1
|
Ma J, Qin X, Le W, Chen X, Wang X, Xu C. Identification of BBC3 as a novel indicator for predicting prostate cancer development and olaparib resistance. Discov Oncol 2024; 15:496. [PMID: 39331229 PMCID: PMC11436583 DOI: 10.1007/s12672-024-01373-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Prostate cancer (PCa) is a commonly occurring malignancy in elderly men. Olaparib, a poly ADP-ribose polymerase inhibitor, is utilized in PCa treatment. However, patients often develop resistance to olaparib after a period of treatment. Genetic alterations may play a significant role in this resistance, but the specific genes involved remain unclear. This study collected RNA-sequence data from the Gene Expression Omnibus database on both olaparib-sensitive and -resistant PCa cells to identify genes crucial for resistance. Subsequently, the enriched pathways of these genes were analyzed, and a protein-protein interaction (PPI) network was constructed to identify hub genes. The effect of these hub genes on PCa occurrence, progression, and prognosis was assessed using data from The Cancer Genome Atlas and Chinese Prostate Cancer Genome and Epigenome Atlas databases. Finally, this study validated our findings in clinical PCa samples and cells. From the GSE189186 dataset, 50 upregulated genes and 2 downregulated genes were identified in olaparib-resistant C4-2B and LNCaP cells. Utilizing the PPI network, eight upregulated genes (BBC3, TP53I3, FDXR, DDB2, CDKN1A, GADD45A, ZMAT3, and SESN1) were identified as hub genes for olaparib-resistant PCa cells. Furthermore, some of these genes were central to PCa occurrence, with BBC3 also influencing progression and prognosis. Importantly, BBC3 expression was upregulated in clinical PCa samples and affected PCa cells sensitive to olaparib, suggesting its potential as a predictive marker for PCa development and olaparib resistance.
Collapse
Affiliation(s)
- Junjie Ma
- Department of Urology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China
| | - Xin Qin
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Wei Le
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Xi Chen
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China
| | - Xiao Wang
- Department of Urology, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing, 314000, Zhejiang, China.
| | - Chengdang Xu
- Department of Urology, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai,, 6000065, China.
| |
Collapse
|
2
|
Gilloteaux J, Jamison JM, Summers JL, Taper HS. Reactivation of nucleases with peroxidation damages induced by a menadione: ascorbate combination devastates human prostate carcinomas: ultrastructural aspects. Ultrastruct Pathol 2024; 48:378-421. [PMID: 39105605 DOI: 10.1080/01913123.2024.2379300] [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: 05/06/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Xenografts of androgen-independent human DU145 prostate metastatic carcinomas implanted in nu/nu male mice have revealed a significant survival after a prooxidant anticancer treatment consisting of a combination of menadione bisulfite and sodium ascorbate (VK3:VC). METHODS Implanted samples of diaphragm carcinomas from longest survived mice from either oral, intraperitoneal (IP), or both oral and IP treatment groups were assessed with light, scanning, and transmission electron microscopy to analyze morphologic damages. RESULTS Compared with previous fine structure data of in vitro untreated carcinomas, the changes induced by oral, IP, and oral with IP VK3:VC treatment dismantled those xenografts with autoschizis, and necrotic atrophy was accomplished by cell's oxidative stress whose injuries were consequent to reactivated deoxyribonucleases and ribonucleases. Tumor destructions resulted from irreversible damages of nucleus components, endoplasmic reticulum, and mitochondria there. Other alterations included those of the cytoskeleton that resulted in characteristic self-excisions named " autoschizis." All these injuries lead resilient cancer cells to necrotic cell death. CONCLUSION The fine structure damages caused by VK3:VC prooxidant combination in the human DU145 prostate xenografts confirmed those shown in vitro and of other cell lines with histochemistry and biomolecular investigations. These devastations incurred without damage to normal tissues; thus, our data brought support for the above combination to assist in the treatment of prostate cancers and other cancers.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- Department of Anatomical Sciences, St Georges' University International School of Medicine, Newcastle upon Tyne, UK
- Department of Anatomical Sciences, NEOMed (NEOUCOM), Rootstown, Ohio, USA
- Department of Medicine, Unit of Research in Molecular Physiology (URPhyM), NARILIS, Université de Namur, Namur, Belgium
| | - James M Jamison
- Department of Urology, Summa Health System, Akron, Ohio, USA
- St Thomas Hospital, The Apatone Development Center, Summa Research Fondation, Akron Ohio, USA
| | - Jack L Summers
- Department of Urology, Summa Health System, Akron, Ohio, USA
- St Thomas Hospital, The Apatone Development Center, Summa Research Fondation, Akron Ohio, USA
| | - Henryk S Taper
- Département des Sciences Pharmaceutiques, Unité de Pharmacocinétique, Métabolisme, Nutrition et Toxicologie, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Chen X, Li H, Xu C, Wang X, Wu G, Li C, Wu D. CYP19A1 is downregulated by BRD4 and suppresses castration-resistant prostate cancer cell invasion and proliferation by decreasing AR expression. Am J Cancer Res 2023; 13:4003-4020. [PMID: 37818065 PMCID: PMC10560923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/17/2023] [Indexed: 10/12/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) is the final stage of prostate cancer (PCa). As the main androgen in males, testosterone, and its androgen receptor (AR) play an important role in CRPC. The enzyme that catalyzes testosterone, aromatase, can be influenced by CYP19A1 activity - thus possibly affecting both AR expression and CRPC. However, the function of CYP19A1 in CRPC remains unclear. Using data derived from public databases and clinical samples, we analyzed the expression of CYP19A1 in PCa and CRPC specimens. The effect of CYP19A1 on cell invasion and proliferation was investigated in vitro and in vivo; while its function in metabolizing testosterone was detected in vitro. The effect of BRD4 on CYP19A1 and AR was investigated by qRT-PCR and western blot; whereas the effect of JQ1 on cells was assessed based on the IC50 value. We found that CYP19A1 was downregulated in CRPC samples and cells which correlated with a decrease in CRPC cell invasion and proliferation, and an increase in AR expression. Inversely, CYP19A1 affected CRPC cell invasion and proliferation by suppressing the expression of AR which may be attributed to the metabolism of testosterone by CYP19A1. Moreover, the BRD4 inhibitor JQ1 induced the CYP19A1 expression and suppressed the AR expression. Following BRD4 knockdown, CYP19A1 showed higher expression while AR expression was decreased. Our findings demonstrated that CYP19A1 could reduce CRPC cell invasion and proliferation by targeting AR, and this process could be regulated by BRD4. CYP19A1 may be a potential therapeutic target and enhance BRD4 inhibition in treating CRPC.
Collapse
Affiliation(s)
- Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Xin'an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Chao Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University Shanghai, The People's Republic of China
| |
Collapse
|
4
|
Silva J, Tavares V, Afonso A, Garcia J, Cerqueira F, Medeiros R. Plasmatic MicroRNAs and Treatment Outcomes of Patients with Metastatic Castration-Resistant Prostate Cancer: A Hospital-Based Cohort Study and In Silico Analysis. Int J Mol Sci 2023; 24:ijms24109101. [PMID: 37240449 DOI: 10.3390/ijms24109101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies among men worldwide. Inevitably, all advanced PCa patients develop metastatic castration-resistant prostate cancer (mCRPC), an aggressive phase of the disease. Treating mCRPC is challenging, and prognostic tools are needed for disease management. MicroRNA (miRNA) deregulation has been reported in PCa, constituting potential non-invasive prognostic biomarkers. As such, this study aimed to evaluate the prognostic potential of nine miRNAs in the liquid biopsies (plasma) of mCRPC patients treated with second-generation androgen receptor axis-targeted (ARAT) agents, abiraterone acetate (AbA) and enzalutamide (ENZ). Low expression levels of miR-16-5p and miR-145-5p in mCRPC patients treated with AbA were significantly associated with lower progression-free survival (PFS). The two miRNAs were the only predictors of the risk of disease progression in AbA-stratified analyses. Low miR-20a-5p levels in mCRPC patients with Gleason scores of <8 were associated with worse overall survival (OS). The transcript seems to predict the risk of death regardless of the ARAT agent. According to the in silico analyses, miR-16-5p, miR-145-5p, and miR-20a-5p seem to be implicated in several processes, namely, cell cycle, proliferation, migration, survival, metabolism, and angiogenesis, suggesting an epigenetic mechanism related to treatment outcome. These miRNAs may represent attractive prognostic tools to be used in mCRPC management, as well as a step further in the identification of new potential therapeutic targets, to use in combination with ARAT for an improved treatment outcome. Despite the promising results, real-world validation is necessary.
