51
|
Yu J, Zhang L, Yan G, Zhou P, Cao C, Zhou F, Li X, Chen Y. Discovery and biological evaluation of novel androgen receptor antagonist for castration-resistant prostate cancer. Eur J Med Chem 2019; 171:265-281. [PMID: 30925341 DOI: 10.1016/j.ejmech.2019.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/14/2019] [Accepted: 03/16/2019] [Indexed: 02/05/2023]
Abstract
Prostate cancer (PC) is the second most common malignancy in men worldwide. Among current therapies, two antiandrogens, Abiraterone Acetate and Enzalutamide (Enza) have become the standard of care for patients with metastatic castration-resistant prostate cancer (mCRPC). Here, we designed and synthesized a new series of nonsteroidal compounds deriving from the hybridization of Abiraterone (Abi) and Enzalutamide, among which compound 4a featuring the diphenylamine scaffold was identified as a potent and cell selective androgen receptor (AR) antagonist. In cell proliferation assays, compound 4a exhibited better antiproliferative activities than Enzalutamide against AR-overexpressing VCaP cells and 22Rv1 cells bearing H874Y-mutated AR. In addition, 4a suppressed the activity of AR-F876L mutant that confers resistance to Enzalutamide and efficiently blocked R1881-induced PSA and FKBP5 gene expression. In competitive binding assay, compound 4a displayed higher binding affinity to AR than Enzalutamide. These results suggest compound 4a as a potential candidate to treat Enza-resistant CRPC.
Collapse
Affiliation(s)
- Jiang Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lanxi Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Guoyi Yan
- Department of Hepatobiliary Pancreatic Surgery, Henan Province People's Hospital, Zhengzhou, 450003, China
| | - Peiting Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Chaoguo Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Fei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xinghai Li
- Hinova Pharmaceuticals Inc, 4th Floor, Building RongYao A, No. 5, Keyuan South Road, Chengdu, 610041, China
| | - Yuanwei Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China; Hinova Pharmaceuticals Inc, 4th Floor, Building RongYao A, No. 5, Keyuan South Road, Chengdu, 610041, China.
| |
Collapse
|
52
|
Yan G, Ru Y, Yan F, Xiong X, Hu W, Pan T, Sun J, Zhang C, Wang Q, Li X. MIIP inhibits the growth of prostate cancer via interaction with PP1α and negative modulation of AKT signaling. Cell Commun Signal 2019; 17:44. [PMID: 31092266 PMCID: PMC6521544 DOI: 10.1186/s12964-019-0355-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/17/2019] [Indexed: 12/09/2022] Open
Abstract
Background Over-activation of phosphatidylinositol 3-kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) signaling pathway is one of important mechanisms to promote castration resistant prostate cancer, the final stage of prostate cancer (PCa). Dysregulation of PP1-meditaed AKT dephosphorylation might contribute to such an event but is not fully understood. As a newly identified tumor suppressor, MIIP exerts its role in various types of cancer but has not been investigated in PCa. Results We first demonstrated that overexpression of migration and invasion inhibitory protein (MIIP) in human PCa cell lines suppresses their growth while knockdown of MIIP does the opposite in vitro. Although MIIP has no effect on the expression of AR and its target genes or the nuclear translocation of AR in AR-positive PCa cells, MIIP overexpression significantly inhibits activation of AKT-mTOR pathway in both AR- positive and negative PCa cells whereas knockdown of MIIP enhances AKT-mTOR signaling. Using Western blot, immunofluorescence co-localization and co-immunoprecipitation analysis, we found that MIIP interacts with PP1α via its C-terminal part but does not affect its protein level. Importantly, silence of PP1α reversed the inhibitory effect of MIIP on AKT phosphorylation and cell growth in PCa cell lines, while MIIP∆C, which is incapable of interacting with PP1α, loses MIIP’s effect, suggesting that MIIP exerts its roles via interaction with PP1α. Further, MIIP overexpression inhibits the growth of both AR- positive and negative PCa xenograft in nude mice. Finally, immunohistochemical staining of PCa tissue microarray showed that MIIP expression level is downregulated in PCa and negatively correlated with Gleason score of PCa. Conclusion We discovered that MIIP is a novel suppressor of oncogenic AKT-mTOR signaling in PCa by facilitating PP1-meditaed AKT dephosphorylation. Our study further emphasized the tumor suppressive role of MIIP and illustrated a novel mechanism. Electronic supplementary material The online version of this article (10.1186/s12964-019-0355-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guang Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,Andrology Department, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Yi Ru
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Fengqi Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,Department of Urology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Xin Xiong
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Wei Hu
- Department of Urology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Tao Pan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jianming Sun
- Andrology Department, Shanghai Seventh People's Hospital, Shanghai, 200137, China
| | - Chi Zhang
- Rehabilitation Department, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200137, China
| | - Qinhao Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xia Li
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
53
|
Braadland PR, Urbanucci A. Chromatin reprogramming as an adaptation mechanism in advanced prostate cancer. Endocr Relat Cancer 2019; 26:R211-R235. [PMID: 30844748 DOI: 10.1530/erc-18-0579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Tumor evolution is based on the ability to constantly mutate and activate different pathways under the selective pressure of targeted therapies. Epigenetic alterations including those of the chromatin structure are associated with tumor initiation, progression and drug resistance. Many cancers, including prostate cancer, present enlarged nuclei, and chromatin appears altered and irregular. These phenotypic changes are likely to result from epigenetic dysregulation. High-throughput sequencing applied to bulk samples and now to single cells has made it possible to study these processes in unprecedented detail. It is therefore timely to review the impact of chromatin relaxation and increased DNA accessibility on prostate cancer growth and drug resistance, and their effects on gene expression. In particular, we focus on the contribution of chromatin-associated proteins such as the bromodomain-containing proteins to chromatin relaxation. We discuss the consequence of this for androgen receptor transcriptional activity and briefly summarize wider gain-of-function effects on other oncogenic transcription factors and implications for more effective prostate cancer treatment.
Collapse
Affiliation(s)
- Peder Rustøen Braadland
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alfonso Urbanucci
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, Oslo, Norway
| |
Collapse
|
54
|
Sternberg CN. Enzalutamide, an oral androgen receptor inhibitor for treatment of castration-resistant prostate cancer. Future Oncol 2019; 15:1437-1457. [PMID: 30848157 DOI: 10.2217/fon-2018-0940] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Androgen receptor (AR) signaling is a key pathway in prostate cancer, and patients are initially treated with androgen deprivation therapy. Patients who have stopped responding to androgen deprivation therapy are considered to have castration-resistant prostate cancer (CRPC), which is still dependent on AR signaling. Enzalutamide, an orally available AR inhibitor, was initially approved by the US FDA for the treatment of patients with metastatic CRPC who have previously received docetaxel. The indication was subsequently extended to include all patients with metastatic CRPC, and most recently to include patients with nonmetastatic CRPC. This review summarizes the body of evidence supporting enzalutamide efficacy and safety in CRPC.
Collapse
Affiliation(s)
- Cora N Sternberg
- Division of Medical Oncology, Weill Cornell Medical Center, New York, NY 10021, USA
| |
Collapse
|
55
|
Molecular Mechanisms Related to Hormone Inhibition Resistance in Prostate Cancer. Cells 2019; 8:cells8010043. [PMID: 30642011 PMCID: PMC6356740 DOI: 10.3390/cells8010043] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Management of metastatic or advanced prostate cancer has acquired several therapeutic approaches that have drastically changed the course of the disease. In particular due to the high sensitivity of prostate cancer cells to hormone depletion, several agents able to inhibit hormone production or binding to nuclear receptor have been evaluated and adopted in clinical practice. However, despite several hormonal treatments being available nowadays for the management of advanced or metastatic prostate cancer, the natural history of the disease leads inexorably to the development of resistance to hormone inhibition. Findings regarding the mechanisms that drive this process are of particular and increasing interest as these are potentially related to the identification of new targetable pathways and to the development of new drugs able to improve our patients' clinical outcomes.
Collapse
|
56
|
Dong L, Zieren RC, Xue W, de Reijke TM, Pienta KJ. Metastatic prostate cancer remains incurable, why? Asian J Urol 2019; 6:26-41. [PMID: 30775246 PMCID: PMC6363601 DOI: 10.1016/j.ajur.2018.11.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/18/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
Metastatic prostate cancer patients present in two ways-with already disseminated disease at the time of presentation or with disease recurrence after definitive local therapy. Androgen deprivation therapy is given as the most effective initial treatment to patients. However, after the initial response, almost all patients will eventually progress despite the low levels of testosterone. Disease at this stage is termed castration resistant prostate cancer (CRPC). Before 2010, the taxane docetaxel was the first and only life prolonging agent for metastatic CRPC (mCRPC). The last decade has witnessed robust progress in CRPC therapeutics development. Abiraterone, enzalutamide, apalutamide and sipuleucel-T have been evaluated as first- and second-line agents in mCRPC patients, while cabazitaxel was approved as a second-line treatment. Radium-223 dichloride was approved in symptomatic patients with bone metastases and no known visceral metastases pre- and post-docetaxel. However, despite significant advances, mCRPC remains a lethal disease. Both primary and acquired resistance have been observed in CRPC patients treated by these new agents. It could be solely cell intrinsic or it is possible that the clonal heterogeneity in treated tumors may result from the adaptive responses to the selective pressures within the tumor microenvironment. The aim of this review is to list current treatment agents of CRPC and summarize recent findings in therapeutic resistance mechanisms.
