hirmand M, Selby B, Seely L, de Bono JS, AFFIRM Investigators Increased survival with enzalutamide in prostate cancer after chemotherapy. The New England Journal of Medicine. 2012;367:1187–1197. doi: 10.1056/NEJMoa1207506.
    1. Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. Journal of Clinical Oncology. 2005;23:8253–8261. doi: 10.1200/JCO.2005.03.4777.
    1. Sharifi N. Minireview: Androgen metabolism in castration-resistant prostate cancer. Molecular Endocrinology. 2013;27:708–714. doi: 10.1210/me.2013-1007.
    1. Sharifi N. Steroid receptors aplenty in prostate cancer. The New England Journal of Medicine. 2014;370:970–971. doi: 10.1056/NEJMcibr1315706.
    1. Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, Wasielewska T, Welsbie D, Chen CD, Higano CS, Beer TM, Hung DT, Scher HI, Jung ME, Sawyers CL. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 2009;324:787–790. doi: 10.1126/science.1168175.

Source: PubMed

3
Sottoscrivi