Collapse
Affiliation(s)
- Jani Silva
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- AquaValor-Centro de Valorização e Transferência de Tecnologia da Água, Rua Dr. Júlio Martins, nº1, 5400-342 Chaves, Portugal
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ana Afonso
- Department of Oncology, Portuguese Institute of Oncology, Rua Dr. António Bernardino Almeida, 4200-072 Porto, Portugal
| | - Juliana Garcia
- AquaValor-Centro de Valorização e Transferência de Tecnologia da Água, Rua Dr. Júlio Martins, nº1, 5400-342 Chaves, Portugal
- Centre for the Research and Technology of Agro-Environment and Biological Sciences (CITAB)/Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal
| | - Fátima Cerqueira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Instituto de Investigação, Inovação e Desenvolvimento Fernando Pessoa (FP-I3ID), Biomedical and Health Sciences (FP-BHS), Universidade Fernando Pessoa, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
- Instituto de Investigação, Inovação e Desenvolvimento Fernando Pessoa (FP-I3ID), Biomedical and Health Sciences (FP-BHS), Universidade Fernando Pessoa, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| |
Collapse
|
5
|
Wasim S, Lee SY, Kim J. Complexities of Prostate Cancer. Int J Mol Sci 2022; 23:14257. [PMID: 36430730 PMCID: PMC9696501 DOI: 10.3390/ijms232214257] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer has a long disease history and a wide variety and uncertainty in individual patients' clinical progress. In recent years, we have seen a revolutionary advance in both prostate cancer patient care and in the research field. The power of deep sequencing has provided cistromic and transcriptomic knowledge of prostate cancer that has not discovered before. Our understanding of prostate cancer biology, from bedside and molecular imaging techniques, has also been greatly advanced. It is important that our current theragnostic schemes, including our diagnostic modalities, therapeutic responses, and the drugs available to target non-AR signaling should be improved. This review article discusses the current progress in the understanding of prostate cancer biology and the recent advances in diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Sobia Wasim
- Department of Neuroscience, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Sang-Yoon Lee
- Department of Neuroscience, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Jaehong Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
6
|
Seale KN, Labriola MK, Jiang X"S, Armstrong A. Remission of progressive metastatic castration-resistant prostate cancer during abiraterone therapy following unilateral adrenalectomy for a functioning adrenal adenoma. BMJ Case Rep 2022; 15:15/10/e251036. [PMID: 36198431 PMCID: PMC9535143 DOI: 10.1136/bcr-2022-251036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Despite advances and introduction of new therapies in the last decade, metastatic castration-resistant prostate cancer (mCRPC) has a poor prognosis. The development of androgen axis-targeted therapies such as abiraterone acetate, enzalutamide and darolutamide can prolong survival in mCPRC; however, resistance remains a barrier to prolonged response, necessitating exploration into resistance mechanisms and locoregional therapies. Here, we describe a patient with mCRPC that was progressing on abiraterone acetate. He was also found to have primary hyperaldosteronism from a functional adrenal adenoma, and thus he had a partial adrenalectomy to remove this tumour. Pathology confirmed an aldosterone-producing adrenal adenoma. After his adrenalectomy, he had a sharp decline in both his PSA (prostate specific antigen) and testosterone levels, and he enjoyed a year-long period of remission after his adrenalectomy. We propose several explanations for his response, the most likely being that his adenoma was producing both aldosterone and androgens. This is a unique case of mCRPC responding to partial adrenalectomy from a functional adrenal adenoma, and it raises insights that warrant further investigation into underlying mechanisms of resistance to androgen-targeted therapies.
Collapse
Affiliation(s)
| | - Matthew K Labriola
- Department of Medicine, Duke University, Durham, North Carolina, USA,Division of Medical Oncology, Duke Cancer Institute, Durham, North Carolina, USA
| | | | - Andrew Armstrong
- Department of Medicine, Duke University, Durham, North Carolina, USA,Division of Medical Oncology, Duke Cancer Institute, Durham, North Carolina, USA,Duke Cancer Institute, Center for Prostate and Urologic Cancer, Durham, North Carolina, USA
| |
Collapse
|
7
|
Yenki P, Adomat HH, Ong CJ. SEMA3C induces androgen synthesis in prostatic stromal cells through paracrine signaling. Prostate 2021; 81:309-317. [PMID: 33503318 DOI: 10.1002/pros.24107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 01/01/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Castration resistant prostate cancer progression is associated with an acquired intratumoral androgen synthesis. Signaling pathways that can upregulate androgen production in prostate tumor microenvironment are not entirely known. In this study, we investigate the potential effect of a secreted signaling protein named semaphorin 3C (SEMA3C) on steroidogenic activities of prostatic stromal cells. METHODS We treated human primary prostate stromal cells (PrSC) with 1uM recombinant SEMA3C protein and androgen precursor named dehydroepiandrosterone (DHEA) 1.7uM. Also, to test SEMA3C's effect on the conversion of DHEA to androgens, we exposed PrSCs to the conditioned media derived from LNCaP cells that were transduced with a lentiviral vector harboring full length SEMA3C gene or empty vector (CM-LNSEMA3C or CM-LNVector ). Then, liquid chromatography-mass spectrometry was performed on steroids isolated from PrSCs media. The messnger RNA expression of steroidogenic enzymes in PrSCs was quantified by quantitative polymerase chain reaction. RESULTS Recombinant SEMA3C had no effect on steroidogenic activities in PrSCs. However, key steroidogenic enzymes expression and androgen synthesis were upregulated in PrSCs treated with CM-LNSEMA3C , compared to those treated with CM-LNVector . These results suggest that steroidogenic activities in PrSCs were upregulated in response to a signaling factor in CM-LNSEMA3C , other than SEMA3C. We hypothesized that SEMA3C overexpression in LNCaP cells affected androgen synthesis in PrSCs through sonic hedgehog (Shh) pathway activation in PrSCs. We verified this effect by blocking Shh signaling with smoothened antagonist. CONCLUSION Based on known ability of Shh signaling pathway to activate steroidogenesis in stromal cells, we suggest that SEMA3C overexpression in LNCaP cells can upregulate Shh which in turn is able to stimulate steroidogenic activities in prostatic stromal cells.
