1
|
Zhang W, Zhu H, Chen Z, Li H, Chen X, Fan Y, Zhou X, Luo Y, Zhang Y, Tang F, Zhang X, Feng Y, Lu T, Wei X, Chen Y, Chen C, Jiao Y. Discovery of Novel Bifunctional Agents as Potent Androgen Receptor Antagonists and Degraders for the Treatment of Enzalutamide-Resistant Prostate Cancer. J Med Chem 2025; 68:8330-8345. [PMID: 40231807 DOI: 10.1021/acs.jmedchem.4c03043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Bifunctional agents that simultaneously antagonize and degrade various AR proteins more effectively block the AR signaling pathway, offering a promising strategy for the treatment of mCRPC patients. Herein, we report the discovery and development of a series of small-molecule AR degraders with 3,8-diazabicyclo[3.2.1]octan scaffold. The optimal compound 20i exhibited potent AR antagonistic and degrading activities, effectively overcoming multiple resistance mechanisms and showing significant antiproliferative effects against enzalutamide-resistant PCa cell lines. Moreover, compound 20i exhibited favorable oral pharmacokinetics and a good safety profile. In the 22Rv1 xenograft models, 20i exhibited potent antitumor activity without obvious toxicity. Taken together, these results demonstrated that 20i might be a potential candidate for the treatment of enzalutamide-resistant PCa.
Collapse
Affiliation(s)
- Wenqiang Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hao Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Zhuolin Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xingru Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yawen Fan
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xiaoyu Zhou
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yi Luo
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yan Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Feng Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Xinhao Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yunrui Feng
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Xian Wei
- Department of pharmacy, Youjiang Medical University for Nationalities, No. 98 ChengXiang Road, Youjiang, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Yadong Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Caiping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yu Jiao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
2
|
Zhang W, Zhou X, Zhu H, Fan Y, Chen Z, Wang C, Chen X, Li H, Lu T, Wei X, Chen Y, Chen C, Jiao Y. Discovery of novel spirocyclic derivates as potent androgen receptor antagonists. Bioorg Med Chem 2025; 120:118082. [PMID: 39892096 DOI: 10.1016/j.bmc.2025.118082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
We report herein the development of a series of novel AR antagonists characterized by a spirocyclic scaffold, employing scaffold hopping and structure-based drug design strategies. Most of the spirocyclic derivatives exhibited enhanced AR antagonistic activity and superior antiproliferative activity against LNCaP cells compared to enzalutamide. Among them, compound 21 demonstrated moderate antiproliferative activity against enzalutamide resistant prostate cancer cell lines and exhibited favorable in vitro metabolic stability. These findings offer valuable insights for the rational design of AR antagonists for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Wenqiang Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Xiaoyu Zhou
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Hao Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009 China
| | - Yawen Fan
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Zhuolin Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Chenxiao Wang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Xingru Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009 China
| | - Xian Wei
- Department of Pharmacy, Youjiang Medical University for Nationalities, No. 98 ChengXiang Road, Youjiang District, Baise 533000 Guangxi Zhuang Autonomous Region, China.
| | - Yadong Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China.
| | - Caiping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009 China.
| | - Yu Jiao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198 China.
| |
Collapse
|
3
|
Chan KC, Basavaraj P, Tsai JC, Viehoever J, Hsieh BY, Li XY, Huang GJ, Huang WC. Evaluating the Therapeutic Effect of Hispidin on Prostate Cancer Cells. Int J Mol Sci 2024; 25:7857. [PMID: 39063105 PMCID: PMC11277327 DOI: 10.3390/ijms25147857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Androgen deprivation therapy (ADT) is the primary treatment for advanced prostate cancer (PCa). However, prolonged ADT inevitably results in therapy resistance with the emergence of the castration-resistant PCa phenotype (CRPC). Hence, there is an urgent need to explore new treatment options capable of delaying PCa progression. Hispidin (HPD) is a natural polyketide primarily derived from plants and fungi. HPD has been shown to have a diverse pharmacological profile, exhibiting anti-inflammatory, antiviral, cardiovascular and neuro-protective activities. However, there is currently no research regarding its properties in the context of PCa treatment. This research article seeks to evaluate the anti-cancer effect of HPD and determine the underlying molecular basis in both androgen-sensitive PCa and CRPC cells. Cell growth, migration, and invasion assays were performed via the MTS method, a wound healing assay and the transwell method. To investigate if HPD affected the expression of proteins, Western blot analysis was conducted. Furthermore, apoptosis was assessed by Annexin V-FITC/PI staining and Western blot analyses. HPD exhibited a favorable pharmaceutical profile to inhibit cell growth; disrupt the cell cycle; attenuate wound healing, migration and invasion; and induce apoptosis in PCa cells in vitro. The mechanistic results demonstrated that HPD reduced AR, MMP-2 and MMP-9 expression and activated the caspase-related pathway, leading to programmed cell death in PCa cells. We showed the anti-cancer effect of HPD on PCa cells and confirmed its feasibility as a novel therapeutic agent. This study provides significant insights into the delineation of the molecular mechanism of HPD in PCa cells and the development of an effective and safe therapy using HPD to eliminate PCa progression.
Collapse
Affiliation(s)
- Kai-Cheng Chan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 40402, Taiwan; (K.-C.C.); (P.B.)
| | - Praveenkumar Basavaraj
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 40402, Taiwan; (K.-C.C.); (P.B.)
| | - Jui-Chen Tsai
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan; (J.-C.T.); (B.-Y.H.); (X.-Y.L.)
| | - Jonathan Viehoever
- International Master’s Program of Biomedical, School of Medicine, China Medical University, Taichung 40402, Taiwan;
| | - Bing-Yan Hsieh
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan; (J.-C.T.); (B.-Y.H.); (X.-Y.L.)
| | - Xin-Yu Li
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan; (J.-C.T.); (B.-Y.H.); (X.-Y.L.)
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Food Nutrition and Healthy Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Wen-Chin Huang
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 40402, Taiwan; (K.-C.C.); (P.B.)
- International Master’s Program of Biomedical, School of Medicine, China Medical University, Taichung 40402, Taiwan;
| |
Collapse
|
4
|
Zhang W, Fan Y, Zhang Y, Feng Y, Luo Y, Zhou X, Chen Z, Wang C, Lu T, Tang F, Chen Y, Li H, Jiao Y. Discovery of novel biphenyl derivatives as androgen receptor degraders for the treatment of enzalutamide-resistant prostate cancer. Bioorg Chem 2024; 148:107433. [PMID: 38754311 DOI: 10.1016/j.bioorg.2024.107433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
Second-generation AR antagonists, such as enzalutamide, are the primary therapeutic agents for advanced prostate cancer. However, the development of both primary and secondary drug resistance leads to treatment failures and patient mortality. Bifunctional agents that simultaneously antagonize and degrade AR block the AR signaling pathway more completely and exhibit excellent antiproliferative activity against wild-type and drug-resistant prostate cancer cells. Here, we reported the discovery and optimization of a series of biphenyl derivatives as androgen receptor antagonists and degraders. These biphenyl derivatives exhibited potent antiproliferative activity against LNCaP and 22Rv1 cells. Our discoveries enrich the diversity of small molecule AR degraders and offer insights for the development of novel AR degraders for the treatment of enzalutamide-resistant prostate cancer.
Collapse
Affiliation(s)
- Wenqiang Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yawen Fan
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yan Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China; State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, PR China; Jiangsu Simcere Pharmaceutical Co, Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, PR China
| | - Yunrui Feng
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Yi Luo
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Xiaoyu Zhou
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Zhuolin Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Chenxiao Wang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Feng Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, PR China; Jiangsu Simcere Pharmaceutical Co, Ltd, 699-18 Xuan Wu Avenue, Nanjing 210042, PR China.
| | - Yadong Chen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Hongmei Li
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Yu Jiao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| |
Collapse
|
5
|
Xiao M, Ha S, Zhu J, Tao W, Fu Z, Wei H, Hou Q, Luo G, Xiang H. Structure-Activity Relationship (SAR) Studies of Novel Monovalent AR/AR-V7 Dual Degraders with Potent Efficacy against Advanced Prostate Cancer. J Med Chem 2024; 67:5567-5590. [PMID: 38512060 DOI: 10.1021/acs.jmedchem.3c02177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Androgen receptor (AR) has been extensively established as a potential therapeutic target for nearly all stages of prostate cancer (PCa). However, acquired resistance to AR-targeted drugs inevitably develops and severely limits their clinical efficacy. Particularly, there currently exists no efficient treatment for patients expressing the constitutively active AR splice variants, such as AR-V7. Herein, we report the structure-activity relationship studies of 55 N-heterocycle-substituted hydantoins, which identified the structural motifs required for AR/AR-V7 degradation. Among them, the most potent compound 27c exhibited selective AR/AR-V7 degradation over other hormone receptors and excellent antiproliferative activities in LNCaP and 22RV1 cells. RNA sequence analysis confirmed that 27c effectively suppressed transcriptional activity of the AR signaling pathway. Importantly, 27c demonstrated potent antitumor efficacy in an enzalutamide-resistant 22RV1 xenograft model. These results highlight the potential of 27c as a promising dual AR/AR-V7 degrader for overcoming drug resistance in advanced PCa expressing AR splice variants.
Collapse
Affiliation(s)
- Maoxu Xiao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Si Ha
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiacheng Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wenxiang Tao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zixuan Fu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hanlin Wei
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qiangqiang Hou
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Guoshun Luo
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
6
|
Structures of Mammeasins P and Q, Coumarin-Related Polysubstituted Benzofurans, from the Thai Medicinal Plant Mammea siamensis (Miq.) T. Anders.: Anti-Proliferative Activity of Coumarin Constituents against Human Prostate Carcinoma Cell Line LNCaP. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A methanol extract of the flowers of Mammea siamensis (Miq.) T. Anders. (Calophyllaceae) showed anti-proliferative activity against human prostate carcinoma LNCaP cells (IC50 = 2.0 µg/mL). Two new coumarin-related polysubstituted benzofurans, mammeasins P (1) and Q (2), and a known polysubstituted coumarin mammea B/AC cyclo F (39) were isolated from the extract along with 44 previously reported polysubstituted coumarin constituents (3–38 and 40–47). The structures of two new compounds (1 and 2) were determined based on their spectroscopic properties derived from the physicochemical evidence including NMR and MS analyses and taking the plausible generative pathway into account. Among the coumarin constituents, mammeasins A (3, IC50 = 1.2 µM) and B (4, 0.63 µM), sugangin B (18, 1.5 µM), kayeassamins E (24, 3.0 µM) and G (26, 3.5 µM), and mammeas E/BA (40, 0.88 µM), E/BB (41, 0.52 µM), and E/BC (42, 0.12 µM) showed relatively potent anti-proliferative activity.
