Synthetic lethality between androgen receptor signalling and the PARP pathway in prostate cancer

Mohammad Asim, Firas Tarish, Heather I Zecchini, Kumar Sanjiv, Eleni Gelali, Charles E Massie, Ajoeb Baridi, Anne Y Warren, Wanfeng Zhao, Christoph Ogris, Leigh-Anne McDuffus, Patrice Mascalchi, Greg Shaw, Harveer Dev, Karan Wadhwa, Paul Wijnhoven, Josep V Forment, Scott R Lyons, Andy G Lynch, Cormac O'Neill, Vincent R Zecchini, Paul S Rennie, Aria Baniahmad, Simon Tavaré, Ian G Mills, Yaron Galanty, Nicola Crosetto, Niklas Schultz, David Neal, Thomas Helleday, Mohammad Asim, Firas Tarish, Heather I Zecchini, Kumar Sanjiv, Eleni Gelali, Charles E Massie, Ajoeb Baridi, Anne Y Warren, Wanfeng Zhao, Christoph Ogris, Leigh-Anne McDuffus, Patrice Mascalchi, Greg Shaw, Harveer Dev, Karan Wadhwa, Paul Wijnhoven, Josep V Forment, Scott R Lyons, Andy G Lynch, Cormac O'Neill, Vincent R Zecchini, Paul S Rennie, Aria Baniahmad, Simon Tavaré, Ian G Mills, Yaron Galanty, Nicola Crosetto, Niklas Schultz, David Neal, Thomas Helleday