Collapse
Affiliation(s)
- Parvin Yenki
- The Vancouver Prostate Center, Vancouver General Hospital, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hans H Adomat
- The Vancouver Prostate Center, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Christopher J Ong
- The Vancouver Prostate Center, Vancouver General Hospital, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Mamouni K, Kallifatidis G, Lokeshwar BL. Targeting Mitochondrial Metabolism in Prostate Cancer with Triterpenoids. Int J Mol Sci 2021; 22:2466. [PMID: 33671107 PMCID: PMC7957768 DOI: 10.3390/ijms22052466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
Metabolic reprogramming is a hallmark of malignancy. It implements profound metabolic changes to sustain cancer cell survival and proliferation. Although the Warburg effect is a common feature of metabolic reprogramming, recent studies have revealed that tumor cells also depend on mitochondrial metabolism. Due to the essential role of mitochondria in metabolism and cell survival, targeting mitochondria in cancer cells is an attractive therapeutic strategy. However, the metabolic flexibility of cancer cells may enable the upregulation of compensatory pathways, such as glycolysis, to support cancer cell survival when mitochondrial metabolism is inhibited. Thus, compounds capable of targeting both mitochondrial metabolism and glycolysis may help overcome such resistance mechanisms. Normal prostate epithelial cells have a distinct metabolism as they use glucose to sustain physiological citrate secretion. During the transformation process, prostate cancer cells consume citrate to mainly power oxidative phosphorylation and fuel lipogenesis. A growing number of studies have assessed the impact of triterpenoids on prostate cancer metabolism, underlining their ability to hit different metabolic targets. In this review, we critically assess the metabolic transformations occurring in prostate cancer cells. We will then address the opportunities and challenges in using triterpenoids as modulators of prostate cancer cell metabolism.
Collapse
Affiliation(s)
- Kenza Mamouni
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Georgios Kallifatidis
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Biological Sciences, Augusta University, Augusta, GA 30912, USA
| | - Bal L. Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA;
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
9
|
Butler W, Huang J. Neuroendocrine cells of the prostate: Histology, biological functions, and molecular mechanisms. PRECISION CLINICAL MEDICINE 2021; 4:25-34. [PMID: 33842835 PMCID: PMC8023015 DOI: 10.1093/pcmedi/pbab003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is a common cause of cancer-related mortality in men worldwide. Although most men are diagnosed with low grade, indolent tumors that are potentially curable, a significant subset develops advanced disease where hormone therapy is required to target the androgen receptor (AR). Despite its initial effect, hormone therapy eventually fails and the tumor progresses to lethal stages even through continued inhibition of AR. This review article focuses on the role of PCa cellular heterogeneity in therapy resistance and disease progression. Although AR-positive luminal-type cells represent the vast majority of PCa cells, there exists a minor component of AR-negative neuroendocrine (NE) cells that are resistant to hormonal therapy and are enriched by the treatment. In addition, it is now well accepted that a significant subset of hormonally treated tumors recur as small cell neuroendocrine carcinoma (SCNC), further highlighting the importance of targeting NE cells in addition to the more abundant luminal-type cancer cells. Although it has been long recognized that NE cells are present in PCa, their underlying function in benign prostate and molecular mechanisms contributing to PCa progression remains poorly understood. In this article, we review the morphology and function of NE cells in benign prostate and PCa as well as underlying molecular mechanisms. In addition, we review the major reported mechanisms for transformation from common adenocarcinoma histology to the highly lethal SCNC, a significant clinical challenge in the management of advanced PCa.
Collapse
Affiliation(s)
- William Butler
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
10
|
Huang H, Liu R, Huang Y, Feng Y, Fu Y, Chen L, Chen Z, Cai Y, Zhang Y, Chen Y. Acetylation-mediated degradation of HSD17B4 regulates the progression of prostate cancer. Aging (Albany NY) 2020; 12:14699-14717. [PMID: 32678070 PMCID: PMC7425433 DOI: 10.18632/aging.103530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/27/2020] [Indexed: 01/09/2023]
Abstract
Steroidogenic enzymes are crucial in prostate cancer (PCa) progression. 17β-Hydroxysteroid dehydrogenase type 4 (HSD17B4), encoded by HSD17B4, lacks catalytic capacity in androgen metabolism. Now the detailed role and molecular mechanism of PCa development are largely unknown. Here we showed that the expression of HSD17B4 was increased in PCa tissues compared to paired paratumor tissues. HSD17B4 knockdown in PCa cells significantly suppressed its proliferation, migration and invasion, while overexpressing HSD17B4 had opposite effects. Mechanistically, we found that the protein level of HSD17B4 was regulated by its acetylation at lysine 669(K669). Dihydroxytestosterone (DHT) treatment increased HSD17B4 acetylation and then promoted its degradation via chaperone-mediated autophagy (CMA). SIRT3 directly interacted with HSD17B4 to inhibit its acetylation and enhance its stability. In addition, we identified CREBBP as a regulator of the K669 acetylation and degradation of HSD17B4, affecting PC cell proliferation, migration and invasion. Notably, in PCa tissues and paired paratumor tissues, the level of HSD17B4 was negatively correlated with its K669 acetylation. Taken together, this study identified a novel role of HSD17B4 in PCa progression and suggested that HSD17B4 and its upstream regulators may be potential therapeutic targets for PCa intervention.
Collapse
Affiliation(s)
- Huichao Huang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Ruijie Liu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China.,Department of Pathology, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Yahui Huang
- Department of Pathology, XuChang Central Hospital, XuChang 461670, China
| | - Yilu Feng
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Lin Chen
- Molecular and Computational Biology Program, Departments of Biological Sciences and Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhuchu Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Yi Cai
- Department of Urology, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, XiangYa Hospital, Central South University, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, XiangYa Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
11
|
Lim J, Amantakul A, Shariff N, Lojanapiwat B, Alip A, Ong TA, Thevarajah S, Ahmayuddin F, Mathew A, Sriplakich S, Vuthiwong J, Chong FLT, Saad M. Clinical outcomes of abiraterone acetate and predictors of its treatment duration in metastatic castration-resistant prostate cancer: Real-world experience in the Southeast Asian cohort. Cancer Med 2020; 9:4613-4621. [PMID: 32374087 PMCID: PMC7333845 DOI: 10.1002/cam4.3101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
It is of much interest to understand the efficacy of abiraterone acetate (AA) in routine clinical practice. We assessed the clinical outcome of AA in patients with metastatic castration-resistant prostate cancer (mCRPC) and determined clinical factors associated with AA treatment duration in real-world setting. This real-world cohort consisted of 93 patients with mCRPC treated with AA in Thailand (58.1%) and Malaysia (41.9%). Primary endpoints were overall survival (OS) and biochemical progression-free survival (bPFS). Secondary endpoints were predictors associated with AA treatment duration evaluated with Cox proportional hazards regression. Around 74% were chemotherapy-naïve. The median AA treatment duration was 10 months (IQR 5.6-17.1). Malaysians had a relatively lower median OS and bPFS (OS 17.8 months; 95% CI 6.4-29.1, bPFS 10.4 months; 95% CI 8.8-12.0) compared to Thais (OS 27.0 months; 95% CI 11.3-42.7, bPFS 14.0 months; 95% CI 5.8-22.2), although it did not achieve statistical significance (P > .05). Patients with longer AA treatment duration (>10 months) had lower risk of death and longer bPFS, compared to those with shorter AA treatment duration (≤10 months) (hazard ratio [HR] 0.10, 95% CI 0.05-0.22 and HR 0.13, 95% CI 0.06-0.25, respectively). Multivariable analysis showed that PSA at AA initiation, presence of PSA response and chemotherapy-naive were independently associated with AA duration (P < .05). Abiraterone acetate is well-tolerated in the Southeast Asian cohort with comparable survival benefits to other Asian populations in real-world setting. Lower PSA levels at AA initiation, presence of PSA response, and chemotherapy-naive were significant in determining AA treatment duration.