Collapse
|
7
|
Dahiya V, Bagchi G. Non-canonical androgen signaling pathways and implications in prostate cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119357. [PMID: 36100060 DOI: 10.1016/j.bbamcr.2022.119357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Androgen signaling is a critical determinant of timely and proper development of all male organs including the prostate. Maturation of prostate and its neoplastic transformation is intricately associated with accurate androgen signaling. Ablation of androgen has therefore been the primary treatment mechanism of Prostate cancer (PCa) patients for several decades. Upon removal, the tumor recedes for a while, yet it reappears soon, in an androgen independent state, untreatable by current therapeutic regimens. Studies reveal that apart from the classical androgen signaling pathway known and targeted for almost a century, there exist several non-canonical pathways, with marked impact on classical androgen signaling and PCa growth. These include non-genomic signaling by androgens via alternate membrane GPCRs, signaling by non-androgens that ultimately impact the androgen signaling pathway, or an integration of non-genomic and genomic response as seen in case of protein kinase A activation. Accurate understanding of these various non-canonical androgen signaling pathways and their influence on the typical androgen signaling pathway can help design important interventions for PCa patients. This review analyses in detail the various non-classical androgen signaling pathways and their impact, if any, on classical mode of androgen action and PCa.
Collapse
Affiliation(s)
- Versha Dahiya
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413
| | - Gargi Bagchi
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413.
| |
Collapse
|
8
|
Gerald T, Raj G. Testosterone and the Androgen Receptor. Urol Clin North Am 2022; 49:603-614. [DOI: 10.1016/j.ucl.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
9
|
Wang A, Luo X, Wang Y, Meng X, Lu Z, Yang Y. Design, Synthesis, and Biological Evaluation of Androgen Receptor Degrading and Antagonizing Bifunctional Steroidal Analogs for the Treatment of Advanced Prostate Cancer. J Med Chem 2022; 65:12460-12481. [PMID: 36070471 DOI: 10.1021/acs.jmedchem.2c01164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) with high mortality has seriously threatened men's health. Bifunctional agents simultaneously degrade and antagonize androgen receptor (AR), display robust AR signaling pathway blockade, and show the therapeutic prospect for mCRPC. Herein, systemic structural modifications on the C-3, C-6, and C-17 positions of galeterone led to the discovery of 67-b with the dual functions of AR antagonism and degradation. In vitro, 67-b exhibited excellent antiproliferative activity and potent AR degradation activity in different PCa cells (LNCaP and 22RV1), as well as outstanding antagonistic activity against wild-type and mutant (W741L, T877A, and F876L) ARs. In vivo, 67-b effectively inhibited the growth of hormone-sensitive organs in the Hershberger assay and exhibited tumor regression in the enzalutamide-resistant (c4-2b-ENZ) xenograft model. These results confirmed 67-b to be a promising AR degrader and antagonist for the treatment of mCRPC patients.
Collapse
Affiliation(s)
- Ao Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xianggang Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yawan Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Xin Meng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhengyu Lu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,School of Pharmacy, University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
10
|
Advances in the Current Understanding of the Mechanisms Governing the Acquisition of Castration-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14153744. [PMID: 35954408 PMCID: PMC9367587 DOI: 10.3390/cancers14153744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Despite aggressive treatment and androgen-deprivation therapy, most prostate cancer patients ultimately develop castration-resistant prostate cancer (CRPC), which is associated with high mortality rates. However, the mechanisms governing the development of CRPC are poorly understood, and androgen receptor (AR) signaling has been shown to be important in CRPC through AR gene mutations, gene overexpression, co-regulatory factors, AR shear variants, and androgen resynthesis. A growing number of non-AR pathways have also been shown to influence the CRPC progression, including the Wnt and Hh pathways. Moreover, non-coding RNAs have been identified as important regulators of the CRPC pathogenesis. The present review provides an overview of the relevant literature pertaining to the mechanisms governing the molecular acquisition of castration resistance in prostate cancer, providing a foundation for future, targeted therapeutic efforts.
Collapse
|
11
|
Chopra H, Bibi S, Goyal R, Gautam RK, Trivedi R, Upadhyay TK, Mujahid MH, Shah MA, Haris M, Khot KB, Gopan G, Singh I, Kim JK, Jose J, Abdel-Daim MM, Alhumaydhi FA, Emran TB, Kim B. Chemopreventive Potential of Dietary Nanonutraceuticals for Prostate Cancer: An Extensive Review. Front Oncol 2022; 12:925379. [PMID: 35903701 PMCID: PMC9315356 DOI: 10.3389/fonc.2022.925379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
There are more than two hundred fifty different types of cancers, that are diagnosed around the world. Prostate cancer is one of the suspicious type of cancer spreading very fast around the world, it is reported that in 2018, 29430 patients died of prostate cancer in the United State of America (USA), and hence it is expected that one out of nine men diagnosed with this severe disease during their lives. Medical science has identified cancer at several stages and indicated genes mutations involved in the cancer cell progressions. Genetic implications have been studied extensively in cancer cell growth. So most efficacious drug for prostate cancer is highly required just like other severe diseases for men. So nutraceutical companies are playing major role to manage cancer disease by the recommendation of best natural products around the world, most of these natural products are isolated from plant and mushrooms because they contain several chemoprotective agents, which could reduce the chances of development of cancer and protect the cells for further progression. Some nutraceutical supplements might activate the cytotoxic chemotherapeutic effects by the mechanism of cell cycle arrest, cell differentiation procedures and changes in the redox states, but in other, it also elevate the levels of effectiveness of chemotherapeutic mechanism and in results, cancer cell becomes less reactive to chemotherapy. In this review, we have highlighted the prostate cancer and importance of nutraceuticals for the control and management of prostate cancer, and the significance of nutraceuticals to cancer patients during chemotherapy.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-milat University, Islamabad, Pakistan
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, China
| | - Rajat Goyal
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
- Maharishi Markandeshwar (MM) College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Rupesh K. Gautam
- Maharishi Markandeshwar (MM) School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, India
| | - Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | - Mohd Hasan Mujahid
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara, India
| | | | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kartik Bhairu Khot
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Gopika Gopan
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Jin Kyu Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jobin Jose
- Department of Pharmaceutics, NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Mangalore, India
| | - Mohamed M. Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
12
|
Yin S, Chen Y, Tong H, Li T, Qin Z, Zhu J, He W. PP2A promotes apoptosis and facilitates docetaxel sensitivity via the PP2A/p‑eIF4B/XIAP signaling pathway in prostate cancer. Oncol Lett 2022; 23:101. [PMID: 35154432 PMCID: PMC8822497 DOI: 10.3892/ol.2022.13221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Serine/threonine protein phosphatase 2A (PP2A) is a protein that has a wide range of biological functions. As prostate cancer progresses from hormone-sensitive prostate cancer to castration-resistant prostate cancer (CRPC), the expression level of PP2A has been found to decrease. The present study aimed to determine the roles that PP2A may play in prostate cancer and its association with the downstream factor, X-linked inhibitor of apoptosis (XIAP). First, the mRNA and protein expression levels of PP2A in LNCaP, DU145 and PC-3 prostate cancer cell lines were measured. Next, the population of PP2A heterodimers was increased using a PP2A agonist, DT061, in the DU145 and PC-3 cell lines. PP2A expression was then knocked down in the LNCaP cell line. Western blot analysis was performed to determine the association between PP2A, phosphorylated (p)-eukaryotic initiation factor 4B (eIF4B) and XIAP. The results revealed that following the increase in PP2A expression, the DU145 and PC-3 cell lines were more sensitive to docetaxel according to Cell Counting Kit-8 assays and had an increased apoptotic rate as assessed by flow cytometry. Conversely, following the transfection of small interfering (si)PP2A into the LNCaP cell line, the sensitivity to docetaxel decreased, as well as the apoptotic rate. In addition, following treatment with the PP2A agonist, DT061, PP2A expression was found to be significantly upregulated, while p-eIF4B and XIAP protein expression levels were significantly downregulated. By contrast, following the transfection of siPP2A into the LNCaP cell line, PP2A protein expression levels were found to be downregulated, while p-eIF4B and XIAP expression levels were significantly upregulated. In conclusion, by affecting the downstream factor XIAP, PP2A may play a key role in promoting apoptosis and facilitating docetaxel sensitivity in prostate cancer cell lines.
Collapse
Affiliation(s)
- Siwen Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yong Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hang Tong
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Tinghao Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zijia Qin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Junlong Zhu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
13
|
Mondal D, Narwani D, Notta S, Ghaffar D, Mardhekar N, Quadri SSA. Oxidative stress and redox signaling in CRPC progression: therapeutic potential of clinically-tested Nrf2-activators. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:96-124. [PMID: 35582006 PMCID: PMC9019181 DOI: 10.20517/cdr.2020.71] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Androgen deprivation therapy (ADT) is the mainstay regimen in patients with androgen-dependent prostate cancer (PCa). However, the selection of androgen-independent cancer cells leads to castrate resistant prostate cancer (CRPC). The aggressive phenotype of CRPC cells underscores the need to elucidate mechanisms and therapeutic strategies to suppress CRPC outgrowth. Despite ADT, the activation of androgen receptor (AR) transcription factor continues via crosstalk with parallel signaling pathways. Understanding of how these signaling cascades are initiated and amplified post-ADT is lacking. Hormone deprivation can increase oxidative stress and the resultant reactive oxygen species (ROS) may activate both AR and non-AR signaling. Moreover, ROS-induced inflammatory cytokines may further amplify these redox signaling pathways to augment AR function. However, clinical trials using ROS quenching small molecule antioxidants have not suppressed CRPC progression, suggesting that more potent and persistent suppression of redox signaling in CRPC cells will be needed. The transcription factor Nrf2 increases the expression of numerous antioxidant enzymes and downregulates the function of inflammatory transcription factors, e.g., nuclear factor kappa B. We documented that Nrf2 overexpression can suppress AR-mediated transcription in CRPC cell lines. Furthermore, two Nrf2 activating agents, sulforaphane (a phytochemical) and bardoxolone-methyl (a drug in clinical trial) suppress AR levels and sensitize CRPC cells to anti-androgens. These observations implicate the benefits of potent Nrf2-activators to suppress the lethal signaling cascades that lead to CRPC outgrowth. This review article will address the redox signaling networks that augment AR signaling during PCa progression to CRPC, and the possible utility of Nrf2-activating agents as an adjunct to ADT.