Collapse
Affiliation(s)
- Liang Dong
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard C. Zieren
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Theo M. de Reijke
- Department of Urology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kenneth J. Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
57
|
Fujita K, Nonomura N. Role of Androgen Receptor in Prostate Cancer: A Review. World J Mens Health 2018; 37:288-295. [PMID: 30209899 PMCID: PMC6704300 DOI: 10.5534/wjmh.180040] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022] Open
Abstract
Androgen receptor (AR) is a steroid receptor transcriptional factor for testosterone and dihydrotestosterone consisting of four main domains, the N-terminal domain, DNA-binding domain, hinge region, and ligand-binding domain. AR plays pivotal roles in prostate cancer, especially castration-resistant prostate cancer (CRPC). Androgen deprivation therapy can suppress hormone-naïve prostate cancer, but prostate cancer changes AR and adapts to survive under castration levels of androgen. These mechanisms include AR point mutations, AR overexpression, changes of androgen biosynthesis, constitutively active AR splice variants without ligand binding, and changes of androgen cofactors. Studies of AR in CRPC revealed that AR was still active in CRPC, and it remains as a potential target to treat CRPC. Enzalutamide is a second-generation antiandrogen effective in patients with CRPC before and after taxane-based chemotherapy. However, CRPC is still incurable and can develop drug resistance. Understanding the mechanisms of this resistance can enable new-generation therapies for CRPC. Several promising new AR-targeted therapies have been developed. Apalutamide is a new Food and Drug Administration-approved androgen agonist binding to the ligand-binding domain, and clinical trials of other new AR-targeted agents binding to the ligand-binding domain or N-terminal domain are underway. This review focuses on the functions of AR in prostate cancer and the development of CRPC and promising new agents against CRPC.
Collapse
Affiliation(s)
- Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
58
|
Bousset L, Rambur A, Fouache A, Bunay J, Morel L, Lobaccaro JMA, Baron S, Trousson A, de Joussineau C. New Insights in Prostate Cancer Development and Tumor Therapy: Modulation of Nuclear Receptors and the Specific Role of Liver X Receptors. Int J Mol Sci 2018; 19:E2545. [PMID: 30154328 PMCID: PMC6164771 DOI: 10.3390/ijms19092545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) incidence has been dramatically increasing these last years in westernized countries. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy is preferred in locally advanced disease in combination with chemotherapy. Unfortunately, PCa goes into a castration-resistant state in the vast majority of the cases, leading to questions about the molecular mechanisms involving the steroids and their respective nuclear receptors in this relapse. Interestingly, liver X receptors (LXRα/NR1H3 and LXRβ/NR1H2) have emerged as new actors in prostate physiology, beyond their historical roles of cholesterol sensors. More importantly LXRs have been proposed to be good pharmacological targets in PCa. This rational has been based on numerous experiments performed in PCa cell lines and genetic animal models pointing out that using selective liver X receptor modulators (SLiMs) could actually be a good complementary therapy in patients with a castration resistant PCa. Hence, this review is focused on the interaction among the androgen receptors (AR/NR3C4), estrogen receptors (ERα/NR3A1 and ERβ/NR3A2), and LXRs in prostate homeostasis and their putative pharmacological modulations in parallel to the patients' support.
Collapse
Affiliation(s)
- Laura Bousset
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amandine Rambur
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Allan Fouache
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Julio Bunay
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Laurent Morel
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Jean-Marc A Lobaccaro
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Silvère Baron
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amalia Trousson
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Cyrille de Joussineau
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| |
Collapse
|
59
|
Abstract
Resistance to steroid hormones presents a serious problem with respect to their mass use in therapy. It may be caused genetically by mutation of genes involved in hormonal signaling, not only steroid receptors, but also other players in the signaling cascade as co-regulators and other nuclear factors, mediating the hormone-born signal. Another possibility is acquired resistance which may develop under long-term steroid treatment, of which a particular case is down regulation of the receptors. In the review recent knowledge is summarized on the mechanism of main steroid hormone action, pointing to already proven or potential sites causing steroid resistance. We have attempted to address following questions: 1) What does stay behind differences among patients as to their response to the (anti)steroid treatment? 2) Why do various tissues/cells respond differently to the same steroid hormone though they contain the same receptors? 3) Are such differences genetically dependent? The main attention was devoted to glucocorticoids as the most frequently used steroid therapeutics. Further, androgen insensitivity is discussed with a particular attention to acquired resistance to androgen deprivation therapy of prostate cancer. Finally the potential causes are outlined of breast and related cancer(s) resistance to antiestrogen therapy.
Collapse
Affiliation(s)
- R Hampl
- Institute of Endocrinology, Prague, Czech Republic.
| | | |
Collapse
|
60
|
Pérez-Ibave DC, Burciaga-Flores CH, Elizondo-Riojas MÁ. Prostate-specific antigen (PSA) as a possible biomarker in non-prostatic cancer: A review. Cancer Epidemiol 2018; 54:48-55. [DOI: 10.1016/j.canep.2018.03.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 12/26/2022]
|
61
|
Chen L, Cao H, Yu C, Feng Y. Lobaplatin inhibits prostate cancer progression in part by impairing AR and ERG signal. Fundam Clin Pharmacol 2018; 32:548-557. [PMID: 29733466 DOI: 10.1111/fcp.12377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/15/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Lei Chen
- Surgical Department I (Urology Department); LONGHUA Hospital Shanghai University of Traditional Chinese Medicine; No. 725 Wanping Road South Xuhui District, Shanghai City 200032 China
| | - Hongwen Cao
- Surgical Department I (Urology Department); LONGHUA Hospital Shanghai University of Traditional Chinese Medicine; No. 725 Wanping Road South Xuhui District, Shanghai City 200032 China
| | - Chao Yu
- Surgical Department I (Urology Department); LONGHUA Hospital Shanghai University of Traditional Chinese Medicine; No. 725 Wanping Road South Xuhui District, Shanghai City 200032 China
| | - Yigeng Feng
- Surgical Department I (Urology Department); LONGHUA Hospital Shanghai University of Traditional Chinese Medicine; No. 725 Wanping Road South Xuhui District, Shanghai City 200032 China
| |
Collapse
|
62
|
Del Re M, Crucitta S, Restante G, Rofi E, Arrigoni E, Biasco E, Sbrana A, Coppi E, Galli L, Bracarda S, Santini D, Danesi R. Pharmacogenetics of androgen signaling in prostate cancer: Focus on castration resistance and predictive biomarkers of response to treatment. Crit Rev Oncol Hematol 2018; 125:51-59. [DOI: 10.1016/j.critrevonc.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/24/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
|
63
|
Urbanucci A, Barfeld SJ, Kytölä V, Itkonen HM, Coleman IM, Vodák D, Sjöblom L, Sheng X, Tolonen T, Minner S, Burdelski C, Kivinummi KK, Kohvakka A, Kregel S, Takhar M, Alshalalfa M, Davicioni E, Erho N, Lloyd P, Karnes RJ, Ross AE, Schaeffer EM, Vander Griend DJ, Knapp S, Corey E, Feng FY, Nelson PS, Saatcioglu F, Knudsen KE, Tammela TLJ, Sauter G, Schlomm T, Nykter M, Visakorpi T, Mills IG. Androgen Receptor Deregulation Drives Bromodomain-Mediated Chromatin Alterations in Prostate Cancer. Cell Rep 2018; 19:2045-2059. [PMID: 28591577 DOI: 10.1016/j.celrep.2017.05.049] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 04/01/2017] [Accepted: 05/12/2017] [Indexed: 12/17/2022] Open
Abstract
Global changes in chromatin accessibility may drive cancer progression by reprogramming transcription factor (TF) binding. In addition, histone acetylation readers such as bromodomain-containing protein 4 (BRD4) have been shown to associate with these TFs and contribute to aggressive cancers including prostate cancer (PC). Here, we show that chromatin accessibility defines castration-resistant prostate cancer (CRPC). We show that the deregulation of androgen receptor (AR) expression is a driver of chromatin relaxation and that AR/androgen-regulated bromodomain-containing proteins (BRDs) mediate this effect. We also report that BRDs are overexpressed in CRPCs and that ATAD2 and BRD2 have prognostic value. Finally, we developed gene stratification signature (BROMO-10) for bromodomain response and PC prognostication, to inform current and future trials with drugs targeting these processes. Our findings provide a compelling rational for combination therapy targeting bromodomains in selected patients in which BRD-mediated TF binding is enhanced or modified as cancer progresses.
Collapse
Affiliation(s)
- Alfonso Urbanucci
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway.
| | - Stefan J Barfeld
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway
| | - Ville Kytölä
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Harri M Itkonen
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel Vodák
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0424 Oslo, Norway
| | - Liisa Sjöblom
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Xia Sheng
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway
| | - Teemu Tolonen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Sarah Minner
- University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Burdelski
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kati K Kivinummi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Annika Kohvakka
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Steven Kregel
- Department of Surgery - Section of Urology, University of Chicago, Chicago, IL 60637, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109-0940, USA
| | - Mandeep Takhar
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Mohammed Alshalalfa
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Elai Davicioni
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Nicholas Erho
- Research and Development, GenomeDx Biosciences, Vancouver, BC V6B 1B8, Canada
| | - Paul Lloyd
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143-0410, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143-0981, USA
| | | | - Ashley E Ross
- Brady Urological Institute, Johns Hopkins Medical Institute, Baltimore, MD 21287, USA
| | - Edward M Schaeffer
- Department of Urology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Tarry 16-703, Chicago, IL 60611-3008, USA
| | - Donald J Vander Griend
- Department of Surgery - Section of Urology, University of Chicago, Chicago, IL 60637, USA
| | - Stefan Knapp
- Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, UK; Institute for Pharmaceutical Chemistry, Goethe-University Frankfurt, Campus Riedberg, Max-von Laue Strasse 9, 60438 Frankfurt am Main, Germany
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Felix Y Feng
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143-0410, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA 94143-0981, USA; Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Urology, University of Washington, Seattle, WA 98195, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, 0316 Oslo, Norway; Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424 Oslo, Norway
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Teuvo L J Tammela
- Prostate Cancer Research Center and Department of Urology, University of Tampere and Tampere University Hospital, 33014 Tampere, Finland
| | - Guido Sauter
- University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg 20095, Germany
| | - Matti Nykter
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University of Technology, 33520 Tampere, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Fimlab Laboratories, Tampere University Hospital, 33520 Tampere, Finland
| | - Ian G Mills
- Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forskningsparken, University of Oslo, 21 0349 Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway; PCUK Movember Centre of Excellence, CCRCB, Queen's University, Belfast BT7 1NN, Northern Ireland, UK.