Collapse
Affiliation(s)
- Jasmine Lim
- Department of SurgeryFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Akara Amantakul
- Department of SurgeryFaculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Nisha Shariff
- Department of Clinical OncologyFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | | | - Adlinda Alip
- Department of Clinical OncologyFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Teng Aik Ong
- Department of SurgeryFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | | | | | - Adeline Mathew
- Department of Radiotherapy and OncologySabah Women and Children HospitalKota KinabaluMalaysia
| | - Supon Sriplakich
- Department of SurgeryFaculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Jaraspong Vuthiwong
- Department of SurgeryFaculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Flora Li Tze Chong
- Department of Radiotherapy and OncologySabah Women and Children HospitalKota KinabaluMalaysia
| | - Marniza Saad
- Department of Clinical OncologyFaculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| |
Collapse
|
12
|
Zedan AH, Osther PJS, Assenholt J, Madsen JS, Hansen TF. Circulating miR-141 and miR-375 are associated with treatment outcome in metastatic castration resistant prostate cancer. Sci Rep 2020; 10:227. [PMID: 31937854 PMCID: PMC6959345 DOI: 10.1038/s41598-019-57101-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/20/2019] [Indexed: 12/31/2022] Open
Abstract
Metastatic castration resistant prostate cancer (mCRPC) is associated with high mortality, where monitoring of disease activity is still a major clinical challenge. The role of microRNAs (miRs) has been widely investigated in prostate cancer with both diagnostic and prognostic potential. The aim of this study was to investigate the relationship between circulating miRs and treatment outcome in mCRPC patients. The relative expression of five miRs (miR-93-5p, -125b-1-5p, -141-3p, -221-3p, and miR-375-3p) was investigated in plasma samples from 84 mCRPC patients; 40 patients were treated with docetaxel (DOC cohort) and 44 patients with abiraterone (ABI cohort). Blood was sampled at baseline before treatment start and at radiological progression. The plasma levels of four miRs; miR-93-5p, -141-3p, -221-3p, and miR-375-3p decreased significantly after treatment initiation in patients receiving docetaxel, and for miR-141-3p and miR-375-3p the level increased again at the time of radiological progression. In the patients treated with abiraterone, the plasma level of miR-221-3p likewise decreased significantly after the first treatment cycle. High baseline levels of both miR-141-3p and miR-375-3p were significantly associated with a shorter time to radiological progression in both cohorts. Additionally, high baseline levels of miR-141-3p and miR-221-3p were significantly associated with a shorter overall survival (OS) in the ABI cohort, while high levels of miR-141-3p and miR-375-3p were significantly associated with shorter OS in the DOC cohort. Plasma levels of miR-141-3p and miR-375-3p may predict time to progression in mCRPC patients treated with docetaxel or abiraterone. The clinical impact of these findings is dependent on validation in larger cohorts.
Collapse
Affiliation(s)
- A H Zedan
- Urological Research Centre, Department of Urology, Vejle Hospital, Vejle, Denmark. .,Department of Oncology, Vejle Hospital, Vejle, Denmark. .,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.
| | - P J S Osther
- Urological Research Centre, Department of Urology, Vejle Hospital, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - J Assenholt
- Department of Biochemistry and Clinical Immunology, Vejle Hospital, Vejle, Denmark
| | - J S Madsen
- Department of Biochemistry and Clinical Immunology, Vejle Hospital, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - T F Hansen
- Department of Oncology, Vejle Hospital, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
13
|
Kaipainen A, Zhang A, da Costa RMG, Lucas J, Marck B, Matsumoto AM, Morrissey C, True LD, Mostaghel EA, Nelson PS. Testosterone accumulation in prostate cancer cells is enhanced by facilitated diffusion. Prostate 2019; 79:1530-1542. [PMID: 31376206 PMCID: PMC6783279 DOI: 10.1002/pros.23874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/14/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Testosterone is a driver of prostate cancer (PC) growth via ligand-mediated activation of the androgen receptor (AR). Tumors that have escaped systemic androgen deprivation, castration-resistant prostate cancers (CRPC), have measurable intratumoral levels of testosterone, suggesting that a resistance mechanism still depends on androgen-simulated growth. However, AR activation requires an optimal intracellular concentration of androgens, a situation challenged by low circulating testosterone concentrations. Notably, PC cells may optimize their androgen levels by regulating the expression of steroid metabolism enzymes that convert androgen precursors into androgens. Here we propose that testosterone entry into the cell could be another control point. METHODS To determine whether testosterone enters cells via a transporter, we performed in vitro 3 H-testosterone uptake assays in androgen-dependent LNCaP and androgen and AR-independent PC3 cells. To determine if the uptake mechanism depended on a concentration gradient, we modified UGT2B17 levels in LNCaP cells and measured androgen levels by liquid-liquid extraction-mass spectrometry. We also analyzed CRPC metastases for expression of AKR1C3 to determine whether this enzyme that converts adrenal androgens to testosterone was present in the tumor stroma (microenvironment) in addition to its expression in the tumor epithelium. RESULTS Testosterone uptake followed a concentration gradient but unlike in passive diffusion, was saturable and temperature-dependent, thus suggesting facilitated transport. Suppression of UGT2B17 to abrogate a testosterone gradient reduced testosterone transport while overexpression of the enzyme enhanced it. The facilitated transport suggests a paracrine route of testosterone uptake for maintaining optimal intracellular levels. We found that AKR1C3 was expressed in the tumor microenvironment of CRPC metastases in addition to epithelial cells and the pattern of relative abundance of the enzyme in epithelium vs stroma varied substantially between the metastatic sites. CONCLUSIONS Our findings suggest that in addition to testosterone transport and metabolism by tumor epithelium, testosterone could also be produced by components of the tumor microenvironment. Facilitated testosterone uptake by tumor cells supports a cell nonautonomous mechanism for testosterone signaling in CRPC.
Collapse
Affiliation(s)
- Arja Kaipainen
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ailin Zhang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Rui M. Gil da Costa
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jared Lucas
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Brett Marck
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108
| | - Alvin M. Matsumoto
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Lawrence D. True
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, Seattle WA 98104
| | - Peter S. Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Medicine, University of Washington, Seattle WA 98104
| |
Collapse
|
14
|
Rice MA, Malhotra SV, Stoyanova T. Second-Generation Antiandrogens: From Discovery to Standard of Care in Castration Resistant Prostate Cancer. Front Oncol 2019; 9:801. [PMID: 31555580 PMCID: PMC6723105 DOI: 10.3389/fonc.2019.00801] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed cancer affecting men in the United States. The prostate is a hormone-dependent gland in which androgen hormones testosterone and dihydrotestosterone bind to and activate the androgen receptor, initiating nuclear translocation of androgen receptor and a subsequent signaling cascade. Due to the androgen dependency of the prostate, androgen deprivation therapies have emerged as first line treatment for aggressive prostate cancer. Such therapies are effective until the point at which prostate cancer, through a variety of mechanisms including but not limited to generation of ligand-independent androgen receptor splice variants, or intratumoral androgen production, overcome hormone deprivation. These cancers are androgen ablation resistant, clinically termed castration resistant prostate cancer (CRPC) and remain incurable. First-generation antiandrogens established androgen receptor blockade as a therapeutic strategy, but these therapies do not completely block androgen receptor activity. Efficacy and potency have been improved by the development of second-generation antiandrogen therapies, which remain the standard of care for patients with CRPC. Four second-generation anti-androgens are currently approved by the Food and Drug Administration (FDA); abiraterone acetate, enzalutamide, and recently approved apalutamide and darolutamide. This review is intended to provide a thorough overview of FDA approved second-generation antiandrogen discovery, treatment application, strategies for combination therapy to overcome resistance, and an insight for the potential future approaches for therapeutic inhibition of androgen receptor.