Collapse
Affiliation(s)
- Debasis Mondal
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| | - Devin Narwani
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| | - Shahnawaz Notta
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| | - Dawood Ghaffar
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| | - Nikhil Mardhekar
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| | - Syed S A Quadri
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37932, USA
| |
Collapse
|
14
|
Pan W, Zhang Z, Kimball H, Qu F, Berlind K, Stopsack KH, Lee GSM, Choueiri TK, Kantoff PW. Abiraterone Acetate Induces CREB1 Phosphorylation and Enhances the Function of the CBP-p300 Complex, Leading to Resistance in Prostate Cancer Cells. Clin Cancer Res 2021; 27:2087-2099. [PMID: 33495313 DOI: 10.1158/1078-0432.ccr-20-4391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/22/2020] [Accepted: 01/19/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Abiraterone acetate (AA), an inhibitor of cytochrome P450 17alpha-hydroxylase/17, 20 lyase, is an FDA-approved drug for advanced prostate cancer. However, not all patients respond to AA, and AA resistance ultimately develops in patients who initially respond. We aimed to identify AA resistance mechanisms in prostate cancer cells. EXPERIMENTAL DESIGN We established several AA-resistant cell lines and performed a comprehensive study on mechanisms involved in AA resistance development. RNA sequencing and phospho-kinase array screenings were performed to discover that the cAMP-response element CRE binding protein 1 (CREB1) was a critical molecule in AA resistance development. RESULTS The drug-resistant cell lines are phenotypically stable without drug selection, and exhibit permanent global gene expression changes. The phosphorylated CREB1 (pCREB1) is increased in AA-resistant cell lines and is critical in controlling global gene expression. Upregulation of pCREB1 desensitized prostate cancer cells to AA, while blocking CREB1 phosphorylation resensitized AA-resistant cells to AA. AA treatment increases intracellular cyclic AMP (cAMP) levels, induces kinases activity, and leads to the phosphorylation of CREB1, which may subsequently augment the essential role of the CBP/p300 complex in AA-resistant cells because AA-resistant cells exhibit a relatively higher sensitivity to CBP/p300 inhibitors. Further pharmacokinetics studies demonstrated that AA significantly synergizes with CBP/p300 inhibitors in limiting the growth of prostate cancer cells. CONCLUSIONS Our studies suggest that AA treatment upregulates pCREB1, which enhances CBP/p300 activity, leading to global gene expression alterations, subsequently resulting in drug resistance development. Combining AA with therapies targeting resistance mechanisms may provide a more effective treatment strategy.
Collapse
Affiliation(s)
- Wenting Pan
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhouwei Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hannah Kimball
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Fangfang Qu
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kyler Berlind
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Konrad H Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gwo-Shu Mary Lee
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
15
|
Fancher AT, Hua Y, Strock CJ, Johnston PA. Assays to Interrogate the Ability of Compounds to Inhibit the AF-2 or AF-1 Transactivation Domains of the Androgen Receptor. Assay Drug Dev Technol 2019; 17:364-386. [PMID: 31502857 DOI: 10.1089/adt.2019.940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the leading cause of cancer and second leading cause of cancer-related death in men in the United States. Twenty percent of patients receiving the standard of care androgen deprivation therapy (ADT) eventually progress to metastatic and incurable castration-resistant prostate cancer (CRPC). Current FDA-approved drugs for CRPC target androgen receptor (AR) binding or androgen production, but only provide a 2- to 5-month survival benefit due to the emergence of resistance. Overexpression of AR coactivators and the emergence of AR splice variants, both promote continued transcriptional activation under androgen-depleted conditions and represent drug resistance mechanisms that contribute to CRPC progression. The AR contains two transactivation domains, activation function 2 (AF-2) and activation function 1 (AF-1), which serve as binding surfaces for coactivators involved in the transcriptional activation of AR target genes. Full-length AR contains both AF-2 and AF-1 surfaces, whereas AR splice variants only have an AF-1 surface. We have recently prosecuted a high-content screening campaign to identify hit compounds that can inhibit or disrupt the protein-protein interactions (PPIs) between AR and transcriptional intermediary factor 2 (TIF2), one of the coactivators implicated in CRPC disease progression. Since an ideal inhibitor/disruptor of AR-coactivator PPIs would target both the AF-2 and AF-1 surfaces, we describe here the development and validation of five AF-2- and three AF-1-focused assays to interrogate and prioritize hits that disrupt both transactivation surfaces. The assays were validated using a test set of seven known AR modulator compounds, including three AR antagonists and one androgen synthesis inhibitor that are FDA-approved ADTs, two investigational molecules that target the N-terminal domain of AR, and an inhibitor of the Hsp90 (heat shock protein) molecular chaperone.
Collapse
Affiliation(s)
- Ashley T Fancher
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yun Hua
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,Head and Neck Cancer, and Skin Cancer Specialized Programs of Research Excellence, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Iwamoto H, Izumi K, Natsagdorj A, Naito R, Makino T, Kadomoto S, Hiratsuka K, Shigehara K, Kadono Y, Narimoto K, Saito Y, Nakagawa-Goto K, Mizokami A. Coffee diterpenes kahweol acetate and cafestol synergistically inhibit the proliferation and migration of prostate cancer cells. Prostate 2019; 79:468-479. [PMID: 30569541 DOI: 10.1002/pros.23753] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 11/27/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Coffee inhibits the progression of prostate cancer; however, the direct mechanism through which coffee acts on prostate cancer cells remains unclear. This study aimed to identify the key compounds of coffee that possess anti-cancer effects and to investigate their mechanisms of action. METHODS The anti-proliferation and anti-migration effects of six potentially active types of coffee compounds, including kahweol acetate, cafestol, caffeine, caffeic acid, chlorogenic acid, and trigonelline hydrochloride, were evaluated using LNCaP, LNCaP-SF, PC-3, and DU145 human prostate cancer cells. The synergistic effects of these compounds were also investigated. Apoptosis-related and epithelial-mesenchymal transition-related proteins, androgen receptor in whole cell and in nucleus, and chemokines were assessed. A xenograft study of SCID mice was performed to examine the in vivo effect of coffee compounds. RESULTS Among the evaluated compounds, only kahweol acetate and cafestol inhibited the proliferation and migration of prostate cancer cells in a dose-dependent manner. The combination treatment involving kahweol acetate and cafestol synergistically inhibited proliferation and migration (combination index <1) with the induction of apoptosis, the inhibition of epithelial-mesenchymal transition, and decrease in androgen receptor, resulting in the reduction of nuclear androgen receptor in androgen receptor-positive cells. Moreover, kahweol acetate and cafestol downregulated CCR2 and CCR5 without an increase in their ligands, CCL2 and CCL5. The xenograft study showed that oral administration of kahweol acetate and cafestol significantly inhibited tumor growth. CONCLUSION Kahweol acetate and cafestol synergistically inhibit the progression of prostate cancer. These coffee compounds may be novel therapeutic candidates for prostate cancer.
Collapse
Affiliation(s)
- Hiroaki Iwamoto
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kouji Izumi
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Ariunbold Natsagdorj
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Renato Naito
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Tomoyuki Makino
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Suguru Kadomoto
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kaoru Hiratsuka
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazuyoshi Shigehara
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Yoshifumi Kadono
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazutaka Narimoto
- Department of Urology, St. Luke's International Hospital, Tokyo, Japan
| | - Yohei Saito
- School of Pharmaceutical Sciences, College of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Kyoko Nakagawa-Goto
- School of Pharmaceutical Sciences, College of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
17
|
Xie W, Stopsack KH, Drouin SJ, Fu H, Pomerantz MM, Mucci LA, Lee GSM, Kantoff PW. Association of genetic variation of the six gene prognostic model for castration-resistant prostate cancer with survival. Prostate 2019; 79:73-80. [PMID: 30141208 PMCID: PMC6476182 DOI: 10.1002/pros.23712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/08/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND We previously identified a blood RNA transcript-based model consisting of six immune or inflammatory response genes (ABL2, SEMA4D, ITGAL, C1QA, TIMP1, and CDKN1A) that was prognostic for survival in cohorts of men with castration-resistant prostate cancer (CRPC). We investigated whether inherited variation in these six genes was associated with overall survival (OS) in men with CRPC. METHODS The test cohort comprised 600 patients diagnosed with CRPC between 1996 and 2011 at Dana-Farber Cancer Institute. Genotyping of 66 tagging single nucleotide polymorphisms (SNPs) spanning the six genes was performed on blood derived DNAs. For the top four SNPs (P < 0.05), validation was conducted in an independent cohort of 223 men diagnosed with CRPC between 2000 and 2014. Multivariable Cox regression adjusting for known prognostic factors estimated hazard ratios (HR) and 95% confidence intervals (CI) of the association of genetic variants with OS. RESULTS Two thirds of patients in both cohorts had metastases at CRPC diagnosis. Median OS from CRPC diagnosis was 3.6 (95%CI 3.3-4.0) years in the test cohort and 4.6 (95%CI 3.8-5.2) years in the validation cohort. Fifty-nine SNPs in Hardy-Weinberg equilibrium were analyzed. The major alleles of rs1318056 and rs1490311 in ABL2, and the minor alleles of rs2073917 and rs3764322 in ITGAL were associated with increased risk of death in the test cohort (adjusted-HRs 1.27-1.39; adjusted-p <0.05; false discovery rate <0.35). In the validation cohort, a similar association with OS was observed for rs1318056 in ABL2 (adjusted-HR 1.44; 95%CI 0.89-2.34) and rs2073917 in ITGAL (adjusted-HR 1.41; 95%CI 0.82-2.42). The associations did not reach statistical significance most likely due to the small sample size of the validation cohort (adjusted-p = 0.142 and 0.209, respectively). Additional eQTL analysis indicated that minor alleles of rs1318056 and rs1490311 in ABL2 are associated with a lower ABL2 expression in blood. CONCLUSIONS These findings corroborate our initial work on the RNA expression of genes involved in immunity and inflammation from blood and clinical outcome and suggest that germline polymorphisms in ABL2 and ITGAL may be associated with the risk of death in men with CRPC. Further studies are needed to validate these findings and to explore their functional mechanisms.