| |
Collapse
|
64
|
Maximov PY, Abderrahman B, Curpan RF, Hawsawi YM, Fan P, Jordan VC. A unifying biology of sex steroid-induced apoptosis in prostate and breast cancers. Endocr Relat Cancer 2018; 25:R83-R113. [PMID: 29162647 PMCID: PMC5771961 DOI: 10.1530/erc-17-0416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 11/21/2017] [Indexed: 12/13/2022]
Abstract
Prostate and breast cancer are the two cancers with the highest incidence in men and women, respectively. Here, we focus on the known biology of acquired resistance to antihormone therapy of prostate and breast cancer and compare laboratory and clinical similarities in the evolution of the disease. Laboratory studies and clinical observations in prostate and breast cancer demonstrate that cell selection pathways occur during acquired resistance to antihormonal therapy. Following sex steroid deprivation, both prostate and breast cancer models show an initial increased acquired sensitivity to the growth potential of sex steroids. Subsequently, prostate and breast cancer cells either become dependent upon the antihormone treatment or grow spontaneously in the absence of hormones. Paradoxically, the physiologic sex steroids now kill a proportion of selected, but vulnerable, resistant tumor cells. The sex steroid receptor complex triggers apoptosis. We draw parallels between acquired resistance in prostate and breast cancer to sex steroid deprivation. Clinical observations and patient trials confirm the veracity of the laboratory studies. We consider therapeutic strategies to increase response rates in clinical trials of metastatic disease that can subsequently be applied as a preemptive salvage adjuvant therapy. The goal of future advances is to enhance response rates and deploy a safe strategy earlier in the treatment plan to save lives. The introduction of a simple evidence-based enhanced adjuvant therapy as a global healthcare strategy has the potential to control recurrence, reduce hospitalization, reduce healthcare costs and maintain a healthier population that contributes to society.
Collapse
Affiliation(s)
- Philipp Y Maximov
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | - Balkees Abderrahman
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | | | - Yousef M Hawsawi
- Department of GeneticsKing Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Ping Fan
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| | - V Craig Jordan
- Department of Breast Medical OncologyMD Anderson Cancer Centre, Houston, Texas, USA
| |
Collapse
|
65
|
|
66
|
Intasqui P, Bertolla RP, Sadi MV. Prostate cancer proteomics: clinically useful protein biomarkers and future perspectives. Expert Rev Proteomics 2017; 15:65-79. [PMID: 29251021 DOI: 10.1080/14789450.2018.1417846] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Although prostate cancer constitutes one of the most important, death-related diseases in the male population, there is still a need for identification of sensitive biomarkers that could precociously detect the disease and differentiate aggressive from indolent cancers, in order to decrease overtreatment. Proteomics research has improved understanding on mechanisms underlying tumorigenesis, cancer cells migration and invasion potential, and castration resistance. This review has focused on proteomic studies of prostate cancer published in the recent years, with a special emphasis on determination of biomarkers for cancer progression and diagnosis. Areas covered: Shotgun and targeted-proteomic studies of prostate cancer in different matrices are reviewed, i.e., prostate tissue, prostate cell lines, blood (serum and plasma), urine, seminal plasma, and exosomes. The most important biomarkers for cancer diagnosis and aggressiveness characterization are highlighted. Expert commentary: In general, results demonstrate alteration in cell cycle control, DNA repair, proteasomal degradation, and metabolic activity. However, these studies suffer from low reproducibility due to heterogeneity of the cancer itself, as well as to techniques utilized for protein identification/quantification. Downstream confirmatory studies in separate cohorts are warranted in order to demonstrate accuracy of these results.
Collapse
Affiliation(s)
- Paula Intasqui
- a Department of Surgery, Division of Urology, Human Reproduction Section , Universidade Federal de São Paulo (UNIFESP) - Sao Paulo Hospital , Sao Paulo , Brazil
| | - Ricardo P Bertolla
- a Department of Surgery, Division of Urology, Human Reproduction Section , Universidade Federal de São Paulo (UNIFESP) - Sao Paulo Hospital , Sao Paulo , Brazil
| | - Marcus Vinicius Sadi
- a Department of Surgery, Division of Urology, Human Reproduction Section , Universidade Federal de São Paulo (UNIFESP) - Sao Paulo Hospital , Sao Paulo , Brazil
| |
Collapse
|
67
|
Höti N, Shah P, Hu Y, Yang S, Zhang H. Proteomics analyses of prostate cancer cells reveal cellular pathways associated with androgen resistance. Proteomics 2017; 17. [PMID: 28116790 DOI: 10.1002/pmic.201600228] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 01/05/2023]
Abstract
While significant advances have been made in the diagnosis and treatment of prostate cancer, each year tens of thousands of men still die from prostate cancer in the United States. Thus, greater understanding of cellular pathways and molecular basis of prostate cancer progression in the development of androgen resistance is needed to treat these lethal phenotypes. To dissect the mechanism of androgen resistance, we utilize a proteomics approach to study the development of androgen resistance in LNCaP prostate cancer cells. Our results showed the predominant involvement of metabolic pathways that were elevated in androgen resistance phenotype. We further found the amplification of PI3K/AKT pathway and the overexpression of proteasome proteins while the mitochondrial oxidation phosphorylation was severely hampered in castration-resistant LNCaP-95 cells compared to LNCaP cells. Interestingly, we also found the induction of Dicer, a cytoplasmic endoribonuclease microRNA regulator in the androgen-ablated LNCaP-95 prostate cancer cells. We verified some of these data by orthogonal methods including Western blot analysis and in castrated animal xenograft studies. To our knowledge, this is the first report showing induced expression of proteasome proteins in androgen ablation prostate cancer cells. If validated in clinical studies, the findings will have significant implications in understanding the complexity of biochemical recurrence in prostate cancer.
Collapse
Affiliation(s)
- Naseruddin Höti
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Punit Shah
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yingwei Hu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Shuang Yang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
68
|
Basile D, Cinausero M, Iacono D, Pelizzari G, Bonotto M, Vitale MG, Gerratana L, Puglisi F. Androgen receptor in estrogen receptor positive breast cancer: Beyond expression. Cancer Treat Rev 2017; 61:15-22. [PMID: 29078133 DOI: 10.1016/j.ctrv.2017.09.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/22/2017] [Accepted: 09/23/2017] [Indexed: 01/22/2023]
Abstract
In recent years, new therapeutic approaches have reshaped the overall strategy of breast cancer (BC) treatment and have markedly improved patient survival. This is, in part, due to novel therapies for estrogen receptor (ER)-positive BC. Unfortunately, many patients present de novo resistance to these therapies or develop an acquired resistance over time. Therefore, research is now focused on discovering new molecular targets to overcome these resistances. Interestingly, preclinical and clinical studies have shown a critical role for the cross-talk between androgen receptor (AR) and ER in luminal-like BC. AR is expressed in >60% of BC and in up to 90% of ERα-positive tumors. Multiple studies suggest that AR is associated with a favorable prognosis. However, AR overexpression and, in particular, the high AR:ER ratio, seem to be involved in resistance to hormonal treatment. In this setting, a group of BCs could benefit from AR-inhibitors; nevertheless, some ER-positive BC patients do not seem to benefit from this strategy. Therefore, it is crucial to identify biomarkers that would enable the selection of patients who might benefit from combination treatment with ER and AR inhibitors.
Collapse
Affiliation(s)
- Debora Basile
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Marika Cinausero
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Donatella Iacono
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Giacomo Pelizzari
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Marta Bonotto
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Maria Grazia Vitale
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy
| | - Lorenzo Gerratana
- Department of Oncology, University Hospital of Udine, Italy; School of Medical Oncology, Department of Medicine, University of Udine, Italy.
| | - Fabio Puglisi
- School of Medical Oncology, Department of Medicine, University of Udine, Italy; Department of Clinical Oncology, CRO Aviano National Cancer Institute, Aviano (PN), Italy
| |
Collapse
|
69
|
Dysregulation of spliceosome gene expression in advanced prostate cancer by RNA-binding protein PSF. Proc Natl Acad Sci U S A 2017; 114:10461-10466. [PMID: 28893982 DOI: 10.1073/pnas.1706076114] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Developing therapeutic approaches are necessary for treating hormone-refractory prostate cancer. Activation of androgen receptor (AR) and its variants' expression along with the downstream signals are mostly important for disease progression. However, the mechanism for marked increases of AR signals and its expression is still unclear. Here, we revealed that various spliceosome genes are aberrantly induced by RNA-binding protein PSF, leading to enhancement of the splicing activities for AR expression. Our high-speed sequence analyses identified global PSF-binding transcripts. PSF was shown to stabilize and activate key long noncoding RNAs and AR-regulated gene expressions in prostate cancer cells. Interestingly, mRNAs of spliceosome-related genes are putative primary targets of PSF. Their gene expressions are up-regulated by PSF in hormone-refractory prostate cancer. Moreover, PSF coordinated these spliceosome proteins to form a complex to promote AR splicing and expression. Thus, targeting PSF and its related pathways implicates the therapeutic possibility for hormone-refractory prostate cancer.
Collapse
|
70
|
Significance of microRNAs in Androgen Signaling and Prostate Cancer Progression. Cancers (Basel) 2017; 9:cancers9080102. [PMID: 28783103 PMCID: PMC5575605 DOI: 10.3390/cancers9080102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 08/01/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022] Open
Abstract
The androgen receptor (AR) plays important roles in prostate cancer development and prostate tumor growth. After binding to androgens, AR functions as a nuclear receptor and translocates to the nucleus to bind to specific AR-binding sites (ARBSs). AR regulates epigenetic factor recruitments to activate its downstream signaling. Although androgen deprivation therapy (ADT) is initially useful for prostate cancer patients, most patients eventually show resistance with hormone-refractory prostate cancers (HRPCs) or castration-resistant prostate cancers (CRPCs). Thus, new therapeutic strategies targeting HRPCs/CRPCs should be very important for clinical medicine as well as prostate cancer biology. Past studies have shown that mechanisms such as AR overexpression, hypersensitivity, variants and reprograming are responsible for developing HRPCs/CRPCs. These findings suggest that AR target genes will be major key factors. In this review article, we focus mainly on the androgen-regulated microRNAs (miRNAs) to summarize the contribution of miRNA-mediated pathways for prostate cancer progression.