Collapse
Affiliation(s)
- Meghan A. Rice
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| | - Sanjay V. Malhotra
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, United States
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
15
|
Pantel K, Hille C, Scher HI. Circulating Tumor Cells in Prostate Cancer: From Discovery to Clinical Utility. Clin Chem 2019; 65:87-99. [DOI: 10.1373/clinchem.2018.287102] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/07/2018] [Indexed: 11/06/2022]
Abstract
Abstract
BACKGROUND
Prostate cancer represents the most common non–skin cancer type in men. Unmet needs include understanding prognosis to determine when intervention is needed and what type, prediction to guide the choice of a systemic therapy, and response indicators to determine whether a treatment is working. Over the past decade, the “liquid biopsy,” characterized by the analysis of tumor cells and tumor cell products such as cell-free nucleic acids (DNA, microRNA) or extracellular vesicles circulating in the blood of cancer patients, has received considerable attention.
CONTENT
Among those biomarkers, circulating tumor cells (CTCs) have been most intensively analyzed in prostate cancer. Here we discuss recent studies on the enumeration and characterization of CTCs in peripheral blood and how this information can be used to develop biomarkers for each of these clinical contexts. We focus on clinical applications in men with metastatic castration-resistant prostate cancer, in whom CTCs are more often detected and at higher numbers, and clinical validation for different contexts of use is most mature.
SUMMARY
The overall goal of CTC-based liquid biopsy testing is to better inform medical decision-making so that patient outcomes are improved.
Collapse
Affiliation(s)
- Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hille
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell College of Medicine, New York, NY
| |
Collapse
|
16
|
Gao X, Dai C, Huang S, Tang J, Chen G, Li J, Zhu Z, Zhu X, Zhou S, Gao Y, Hou Z, Fang Z, Xu C, Wang J, Wu D, Sharifi N, Li Z. Functional Silencing of HSD17B2 in Prostate Cancer Promotes Disease Progression. Clin Cancer Res 2018; 25:1291-1301. [PMID: 30228209 DOI: 10.1158/1078-0432.ccr-18-2392] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/08/2018] [Accepted: 09/14/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Steroidogenic enzymes are essential for prostate cancer development. Enzymes inactivating potent androgens were not investigated thoroughly, which leads to limited interference strategies for prostate cancer therapy. Here we characterized the clinical relevance, significance, and regulation mechanism of enzyme HSD17B2 in prostate cancer development. EXPERIMENTAL DESIGN HSD17B2 expression was detected with patient specimens and prostate cancer cell lines. Function of HSD17B2 in steroidogenesis, androgen receptor (AR) signaling, and tumor growth was investigated with prostate cancer cell lines and a xenograft model. DNA methylation and mRNA alternative splicing were investigated to unveil the mechanisms of HSD17B2 regulation. RESULTS HSD17B2 expression was reduced as prostate cancer progressed. 17βHSD2 decreased potent androgen production by converting testosterone (T) or dihydrotestosterone (DHT) to each of their upstream precursors. HSD17B2 overexpression suppressed androgen-induced cell proliferation and xenograft growth. Multiple mechanisms were involved in HSD17B2 functional silencing including DNA methylation and mRNA alternative splicing. DNA methylation decreased the HSD17B2 mRNA level. Two new catalytic-deficient isoforms, generated by alternative splicing, bound to wild-type 17βHSD2 and promoted its degradation. Splicing factors SRSF1 and SRSF5 participated in the generation of new isoforms. CONCLUSIONS Our findings provide evidence of the clinical relevance, significance, and regulation of HSD17B2 in prostate cancer progression, which might provide new strategies for clinical management by targeting the functional silencing mechanisms of HSD17B2.See related commentary by Mostaghel, p. 1139.
Collapse
Affiliation(s)
- Xiaomei Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Charles Dai
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Shengsong Huang
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jingjie Tang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Guoyuan Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China
| | - Jianneng Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ziqi Zhu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Xuyou Zhu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Shuirong Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Yuanyuan Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Zemin Hou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Zijun Fang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Chengdang Xu
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Jianyang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China.,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China.
| | - Nima Sharifi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio. .,Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Zhenfei Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, P.R. China. .,CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P.R. China
| |
Collapse
|
17
|
Del Re M, Crucitta S, Restante G, Rofi E, Arrigoni E, Biasco E, Sbrana A, Coppi E, Galli L, Bracarda S, Santini D, Danesi R. Pharmacogenetics of androgen signaling in prostate cancer: Focus on castration resistance and predictive biomarkers of response to treatment. Crit Rev Oncol Hematol 2018; 125:51-59. [DOI: 10.1016/j.critrevonc.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/24/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
|
18
|
Migita T, Takayama KI, Urano T, Obinata D, Ikeda K, Soga T, Takahashi S, Inoue S. ACSL3 promotes intratumoral steroidogenesis in prostate cancer cells. Cancer Sci 2017; 108:2011-2021. [PMID: 28771887 PMCID: PMC5623750 DOI: 10.1111/cas.13339] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/06/2017] [Accepted: 07/30/2017] [Indexed: 01/12/2023] Open
Abstract
Long‐chain acyl‐coenzyme A (CoA) synthetase 3 (ACSL3) is an androgen‐responsive gene involved in the generation of fatty acyl‐CoA esters. ACSL3 is expressed in both androgen‐sensitive and castration‐resistant prostate cancer (CRPC). However, its role in prostate cancer remains elusive. We overexpressed ACSL3 in androgen‐dependent LNCaP cells and examined the downstream effectors of ACSL3. Furthermore, we examined the role of ACSL3 in the androgen metabolism of prostate cancer. ACSL3 overexpression led to upregulation of several genes such as aldo‐keto reductase 1C3 (AKR1C3) involved in steroidogenesis, which utilizes adrenal androgen dehydroepiandrosterone sulfate (DHEAS) as substrate, and downregulated androgen‐inactivating enzyme UDP‐glucuronosyltransferase 2 (UGT2B). Exposure to DHEAS significantly increased testosterone levels and cell proliferative response in ACSL3‐overexpressing cells when compared to that in control cells. A public database showed that ACSL3 level was higher in CRPC than in hormone‐sensitive prostate cancer. CRPC cells showed an increased expression of ACSL3 and an expression pattern of AKR1C3 and UGT2B similar to ACSL3‐overexpressing cells. DHEAS stimulation significantly promoted the proliferation of CRPC cells when compared to that of LNCaP cells. These findings suggest that ACSL3 contributes to the growth of CRPC through intratumoral steroidogenesis (i.e. promoting androgen synthesis from DHEAS and preventing the catabolism of active androgens).
Collapse
Affiliation(s)
- Toshiro Migita
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken-Ichi Takayama
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomohiko Urano
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Daisuke Obinata
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Kazutaka Ikeda
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan.,RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Inoue
- Departments of Anti-Aging Medicine and Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
19
|
CX4945 suppresses the growth of castration-resistant prostate cancer cells by reducing AR-V7 expression. World J Urol 2017; 35:1213-1221. [PMID: 28105499 DOI: 10.1007/s00345-016-1996-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/21/2016] [Indexed: 02/08/2023] Open
Abstract
PURPOSE The aberrant expression of casein kinase 2 (CK2) has been reported to be involved in the tumorigenesis and progression of prostate cancer. The inhibition of CK2 activity represses androgen-dependent prostate cancer cells by attenuating the androgen receptor (AR) signaling pathway. In this study, we examined the effect of CK2 inhibition in castration-resistant prostate cancer (CRPC) cells, in which AR variants (ARVs) play a predominant role. METHODS A newly synthetic CK2 selective inhibitor CX4945 was utilized to study the effect of CK2 inhibition in CRPC cells by CCK8 assay and colony formation assay. Protein and mRNA levels of full-length AR (AR-FL) and AR-V7 were determined by qPCR and western blot, respectively. The nuclear translocation of p50 and p65 was assessed to reflect the activity of the NF-κB pathway. RESULTS CX4945 reduced the proliferation of CRPC cells in a dose-dependent and time-dependent manner. AR-V7 rather than AR-FL was downregulated by CX4945 in both the mRNA and protein level. Furthermore, CX4945 could restore the sensitivity of CRPC cells to bicalutamide. The analysis of possible mechanisms demonstrated that the inhibition of CK2 diminished the phosphorylation of p65 at ser529 and thus attenuated the activity of the NF-κB pathway. CONCLUSION The inhibition of CK2 by CX4945 can repress the viability of CRPC cells and restore their sensitivity to anti-androgen therapy by suppressing AR-V7. This finding presents a potential option for the treatment of prostate cancer, especially CRPC.