Collapse
Affiliation(s)
- Wanling Xie
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Konrad H. Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Sarah J Drouin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Henry Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Mark M. Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Lorelei A. Mucci
- Harvard T. H Chan Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02215
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
- Correspondence: Philip W. Kantoff, Phone: 212-639-5851; Fax: 929-321-5023; . Gwo-Shu Mary Lee, Phone: 617-632-5088;
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Correspondence: Philip W. Kantoff, Phone: 212-639-5851; Fax: 929-321-5023; . Gwo-Shu Mary Lee, Phone: 617-632-5088;
| |
Collapse
|
18
|
Ma MW, Gao XS, Yu HL, Qi X, Sun SQ, Wang D. Cordyceps sinensis Promotes the Growth of Prostate Cancer Cells. Nutr Cancer 2018; 70:1166-1172. [PMID: 30273008 DOI: 10.1080/01635581.2018.1504091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Ming-wei Ma
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xian-shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Hong-liang Yu
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, The Affiliate Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xin Qi
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Shao-qian Sun
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Dian Wang
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
19
|
Lian X, Gu J, Gao B, Li Y, Damodaran C, Wei W, Fu Y, Cai L. Fenofibrate inhibits mTOR-p70S6K signaling and simultaneously induces cell death in human prostate cancer cells. Biochem Biophys Res Commun 2018; 496:70-75. [PMID: 29305864 DOI: 10.1016/j.bbrc.2017.12.168] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/31/2017] [Indexed: 02/07/2023]
Abstract
Fenofibrate is the most widely used lipid-lowering drug, but it seems to have anti-tumor effects in several tumor cell lines. However, there are only a few reports on its effects on human prostate cancer cells. Thus, we investigated the anti-proliferative effects of fenofibrate on human prostate cancer cells and potential mechanisms. The methods used include cell viability analysis with an MTT assay, as well as apoptosis and related signaling pathway analyses with flow cytometry and Western blotting. Fenofibrate inhibited PC-3 cell growth in dose- and time-dependent manners. The fenofibrate-induced cell death is predominantly apoptotic death that is mediated by both the caspase-3 activation and apoptosis-inducing factor (AIF) signaling pathways. Fenofibrate also increased the expression of Bad and decreased the expression of Bcl-2 and Survivin. Mechanistically, fenofibrate-induced cell death was associated with decreased p-p70S6K and the mammalian target of rapamycin (mTOR) phosphorylation levels. When further exploring the upstream mediators of mTOR/p70S6K, we found that fenofibrate increased p38 MAPK and AMPK phosphorylation but did not significantly change the phosphorylation levels of PI3K, AKT, and JNK. However, the inhibition of either p38 MAPK or AMPK with their specific inhibitor did not change the effect of fenofibrate-induced cell death. These findings suggested that fenofibrate indeed significantly inhibited the proliferation of PC-3 cells via apoptotic action, which is associated with the inactivation of the mTOR/p70S6K-dependent cell survival pathway. Although the mechanisms by which fenofibrate inactivates this pathway remains unclear, this study reveals great potential for its use for the clinical treatment of prostate cancers.
Collapse
Affiliation(s)
- Xin Lian
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China; Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Junlian Gu
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Baoshan Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yan Li
- Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Wei Wei
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yaowen Fu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA; Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
20
|
Chopra H, Khan Z, Contreras J, Wang H, Sedrak A, Zhu Y. Activation of p53 and destabilization of androgen receptor by combinatorial inhibition of MDM2 and MDMX in prostate cancer cells. Oncotarget 2017; 9:6270-6281. [PMID: 29464071 PMCID: PMC5814211 DOI: 10.18632/oncotarget.23569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 10/13/2017] [Indexed: 01/22/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) frequently develops after initial standard radiation and androgen deprivation therapy, leaving patients with limited further treatment options. Androgen receptor (AR) is a transcription factor that plays a key role in the initiation and progression of prostate cancer. p53, a major tumor suppressor that is rarely mutated in early-stages of prostate cancer, is often deregulated during prostate cancer progression. Here, we report an unusual co-amplification of MDM2 and MDMX, two crucial negative regulators of p53, in CRPC datasets. We demonstrate that combinatorial inhibition of MDM2 and MDMX, with nutlin-3 and NSC207895 respectively, has a profound inhibitory effect on cell proliferation of androgen-responsive, wild-type TP53 gene carrying prostate cancer cells LNCaP and 22Rv1. We further show that the combinatorial inhibition of MDM2 and MDMX not only activates p53, but also decreases cellular levels of AR and represses its function. Additionally, co-expression of MDM2 and MDMX stabilizes AR. Together, our results indicate that combinatorial inhibition of MDM2 and MDMX may offer a novel compelling strategy for prostate cancer therapy.
Collapse
Affiliation(s)
- Harman Chopra
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Zara Khan
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Jamie Contreras
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Herui Wang
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Abanob Sedrak
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Yan Zhu
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
21
|
Stossi F, Dandekar RD, Bolt MJ, Newberg JY, Mancini MG, Kaushik AK, Putluri V, Sreekumar A, Mancini MA. High throughput microscopy identifies bisphenol AP, a bisphenol A analog, as a novel AR down-regulator. Oncotarget 2017; 7:16962-74. [PMID: 26918604 PMCID: PMC4941363 DOI: 10.18632/oncotarget.7655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 01/17/2016] [Indexed: 01/12/2023] Open
Abstract
Prostate cancer remains a deadly disease especially when patients become resistant to drugs that target the Androgen Receptor (AR) ligand binding domain. At this stage, patients develop recurring castrate-resistant prostate cancers (CRPCs). Interestingly, CRPC tumors maintain dependency on AR for growth; moreover, in CRPCs, constitutively active AR splice variants (e.g., AR-V7) begin to be expressed at higher levels. These splice variants lack the ligand binding domain and are rendered insensitive to current endocrine therapies. Thus, it is of paramount importance to understand what regulates the expression of AR and its splice variants to identify new therapeutic strategies in CRPCs. Here, we used high throughput microscopy and quantitative image analysis to evaluate effects of selected endocrine disruptors on AR levels in multiple breast and prostate cancer cell lines. Bisphenol AP (BPAP), which is used in chemical and medical industries, was identified as a down-regulator of both full length AR and the AR-V7 splice variant. We validated its activity by performing time-course, dose-response, Western blot and qPCR analyses. BPAP also reduced the percent of cells in S phase, which was accompanied by a ~60% loss in cell numbers and colony formation in anchorage-independent growth assays. Moreover, it affected mitochondria size and cell metabolism. In conclusion, our high content analysis-based screening platform was used to classify the effect of compounds on endogenous ARs, and identified BPAP as being capable of causing AR (both full-length and variants) down-regulation, cell cycle arrest and metabolic alterations in CRPC cell lines.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Radhika D Dandekar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael J Bolt
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justin Y Newberg
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maureen G Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Akash K Kaushik
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vasanta Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Arun Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Chakraborty S, Kumar A, Butt NA, Zhang L, Williams R, Rimando AM, Biswas PK, Levenson AS. Molecular insight into the differential anti-androgenic activity of resveratrol and its natural analogs: in silico approach to understand biological actions. MOLECULAR BIOSYSTEMS 2017; 12:1702-9. [PMID: 27063447 DOI: 10.1039/c6mb00186f] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The androgen receptor (AR) is a therapeutic target for the treatment of prostate cancer. Androgen receptor reactivation during the androgen-independent stage of prostate cancer is mediated by numerous mechanisms including expression of AR mutants and splice variants that become non-responsive to conventional anti-androgenic agents. Resveratrol and its natural analogs exhibit varying degrees of anti-androgenic effects on tumor growth suppression in prostate cancer. However, the structural basis for the observed differential activity remains unknown. Here, anti-androgenic activities of resveratrol and its natural analogs, namely, pterostilbene, piceatannol and trimethoxy-resveratrol were studied in LNCaP cells expressing T877A mutant AR and atomistic simulations were employed to establish the structure activity relationship. Interestingly, essential hydrogen bonding contacts and the binding energies of resveratrol analogs with AR ligand binding domain (LBD), emerge as key differentiating factors for varying anti-androgenic action. Among all the analogs, pterostilbene exhibited strongest anti-androgenic activity and its binding energy and hydrogen bonding interactions pattern closely resembled pure anti-androgen, flutamide. Principal component analysis of our simulation studies revealed that androgenic compounds bind more strongly to AR LBD compared to anti-androgenic compounds and provide conformational stabilization of the receptor in essential subspace. The present study provides critical insight into the structure-activity relationship of the anti-androgenic action of resveratrol analogs, which can be translated further to design novel highly potent anti-androgenic stilbenes.
Collapse
Affiliation(s)
- Sandipan Chakraborty
- Laboratory of Computational Biophysics & Bioengineering, Department of Physics, Tougaloo College, Tougaloo, MS 39174, USA. and Department of Physical Chemistry, Indian Association for the Cultivation of Science, Kolkata, India
| | - Avinash Kumar
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39202, USA
| | - Nasir A Butt
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39202, USA and Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39202, USA
| | - Liangfen Zhang
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39202, USA
| | - Raquema Williams
- Laboratory of Computational Biophysics & Bioengineering, Department of Physics, Tougaloo College, Tougaloo, MS 39174, USA.
| | - Agnes M Rimando
- United States Department of Agriculture, Agriculture Research Service, Natural Products Utilization Research Unit, University, MS 38677, USA
| | - Pradip K Biswas
- Laboratory of Computational Biophysics & Bioengineering, Department of Physics, Tougaloo College, Tougaloo, MS 39174, USA.
| | - Anait S Levenson
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39202, USA and Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39202, USA
| |
Collapse
|
23
|
Baciarello G, Sternberg CN. Treatment of metastatic castration-resistant prostate cancer (mCRPC) with enzalutamide. Crit Rev Oncol Hematol 2016; 106:14-24. [PMID: 27637350 DOI: 10.1016/j.critrevonc.2016.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/23/2016] [Accepted: 07/12/2016] [Indexed: 01/15/2023] Open
Abstract
Prostate cancer is initially responsive to androgen deprivation therapy, but most patients eventually develop castration-resistant disease. Enzalutamide is an androgen receptor (AR) inhibitor that targets several steps in the AR signaling pathway and has shown significant efficacy in the treatment of metastatic castration-resistant prostate cancer in patients with or without prior chemotherapy. To provide optimal treatment, it is important to understand the implications of enzalutamide use in the context of other therapies, as recent findings have suggested cross-resistance occurs between and within drug classes. Mutations and splice variants of AR also impact the course of prostate cancer. Future strategies involving enzalutamide should account for previous exposure to taxanes or antiandrogen therapies and the presence of AR variants that could affect efficacy.
Collapse
Affiliation(s)
- Giulia Baciarello
- Department of Medical Oncology, San Camillo and Forlanini Hospitals, Padiglione Flajani, Circonvallazione Gianicolense 87, Rome, 00152, Italy.
| | - Cora N Sternberg
- Department of Medical Oncology, San Camillo and Forlanini Hospitals, Padiglione Flajani, Circonvallazione Gianicolense 87, Rome, 00152, Italy.
| |
Collapse
|
24
|
Purushottamachar P, Kwegyir-Afful AK, Martin M, Ramamurthy V, Ramalingam S, Njar VCO. Identification of Novel Steroidal Androgen Receptor Degrading Agents Inspired by Galeterone 3β-Imidazole Carbamate. ACS Med Chem Lett 2016; 7:708-13. [PMID: 27437082 PMCID: PMC4948004 DOI: 10.1021/acsmedchemlett.6b00137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/23/2016] [Indexed: 01/03/2023] Open
Abstract
Degradation of all forms of androgen receptors (ARs) is emerging as an advantageous therapeutic paradigm for the effective treatment of prostate cancer. In continuation of our program to identify and develop improved efficacious novel small-molecule agents designed to disrupt AR signaling through enhanced AR degradation, we have designed, synthesized, and evaluated novel C-3 modified analogues of our phase 3 clinical agent, galeterone (5). Concerns of potential in vivo stability of our recently discovered more efficacious galeterone 3β-imidazole carbamate (6) led to the design and synthesis of new steroidal compounds. Two of the 11 compounds, 3β-pyridyl ether (8) and 3β-imidazole (17) with antiproliferative GI50 values of 3.24 and 2.54 μM against CWR22Rv1 prostate cancer cell, are 2.75- and 3.5-fold superior to 5. In addition, compounds 8 and 17 possess improved (∼4-fold) AR-V7 degrading activities. Importantly, these two compounds are expected to be metabolically stable, making them suitable for further development as new therapeutics against all forms of prostate cancer.