Collapse
|
71
|
Tan G, Qiu M, Chen L, Zhang S, Ke L, Liu J. JS-K, a nitric oxide pro-drug, regulates growth and apoptosis through the ubiquitin-proteasome pathway in prostate cancer cells. BMC Cancer 2017; 17:376. [PMID: 28549433 PMCID: PMC5446692 DOI: 10.1186/s12885-017-3351-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/12/2017] [Indexed: 01/17/2023] Open
Abstract
Background In view of the fact that JS-K might regulate ubiquitin E3 ligase and that ubiquitin E3 ligase plays an important role in the mechanism of CRPC formation, the goal was to investigate the probable mechanism by which JS-K regulates prostate cancer cells. Methods Proliferation inhibition by JS-K on prostate cancer cells was examined usingCCK-8 assays. Caspase 3/7 activity assays and flow cytometry were performed to examine whether JS-K induced apoptosis in prostate cancer cells. Western blotting and co-immunoprecipitation analyses investigated JS-K’s effects on the associated apoptosis mechanism. Real time-PCR and Western blotting were performed to assess JS-K’s effect on transcription of specific AR target genes. Western blotting was also performed to detect Siah2 and AR protein concentrations and co-immunoprecipitation to detect interactions of Siah2 and AR, NCoR1 and AR, and p300 and AR. Results JS-K inhibited proliferation and induced apoptosis in prostate cancer cells. JS-K increased p53 and Mdm2 concentrations and regulated the caspase cascade reaction-associated protein concentrations. JS-K inhibited transcription of AR target genes and down-regulated PSA protein concentrations. JS-K inhibited Siah2 interactions and also inhibited the ubiquitination of AR. With further investigation, JS-K was found to stabilize AR and NCoR1 interactions and diminish AR and p300 interactions. Conclusions The present results suggested that JS-K might have been able to inhibit proliferation and induce apoptosis via regulation of the ubiquitin-proteasome degradation pathway, which represented a promising platform for the development of new compounds for PCa treatments. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3351-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guobin Tan
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Mingning Qiu
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Lieqian Chen
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Sai Zhang
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Longzhi Ke
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical College, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
72
|
Mechanisms of resistance to systemic therapy in metastatic castration-resistant prostate cancer. Cancer Treat Rev 2017; 57:16-27. [PMID: 28527407 DOI: 10.1016/j.ctrv.2017.04.008] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 12/22/2022]
Abstract
Patients with metastatic castration-resistant prostate cancer (mCPRC) now have an unprecedented number of approved treatment options, including chemotherapies (docetaxel, cabazitaxel), androgen receptor (AR)-targeted therapies (enzalutamide, abiraterone), a radioisotope (radium-223) and a cancer vaccine (sipuleucel-T). However, the optimal treatment sequencing pathway is unknown, and this problem is exacerbated by the issues of primary and acquired resistance. This review focuses on mechanisms of resistance to AR-targeted therapies and taxane-based chemotherapy. Patients treated with abiraterone, enzalutamide, docetaxel or cabazitaxel may present with primary resistance, or eventually acquire resistance when on treatment. Multiple resistance mechanisms to AR-targeted agents have been proposed, including: intratumoral androgen production, amplification, mutation, or expression of AR splice variants, increased steroidogenesis, upregulation of signals downstream of the AR, and development of androgen-independent tumor cells. Known mechanisms of resistance to chemotherapy are distinct, and include: tubulin alterations, increased expression of multidrug resistance genes, TMPRSS2-ERG fusion genes, kinesins, cytokines, and components of other signaling pathways, and epithelial-mesenchymal transition. Utilizing this information, biomarkers of resistance/response have the potential to direct treatment decisions. Expression of the AR splice variant AR-V7 may predict resistance to AR-targeted agents, but available biomarker assays are yet to be prospectively validated in the clinic. Ongoing prospective trials are evaluating the sequential use of different drugs, or combination regimens, and the results of these studies, combined with a deeper understanding of mechanisms of primary and acquired resistance to treatment, have the potential to drive future treatment decisions in mCRPC.
Collapse
|
73
|
Barfeld SJ, Urbanucci A, Itkonen HM, Fazli L, Hicks JL, Thiede B, Rennie PS, Yegnasubramanian S, DeMarzo AM, Mills IG. c-Myc Antagonises the Transcriptional Activity of the Androgen Receptor in Prostate Cancer Affecting Key Gene Networks. EBioMedicine 2017; 18:83-93. [PMID: 28412251 PMCID: PMC5405195 DOI: 10.1016/j.ebiom.2017.04.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 04/04/2017] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men. The androgen receptor (AR), a ligand-activated transcription factor, constitutes the main drug target for advanced cases of the disease. However, a variety of other transcription factors and signaling networks have been shown to be altered in patients and to influence AR activity. Amongst these, the oncogenic transcription factor c-Myc has been studied extensively in multiple malignancies and elevated protein levels of c-Myc are commonly observed in PCa. Its impact on AR activity, however, remains elusive. In this study, we assessed the impact of c-Myc overexpression on AR activity and transcriptional output in a PCa cell line model and validated the antagonistic effect of c-MYC on AR-targets in patient samples. We found that c-Myc overexpression partially reprogrammed AR chromatin occupancy and was associated with altered histone marks distribution, most notably H3K4me1 and H3K27me3. We found c-Myc and the AR co-occupy a substantial number of binding sites and these exhibited enhancer-like characteristics. Interestingly, c-Myc overexpression antagonised clinically relevant AR target genes. Therefore, as an example, we validated the antagonistic relationship between c-Myc and two AR target genes, KLK3 (alias PSA, prostate specific antigen), and Glycine N-Methyltransferase (GNMT), in patient samples. Our findings provide unbiased evidence that MYC overexpression deregulates the AR transcriptional program, which is thought to be a driving force in PCa. c-MYC and AR share one third of chromatin binding with enhancer-like features. c-MYC can repress the expression of a subset prostate cancer biomarkers, including PSA. c-MYC and AR antagonize the expression of, Glycine N-Methyltransferase (GNMT), responsible for sarcosine biosynthesis.
Prostate cancer is a heterogeneous disease. The most frequently used biomarker in clinical setting, a well described androgen receptor target gene, PSA, still performs poorly in stratifying patients at real risk of death due to the disease. Despite this, therapeutic approaches focus on suppressing androgen receptor signaling. However, this is only one of the recurrent alterations found in patients. This study focuses on c-MYC and the effects of its deregulation in advanced prostate cancer. We find that there is an inverse relationship between established biomarkers expression, including PSA. This inverse relationship could be used in clinics to select beneficial therapeutic approaches for a subset of prostate cancer cases.
Collapse
Affiliation(s)
- Stefan J Barfeld
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway.
| | - Alfonso Urbanucci
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Harri M Itkonen
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Canada
| | | | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Paul S Rennie
- The Vancouver Prostate Centre, University of British Columbia, Canada
| | | | - Angelo M DeMarzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ian G Mills
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; PCUK/Movember Centre of Excellence, CCRCB, Queen's University, Belfast, UK.
| |
Collapse
|
74
|
Shiota M, Yokomizo A, Takeuchi A, Kashiwagi E, Dejima T, Inokuchi J, Tatsugami K, Uchiumi T, Eto M. Protein kinase C regulates Twist1 expression via NF-κB in prostate cancer. Endocr Relat Cancer 2017; 24:171-180. [PMID: 28223364 DOI: 10.1530/erc-16-0384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/21/2017] [Indexed: 11/08/2022]
Abstract
The progression of prostate cancer to metastatic and castration-resistant disease represents a critical step. We previously showed that protein kinase C (PKC) activation followed by Twist1 and androgen receptor (AR) induction played a critical role in castration resistance, but the precise molecular mechanism remains unknown. This study aimed to elucidate the relevant molecular mechanism, focusing on NF-κB transcription factor. We examined the activity of NF-κB after PKC inhibition, and the expression of Twist1 and AR after inhibition of NF-κB in human prostate cancer cells. We also investigated the status of PKC/NF-κB after inhibition of AR signaling in cells resistant to hormonal therapy. As a result, inhibition of PKC signaling using knockdown and small-molecule inhibition of PKC suppressed RelA activity, while blocking NF-κB suppressed Twist1 and AR expression. Conversely, inhibition of AR signaling by androgen depletion and the novel antiandrogen enzalutamide induced PKC and RelA activation, resulting in Twist1/AR induction at the transcript level. Moreover, inhibition of NF-κB signaling prevented enzalutamide-induced Twist1 and AR induction. Finally, NF-κB was activated in both castration-resistant and enzalutamide-resistant cells. In conclusion, NF-κB signaling was responsible for Twist1 upregulation by PKC in response to AR inhibition, resulting in aberrant activation of AR. NF-κB signaling thus appears to play a critical role in promoting both castration resistance and enzalutamide resistance in PKC/Twist1 signaling in prostate cancer.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Yokomizo
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ario Takeuchi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Kashiwagi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Dejima
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Inokuchi
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsunori Tatsugami
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory MedicineGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of UrologyGraduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
75
|
Xie H, Li C, Dang Q, Chang LS, Li L. Infiltrating mast cells increase prostate cancer chemotherapy and radiotherapy resistances via modulation of p38/p53/p21 and ATM signals. Oncotarget 2016; 7:1341-53. [PMID: 26625310 PMCID: PMC4811464 DOI: 10.18632/oncotarget.6372] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/06/2015] [Indexed: 12/11/2022] Open
Abstract
Early studies indicated that mast cells in prostate tumor microenvironment might influence prostate cancer (PCa) progression. Their impacts to PCa therapy, however, remained unclear. Here we found PCa could recruit more mast cells than normal prostate epithelial cells then alter PCa chemotherapy and radiotherapy sensitivity, leading to PCa more resistant to these therapies. Mechanism dissection revealed that infiltrated mast cells could increase p21 expression via modulation of p38/p53 signals, and interrupting p38-p53 signals via siRNAs of p53 or p21 could reverse mast cell-induced docetaxel chemotherapy resistance of PCa. Furthermore, recruited mast cells could also increase the phosphorylation of ATM at ser-1981 site, and inhibition of ATM activity could reverse mast cell-induced radiotherapy resistance. The in vivo mouse model with xenografted PCa C4-2 cells co-cultured with mast cells also confirmed that mast cells could increase PCa chemotherapy resistance via activating p38/p53/p21 signaling. Together, our results provide a new mechanism showing infiltrated mast cells could alter PCa chemotherapy and radiotherapy sensitivity via modulating the p38/p53/p21 signaling and phosphorylation of ATM. Targeting this newly identified signaling may help us better suppress PCa chemotherapy and radiotherapy resistance.