Collapse
|
20
|
Yaldo A, Wen L, Ogbonnaya A, Valderrama A, Kish J, Eaddy M, Kreilick C, Tangirala K, Shields K. Opioid Use Among Metastatic Prostate Cancer Patients With Skeletal-related Events. Clin Ther 2016; 38:1880-9. [PMID: 27478111 DOI: 10.1016/j.clinthera.2016.06.018] [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: 03/08/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/12/2022]
Abstract
PURPOSE The development of skeletal-related events (SREs) (pathologic fracture, need for surgery and/or radiation to bone, spinal cord compression, and hypercalcemia of malignancy) in metastatic prostate cancer (MPC) is associated with worsened pain and compromised quality of life. Opioids are frequently used throughout the course of SRE treatment. This study describes the treatment patterns and incremental use of opioids in MPC patients diagnosed with SREs. METHODS PC patients with bone metastases newly diagnosed with an SRE between January 1, 2005, and September 30, 2014, were identified using MarketScan Commercial and Medicare databases. Included patients were aged ≥40 years, had medical/pharmacy benefits for ≥12 months before (preindex) and ≥6 months after (postindex) diagnosis, and were without evidence of other primary cancers. Patients were categorized as nonusers of opioids (<10 days), short-term users (≥10 and <60 days), or long-term users (≥60 days) and further by SRE type. Opioid type, proportion of time on opioids, morphine-equivalent dose, adjuvant medications, and radiation use before and after SRE diagnosis were evaluated. FINDINGS A total of 1071 eligible patients were identified (mean age, 71 years; 10.8% had chronic pain at baseline). The most common SRE types present were radiation (60.2%), radiation and bone surgery (15.0%), pathologic fracture (7.2%), and bone surgery (6.5%). Opioid use increased from 49.9% preindex to 53.3% postindex (P < 0.0001). The proportion of time on opioids doubled after SRE (pre, 0.3 vs post, 0.6; P < 0.0001). A greater percentage of patients used only opioids after an SRE (pre, 11.0%; post, 46.1% [P < 0.0001]), while a lesser percentage of patients used only radiation after an SRE (pre, 36.0%; post, 4.7% [P < 0.0001]). An increase was observed in patients using neither radiation nor opioids (pre, 14.5%; post, 42.0% [P < 0.0001]). An increase of ~50% was noted in long-term opioid users (from 22.1% to 32.1%). The use of monotherapy with a short-acting opioid decreased (pre, 35.1%; post, 32.5% [P < 0.0001]), while use of mixed opioids increased (pre, 13.7%; post, 19.1% [P < 0.0001]). Mean morphine-equivalent dose increased from pre- to post-SRE (9.1 vs 13.1 mg). Bisphosphonate and NSAID users decreased from before to after an SRE diagnosis (bisphosphonates, 40.2% vs 8.6%; NSAIDs, 26.7% vs 17.5% [both, P < 0.0001]). IMPLICATIONS Long-term opioid use and dose were significantly increased after SRE development in MPC. The high percentage of patients not treated with an opioid or radiation potentially supports the need for additional treatment options for controlling pain if medically necessary and/or to prevent SREs.
Collapse
Affiliation(s)
- Avin Yaldo
- Bayer HealthCare Pharmaceuticals, Wayne, New Jersey
| | - Lonnie Wen
- Bayer HealthCare Pharmaceuticals, Wayne, New Jersey
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Takeda K, Hara N, Nishiyama T, Tasaki M, Ishizaki F, Tomita Y. Corepressive function of nuclear receptor coactivator 2 in androgen receptor of prostate cancer cells treated with antiandrogen. BMC Cancer 2016; 16:332. [PMID: 27225190 PMCID: PMC4880970 DOI: 10.1186/s12885-016-2378-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 05/23/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recruitment of cofactors in the interaction of the androgen receptor (AR) and AR ligands plays a critical role in determining androgenic/antiandrogenic effects of the AR ligand on signaling, but the functions of key cofactors, including nuclear receptor coactivator (NCOA), remain poorly understood in prostate cancer cells treated with AR ligands. METHODS We examined prostate cancer cell lines LNCaP and VCaP expressing mutated and wild-type ARs, respectively, to clarify the significance of NCOAs in the effect of antiandrogens. Hydroxyflutamide showed antagonistic activity against VCaP and an agonistic effect on LNCaP. Bicalutamide served as an antagonist for both. We analyzed mRNA transcription and protein expression of NCOAs in these cells pretreated with dihydrotestosterone and thereafter treated with the mentioned antiandrogens. Transcriptional silencing of candidate NCOAs and AR was performed using small interfering RNA (siRNA). Cell proliferation was evaluated with MTT assay. RESULTS LNCaP treated with bicalutamide showed an about four-fold increase in the expression of NCOA2 mRNA compared to those pretreated with dihydrotestosterone alone (P <0.01). In VCaP pretreated with dihydrotestosterone, transcriptions of NCOA2 and NCOA7 were slightly increased with bicalutamide (1.96- and 2.42-fold, respectively) and hydroxyflutamide (1.33-fold in both). With Western blotting, the expression of NCOA2 protein also increased in LNCaP cells treated with bicalutamide compared with that in control cells pretreated with dihydrotestosterone alone. Following silencing with siRNA for NCOA2, PSA levels in media with LNCaP receiving bicalutamide were elevated compared with those in non-silencing controls (101.6 ± 4.2 vs. 87.8 ± 1.4 ng/mL, respectively, P =0.0495). In LNCaP cells treated with dihydrotestosterone and bicalutamide, NCOA2-silencing was associated with a higher proliferation activity compared with non-silencing control and AR-silencing. CONCLUSION NCOA2, which has been thought to be recruited as a coactivator, possibly plays a corepressive role in AR of prostate cancer cells when treated with antiandrogens, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Keisuke Takeda
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Noboru Hara
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan. .,Division of Molecular Oncology, Department of Signal Transduction Research, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan. .,, Asahimachi 1, Niigata, 951-8510, Japan.