Abstract

Emerging data demonstrate homologous recombination (HR) defects in castration-resistant prostate cancers, rendering these tumours sensitive to PARP inhibition. Here we demonstrate a direct requirement for the androgen receptor (AR) to maintain HR gene expression and HR activity in prostate cancer. We show that PARP-mediated repair pathways are upregulated in prostate cancer following androgen-deprivation therapy (ADT). Furthermore, upregulation of PARP activity is essential for the survival of prostate cancer cells and we demonstrate a synthetic lethality between ADT and PARP inhibition in vivo. Our data suggest that ADT can functionally impair HR prior to the development of castration resistance and that, this potentially could be exploited therapeutically using PARP inhibitors in combination with androgen-deprivation therapy upfront in advanced or high-risk prostate cancer.Tumours with homologous recombination (HR) defects become sensitive to PARPi. Here, the authors show that androgen receptor (AR) regulates HR and AR inhibition activates the PARP pathway in vivo, thus inhibition of both AR and PARP is required for effective treatment of high risk prostate cancer.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
AR signalling regulates homologous recombination (HR) and the DNA damage response. a Heat map showing expression of homologous recombination regulators following AR RNAi knockdown, AR transcript and known AR targets/controls are shown mined from the microarray data from LNCaP cells. b Time course of formation of RAD51 foci showing high content cytometry-based quantification of RAD51 nuclear foci number per nucleus in ‘low AR; and ‘high AR’ C4-2 cells in response to 10 Gy radiation. Statistical significance was determined using the Holm–Sidak method, with alpha = 5.000% (n = 3). Scale bar = 1 μm. c Quantification of gene conversion assay from analysis of GFP-positive C4-2-DRGFP cells transfected with non-targeting control siRNA (siNT) or siAR along with ISce1 endonuclease for 72 h followed by detection of GFP-positive cells by flow cytometry. P-value by two-sided Student’s t-test (n = 3); the data represent mean ± SEM. d Foci analysis time course showing high content cytometry-based quantification of the number of γH2AX foci in ‘low AR’ and ‘high AR’ C4-2 cell nuclei in response to radiation (10 Gy). Statistical significance determined using the Holm–Sidak method, with alpha = 5.000%. (n = 3). Scale bar = 1 μm. e High content cytometry analysis of MRN foci in ‘high AR’ and ‘low AR’ C4-2 cells treated with hydroxyurea (HU). Upper panel shows ‘high AR’ and ‘low AR’ nuclei with MRN foci (red dots), lower panel shows a histogram with means of the errors; P-value by two-sided Student’s t-test (n = 2). f Micrographs from biopsies showing examples of Ki67- and RAD51 positive cells and Ki67 positive alone. Scale bar = 5 μm. g Graph showing percentage of RAD51-positive cells in the population of Ki67-positive cells in biopsies after indicated treatment. Error bars shows SEM and ** = P < 0.01, Mann–Whitney test
Fig. 2
Fig. 2
Androgen deprivation therapy triggers PARP activation. a Western blot showing C4-2 cells treated for 72 h with indicated agents (10 µM). b Western blot showing C4-2 cells transfected with indicated siRNA (20 nM). c Immunofluorescence microscopy images showing relative intensities of PAR Poly(ADP Ribose) and PARP1 (Poly ADP-ribose polymerase 1) in PCa tissue pre- (pre-castration or AR+) and post leuprolide (post-castration or AR-) treatment (8 weeks). Scale bar = 100 μm. d Histogram shows PARP activity in PCa patients pre- and post-leuprolide treatment (One sample t-test, two-tailed, test value 100)
Fig. 3
Fig. 3
Synthetic lethality between AR and PARP pathways in PCa. a, b Viable fraction assessed by MTS assay, of C4-2 (a) and (b) LN3 cells treated with the indicated doses of Olaparib and/or enzalutamide or bicalutamide (10 μM) for 7 days or until 95% confluence was reached; P-value by two-sided Student’s t-test; bars show mean ± SEM. c Live cell imaging confluence analysis (Incucyte) of C4-2 cells treated with Olaparib (1 μM), enzalutamide (10 μM), bicalutamide (10 μM) or combined treatment, statistical significance calculated by two-way ANOVA. d Live cell imaging confluence analysis (Incucyte) of ‘high AR’ and ‘low AR’ C4-2 cells treated with doxycycline and/or Olaparib (1 μM), statistical significance calculated using two-way ANOVA. e, f Clonogenic survival assay for C4-2 cells with either inducible shNT (e) or shAR (f); cells were treated with doxycycline and 1 μM Olaparib, P-value by two-sided Student’s t-test; bars show mean ± SEM. All experiments were independently performed in triplicates. The data represent means ± SEM. P-values from significant two-sided Student’s t-tests are given (* = P < 0.05)
Fig. 4
Fig. 4
Dual inhibition of AR and PARP1/2 function represses PCa growth. a Tumour xenografts of C4-2 cells. NSG mice were treated with DMSO (vehicle) or bicalutamide and/or Olaparib for 6 weeks, statistical significance calculated using two-way ANOVA. b Tumour xenografts of PC3-ctl or PC3-AR cells, NSG mice were administered with DMSO (vehicle) or Olaparib as indicated, P-value by two-sided Student’s t-test. c, d Quantification of ki67 expression in ex vivo culture of human PCa treated with (c) bicalutamide (10 μM) or (d) Enzalutamide (10 μM) and/or Olaparib (2 μM) for 72 h, significance calculated by Mann–Whitney U test