Collapse
Affiliation(s)
- Hongjun Xie
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chong Li
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Qiang Dang
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Luke S Chang
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Lei Li
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
76
|
The flavonoid fisetin as an anticancer agent targeting the growth signaling pathways. Eur J Pharmacol 2016; 789:8-16. [DOI: 10.1016/j.ejphar.2016.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 12/29/2022]
|
77
|
Armstrong CM, Gao AC. Adaptive pathways and emerging strategies overcoming treatment resistance in castration resistant prostate cancer. Asian J Urol 2016. [PMID: 28642838 PMCID: PMC5477778 DOI: 10.1016/j.ajur.2016.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The therapies available for prostate cancer patients whom progress from hormone-sensitive to castration resistant prostate cancer include both systemic drugs, including docetaxel and cabazitaxel, and drugs that inhibit androgen signaling such as enzalutamide and abiraterone. Unfortunately, it is estimated that up to 30% of patients have primary resistance to these treatments and over time even those who initially respond to therapy will eventually develop resistance and their disease will continue to progress regardless of the presence of the drug. Determining the mechanisms involved in the development of resistance to these therapies has been the area of intense study and several adaptive pathways have been uncovered. Androgen receptor (AR) mutations, expression of AR-V7 (or other constitutively active androgen receptor variants), intracrine androgen production and overexpression of androgen synthesis enzymes such as Aldo-Keto Reductase Family 1, Member C3 (AKR1C3) are among the many mechanisms associated with resistance to anti-androgens. In regards to the taxanes, one of the key contributors to drug resistance is increased drug efflux through ATP Binding Cassette Subfamily B Member 1 (ABCB1). Targeting these resistance mechanisms using different strategies has led to various levels of success in overcoming resistance to current therapies. For instance, targeting AR-V7 with niclosamide or AKR1C3 with indomethacin can improve enzalutamide and abiraterone treatment. ABCB1 transport activity can be inhibited by the dietary constituent apigenin and antiandrogens such as bicalutamide which in turn improves response to docetaxel. A more thorough understanding of how drug resistance develops will lead to improved treatment strategies. This review will cover the current knowledge of resistance mechanisms to castration resistant prostate cancer therapies and methods that have been identified which may improve treatment response.
Collapse
Affiliation(s)
| | - Allen C Gao
- Department of Urology, University of California, Davis, Sacramento, CA, USA.,Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA.,VA Northern California Health Care System, Sacramento, CA, USA
| |
Collapse
|
78
|
Johnson J, Skoda EM, Zhou J, Parrinello E, Wang D, O’Malley K, Eyer BR, Kazancioglu M, Eisermann K, Johnston PA, Nelson JB, Wang Z, Wipf P. Small Molecule Antagonists of the Nuclear Androgen Receptor for the Treatment of Castration-Resistant Prostate Cancer. ACS Med Chem Lett 2016; 7:785-90. [PMID: 27563404 PMCID: PMC4983742 DOI: 10.1021/acsmedchemlett.6b00186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/27/2016] [Indexed: 01/29/2023] Open
Abstract
After a high-throughput screening campaign identified thioether 1 as an antagonist of the nuclear androgen receptor, a zone model was developed for structure-activity relationship (SAR) purposes and analogues were synthesized and evaluated in a cell-based luciferase assay. A novel thioether isostere, cyclopropane (1S,2R)-27, showed the desired increased potency and structural properties (stereospecific SAR response, absence of a readily oxidized sulfur atom, low molecular weight, reduced number of flexible bonds and polar surface area, and drug-likeness score) in the prostate-specific antigen luciferase assay in C4-2-PSA-rl cells to qualify as a new lead structure for prostate cancer drug development.
Collapse
Affiliation(s)
- James
K. Johnson
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Erin M. Skoda
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Jianhua Zhou
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Erica Parrinello
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Dan Wang
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Katherine O’Malley
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Benjamin R. Eyer
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Mustafa Kazancioglu
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Kurtis Eisermann
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Paul A. Johnston
- Department
of Pharmaceutical Sciences, University of
Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Joel B. Nelson
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Zhou Wang
- Department
of Urology, University of Pittsburgh School
of Medicine, Pittsburgh, Pennsylvania 15232, United States
| | - Peter Wipf
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Pharmaceutical Sciences, University of
Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
79
|
Imamura Y, Sadar MD. Androgen receptor targeted therapies in castration-resistant prostate cancer: Bench to clinic. Int J Urol 2016; 23:654-65. [PMID: 27302572 PMCID: PMC6680212 DOI: 10.1111/iju.13137] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/05/2016] [Indexed: 12/11/2022]
Abstract
The androgen receptor is a transcription factor and validated therapeutic target for prostate cancer. Androgen deprivation therapy remains the gold standard treatment, but it is not curative, and eventually the disease will return as lethal castration‐resistant prostate cancer. There have been improvements in the therapeutic landscape with new agents approved, such as abiraterone acetate, enzalutamide, sipuleucel‐T, cabazitaxel and Ra‐223, in the past 5 years. New insight into the mechanisms of resistance to treatments in advanced disease is being and has been elucidated. All current androgen receptor‐targeting therapies inhibit the growth of prostate cancer by blocking the ligand‐binding domain, where androgen binds to activate the receptor. Persuasive evidence supports the concept that constitutively active androgen receptor splice variants lacking the ligand‐binding domain are one of the resistant mechanisms underlying advanced disease. Transcriptional activity of the androgen receptor requires a functional AF‐1 region in its N‐terminal domain. Preclinical evidence proved that this domain is a druggable target to forecast a potential paradigm shift in the management of advanced prostate cancer. This review presents an overview of androgen receptor‐related mechanisms of resistance as well as novel therapeutic agents to overcome resistance that is linked to the expression of androgen receptor splice variants in castration‐resistant prostate cancer.
Collapse
Affiliation(s)
- Yusuke Imamura
- Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Marianne D Sadar
- Genome Sciences Center, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| |
Collapse
|
80
|
Khanna A, Rane JK, Kivinummi KK, Urbanucci A, Helenius MA, Tolonen TT, Saramäki OR, Latonen L, Manni V, Pimanda JE, Maitland NJ, Westermarck J, Visakorpi T. CIP2A is a candidate therapeutic target in clinically challenging prostate cancer cell populations. Oncotarget 2016; 6:19661-70. [PMID: 25965834 PMCID: PMC4637312 DOI: 10.18632/oncotarget.3875] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/03/2015] [Indexed: 12/15/2022] Open
Abstract
Residual androgen receptor (AR)-signaling and presence of cancer stem-like cells (SCs) are the two emerging paradigms for clinically challenging castration-resistant prostate cancer (CRPC). Therefore, identification of AR-target proteins that are also overexpressed in the cancer SC population would be an attractive therapeutic approach. Our analysis of over three hundred clinical samples and patient-derived prostate epithelial cultures (PPECs), revealed Cancerous inhibitor of protein phosphatase 2A (CIP2A) as one such target. CIP2A is significantly overexpressed in both hormone-naïve prostate cancer (HN-PC) and CRPC patients. CIP2A is also overexpressed, by 3- and 30-fold, in HN-PC and CRPC SCs respectively. In vivo binding of the AR to the intronic region of CIP2A and its functionality in the AR-moderate and AR-high expressing LNCaP cell-model systems is also demonstrated. Further, we show that AR positively regulates CIP2A expression, both at the mRNA and protein level. Finally, CIP2A depletion reduced cell viability and colony forming efficiency of AR-independent PPECs as well as AR-responsive LNCaP cells, in which anchorage-independent growth is also impaired. These findings identify CIP2A as a common denominator for AR-signaling and cancer SC functionality, highlighting its potential therapeutic significance in the most clinically challenging prostate pathology: castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Anchit Khanna
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland.,Adult Cancer Program, The Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Jayant K Rane
- YCR Cancer Research Unit, Department of Biology, The University of York, Heslington, United Kingdom
| | - Kati K Kivinummi
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland.,Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - Alfonso Urbanucci
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland.,Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Cancer Prevention, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Merja A Helenius
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Teemu T Tolonen
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Outi R Saramäki
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Leena Latonen
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Visa Manni
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| | - John E Pimanda
- Adult Cancer Program, The Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Norman J Maitland
- YCR Cancer Research Unit, Department of Biology, The University of York, Heslington, United Kingdom
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Pathology, University of Turku, Turku, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center (PCRC), Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
81
|
Inhibition of the hexosamine biosynthetic pathway promotes castration-resistant prostate cancer. Nat Commun 2016; 7:11612. [PMID: 27194471 PMCID: PMC4874037 DOI: 10.1038/ncomms11612] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/13/2016] [Indexed: 01/22/2023] Open
Abstract
The precise molecular alterations driving castration-resistant prostate cancer (CRPC) are not clearly understood. Using a novel network-based integrative approach, here, we show distinct alterations in the hexosamine biosynthetic pathway (HBP) to be critical for CRPC. Expression of HBP enzyme glucosamine-phosphate N-acetyltransferase 1 (GNPNAT1) is found to be significantly decreased in CRPC compared with localized prostate cancer (PCa). Genetic loss-of-function of GNPNAT1 in CRPC-like cells increases proliferation and aggressiveness, in vitro and in vivo. This is mediated by either activation of the PI3K-AKT pathway in cells expressing full-length androgen receptor (AR) or by specific protein 1 (SP1)-regulated expression of carbohydrate response element-binding protein (ChREBP) in cells containing AR-V7 variant. Strikingly, addition of the HBP metabolite UDP-N-acetylglucosamine (UDP-GlcNAc) to CRPC-like cells significantly decreases cell proliferation, both in-vitro and in animal studies, while also demonstrates additive efficacy when combined with enzalutamide in-vitro. These observations demonstrate the therapeutic value of targeting HBP in CRPC. The molecular alterations driving anti-androgen resistance in prostate cancer are unclear. Here, the authors show, using a network-based approach, that inhibition of the hexosamine biosynthetic pathway is necessary to develop resistance and that increasing the activity of the pathway enhances the anti-androgen response.