| | - Tsutomu Nishiyama
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masayuki Tasaki
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Fumio Ishizaki
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoshihiko Tomita
- Division of Urology, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
22
|
Soh SF, Yin X, Sun J, Li J, Yong EL, Wei Q, Gong Y. Simultaneous determination of multiple androgens in mice organs with liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal 2015; 115:457-66. [DOI: 10.1016/j.jpba.2015.07.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/09/2015] [Accepted: 07/26/2015] [Indexed: 12/23/2022]
|
23
|
Chu FM, Sartor O, Gomella L, Rudo T, Somerville MC, Hereghty B, Manyak MJ. A randomised, double-blind study comparing the addition of bicalutamide with or without dutasteride to GnRH analogue therapy in men with non-metastatic castrate-resistant prostate cancer. Eur J Cancer 2015; 51:1555-69. [DOI: 10.1016/j.ejca.2015.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/01/2015] [Accepted: 04/28/2015] [Indexed: 01/29/2023]
|
24
|
Schalken J, Fitzpatrick JM. Enzalutamide: targeting the androgen signalling pathway in metastatic castration-resistant prostate cancer. BJU Int 2015; 117:215-25. [PMID: 25818596 PMCID: PMC4744713 DOI: 10.1111/bju.13123] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significant progress has been made in the understanding of the underlying cancer biology of castration‐resistant prostate cancer (CRPC) with the androgen receptor (AR) signalling pathway remaining implicated throughout the prostate cancer disease continuum. Reactivation of the AR signalling pathway is considered to be a key driver of CRPC progression and, as such, the AR is a logical target for therapy in CRPC. The objective of this review was to understand the importance of AR signalling in the treatment of patients with metastatic CRPC (mCRPC) and to discuss the clinical benefits associated with inhibition of the AR signalling pathway. A search was conducted to identify articles relating to the role of AR signalling in CRPC and therapies that inhibit the AR signalling pathway. Current understanding of prostate cancer has identified the AR signalling pathway as a logical target for the treatment of CRPC. Available therapies that inhibit the AR signalling pathway include AR blockers, androgen biosynthesis inhibitors, and AR signalling inhibitors. Enzalutamide, the first approved AR signalling inhibitor, has a novel mode of action targeting AR signalling at three key stages. The direct mode of action of enzalutamide has been shown to translate into clinical responses in patients with mCRPC. In conclusion, the targeting of the AR signalling pathway in patients with mCRPC results in numerous clinical benefits. As the number of treatment options increase, more trials evaluating the sequencing and combination of treatments are required. This review highlights the continued importance of targeting a key driver in the progression of CRPC, AR signalling, and the clinical benefits associated with inhibition of the AR signalling pathway in the treatment of patients with CRPC.
Collapse
Affiliation(s)
- Jack Schalken
- Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - John M Fitzpatrick
- Department of Urology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Abstract
Bone metastases develop in most patients with metastatic castration-resistant prostate cancer (mCRPC). They affect the structural integrity of bone, manifesting as pain and skeletal-related events (SREs), and are the primary cause of patient disability, reduced quality of life (QOL) and death. Understanding the pathophysiology of bone metastases resulted in the development of agents that improve clinical outcome, suggesting that managing both the systemic disease and associated bone events is important. Historically, the treatment of CRPC bone metastases with early radiopharmaceuticals and external beam radiation therapy was largely supportive; however, now, zoledronic acid and denosumab are integral to the therapeutic strategy for mCRPC. These agents substantially reduce skeletal morbidity and improve patient QOL. Radium-223 dichloride is the first bone-targeting agent to show improved survival and reduced pain and symptomatic skeletal events in patients with mCRPC without visceral disease. Five other systemic agents are currently approved for use in mCRPC based on their ability to improve survival. These include the cytotoxic drugs docetaxel and cabazitaxel, the hormone-based therapies, abiraterone and enzalutamide, and the immunotherapeutic vaccine sipuleucel-T. Abiraterone and enzalutamide are able to reduce SREs and improve survival in this setting. Novel agents targeting tumour and bone cells are under clinical development.
Collapse
|
26
|
Labrie F. Combined blockade of testicular and locally made androgens in prostate cancer: a highly significant medical progress based upon intracrinology. J Steroid Biochem Mol Biol 2015; 145:144-56. [PMID: 24925260 DOI: 10.1016/j.jsbmb.2014.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/15/2014] [Accepted: 05/20/2014] [Indexed: 10/25/2022]
Abstract
Recently two drugs, namely the antiandrogen MDV-3100 and the inhibitor of 17α-hydroxylase abiraterone have been accepted by the FDA for the treatment of castration-resistant prostate cancer (CRPC) with or without previous chemotherapy, with a prolongation of overall survival of 2.2-4.8 months. While medical (GnRH agonist) or surgical castration reduces the serum levels of testosterone by about 97%, an important concentration of testosterone and dihydrotestosterone remains in the prostate and activates the androgen receptor (AR), thus offering an explanation for the positive data obtained in CRPC. In fact, explanation of the response observed with MDV-3100 or enzalutamide in CRPC is essentially a blockade of the action or formation of intraprostatic androgens. In addition to the inhibition of the action or formation of androgens made locally by the mechanisms of intracrinology, increased AR levels and AR mutations can be involved, especially in very advanced disease. Future developments look at more efficient inhibitors of the action or formation of intraprostatic androgens and starting treatment earlier when blockade of androgens can exert long-term control and even cure prostate cancer treated at a stage before the appearance of metastases. This article is part of a Special Issue entitled 'Essential role of DHEA'.
Collapse
|
27
|
Kosaka T, Miyajima A, Yasumizu Y, Miyazaki Y, Kikuchi E, Oya M. Limited in vitro efficacy of CYP17A1 inhibition on human castration resistant prostate cancer. Steroids 2014; 92:39-44. [PMID: 25150014 DOI: 10.1016/j.steroids.2014.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 07/17/2014] [Accepted: 07/26/2014] [Indexed: 01/02/2023]
Abstract
Although accumulating evidence indicates high expression of CYP17A1(P45017A1) allows castration resistant prostate cancer (CRPC) to maintain high intratumoral androgen levels, the potential P45017A1 activity has not been characterized yet. The aim of this study was to examine the potential CYP17A1 activity including 17α-hydroxylase and 17,20-lyase activities in human CRPC and the effect of a CYP17A inhibitor. We used three human CRPC cell lines: C4-2 and C4-2AT6 which was established from C4-2 under androgen ablation conditions for 6months, and PC3. To ascertain the potential CYP17A1 activity, we cultured with the steroid precursors: (13)C-[2,3,4]-progesterone (13C-Prog), and analyzed the sequential biosynthesis (13)C-[2,3,4]-17-hydroxyprogesterone (13C-17OHP) and (13)C-[2,3,4]-androstenedione(13C-Adione) by liquid chromatography/mass spectrometry (LC/MS/MS).The C4-2AT6 cells showed significantly higher CYP17A1 expression than C4-2 cells (p<0.001). LC/MS/MS analysis enabled us to detect the 13C-17-OHP and 13C-A-dione in these cell lines. The concentration ratio of 13C-Adione/13C-17OHP (Adione-17OHP ratio), which is thought to reflect the differences between 17-hydroxylase and 17,20-lyase activities, was then determined. The Adione-17OHP ratio in C4-2AT6 cells was significantly higher than that of C4-2 cells (p<0.001). Abiraterone were able to inhibit the CYP17A activities, although abiraterone did not have anti-proliferative effects on C4-2 and C4-2AT6 cells at clinically achievable concentrations of <1000nM in vitro. The present study clearly demonstrates CRPC have the dual activities of CYP17A1 mediated by 17-hydroxylase activity and 17,20-lyase activity. Abiraterone doesn't have an in vitro anti-proliferative efficacy in CRPC cells, suggesting limited efficacy in vitro.
Collapse
Affiliation(s)
- Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Akira Miyajima
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yasumasa Miyazaki
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Eiji Kikuchi
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
28
|
Kosaka T, Miyajima A, Oya M. Is DHT Production by 5α-Reductase Friend or Foe in Prostate Cancer? Front Oncol 2014; 4:247. [PMID: 25279351 PMCID: PMC4165281 DOI: 10.3389/fonc.2014.00247] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/29/2014] [Indexed: 01/01/2023] Open
Abstract
The first advance in the history of studies on prostate cancer (PCa) and androgens was the development of treatment with castration and administration of estrogen by Charles B. Huggins, who won the Nobel Prize in Physiology and Medicine. Since then, and for 70 years, androgen deprivation therapy has been the standard therapy for advanced PCa and the center of studies on PCa. However, recent advances have shed light on the relationship between androgens and the development or the progression of PCa. The use of 5AR inhibitors to prevent progression of PCa continues to be widely discussed. Discussion has been fueled by the findings of two large randomized, placebo-controlled trials: the Prostate Cancer Prevention Trial with finasteride and the Reduction by Dutasteride of Prostate Cancer Events trial. Does the development of PCa or progression to castration-resistant PCa depend on dihydrotestosterone (DHT)? Here, we summarize and discuss recent topics of local androgen production of DHT in PCa.