Collapse
Affiliation(s)
- Puranik Purushottamachar
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Andrew K. Kwegyir-Afful
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Marlena
S. Martin
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Vidya
P. Ramamurthy
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Senthilmurugan Ramalingam
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Vincent C. O. Njar
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| |
Collapse
|
25
|
Zarif JC, Miranti CK. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal 2016; 28:348-356. [PMID: 26829214 PMCID: PMC4788534 DOI: 10.1016/j.cellsig.2016.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.
Collapse
Affiliation(s)
- Jelani C Zarif
- The James Buchanan Brady Urological Institute at The Johns Hopkins University School of Medicine Baltimore, MD 21287, United States
| | - Cindy K Miranti
- Lab of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, United States.
| |
Collapse
|
26
|
The inhibitory effects of AR/miR-190a/YB-1 negative feedback loop on prostate cancer and underlying mechanism. Sci Rep 2015; 5:13528. [PMID: 26314494 PMCID: PMC4551971 DOI: 10.1038/srep13528] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/29/2015] [Indexed: 01/13/2023] Open
Abstract
Prostate cancer at advanced stages including metastatic and castration-resistant cancer remains incurable due to the lack of effective therapies. MiR-190a belongs to the small noncoding RNA family and has an important role in breast cancer metastasis. However, it is still unknown whether miR-190a plays a role in prostate cancer development. Herein, we first observed AR/miR-190a/YB-1 forms an auto-regulatory negative feedback loop in prostate cancer: miR-190a expression was down-regulated by AR activation; YB-1 functions are as an AR activator; miR-190a inhibited AR expression and transactivation through direct binding to 3′UTR of YB-1 gene. MiR-190a contributes the human prostate cancer cell growth through AR-dependent signaling. Moreover, we examined the expression of miR-190a and observed a significant decrease in human prostate cancers. Reduced expression of miR-190a was inversely correlated to AR levels of prostate cancer patients, and patients with higher miR-190a expression in their tumor have improved tumor-free survival. Taken together, our findings identified a biochemical and functional link between miR-190a with reduced expression in advanced prostate cancer, YB-1 and AR signaling in prostate cancer.
Collapse
|
27
|
Wang X, Huang Y, Christie A, Bowden M, Lee GSM, Kantoff PW, Sweeney CJ. Cabozantinib Inhibits Abiraterone's Upregulation of IGFIR Phosphorylation and Enhances Its Anti-Prostate Cancer Activity. Clin Cancer Res 2015; 21:5578-87. [PMID: 26289068 DOI: 10.1158/1078-0432.ccr-15-0824] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Abiraterone improves the overall survival of men with metastatic castration-resistant prostate cancer. However, de novo or adaptive resistance to abiraterone limits its activity. Rational combinations of drugs with different mechanisms of action that overcome resistance mechanisms may improve the efficacy of therapy. To that end, we studied the molecular and phenotypic effects of the combination of cabozantinib plus abiraterone. EXPERIMENTAL DESIGN Three prostate cancer cell lines were used to interrogate the in vitro molecular and antiproliferative effects of the single agents and combination of cabozantinib and abiraterone. The in vivo impact of the combination was assessed using the LAPC4-CR xenograft mouse model. RESULTS In vitro proliferation studies demonstrated single-agent doses between 2 μmol/L and 10 μmol/L for abiraterone and cabozantinib inhibit prostate cancer cell proliferation in a dose-dependent manner, and the anticancer activity of abiraterone is enhanced when combined with cabozantinib. In vivo LAPC4-CR xenograft mouse studies also showed that cabozantinib can improve the antitumor activity of abiraterone. Cabozantinib, a multiple receptor tyrosine kinase inhibitor, enhances the ability of abiraterone to inhibit AR activity in a cell line-dependent manner. In addition, our cell line studies demonstrate abiraterone-stimulated insulin-like growth factor I receptor (IGFIR) phosphorylation with downstream activation of MEK1/2 and ERK1/2, and that this potential adaptive resistance mechanism was inhibited by cabozantinib. CONCLUSIONS Cabozantinib can enhance the efficacy of abiraterone by blocking multiple compensatory survival mechanisms, including IGFIR activation, and supports the assessment of the combination in a clinical trial.
Collapse
Affiliation(s)
- Xiaodong Wang
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ying Huang
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda Christie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michaela Bowden
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gwo-Shu Mary Lee
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Philip W Kantoff
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Christopher J Sweeney
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
28
|
Liang M, Adisetiyo H, Liu X, Liu R, Gill P, Roy-Burman P, Jones JO, Mulholland DJ. Identification of Androgen Receptor Splice Variants in the Pten Deficient Murine Prostate Cancer Model. PLoS One 2015. [PMID: 26196517 PMCID: PMC4510390 DOI: 10.1371/journal.pone.0131232] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Androgen receptor (AR) variants are associated with resistance to anti androgen therapy both in human prostate cancer cell lines and clinical samples. These observations support the hypothesis that AR isoform accumulation is a consequence of selective therapeutic pressure on the full length AR. The Pten deficient prostate cancer model proceeds with well-defined kinetics including progression to castration resistant prostate cancer (CRPC). While surgical castration and enzalutamide treatments yield an initial therapeutic response, Pten-/-epithelia continue to proliferate yielding locally invasive primary tumor pathology. That most epithelium remains AR positive, but ligand independent, suggests the presence of oncogenic AR variants. To address this hypothesis, we have used a panel of recently described Pten-/- tumor cell lines derived from both from hormone intact (E4, E8) and castrated Pten mutants (cE1, cE2) followed by RACE PCR to identify and characterize three novel truncated, amino terminus containing AR variants (mAR-Va, b, c). Variants appear not only conserved throughout progression but are correlated with nearly complete loss of full length AR (AR-FL) at castrate androgen levels. The overexpression of variants leads to enhanced transcriptional activity of AR while knock down studies show reduced transcriptional output. Collectively, the identification of truncated AR variants in the conditional PTEN deletion model supports a role for maintaining the CRPC phenotype and provides further therapeutic applications of this preclinical model.
Collapse
Affiliation(s)
- Mengmeng Liang
- Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Helty Adisetiyo
- Children’s Hospital of Los Angeles, Los Angeles, California, United States of America
| | - Xiuqing Liu
- St. Luke's Hospital, Internal medicine resident, Chesterfield, Missouri, United States of America
| | - Ren Liu
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Parkash Gill
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Pradip Roy-Burman
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, United States of America
| | - Jeremy O. Jones
- Beckman Research Institute, City of Hope, Duarte, California, United States of America
- * E-mail: (DJM); (JJ)
| | - David J. Mulholland
- Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail: (DJM); (JJ)
| |
Collapse
|
29
|
Greasley R, Khabazhaitajer M, Rosario DJ. A profile of enzalutamide for the treatment of advanced castration resistant prostate cancer. Cancer Manag Res 2015; 7:153-64. [PMID: 26109877 PMCID: PMC4472073 DOI: 10.2147/cmar.s50585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Recent advances in understanding the mechanisms underlying the development and progression of castration resistant prostate cancer from androgen-sensitive prostate cancer have provided new avenues exploring efficacious therapies in a disease which is the second leading cause of cancer deaths among men in the western world. In the evolution of second generation anti-androgens, enzalutamide, a novel androgen-receptor signaling inhibitor, has emerged targeting multiple steps within the androgenic stimulation pathway. This review discusses what is currently known of the mechanisms surrounding castration resistant prostate cancer development and the current human clinical trials to determine whether enzalutamide presents a new hope for men with advanced prostate cancer. The issues of therapy resistance, withdrawal effects and cross-resistance are briefly touched upon.
Collapse
Affiliation(s)
- Rosa Greasley
- The Department of Oncology, The University of Sheffield, Sheffield, UK
| | | | - Derek J Rosario
- The Department of Urology, Sheffield Teaching Hospitals, The Royal Hallamshire Hospital, Sheffield, UK
| |
Collapse
|
30
|
Doucet L, Terrisse S, Gauthier H, Pouessel D, Le Maignan C, Teixeira L, Culine S. Bases de biologie moléculaire du cancer de la prostate résistant à la castration. Bull Cancer 2015; 102:497-500. [DOI: 10.1016/j.bulcan.2015.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
|
31
|
Baicalein inhibits prostate cancer cell growth and metastasis via the caveolin-1/AKT/mTOR pathway. Mol Cell Biochem 2015; 406:111-9. [PMID: 25957503 PMCID: PMC4502300 DOI: 10.1007/s11010-015-2429-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/30/2015] [Indexed: 01/22/2023]
Abstract
Prostate cancer (PCa) is lethal type of genitourinary cancer due to its high morbidity and gradual resistance to androgen deprivation therapy. Accumulating evidence has recently suggested that the daily intake of flavonoids is negatively correlated with the risk of cancer. In this study, we aimed to investigate the potential effects of baicalein on androgen-independent PCa cells and the underlying mechanisms through which baicalein exerts its actions. Cell viability and flow cytometric apoptosis assays indicated that baicalein potently suppressed the growth and induced the apoptosis of DU145 and PC-3 cells in a time- and dose-dependent manner. Consistently, the inhibitory effects of baicalein on migration and invasion were also observed in vitro. Mechanistically, we found that baicalein can suppress caveolin-1 and the phosphorylation of AKT and mTOR in a time- and dose-dependent manner. Moreover, the inhibition of the activation of AKT with LY294002 significantly promoted the apoptosis and metastasis induced by baicalein. In conclusion, these findings suggested that baicalein can induce apoptosis and inhibit metastasis of androgen-independent PCa cells through inhibition of the caveolin-1/AKT/mTOR pathway, which implies that baicalein may be a potential therapeutic agent for the treatment of androgen-independent prostate cancer patients.
Collapse
|
32
|
Fu X, Ren L, Chen J, Liao K, Fu Y, Qian X, Xiao J. Characterization of the roles of suppressor of cytokine signaling-3 in prostate cancer development and progression. Asia Pac J Clin Oncol 2015; 11:106-13. [PMID: 25899712 DOI: 10.1111/ajco.12357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2015] [Indexed: 01/01/2023]
Abstract
As negative feedback regulators of cytokine signaling, suppressor of cytokine signaling proteins are induced by interleukins and various peptide hormones and may prevent sustained activation of signaling pathways. In particular, suppressor of cytokine signaling-3 (SOCS-3) plays pivotal roles in the development and progression of various cancers and exerts pleiotropic effects on cell proliferation and apoptosis. In recent years, abnormal expression of SOCS-3 and its multiple functions have been extensively investigated in human carcinomas, particularly in prostate cancer. SOCS-3 can act as an oncogene or a tumor suppressor depending on the cellular context. In this review, we focus on the role of SOCS-3 in prostate cancer development and prognosis, as well as the potential of SOCS-3 as a therapeutic target and diagnostic marker.