Collapse
|
82
|
Zito Marino F, Rocco G, Morabito A, Mignogna C, Intartaglia M, Liguori G, Botti G, Franco R. A new look at the ALK gene in cancer: copy number gain and amplification. Expert Rev Anticancer Ther 2016; 16:493-502. [PMID: 26943457 DOI: 10.1586/14737140.2016.1162098] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To date, ALK-rearrangement is a molecular target in several cancers, i.e. NSCLC. The dramatic benefits of crizotinib have prompted research into identifying other possible patients carrying ALK gene alterations with possible clinical significance. The ALK gene is involved not only in several rearrangements but also in other alterations such as amplification. ALK-amplification (ALK-A) is a common genetic event in several cancers, generally associated with poor outcome and more aggressive behaviour. Here we review the role of ALK-A in cancer as a prognostic and predictive biomarker. Furthermore, several critical issues regarding ALK-A in relation to; methods of detection, acquired resistance and ALK second generation inhibitors are analyzed. We conclude that ALK-A could be an intriguing alteration in the context of targeted therapy.
Collapse
Affiliation(s)
- Federica Zito Marino
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Gaetano Rocco
- b Division of Thoracic Surgery, Department of Thoracic Surgical and Medical Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Alessandro Morabito
- c Medical Oncology Unit, Department of Thoracic Surgical and Medical Oncology , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Chiara Mignogna
- d Department of Heath Science, Pathology Unit , University 'Magna Graecia' of Catanzaro , Catanzaro , Italy
| | - Martina Intartaglia
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Giuseppina Liguori
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Gerardo Botti
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy
| | - Renato Franco
- a Pathology Unit , Istituto Nazionale Tumori 'Fondazione G. Pascale'-IRCCS , Naples , Italy.,e Pathology Unit , Second University of Naples - SUN , Naples , Italy
| |
Collapse
|
83
|
Shiota M, Yokomizo A, Takeuchi A, Kiyoshima K, Inokuchi J, Tatsugami K, Shiga KI, Koga H, Yamaguchi A, Naito S, Eto M. Co-introduction of a steroid with docetaxel chemotherapy for metastatic castration-resistant prostate cancer affects PSA flare. BJU Int 2016; 118:880-884. [DOI: 10.1111/bju.13483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Masaki Shiota
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Akira Yokomizo
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Ario Takeuchi
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Keijiro Kiyoshima
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Junichi Inokuchi
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Katsunori Tatsugami
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | | | - Hirofumi Koga
- Division of Urology; Harasanshin Hospital; Fukuoka Japan
| | | | - Seiji Naito
- Division of Urology; Harasanshin Hospital; Fukuoka Japan
| | - Masatoshi Eto
- Department of Urology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|
84
|
Lallous N, Volik SV, Awrey S, Leblanc E, Tse R, Murillo J, Singh K, Azad AA, Wyatt AW, LeBihan S, Chi KN, Gleave ME, Rennie PS, Collins CC, Cherkasov A. Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients. Genome Biol 2016; 17:10. [PMID: 26813233 PMCID: PMC4729137 DOI: 10.1186/s13059-015-0864-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/29/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The androgen receptor (AR) is a pivotal drug target for the treatment of prostate cancer, including its lethal castration-resistant (CRPC) form. All current non-steroidal AR antagonists, such as hydroxyflutamide, bicalutamide, and enzalutamide, target the androgen binding site of the receptor, competing with endogenous androgenic steroids. Several AR mutations in this binding site have been associated with poor prognosis and resistance to conventional prostate cancer drugs. In order to develop an effective CRPC therapy, it is crucial to understand the effects of these mutations on the functionality of the AR and its ability to interact with endogenous steroids and conventional AR inhibitors. RESULTS We previously utilized circulating cell-free DNA (cfDNA) sequencing technology to examine the AR gene for the presence of mutations in CRPC patients. By modifying our sequencing and data analysis approaches, we identify four additional single AR mutations and five mutation combinations associated with CRPC. Importantly, we conduct experimental functionalization of all the AR mutations identified by the current and previous cfDNA sequencing to reveal novel gain-of-function scenarios. Finally, we evaluate the effect of a novel class of AR inhibitors targeting the binding function 3 (BF3) site on the activity of CRPC-associated AR mutants. CONCLUSIONS This work demonstrates the feasibility of a prognostic and/or diagnostic platform combining the direct identification of AR mutants from patients' serum, and the functional characterization of these mutants in order to provide personalized recommendations regarding the best future therapy.
Collapse
Affiliation(s)
- Nada Lallous
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Stanislav V Volik
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
- Laboratory for Advanced Genome Analysis (LAGA), Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Shannon Awrey
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Eric Leblanc
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Ronnie Tse
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Josef Murillo
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Kriti Singh
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Arun A Azad
- Department of Medical Oncology, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada.
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Stephane LeBihan
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
- Laboratory for Advanced Genome Analysis (LAGA), Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Kim N Chi
- Department of Medical Oncology, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada.
| | - Martin E Gleave
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Paul S Rennie
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Colin C Collins
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
- Laboratory for Advanced Genome Analysis (LAGA), Vancouver Prostate Centre, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak St., Vancouver, BC, V6H 3Z6, Canada.
| |
Collapse
|
85
|
Takayama KI, Suzuki T, Tsutsumi S, Fujimura T, Urano T, Takahashi S, Homma Y, Aburatani H, Inoue S. RUNX1, an androgen- and EZH2-regulated gene, has differential roles in AR-dependent and -independent prostate cancer. Oncotarget 2016; 6:2263-76. [PMID: 25537508 PMCID: PMC4385850 DOI: 10.18632/oncotarget.2949] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/09/2015] [Indexed: 12/03/2022] Open
Abstract
Androgen receptor (AR) signaling is essential for the development of prostate cancer. Here, we report that runt-related transcription factor (RUNX1) could be a key molecule for the androgen-dependence of prostate cancer. We found RUNX1 is a target of AR and regulated positively by androgen. Our RUNX1 ChIP-seq analysis indicated that RUNX1 is recruited to AR binding sites by interacting with AR. In androgen-dependent cancer, loss of RUNX1 impairs AR-dependent transcription and cell growth. The RUNX1 promoter is bound by enhancer of zeste homolog 2 (EZH2) and is negatively regulated by histone H3 lysine 27 (K27) trimethylation. Repression of RUNX1 is important for the growth promotion ability of EZH2 in AR-independent cells. In clinical prostate cancer samples, the RUNX1 expression level is negatively associated with EZH2 and that RUNX1 loss correlated with poor prognosis. These results indicated the significance of RUNX1 for androgen-dependency and that loss of RUNX1 could be a key step for the progression of prostate cancer.
Collapse
Affiliation(s)
- Ken-ichi Takayama
- Department of Anti-Aging Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Geriatric Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Suzuki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shuichi Tsutsumi
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Tetsuya Fujimura
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomohiko Urano
- Department of Anti-Aging Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Geriatric Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Yukio Homma
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Satoshi Inoue
- Department of Anti-Aging Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Geriatric Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| |
Collapse
|
86
|
Ran F, Xing H, Liu Y, Zhang D, Li P, Zhao G. Recent Developments in Androgen Receptor Antagonists. Arch Pharm (Weinheim) 2015; 348:757-775. [PMID: 26462013 DOI: 10.1002/ardp.201500187] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
The androgen receptor (AR), a ligand-dependent transcription factor that regulates the expression of a series of downstream target genes after the binding of androgens, has been a target for the discovery of drugs used to treat prostate cancer. Prostate cancer always progresses to castration-resistant prostate cancer after a period of androgen deprivation therapy. Thus, developing potent androgen receptor antagonists for the therapy of castration-resistant prostate cancer possesses great significance. This review summarizes the preclinical development of androgen receptor antagonists, conventional androgen receptor antagonists that competitively bind to the ligand binding domain of the androgen receptor and coactivator antagonists of the androgen receptor, including both activation function-2 antagonists and binding function-3 antagonists. We hope that this review can help other researchers find new scaffolds and sites for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Fansheng Ran
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| | - Hualu Xing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| | - Yang Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| | - Daoguang Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| | - Pengzhan Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| | - Guisen Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, P. R. China
| |
Collapse
|
87
|
O'Neill D, Jones D, Wade M, Grey J, Nakjang S, Guo W, Cork D, Davies BR, Wedge SR, Robson CN, Gaughan L. Development and exploitation of a novel mutant androgen receptor modelling strategy to identify new targets for advanced prostate cancer therapy. Oncotarget 2015; 6:26029-40. [PMID: 26267320 PMCID: PMC4694883 DOI: 10.18632/oncotarget.4347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/08/2015] [Indexed: 11/25/2022] Open
Abstract
The persistence of androgen receptor (AR) signalling in castrate-resistant prostate cancer (CRPC) highlights the unmet clinical need for the development of more effective AR targeting therapies. A key mechanism of therapy-resistance is by selection of AR mutations that convert anti-androgens to agonists enabling the retention of androgenic signalling in CRPC. To improve our understanding of these receptors in advanced disease we developed a physiologically-relevant model to analyse the global functionality of AR mutants in CRPC. Using the bicalutamide-activated AR(W741L/C) mutation as proof of concept, we demonstrate that this mutant confers an androgenic-like signalling programme and growth promoting phenotype in the presence of bicalutamide. Transcriptomic profiling of AR(W741L) highlighted key genes markedly up-regulated by the mutant receptor, including TIPARP, RASD1 and SGK1. Importantly, SGK1 expression was found to be highly expressed in the KUCaP xenograft model and a CRPC patient biopsy sample both of which express the bicalutamide-activated receptor mutant. Using an SGK1 inhibitor, AR(W741L) transcriptional and growth promoting activity was reduced indicating that exploiting functional distinctions between receptor isoforms in our model may provide new and effective therapies for CRPC patients.