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA. Cancer. J. Clin. 2015;65:5–29. doi: 10.3322/caac.21254.
    1. Chen CD, et al. Molecular determinants of resistance to antiandrogen therapy. Nat. Med. 2004;10:33–39. doi: 10.1038/nm972.
    1. Attard G, et al. Selective inhibition of CYP17 with abiraterone acetate is highly active in the treatment of castration-resistant prostate cancer. J. Clin. Oncol. 2009;27:3742–3748. doi: 10.1200/JCO.2008.20.0642.
    1. Asim, M., et al. Choline kinase alpha as an androgen receptor chaperone and prostate cancer therapeutic target. J. Natl. Cancer. Inst. 108, djv371 (2016).
    1. Massie CE, et al. The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO. J. 2011;30:2719–2733. doi: 10.1038/emboj.2011.158.
    1. Sharma A, et al. The retinoblastoma tumor suppressor controls androgen signaling and human prostate cancer progression. J. Clin. Invest. 2010;120:4478–4492. doi: 10.1172/JCI44239.
    1. Goodwin JF, et al. A hormone-DNA repair circuit governs the response to genotoxic insult. Cancer Discov. 2013;3:1254–1271. doi: 10.1158/-13-0108.
    1. Polkinghorn WR, et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov. 2013;3:1245–1253. doi: 10.1158/-13-0172.
    1. Al-Ubaidi FL, et al. Castration therapy results in decreased Ku70 levels in prostate cancer. Clin. Cancer. Res. 2013;19:1547–1556. doi: 10.1158/1078-0432.CCR-12-2795.
    1. Tarish FL, et al. Castration radiosensitizes prostate cancer tissue by impairing DNA double-strand break repair. Sci. Transl. Med. 2015;7:312re311. doi: 10.1126/scitranslmed.aac5671.
    1. Robinson D, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–1228. doi: 10.1016/j.cell.2015.05.001.
    1. Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol. Oncol. 2011;5:387–393. doi: 10.1016/j.molonc.2011.07.001.
    1. Bryant HE, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–917. doi: 10.1038/nature03443.
    1. Farmer H, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917–921. doi: 10.1038/nature03445.
    1. VanderWeele DJ, Paner GP, Fleming GF, Szmulewitz RZ. Sustained Complete response to cytotoxic therapy and the PARP inhibitor veliparib in metastatic castration-resistant prostate cancer - a case report. Front. Oncol. 2015;5:169. doi: 10.3389/fonc.2015.00169.
    1. Mateo J, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 2015;373:1697–1708. doi: 10.1056/NEJMoa1506859.
    1. D’Amico AV, Chen MH, Renshaw AA, Loffredo M, Kantoff PW. Androgen suppression and radiation vs radiation alone for prostate cancer: a randomized trial. JAMA. 2008;299:289–295.
    1. Mason MD, et al. Final Report of the Intergroup Randomized study of combined androgen-deprivation therapy plus radiotherapy versus androgen-deprivation therapy alone in locally advanced prostate cancer. J. Clin. Oncol. 2015;33:2143–2150. doi: 10.1200/JCO.2014.57.7510.
    1. Cheng H, Snoek R, Ghaidi F, Cox ME, Rennie PS. Short hairpin RNA knockdown of the androgen receptor attenuates ligand-independent activation and delays tumor progression. Cancer Res. 2006;66:10613–10620. doi: 10.1158/0008-5472.CAN-06-0028.
    1. Gottipati P, et al. Poly(ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res. 2010;70:5389–5398. doi: 10.1158/0008-5472.CAN-09-4716.
    1. Spratt DE, et al. Androgen receptor upregulation mediates radioresistance after ionizing radiation. Cancer. Res. 2015;75:4688–4696. doi: 10.1158/0008-5472.CAN-15-0892.
    1. Bryant HE, et al. PARP is activated at stalled forks to mediate Mre11-dependent replication restart and recombination. EMBO J. 2009;28:2601–2615. doi: 10.1038/emboj.2009.206.
    1. Strom CE, et al. Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP inhibition traps a single-strand intermediate. Nucleic. Acids. Res. 2011;39:3166–3175. doi: 10.1093/nar/gkq1241.
    1. Murai J, et al. Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. Cancer Res. 2012;72:5588–5599. doi: 10.1158/0008-5472.CAN-12-2753.
    1. Rosen EM, Fan S, Isaacs C. BRCA1 in hormonal carcinogenesis: basic and clinical research. Endocr. Relat. Cancer. 2005;12:533–548. doi: 10.1677/erc.1.00972.
    1. Pettaway CA, et al. Selection of highly metastatic variants of different human prostatic carcinomas using orthotopic implantation in nude mice. Clin. Cancer. Res. 1996;2:1627–1636.
    1. Ruifrok AC, Johnston DA. Quantification of histochemical staining by color deconvolution. Anal. Quant. Cytol. Histol. 2001;23:291–299.
    1. Bostwick DG. Grading prostate cancer. Am. J. Clin. Pathol. 1994;102:S38–S56.
    1. Semrau S, et al. FuseFISH: robust detection of transcribed gene fusions in single cells. Cell Rep. 2014;6:18–23. doi: 10.1016/j.celrep.2013.12.002.
    1. Annaratone L, et al. Quantification of HER2 and estrogen receptor heterogeneity in breast cancer by single-molecule RNA fluorescence in situ hybridization. Oncotarget. 2017;8:18680–18698.

Source: PubMed

3
Se inscrever