Collapse
Affiliation(s)
- Xian Fu
- Department of Urology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Liu C, Chen Z, Hu X, Wang L, Li C, Xue J, Zhang P, Chen W, Jiang A. MicroRNA-185 downregulates androgen receptor expression in the LNCaP prostate carcinoma cell line. Mol Med Rep 2015; 11:4625-32. [PMID: 25673182 DOI: 10.3892/mmr.2015.3332] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 01/02/2015] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate whether microRNA (miR)‑185 downregulated androgen receptor expression in the LNCaP prostate carcinoma cell line. Human prostate cancer (PCa) LNCaP cells were cultured and transfected with synthetic has‑miR‑185 mimic or inhibitor. The transfected cells were subsequently evaluated with a viability assay, nuclear staining, reverse transcription quantitative polymerase chain reaction (RT‑qPCR), dual luciferase assay and western blot analysis. The results of the western blot analysis and RT‑qPCR indicated that transfection with an miR‑185 mimic markedly reduced the androgen receptor (AR) protein expression levels in LNCaP cells, whereas transfection with an miR‑185 inhibitor increased the protein expression of AR in the LNCaP cells. The results of the luciferase reporter assay demonstrated that the predicted target site in the AR 3' untranslated regions was a specific functional binding site for miR‑185, and that AR was a direct target of miR‑185. In addition, downregulation of AR by miR‑185 impaired the interaction between AR and androgen response element, and downregulated the expression of the AR target gene prostate specific antigen. Data also suggested that the downregulation of AR mediated by miR‑185, inhibited the proliferation and induced the apoptosis of the LNCaP cells. Therefore, the results of the present study suggested that miR‑185 may be a potential negative modulator of AR‑mediated signaling and may act as a tumor suppressor in prostate cancer cells.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhaobo Chen
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lina Wang
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chaoyang Li
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jing Xue
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Pengju Zhang
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Anli Jiang
- Department of Biochemistry and Molecular Biology, Medical School of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
34
|
Kong D, Sethi S, Li Y, Chen W, Sakr WA, Heath E, Sarkar FH. Androgen receptor splice variants contribute to prostate cancer aggressiveness through induction of EMT and expression of stem cell marker genes. Prostate 2015; 75:161-74. [PMID: 25307492 PMCID: PMC4270852 DOI: 10.1002/pros.22901] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND The mechanism(s) by which androgen receptor (AR) splice variants contribute to castration-resistant prostate cancer (CRPC) is still lacking. METHODS Expressions of epithelial-to-mesenchymal transition (EMT) and stem cell markers were molecularly tested using prostate cancer (PCa) cells transfected with AR and AR3 (also known as AR-V7) plasmids or siRNA, and also cultured cells under androgen deprivation therapy (ADT) condition. Cell migration, clonogenicity, sphere-forming capacity was assessed using PCa cells under all experimental conditions and 3,3'-diindolylmethane (DIM; BR-DIM) treatment. Human PCa samples from BR-DIM untreated or treated patients were also used for assessing the expression of AR3 and stem cell markers. RESULTS Overexpression of AR led to the induction of EMT phenotype, while overexpression of AR3 not only induced EMT but also led to the expression of stem cell signature genes. More importantly, ADT enhanced the expression of AR and AR3 concomitant with up-regulated expression of EMT and stem cell marker genes. Dihydrotestosterone (DHT) treatment decreased the expression of AR and AR3, and reversed the expression of these EMT and stem cell marker genes. BR-DIM administered to PCa patients prior to radical prostatectomy inhibited the expression of cancer stem cell markers consistent with inhibition of self-renewal of PCa cells after BR-DIM treatment. CONCLUSION AR variants could contribute to PCa progression through induction of EMT and acquisition of stem cell characteristics, which could be attenuated by BR-DIM, suggesting that BR-DIM could become a promising agent for the prevention of CRPC and/or for the treatment of PCa.
Collapse
Affiliation(s)
- Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | | | | | | | | | | | | |
Collapse
|
35
|
Schalken J, Dijkstra S, Baskin-Bey E, van Oort I. Potential utility of cancer-specific biomarkers for assessing response to hormonal treatments in metastatic prostate cancer. Ther Adv Urol 2014; 6:245-52. [PMID: 25435918 DOI: 10.1177/1756287214545328] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer is the second leading cause of cancer death in men and there is an urgent clinical need to improve its detection and treatment. The introduction of prostate-specific antigen (PSA) as a biomarker for prostate cancer several decades ago represented an important step forward in our ability to diagnose this disease and offers the potential for earlier and more effective treatment. PSA measurements are now routinely conducted alongside digital rectal examination, with raised PSA levels leading to biopsy. PSA is also used to monitor disease and assess therapeutic response. However, there are some important limitations to its use, not least its lack of specificity for prostate cancer, and increased PSA screening may have resulted in overdiagnosis and overtreatment of early, low-risk prostate cancer. Therefore, there is a need for more specific and sensitive biomarkers for the diagnosis and monitoring of prostate cancer and treatment response; in particular, biomarkers of response to hormonal treatments in prostate cancer and predictive biomarkers to identify who is most likely to respond to these treatments. Here we review the current utilization of PSA and data on potentially more specific and sensitive biomarkers for the diagnosis and monitoring of prostate cancer: prostate cancer antigen 3 (PCA3) and the TMPRSS2-ERG fusion gene. A description of the design of an ongoing study of the 6-month extended release formulation of leuprorelin acetate (Eligard(®) 45 mg) will provide preliminary data on the potential utility of these new biomarkers for detecting therapeutic response after hormonal therapy.
Collapse
Affiliation(s)
- Jack Schalken
- Department of Urology, Radboud University Medical Centre, Postbus 9101, 6500 HB, Nijmegen, The Netherlands
| | - Siebren Dijkstra
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Inge van Oort
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Kim JS, Roberts JM, Bingman WE, Shao L, Wang J, Ittmann MM, Weigel NL. The prostate cancer TMPRSS2:ERG fusion synergizes with the vitamin D receptor (VDR) to induce CYP24A1 expression-limiting VDR signaling. Endocrinology 2014; 155:3262-73. [PMID: 24926821 PMCID: PMC5377584 DOI: 10.1210/en.2013-2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A number of preclinical studies have shown that the activation of the vitamin D receptor (VDR) reduces prostate cancer (PCa) cell and tumor growth. The majority of human PCas express a transmembrane protease serine 2 (TMPRSS2):erythroblast transformation-specific (ETS) fusion gene, but most preclinical studies have been performed in PCa models lacking TMPRSS2:ETS in part due to the limited availability of model systems expressing endogenous TMPRSS2:ETS. The level of the active metabolite of vitamin D, 1α,25-dihydroxyvitamin D3 (1,25D), is controlled in part by VDR-dependent induction of cytochrome P450, family 24, subfamily 1, polypeptide1 (CYP24A1), which metabolizes 1,25D to an inactive form. Because ETS factors can cooperate with VDR to induce rat CYP24A1, we tested whether TMPRSS2:ETS would cause aberrant induction of human CYP24A1 limiting the activity of VDR. In TMPRSS2:ETS positive VCaP cells, depletion of TMPRSS2:ETS substantially reduced 1,25D-mediated CYP24A1 induction. Artificial expression of the type VI+72 TMPRSS2:ETS isoform in LNCaP cells synergized with 1,25D to greatly increase CYP24A1 expression. Thus, one of the early effects of TMPRSS2:ETS in prostate cells is likely a reduction in intracellular 1,25D, which may lead to increased proliferation. Next, we tested the net effect of VDR action in TMPRSS2:ETS containing PCa tumors in vivo. Unlike previous animal studies performed on PCa tumors lacking TMPRSS2:ETS, EB1089 (seocalcitol) (a less calcemic analog of 1,25D) did not inhibit the growth of TMPRSS2:ETS containing VCaP tumors in vivo, suggesting that the presence of TMPRSS2:ETS may limit the growth inhibitory actions of VDR. Our findings suggest that patients with TMPRSS2:ETS negative tumors may be more responsive to VDR-mediated growth inhibition and that TMPRSS2:ETS status should be considered in future clinical trials.
Collapse
Affiliation(s)
- Jung-Sun Kim
- Departments of Molecular and Cellular Biology (J.-S.K., J.M.R., W.E.B., N.L.W.) and Pathology and Immunology (L.S., J.W., M.M.I.), Baylor College of Medicine, Houston, Texas 77030
| | | | | | | | | | | | | |
Collapse
|
37
|
Richmond O, Ghotbaddini M, Allen C, Walker A, Zahir S, Powell JB. The aryl hydrocarbon receptor is constitutively active in advanced prostate cancer cells. PLoS One 2014; 9:e95058. [PMID: 24755659 PMCID: PMC3995675 DOI: 10.1371/journal.pone.0095058] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/23/2014] [Indexed: 11/30/2022] Open
Abstract
Background Distant prostate cancers are commonly hormone refractory and exhibit increased growth no longer inhibited by androgen deprivation therapy. Understanding all molecular mechanisms contributing to uncontrolled growth is important to obtain effective treatment strategies for hormone refractory prostate cancers (HRPC). The aryl hydrocarbon receptor (AhR) affects a number of biological processes including cell growth and differentiation. Several studies have revealed that exogenous AhR ligands inhibit cellular proliferation but recent evidence suggests AhR may possess intrinsic functions that promote cellular proliferation in the absence of exogenous ligands. Methods/Results qRT-PCR and western blot analysis was used to determine AhR mRNA and protein expression in hormone sensitive LNCaP cells as well as hormone refractory DU145, PC3 and PC3M prostate cancer cell lines. LNCaP cells express AhR mRNA and protein at a much lower level than the hormone refractory cell models. Cellular fractionation and immunocytochemistry revealed nuclear localization of AhR in the established hormone refractory cell lines while LNCaP cells are devoid of nuclear AhR protein. qRT-PCR analysis used to assess basal CYP1B1 levels and a xenobiotic responsive element binding assay confirmed ligand independent transcriptional activity of AhR in DU145, PC3 and PC3M cells. Basal CYP1B1 levels were decreased by treatment with specific AhR inhibitor, CH223191. An in vitro growth assay revealed that CH223191 inhibited growth of DU145, PC3 and PC3M cells in an androgen depleted environment. Immunohistochemical staining of prostate cancer tissues revealed increased nuclear localization of AhR in grade 2 and grade 3 cancers compared to the well differentiated grade 1 cancers. Conclusions Together, these results show that AhR is constitutively active in advanced prostate cancer cell lines that model hormone refractory prostate cancer. Chemical ablation of AhR signaling can reduce the growth of advanced prostate cancer cells, an effect not achieved with androgen receptor inhibitors or growth in androgen depleted media.