Collapse
Affiliation(s)
- Daniel O'Neill
- AstraZeneca, Innovative Medicines, Discovery Sciences, Cambridge Science Park, Cambridge, CB4 0WG, UK
| | - Dominic Jones
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Mark Wade
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - James Grey
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Sirintra Nakjang
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Wenrui Guo
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - David Cork
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Barry R. Davies
- AstraZeneca, Innovative Medicines, Discovery Sciences, Cambridge Science Park, Cambridge, CB4 0WG, UK
| | - Steve R. Wedge
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Craig N. Robson
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Luke Gaughan
- Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| |
Collapse
|
88
|
Takayama KI, Suzuki T, Tsutsumi S, Fujimura T, Takahashi S, Homma Y, Urano T, Aburatani H, Inoue S. Integrative analysis of FOXP1 function reveals a tumor-suppressive effect in prostate cancer. Mol Endocrinol 2015; 28:2012-24. [PMID: 25329375 DOI: 10.1210/me.2014-1171] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The transcriptional network of the androgen receptor (AR), a key molecule of prostate cancer, is frequently modulated by interactions with other transcriptional factors such as forkhead box protein A1 (FOXA1). However, global regulatory mechanisms of AR signaling mediated by such factors have not been well investigated. Here we conducted a chromatin immunoprecipitation sequence analysis, which revealed that another FOX family, FOXP1, is specifically regulated by both AR and FOXA1. We also found that FOXP1 acts as a tumor suppressor in prostate cancer through inhibiting cell proliferation and migration. We generated an extensive global map of FOXP1 binding sites and found that FOXP1 is directly involved in AR-mediated transcription. We demonstrated that FOXP1 has a repressive effect on AR-induced transcriptional activity or histone modification in enhancer regions. Moreover, by a global analysis of androgen-mediated transcriptional networks, we observed enrichment of FOXP1 binding genes in the gene cluster negatively regulated by FOXP1. Evaluation of FOXP1 expression in clinical samples indicated that the decreased expression of FOXP1 is another prognostic factor of prostate cancer. Taken together, our results suggest a novel mechanism in which AR-induced FOXP1 functions as a direct modulator of the AR and FOXA1 centric global transcriptional network.
Collapse
Affiliation(s)
- Ken-Ichi Takayama
- Departments of Anti-Aging Medicine (K.T., T.U., S.I.) and Geriatric Medicine (K.T., T.U., S.I.) and Department of Urology (T.F., Y.H.), Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Pathology (T.S.), Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan; Genome Science Division (Sh.T., H.A.), Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan; Department of Urology (Sa.T.), Nihon University School of Medicine, Itabashi-ku, Tokyo 173-0032, Japan; and Division of Gene Regulation and Signal Transduction (S.I.), Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
YAP1 and AR interactions contribute to the switch from androgen-dependent to castration-resistant growth in prostate cancer. Nat Commun 2015; 6:8126. [PMID: 28230103 DOI: 10.1038/ncomms9126] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 07/21/2015] [Indexed: 12/25/2022] Open
Abstract
The transcriptional co-activator Yes-associated protein 1 (YAP1), a key nuclear effector of the Hippo pathway, is a potent oncogene, and yet, the interaction between YAP1 and androgen receptor (AR) remains unexplored. Here we identify YAP1 as a physiological binding partner and positive regulator of AR in prostate cancer. YAP1 and AR co-localize and interact with each other predominantly within cell nuclei by an androgen-dependent mechanism in a hormone naive and an androgen-independent mechanism in castration-resistant prostate cancer cells. The growth suppressor MST1 kinase modulates androgen-dependent and -independent nuclear YAP1-AR interactions through directly regulating YAP1 nuclear accumulation. Disruption of YAP1 signalling by genetic (RNAi) and pharmacological (Verteporfin) approaches suppresses AR-dependent gene expression and prostate cancer cell growth. These findings indicate that the YAP1-AR axis may have a critical role in prostate cancer progression and serves as a viable drug target.
Collapse
|
90
|
Batth IS, Yun H, Kumar AP. Recepteur d'origine nantais (RON), more than a kinase: Role in castrate-resistant prostate cancer. Mol Carcinog 2015; 54:937-46. [PMID: 26152593 DOI: 10.1002/mc.22354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/20/2015] [Accepted: 05/28/2015] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCA) is the second leading cause of cancer-related deaths in men in the United States. It is natural for a hormone-driven malignancy such as prostate cancer that androgen deprivation therapy (ADT) would be the preferred treatment for clinical disease management. However, after initial treatment response a vast majority of patients develop metastatic castrate-resistant prostate cancer (CRPC), which is fatal. While great headway has been made to understand the possible mechanisms that drive castrate-resistant disease, a bonafide cure remains elusive. Reactivation of androgen receptor (AR) signaling partly contributes to the emergence of CRPC. Here we briefly examine some of the known mechanisms of AR reactivation including intratumoral synthesis of androgens, modulation of AR coregulators, and AR variants with constitutive activity as well as activation of receptor tyrosine kinases. We primarily focus on the emerging dual function of the receptor tyrosine kinase (recepteur d'origine nantais; RON) as a traditional tyrosine kinase and transcription factor. We further discuss activation of RON as an alternate mechanism in the development of CRPC and available therapeutic approaches for clinical management of CRPC by combined inhibition of RON and AR.
Collapse
Affiliation(s)
- Izhar Singh Batth
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas
| | - Huiyoung Yun
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas
| | - Addankl P Kumar
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas.,Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas.,South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
91
|
Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies. Sci Rep 2015; 5:12007. [PMID: 26137992 PMCID: PMC4490394 DOI: 10.1038/srep12007] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/29/2015] [Indexed: 12/12/2022] Open
Abstract
Activation of androgen receptor (AR) is crucial for prostate cancer growth. Remarkably, also castration-resistant prostate cancer (CRPC) is dependent on functional AR, and several mechanisms have been proposed to explain the addiction. Known causes of CRPC include gene amplification and overexpression as well as point mutations of AR. We report here the pharmacological profile of ODM-201, a novel AR inhibitor that showed significant antitumor activity and a favorable safety profile in phase 1/2 studies in men with CRPC. ODM-201 is a full and high-affinity AR antagonist that, similar to second-generation antiandrogens enzalutamide and ARN-509, inhibits testosterone-induced nuclear translocation of AR. Importantly, ODM-201 also blocks the activity of the tested mutant ARs arising in response to antiandrogen therapies, including the F876L mutation that confers resistance to enzalutamide and ARN-509. In addition, ODM-201 reduces the growth of AR-overexpressing VCaP prostate cancer cells both in vitro and in a castration-resistant VCaP xenograft model. In contrast to other antiandrogens, ODM-201 shows negligible brain penetrance and does not increase serum testosterone levels in mice. In conclusion, ODM-201 is a potent AR inhibitor that overcomes resistance to AR-targeted therapies by antagonizing both overexpressed and mutated ARs. ODM-201 is currently in a phase 3 trial in CRPC.
Collapse
|
92
|
Modena A, Ciccarese C, Fantinel E, Bimbatti D, Tortora G, Massari F. Metastatic castration-resistant prostate cancer: targeting the mechanisms of resistance to abiraterone acetate and enzalutamide. Expert Rev Anticancer Ther 2015; 15:1037-48. [DOI: 10.1586/14737140.2015.1063423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
93
|
Pantano F, Iuliani M, Zoccoli A, Fioramonti M, De Lisi D, Fioroni I, Ribelli G, Santoni M, Vincenzi B, Tonini G, Santini D. Emerging drugs for the treatment of bone metastasis. Expert Opin Emerg Drugs 2015; 20:637-51. [PMID: 26113304 DOI: 10.1517/14728214.2015.1062876] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Bone metastases are virtually incurable resulting in significant disease morbidity, reduced quality of life and mortality. Bone provides a unique microenvironment whose local interactions with tumor cells offer novel targets for therapeutic interventions. Increased understanding of the pathogenesis of bone disease has led to the discovery and clinical utility of bone-targeted agents other than bisphosphonates and denosumab, currently, the standard of care in this setting. AREAS COVERED In this review, we present the recent advances in molecular targeted therapies focusing on therapies that inhibit bone resorption and/or stimulate bone formation and novel anti-tumoral agents that exerts significant effects on skeletal metastases, nowadays available in clinical practice or in phase of development. EXPERT OPINION New emergent bone target therapies radium-223, mTOR inhibitors, anti-androgens have demonstrated the ability to increase overall survival in bone metastatic patients, other compounds, such as ET-1 and SRC inhibitors, up to now failed to clearly confirm in clinical trials their promising preclinical data.
Collapse
Affiliation(s)
- Francesco Pantano
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Michele Iuliani
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Alice Zoccoli
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Marco Fioramonti
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Delia De Lisi
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Iacopo Fioroni
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Giulia Ribelli
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Matteo Santoni
- b 2 Università Politecnica delle Marche, AOU Ospedali Riuniti, Medical Oncology Department , Ancona, Italy
| | - Bruno Vincenzi
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Giuseppe Tonini
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| | - Daniele Santini
- a 1 Campus Bio-Medico University of Rome, Medical Oncology Department , Via Alvaro del Portillo 200, 00128 Rome, Italy +39 062 254 191 17 ; +39 062 254 119 33;
| |
Collapse
|
94
|
Abstract
When the National Institutes of Health Mouse Models of Human Cancer Consortium initiated the Prostate Steering Committee 15 years ago, there were no genetically engineered mouse (GEM) models of prostate cancer (PCa). Today, a PubMed search for "prostate cancer mouse model" yields 3,200 publications and this list continues to grow. The first generation of GEM utilized the newly discovered and characterized probasin promoter driving viral oncogenes such as Simian virus 40 large T antigen to yield the LADY and TRAMP models. As the PCa research field has matured, the second generation of models has incorporated the single and multiple molecular changes observed in human disease, such as loss of PTEN and overexpression of Myc. Application of these models has revealed that mice are particularly resistant to developing invasive PCa, and once they achieve invasive disease, the PCa rarely resembles human disease. Nevertheless, these models and their application have provided vital information on human PCa progression. The aim of this review is to provide a brief primer on mouse and human prostate histology and pathology, provide descriptions of mouse models, as well as attempt to answer the age old question: Which GEM model of PCa is the best for my research question?
Collapse
|
95
|
Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, Anderson SA, McConeghy B, Shukin R, Bazov J, Youngren J, Paris P, Thomas G, Small EJ, Wang Y, Gleave ME, Collins CC, Chi KN. Androgen Receptor Gene Aberrations in Circulating Cell-Free DNA: Biomarkers of Therapeutic Resistance in Castration-Resistant Prostate Cancer. Clin Cancer Res 2015; 21:2315-24. [PMID: 25712683 DOI: 10.1158/1078-0432.ccr-14-2666] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/05/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE Although novel agents targeting the androgen-androgen receptor (AR) axis have altered the treatment paradigm of metastatic castration-resistant prostate cancer (mCRPC), development of therapeutic resistance is inevitable. In this study, we examined whether AR gene aberrations detectable in circulating cell-free DNA (cfDNA) are associated with resistance to abiraterone acetate and enzalutamide in mCRPC patients. EXPERIMENTAL DESIGN Plasma was collected from 62 mCRPC patients ceasing abiraterone acetate (n = 29), enzalutamide (n = 19), or other agents (n = 14) due to disease progression. DNA was extracted and subjected to array comparative genomic hybridization (aCGH) for chromosome copy number analysis, and Roche 454 targeted next-generation sequencing of exon 8 in the AR. RESULTS On aCGH, AR amplification was significantly more common in patients progressing on enzalutamide than on abiraterone or other agents (53% vs. 17% vs. 21%, P = 0.02, χ(2)). Missense AR exon 8 mutations were detected in 11 of 62 patients (18%), including the first reported case of an F876L mutation in an enzalutamide-resistant patient and H874Y and T877A mutations in 7 abiraterone-resistant patients. In patients switched onto enzalutamide after cfDNA collection (n = 39), an AR gene aberration (copy number increase and/or an exon 8 mutation) in pretreatment cfDNA was associated with adverse outcomes, including lower rates of PSA decline ≥ 30% (P = 0.013, χ(2)) and shorter time to radiographic/clinical progression (P = 0.010, Cox proportional hazards regression). CONCLUSIONS AR gene aberrations in cfDNA are associated with resistance to enzalutamide and abiraterone in mCRPC. Our data illustrate that genomic analysis of cfDNA is a minimally invasive method for interrogating mechanisms of therapeutic resistance in mCRPC.
Collapse
Affiliation(s)
- Arun A Azad
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | - Anne Haegert
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Robert H Bell
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Brian McConeghy
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Robert Shukin
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jenny Bazov
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jack Youngren
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | - Pamela Paris
- Department of Urology, UCSF, UCSF Diller Comprehensive Cancer Center, San Francisco, California
| | - George Thomas
- Department of Pathology and Laboratory Medicine, OHSU Knight Cancer Institute, Portland, Oregon
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. University of British Columbia, Vancouver, British Columbia, Canada.
| | - Kim N Chi
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
96
|
Alioui A, Celhay O, Baron S, Lobaccaro JMA. Lipids and prostate cancer adenocarcinoma. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/clp.14.51] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
97
|
Mitani Y, Rao PH, Maity SN, Lee YC, Ferrarotto R, Post JC, Licitra L, Lippman SM, Kies MS, Weber RS, Caulin C, Lin SH, El-Naggar AK. Alterations associated with androgen receptor gene activation in salivary duct carcinoma of both sexes: potential therapeutic ramifications. Clin Cancer Res 2014; 20:6570-81. [PMID: 25316813 DOI: 10.1158/1078-0432.ccr-14-1746] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the molecular events associated with the activation of androgen receptor (AR) as a potential therapeutic target in patients with salivary duct carcinoma (SDC). EXPERIMENTAL DESIGN Comprehensive molecular and expression analysis of the AR gene in 35 tumor specimens (20 males and 15 females) and cell lines derived from SDC using Western blotting and RT-PCR, FISH analysis, and DNA sequencing was conducted. In vitro and in vivo animal studies were also performed. RESULTS AR expression was detected in 70% of the tumors and was mainly nuclear and homogenous in both male and female SDCs, although variable cytoplasmic and/or nuclear localization was also found. We report the identification of ligand-independent AR splice variants, mutations, and extra AR gene copy in primary untreated SDC tumors. In contrast to prostate cancer, no AR gene amplification was observed. In vitro knockdown of AR in a female derived SDC cell line revealed marked growth inhibition in culture and in vivo androgen-independent tumor growth. CONCLUSIONS Our study provides new detailed information on the molecular and structural alterations associated with AR gene activation in SDC and sheds more light on the putative functional role of AR in SDC cells. On the basis of these data, we propose that patients with SDC (male and female) can be stratified for hormone-based therapy in future clinical trials.
Collapse
Affiliation(s)
- Yoshitsugu Mitani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pulivarthi H Rao
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Sankar N Maity
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julian C Post
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lisa Licitra
- Head and Neck Cancer Medical Oncology Unit, Department of Medical Oncology, Fondazione IRCCS "Istituto Nazionale dei Tumori," Milan, Italy
| | - Scott M Lippman
- Moores Cancer Center, University of California San Diego, San Diego, California
| | - Merrill S Kies
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Randal S Weber
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos Caulin
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adel K El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
98
|
Increased expression of CYP17A1 indicates an effective targeting of the androgen receptor axis in castration resistant prostate cancer (CRPC). SPRINGERPLUS 2014; 3:574. [PMID: 25332874 PMCID: PMC4193971 DOI: 10.1186/2193-1801-3-574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/26/2014] [Indexed: 01/18/2023]
Abstract
Recent breakthrough therapies targeting androgen receptor signalling in castration resistant prostate cancer (CRPC) involve multifunctional androgen receptor (AR) blockade and exhaustive androgen deprivation. Nevertheless, limitations to an enduring effectiveness of new drugs are anticipated in resistance mechanisms occurring under such treatments. In this study we used CRPC cell models VCaP and LNCaP as well as AR-negative PC-3- and non-neoplastic epithelial BPH-1-cells treated with 5, 10 or 25 μmol/L abiraterone hydrolyzed from abiraterone acetate (AA). The origin of CYP17A1 up-regulation under AA treatment was investigated in CRPC cell models by qRT-PCR and western-blot procedures. AA treatments of AR positive CRPC cell models led to decreased expression of androgen regulated genes such as PSA. In these cells diminished expression of androgen regulated genes was accompanied by an up-regulation of CYP17A1 expression within short-term treatments. No such effects became evident in AR-negative PC-3 cells. AR directed siRNA (siAR) used in VCaP cells significantly reduced mRNA expression and AR protein abundance. Such interference with AR signalling in the absence of abiraterone acetate also caused a marked up-regulation of CYP17A1 expression. Down-regulation of androgen regulated genes occurs in spite of an elevated expression of CYP17A1, the very target enzyme for this drug. CYP17A1 up-regulation already takes place within such short treatments with AA and does not require adaptation events over several cell cycles. CYP17A1 is also up-regulated in the absence of AA when AR signalling is physically eliminated by siAR. These results reveal an immediate counter-regulation of CYP17A1 expression whenever AR-signalling is inhibited adequately but not a persisting adaptation yielding drug resistance.
Collapse
|
99
|
Abstract
The majority of prostate cancers are hormone-dependent at diagnosis highlighting the central role of androgen signalling in this disease. Surprisingly, most forms of castration-resistant prostate cancer (CRPC) are still dependent on the androgen receptor (AR) for survival. Therefore, the advent of new AR-targeting drugs, such as enzalutamide, is certainly beneficial for the many patients with metastatic CRPC. Indeed, this compound provides a substantial survival benefit-but it is not curative. This Perspectives article describes the different ways through which cancer cells can become resistant to enzalutamide, such as AR truncation and other mutations, as well as by-pass of the AR dependence of prostate cancer cells through expression of the glucocorticoid receptor. The clinical relevance of these mechanisms and emerging questions concerning new therapeutic regimens in the treatment of metastatic CRPC are being discussed.
Collapse
|
100
|
Abstract
Prostate cancer is the commonest, non-cutaneous cancer in men. At present, there is no cure for the advanced, castration-resistant form of the disease. Estrogen has been shown to be important in prostate carcinogenesis, with evidence resulting from epidemiological, cancer cell line, human tissue and animal studies. The prostate expresses both estrogen receptor alpha (ERA) and estrogen receptor beta (ERB). Most evidence suggests that ERA mediates the harmful effects of estrogen in the prostate, whereas ERB is tumour suppressive, but trials of ERB-selective agents have not translated into improved clinical outcomes. The role of ERB in the prostate remains unclear and there is increasing evidence that isoforms of ERB may be oncogenic. Detailed study of ERB and ERB isoforms in the prostate is required to establish their cell-specific roles, in order to determine if therapies can be directed towards ERB-dependent pathways. In this review, we summarise evidence on the role of ERB in prostate cancer and highlight areas for future research.
Collapse
Affiliation(s)
- Adam W Nelson
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Wayne D Tilley
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - David E Neal
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| | - Jason S Carroll
- Cancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UKCancer Research UKCambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UKDepartment of UrologyAddenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UKDame Roma Mitchell Cancer Research LaboratoriesFaculty of Health Sciences, School of Medicine, The University of Adelaide, Level 4, Hanson Institute Building, DX Number 650 801, Adelaide, South Australia 5000, AustraliaDepartment of OncologyUniversity of Cambridge, Cambridge CB2 2QQ, UK
| |
Collapse
|