Collapse
Affiliation(s)
- Oliver Richmond
- Clark Atlanta University Center for Cancer Research and Therapeutic Development (CCRTD), Atlanta, Georgia, United States of America
| | - Maryam Ghotbaddini
- Clark Atlanta University Center for Cancer Research and Therapeutic Development (CCRTD), Atlanta, Georgia, United States of America
| | - Cidney Allen
- Clark Atlanta University Department of Biological Sciences, Atlanta, Georgia, United States of America
| | - Alice Walker
- Clark Atlanta University Center for Cancer Research and Therapeutic Development (CCRTD), Atlanta, Georgia, United States of America
| | - Shokouh Zahir
- Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Joann B. Powell
- Clark Atlanta University Center for Cancer Research and Therapeutic Development (CCRTD), Atlanta, Georgia, United States of America
- Clark Atlanta University Department of Biological Sciences, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
38
|
Mahmoud AM, Yang W, Bosland MC. Soy isoflavones and prostate cancer: a review of molecular mechanisms. J Steroid Biochem Mol Biol 2014; 140:116-32. [PMID: 24373791 PMCID: PMC3962012 DOI: 10.1016/j.jsbmb.2013.12.010] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 12/06/2013] [Accepted: 12/10/2013] [Indexed: 02/08/2023]
Abstract
Soy isoflavones are dietary components for which an association has been demonstrated with reduced risk of prostate cancer (PCa) in Asian populations. However, the exact mechanism by which these isoflavones may prevent the development or progression of PCa is not completely understood. There are a growing number of animal and in vitro studies that have attempted to elucidate these mechanisms. The predominant and most biologically active isoflavones in soy products, genistein, daidzein, equol, and glycetin, inhibit prostate carcinogenesis in some animal models. Cell-based studies show that soy isoflavones regulate genes that control cell cycle and apoptosis. In this review, we discuss the literature relevant to the molecular events that may account for the benefit of soy isoflavones in PCa prevention or treatment. These reports show that although soy isoflavone-induced growth arrest and apoptosis of PCa cells are plausible mechanisms, other chemo protective mechanisms are also worthy of consideration. These possible mechanisms include antioxidant defense, DNA repair, inhibition of angiogenesis and metastasis, potentiation of radio- and chemotherapeutic agents, and antagonism of estrogen- and androgen-mediated signaling pathways. Moreover, other cells in the cancer milieu, such as the fibroblastic stromal cells, endothelial cells, and immune cells, may be targeted by soy isoflavones, which may contribute to soy-mediated prostate cancer prevention. In this review, these mechanisms are discussed along with considerations about the doses and the preclinical models that have been used.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Wancai Yang
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Maarten C Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
39
|
Tan JA, Bai S, Grossman G, Titus MA, Harris Ford O, Pop EA, Smith GJ, Mohler JL, Wilson EM, French FS. Mechanism of androgen receptor corepression by CKβBP2/CRIF1, a multifunctional transcription factor coregulator expressed in prostate cancer. Mol Cell Endocrinol 2014; 382:302-313. [PMID: 24103312 PMCID: PMC3880566 DOI: 10.1016/j.mce.2013.09.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 09/27/2013] [Indexed: 01/12/2023]
Abstract
The transcription factor coregulator Casein kinase IIβ-binding protein 2 or CR6-interacting factor 1 (CKβBP2/CRIF1) binds the androgen receptor (AR) in prostate cancer cells and in response to dihydrotestosterone localizes with AR on the prostate-specific antigen gene enhancer, but does not bind DNA suggesting CKβBP2/CRIF1 localization in chromatin is determined by AR. In this study we show also that CKβBP2/CRIF1 inhibits wild-type AR and AR N-terminal transcriptional activity, binds to the AR C-terminal region, inhibits interaction of the AR N- and C-terminal domains (N/C interaction) and competes with p160 coactivator binding to the AR C-terminal domain, suggesting CKβBP2/CRIF1 interferes with AR activation functions 1 and 2. CKβBP2/CRIF1 is expressed mainly in stromal cells of benign prostatic hyperplasia and in stroma and epithelium of prostate cancer. CKβBP2/CRIF1 protein is increased in epithelium of androgen-dependent prostate cancer compared to benign prostatic hyperplasia and decreased slightly in castration recurrent epithelium compared to androgen-dependent prostate cancer. The multifunctional CKβBP2/CRIF1 is a STAT3 interacting protein and reported to be a coactivator of STAT3. CKβBP2/CRIF1 is expressed with STAT3 in prostate cancer where STAT3 may help to offset the AR repressor effect of CKβBP2/CRIF1 and allow AR regulation of prostate cancer growth.
Collapse
Affiliation(s)
- Jiann-An Tan
- Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States
| | - Suxia Bai
- Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States
| | - Gail Grossman
- Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States
| | - Mark A Titus
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, United States
| | - O Harris Ford
- Lineberger Comprehensive Cancer Center, University of North Carolina, School of Medicine, Chapel Hill, NC, United States
| | - Elena A Pop
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, United States
| | - Gary J Smith
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, United States
| | - James L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, School of Medicine, Chapel Hill, NC, United States; Department of Urology, University of Buffalo, School of Medicine and Biotechnology, Buffalo, NY, United States
| | - Elizabeth M Wilson
- Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, School of Medicine, Chapel Hill, NC, United States; Department of Biochemistry and Biophysics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States
| | - Frank S French
- Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina, School of Medicine, Chapel Hill, NC, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
40
|
Chan SC, Dehm SM. Constitutive activity of the androgen receptor. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:327-66. [PMID: 24931201 DOI: 10.1016/b978-0-12-417197-8.00011-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer in the United States. The androgen receptor (AR) signaling axis is central to all stages of PCa pathophysiology and serves as the main target for endocrine-based therapy. The most advanced stage of the disease, castration-resistant prostate cancer (CRPC), is presently incurable and accounts for most PCa mortality. In this chapter, we highlight the mechanisms by which the AR signaling axis can bypass endocrine-targeted therapies and drive progression of CRPC. These mechanisms include alterations in growth factor, cytokine, and inflammatory signaling pathways, altered expression or activity of transcriptional coregulators, AR point mutations, and AR gene amplification leading to AR protein overexpression. Additionally, we will discuss the mechanisms underlying the synthesis of constitutively active AR splice variants (AR-Vs) lacking the COOH-terminal ligand-binding domain, as well as the role and regulation of AR-Vs in supporting therapeutic resistance in CRPC. Finally, we summarize the ongoing development of inhibitors targeting discrete AR functional domains as well as the status of new biomarkers for monitoring the AR signaling axis in patients.
Collapse
Affiliation(s)
- Siu Chiu Chan
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Scott M Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
41
|
Shafi AA, Yen AE, Weigel NL. Androgen receptors in hormone-dependent and castration-resistant prostate cancer. Pharmacol Ther 2013; 140:223-38. [PMID: 23859952 DOI: 10.1016/j.pharmthera.2013.07.003] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 01/18/2023]
Abstract
In the United States, prostate cancer (PCa) is the most commonly diagnosed non-cutaneous cancer in males and the second leading cause of cancer-related death for men. The prostate is an androgen-dependent organ and PCa is an androgen-dependent disease. Androgen action is mediated by the androgen receptor (AR), a hormone activated transcription factor. The primary treatment for metastatic PCa is androgen deprivation therapy (ADT). For the most part, tumors respond to ADT, but most become resistant to therapy within two years. There is persuasive evidence that castration resistant (also termed castration recurrent) PCa (CRPC) remains AR dependent. Recent studies have shown that there are numerous factors that contribute to AR reactivation despite castrate serum levels of androgens. These include changes in AR expression and structure through gene amplification, mutation, and alternative splicing. Changes in steroid metabolism, cell signaling, and coregulator proteins are also important contributors to AR reactivation in CRPC. Most AR targeted therapies have been directed at the hormone binding domain. The finding that constitutively active AR splice variants that lack the hormone binding domain are frequently expressed in CRPC highlights the need to develop therapies that target other portions of AR. In this review, the role of AR in normal prostate, in PCa, and particularly the mechanisms for its reactivation subsequent to ADT are summarized. In addition, recent clinical trials and novel approaches to target AR are discussed.
Collapse
Affiliation(s)
- Ayesha A Shafi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, M515, One Baylor Plaza, Houston, TX 77030, USA
| | | | | |
Collapse
|
42
|
Molecular circuit involving KLK4 integrates androgen and mTOR signaling in prostate cancer. Proc Natl Acad Sci U S A 2013; 110:E2572-81. [PMID: 23798432 DOI: 10.1073/pnas.1304318110] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The androgen receptor (AR) and the phosphoinositide 3-kinase (PI3K)/protein kinase B/mammalian target of rapamycin (mTOR) signaling are two of the major proliferative pathways in a number of tissues and are the main therapeutic targets in various disorders, including prostate cancer (PCa). Previous work has shown that there is reciprocal feedback regulation of PI3K and AR signaling in PCa, suggesting that cotargeting both pathways may enhance therapeutic efficacy. Here we show that proteins encoded by two androgen-regulated genes, kallikrein related peptidase 4 (KLK4) and promyelocytic leukemia zinc finger (PLZF), integrate optimal functioning of AR and mTOR signaling in PCa cells. KLK4 interacts with PLZF and decreases its stability. PLZF in turn interacts with AR and inhibits its function as a transcription factor. PLZF also activates expression of regulated in development and DNA damage responses 1, an inhibitor of mTORC1. Thus, a unique molecular switch is generated that regulates both AR and PI3K signaling. Consistently, KLK4 knockdown results in a significant decline in PCa cell proliferation in vitro and in vivo, decreases anchorage-independent growth, induces apoptosis, and dramatically sensitizes PCa cells to apoptosis-inducing agents. Furthermore, in vivo nanoliposomal KLK4 siRNA delivery in mice bearing PCa tumors results in profound remission. These results demonstrate that the activities of AR and mTOR pathways are maintained by KLK4, which may thus be a viable target for therapy.
Collapse
|
43
|
Shafi AA, Cox MB, Weigel NL. Androgen receptor splice variants are resistant to inhibitors of Hsp90 and FKBP52, which alter androgen receptor activity and expression. Steroids 2013; 78:548-54. [PMID: 23380368 PMCID: PMC3640750 DOI: 10.1016/j.steroids.2012.12.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/20/2012] [Accepted: 12/28/2012] [Indexed: 01/18/2023]
Abstract
Androgen ablation therapy is the most common treatment for advanced prostate cancer (PCa), but most patients will develop castration-resistant prostate cancer (CRPC), which has no cure. CRPC is androgen-depletion resistant but androgen receptor (AR) dependent. AR is a nuclear receptor whose transcriptional activity is regulated by hormone binding to the ligand-binding domain (LBD). Constitutively active AR splice variants that lack LBDs often are expressed in CRPC. The expression of these variants indicates that methods to inhibit AR activity that do not rely on inactivating the LBD are needed. Heat shock protein 90 (Hsp90), a potential therapeutic target in PCa, is an AR chaperone crucial for proper folding, hormone binding and transcriptional activity of AR. We generated LNCaP cell lines with regulated expression of the AR-V7 variant as well as a cell line expressing artificially truncated AR (termed AR-NTD) to characterize splice variant function. Using an Hsp90 inhibitor, Geldanamycin (GA), and an AR-Hsp90-FKBP52 specific inhibitor, MJC13, we sought to determine if the AR variants also require Hsp90 and associated co-chaperone, FKBP52, for their activity. GA inhibits AR transcriptional activity but has little effect on AR-V7 activity. Moreover, GA decreases the stability of AR protein, with no effect on AR-V7 levels. Full-length AR activity is strongly inhibited by MJC13 while AR-V7 is unaffected. Thus, the variants are resistant to inhibitors of the Hsp90-AR heterocomplex. Although Hsp90 inhibitors will continue to inhibit growth promoting kinases and signaling through activated full-length AR in CRPC, AR signaling through variants will be retained.
Collapse
Affiliation(s)
- Ayesha A. Shafi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, M515, One Baylor Plaza, Houston, TX 77030, USA
| | - Marc B. Cox
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Nancy L. Weigel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, M515, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
44
|
Bastien D, Hanna R, Leblanc V, Asselin É, Bérubé G. Synthesis and preliminary in vitro biological evaluation of 7α-testosterone-chlorambucil hybrid designed for the treatment of prostate cancer. Eur J Med Chem 2013; 64:442-7. [PMID: 23665800 DOI: 10.1016/j.ejmech.2013.04.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/12/2013] [Accepted: 04/13/2013] [Indexed: 11/17/2022]
Abstract
The synthesis of 7α-testosterone-chlorambucil hybrid is reported. This compound is made from testosterone in a 6 step reaction sequence and with 23% overall yield. An alternative convergent reaction sequence yielded the same hybrid through a Grubbs metathesis reaction between chlorambucil allyl ester and 7α-allyltestosterone with 35% overall yield. MTT assays showed that the hybrid is selective towards hormone-dependent prostate cancer cell line (LNCaP (AR+)) and shows similar activity than the parent drug, chlorambucil. Thus, the new hybrid shows promising potential for drug targeting of hormone-dependent prostate cancer through its capacity of delivering chlorambucil directly to the site of treatment. This could extend the use of chlorambucil to prostate cancer in the future.
Collapse
Affiliation(s)
- Dominic Bastien
- Département de Chimie et Physique, Université du Québec à Trois-Rivières, C.P. 500, Trois-Rivières, Québec, Canada G9A 5H7.
| | | | | | | | | |
Collapse
|
45
|
Arlen PM. Prostate cancer vaccines: an old yet novel target, the androgen receptor. Expert Rev Vaccines 2013; 12:249-51. [PMID: 23496664 DOI: 10.1586/erv.13.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer vaccines have been utilized as a therapeutic modality to treat prostate cancer in clinical studies for several decades. Recently with the approval of sipuleucel-T, vaccines have now been accepted as standard therapy for this disease. The androgen receptorhas long been recognized as a therapeutic target for the treatment of patients with locally advanced as well as metastatic disease. Recent preclinical studies described by Olson et al. have focused on the androgen receptor as a target for prostate cancer immunotherapy. They have developed and tested a DNA vaccine targeting the ligand-binding domain of the androgen receptor and have demonstrated in animal studies the ability to elicit T-cell responses towards the vaccine that have resulted in both antitumor activity as well as increased survival in the animal models described.
Collapse
Affiliation(s)
- Philip M Arlen
- Precision Biologics, Inc., 9700 Great Seneca Hwy, Suite 321, Rockville, MD 20850, USA.
| |
Collapse
|
46
|
Lu TL, Huang YF, You LR, Chao NC, Su FY, Chang JL, Chen CM. Conditionally ablated Pten in prostate basal cells promotes basal-to-luminal differentiation and causes invasive prostate cancer in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:975-91. [PMID: 23313138 DOI: 10.1016/j.ajpath.2012.11.025] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 10/27/2012] [Accepted: 11/07/2012] [Indexed: 12/22/2022]
Abstract
Prostate glands comprise two major epithelial cell types: luminal and basal. Luminal cells have long been considered the cellular origin of prostate cancer (CaP). However, recent evidence from a prostate regeneration assay suggests that prostate basal cells can also give rise to CaP. Here, we characterize Pten-deficient prostate lesions arising from keratin 5-expressing basal cells in a temporally controlled system in mice. Pten-deficient prostate lesions arising from basal cells exhibited luminal phenotypes with higher invasiveness, and the cell fate of Pten-deficient basal cells was traced to neoplastic luminal cells. After temporally ablating Pten in keratin 8-expressing luminal cells, luminal-derived Pten-deficient prostate tumors exhibited slower disease progression, compared with basal-derived tumors, within 13 weeks after Pten ablation. Cellular proliferation was significantly increased in basal-derived versus luminal-derived Pten-deficient prostate lesions. Increased tumor invasion into the smooth muscle layer and aberrantly regulated aggressive signatures (Smad4 and Spp1) were identified exclusively in basal-derived Pten-deficient lesions. Interestingly, p63-expressing cells, which represent basal stem and progenitor cells of basal-derived Pten-deficient prostate lesions, were significantly increased, relative to cells of the luminal-derived prostate lesion. Furthermore, castration did not suppress cellular proliferation of either basal-derived or luminal-derived Pten-deficient prostate tumors. Taken together, our data suggest that, although prostate malignancy can originate from both basal and luminal populations, these two populations differ in aggressive potential.
Collapse
Affiliation(s)
- Tsai-Ling Lu
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
47
|
Sheikh H, Abdulghani J, Ali S, Sinha R, Lipton A. Impact of Genetic Targets on Prostate Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:359-83. [DOI: 10.1007/978-1-4614-6176-0_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
48
|
Targeted disruption of the p160 coactivator interface of androgen receptor (AR) selectively inhibits AR activity in both androgen-dependent and castration-resistant AR-expressing prostate cancer cells. Int J Biochem Cell Biol 2012; 45:763-72. [PMID: 23270728 DOI: 10.1016/j.biocel.2012.12.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/24/2012] [Accepted: 12/13/2012] [Indexed: 10/27/2022]
Abstract
The evidence that androgen blockade-resistant prostate cancer, termed castration resistant, remains androgen receptor (AR) dependent is compelling. AR is re-activated through multiple mechanisms including expression of constitutively active splice variants that lack hormone binding domains (HBDs). This highlights the need to develop therapies that target regions other than the HBD. Because the p160 coactivators interact most strongly with the amino-terminus of AR, we examined the consequences of disrupting this interaction. We identified two overlapping SRC-1 peptides that interact with AR, but not with progesterone receptor. These peptides reduce AR and AR variant AR-V7 dependent induction of an AR responsive reporter. Using mammalian two hybrid assays, we found that the peptides interrupt the AR/SRC-1, AR/SRC-2 and AR N/C interactions, but not SRC-1/CARM-1 interactions. Consistent with the SRC-1 dependence of induced, but not repressed genes, in LNCaP cells, the peptides inhibited hormone dependent induction of endogenous target genes including PSA and TMPRSS2, but did not block AR dependent repression of UGT2B17 or inhibit vitamin D receptor activity. Simultaneous detection of SRC-1 peptides and PSA by double immunofluorescence in transfected LNCaP cells clearly demonstrated a strong reduction in PSA levels in cells expressing the peptides. The peptides also inhibited the AR dependent expression of PSA in castration resistant C4-2 cells. Moreover they inhibited androgen dependent proliferation of LNCaP cells and proliferation of C4-2 cells in androgen depleted medium without affecting AR negative PC-3 cells. Thus, the p160 coactivator binding site is a novel potential therapeutic target to inhibit AR activity.
Collapse
|
49
|
Kitagishi Y, Kobayashi M, Matsuda S. Defective DNA repair systems and the development of breast and prostate cancer (review). Int J Oncol 2012; 42:29-34. [PMID: 23151935 DOI: 10.3892/ijo.2012.1696] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/29/2012] [Indexed: 11/06/2022] Open
Abstract
Genetic defects in DNA repair and DNA damage response genes often lead to an increase in cancer incidence. The role of defects is also associated with the modulation of hormone signaling pathways. A number of studies have suggested a role for estrogen in the regulation of DNA repair activity. Furthermore, mutations or epigenetic silencing in DNA repair genes have been associated with the sensitivity of cancers to hormonal therapy. The molecular basis for the progression of cancers from hormone-dependent to hormone-independent remains a critical issue in the management of these types of cancer. In the present review, we aimed to summarize the function of DNA repair molecules from the viewpoint of carcinogenesis and hormone-related cell modulation, providing a comprehensive view of the molecular mechanisms by which hormones may exert their effects on the regulation of tumor progression.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Environmental Health Science, Nara Women's University, Nara, Japan
| | | | | |
Collapse
|
50
|
Olson BM, Johnson LE, McNeel DG. The androgen receptor: a biologically relevant vaccine target for the treatment of prostate cancer. Cancer Immunol Immunother 2012; 62:585-96. [PMID: 23108626 DOI: 10.1007/s00262-012-1363-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 10/12/2012] [Indexed: 10/27/2022]
Abstract
The androgen receptor (AR) plays an essential role in the development and progression of prostate cancer. However, while it has long been the primary molecular target of metastatic prostate cancer therapies, it has not been explored as an immunotherapeutic target. In particular, the AR ligand-binding domain (LBD) is a potentially attractive target, as it has an identical sequence among humans as well as among multiple species, providing a logical candidate for preclinical evaluation. In this report, we evaluated the immune and anti-tumor efficacy of a DNA vaccine targeting the AR LBD (pTVG-AR) in relevant rodent preclinical models. We found immunization of HHDII-DR1 mice, which express human HLA-A2 and HLA-DR1, with pTVG-AR augmented AR LBD HLA-A2-restricted peptide-specific, cytotoxic immune responses in vivo that could lyse human prostate cancer cells. Using an HLA-A2-expressing autochthonous model of prostate cancer, immunization with pTVG-AR augmented HLA-A2-restricted immune responses that could lyse syngeneic prostate tumor cells and led to a decrease in tumor burden and an increase in overall survival of tumor-bearing animals. Finally, immunization decreased prostate tumor growth in Copenhagen rats that was associated with a Th1-type immune response. These data show that the AR is as a prostate cancer immunological target antigen and that a DNA vaccine targeting the AR LBD is an attractive candidate for clinical evaluation.
Collapse
Affiliation(s)
- Brian M Olson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI 53705, USA.
| | | | | |
Collapse
|