Collapse
Affiliation(s)
- Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Akira Miyajima
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Schmidt A, Meissner RS, Gentile MA, Chisamore MJ, Opas EE, Scafonas A, Cusick TE, Gambone C, Pennypacker B, Hodor P, Perkins JJ, Bai C, Ferraro D, Bettoun DJ, Wilkinson HA, Alves SE, Flores O, Ray WJ. Identification of an anabolic selective androgen receptor modulator that actively induces death of androgen-independent prostate cancer cells. J Steroid Biochem Mol Biol 2014; 143:29-39. [PMID: 24565564 DOI: 10.1016/j.jsbmb.2014.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/10/2014] [Accepted: 02/14/2014] [Indexed: 11/26/2022]
Abstract
Prostate cancer (PCa) initially responds to inhibition of androgen receptor (AR) signaling, but inevitably progresses to hormone ablation-resistant disease. Much effort is focused on optimizing this androgen deprivation strategy by improving hormone depletion and AR antagonism. However we found that bicalutamide, a clinically used antiandrogen, actually resembles a selective AR modulator (SARM), as it partially regulates 24% of endogenously 5α-dihydrotestosterone (DHT)-responsive genes in AR(+) MDA-MB-453 breast cancer cells. These data suggested that passive blocking of all AR functions is not required for PCa therapy. Hence, we adopted an active strategy that calls for the development of novel SARMs, which induce a unique gene expression profile that is intolerable to PCa cells. Therefore, we screened 3000 SARMs for the ability to arrest the androgen-independent growth of AR(+) 22Rv1 and LNCaP PCa cells but not AR(-) PC3 or DU145 cells. We identified only one such compound; the 4-aza-steroid, MK-4541, a potent and selective SARM. MK-4541 induces caspase-3 activity and cell death in both androgen-independent, AR(+) PCa cell lines but spares AR(-) cells or AR(+) non-PCa cells. This activity correlates with its promoter context- and cell-type dependent transcriptional effects. In rats, MK-4541 inhibits the trophic effects of DHT on the prostate, but not the levator ani muscle, and triggers an anabolic response in the periosteal compartment of bone. Therefore, MK-4541 has the potential to effectively manage prostatic hypertrophic diseases owing to its antitumor SARM-like mechanism, while simultaneously maintaining the anabolic benefits of natural androgens.
Collapse
MESH Headings
- Anabolic Agents/chemistry
- Anabolic Agents/pharmacology
- Androgen Receptor Antagonists/pharmacology
- Androgens/pharmacology
- Animals
- Apoptosis/drug effects
- Azasteroids/chemistry
- Azasteroids/pharmacology
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carbamates/chemistry
- Carbamates/pharmacology
- Cell Proliferation/drug effects
- Combinatorial Chemistry Techniques
- Female
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
- Real-Time Polymerase Chain Reaction
- Receptors, Androgen/chemistry
- Receptors, Androgen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Azriel Schmidt
- Departments of Molecular Endocrinology, West Point, PA 19486, USA.
| | | | | | | | - Evan E Opas
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - Angela Scafonas
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - Tara E Cusick
- Departments of Molecular Endocrinology, West Point, PA 19486, USA; Departments of Medicinal Chemistry, West Point, PA 19486, USA; Departments of Molecular Profiling Merck & Co., West Point, PA 19486, USA
| | - Carlo Gambone
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | | | - Paul Hodor
- Departments of Molecular Profiling Merck & Co., West Point, PA 19486, USA
| | - James J Perkins
- Departments of Medicinal Chemistry, West Point, PA 19486, USA
| | - Chang Bai
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - Damien Ferraro
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - David J Bettoun
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | | | - Stephen E Alves
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - Osvaldo Flores
- Departments of Molecular Endocrinology, West Point, PA 19486, USA
| | - William J Ray
- Departments of Molecular Endocrinology, West Point, PA 19486, USA.
| |
Collapse
|
30
|
Milecki T, Antczak A, Kwias Z, Milecki P. The use of luteinizing hormone-releasing hormone analogues is still an indispensable element of therapy in castrate-resistant prostate cancer. Contemp Oncol (Pozn) 2014; 18:85-9. [PMID: 24966789 PMCID: PMC4068814 DOI: 10.5114/wo.2014.42723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/22/2014] [Accepted: 03/26/2014] [Indexed: 11/17/2022] Open
Abstract
Metastatic prostate cancer, which shows progression despite castration testosterone levels, was previously defined as hormone-refractory. This definition has recently been changed to the one presently used - castrate-resistant prostate cancer. Numerous fundamental studies have provided evidence that the development of hormone-refractory prostate cancer is constantly dependent on the concentration of androgens. The aim of the metastatic castrate-resistant prostate cancer (mCRPC) treatment is currently to obtain the lowest possible androgen concentration. The effectiveness of such management has been proven by the results of clinical studies on the latest hormonal and chemotherapeutic medications. In the last two decades, new effective chemotherapeutics have become available on the market: abiraterone, enzalutamide, docetaxel, cabazitaxel, zoldronic acid, denosumab and alpharadin They significantly contribute to extending patients' survival and to improving their quality of life. Therefore, the question arises whether using luteinizing hormone-releasing hormone (LHRH) analogues is still a necessary element of the therapy. A detailed analysis of study regimens involving the above-mentioned medications and of available publications supports the view that LHRH analogues are the basic strategy in the treatment of patients with mCRPC. All clinical trials evaluating new therapies still followed the principle of obtaining castration testosterone levels as a result of using LHRH analogues simultaneously with the new medications.
Collapse
Affiliation(s)
- Tomasz Milecki
- Department and Clinic of Urology and Urologic Oncology, Poznan University of Medical Science, Poland
| | - Andrzej Antczak
- Department and Clinic of Urology and Urologic Oncology, Poznan University of Medical Science, Poland
| | - Zbigniew Kwias
- Department and Clinic of Urology and Urologic Oncology, Poznan University of Medical Science, Poland
| | - Piotr Milecki
- Department of Radiotherapy, Greater Poland Cancer Centre, Poznan, Poland
- Chair and Department of Electroradiology, Poznan University of Medical Sciences, Poland
| |
Collapse
|
31
|
Fitzgerald KA, Evans JC, McCarthy J, Guo J, Prencipe M, Kearney M, Watson WR, O'Driscoll CM. The role of transcription factors in prostate cancer and potential for future RNA interference therapy. Expert Opin Ther Targets 2014; 18:633-49. [DOI: 10.1517/14728222.2014.896904] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Lim M, Otto-Duessel M, He M, Su L, Nguyen D, Chin E, Alliston T, Jones JO. Ligand-independent and tissue-selective androgen receptor inhibition by pyrvinium. ACS Chem Biol 2014; 9:692-702. [PMID: 24354286 PMCID: PMC3962707 DOI: 10.1021/cb400759d] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pyrvinium pamoate (PP) is a potent noncompetitive inhibitor of the androgen receptor (AR). Using a novel method of target identification, we demonstrate that AR is a direct target of PP in prostate cancer cells. We demonstrate that PP inhibits AR activity via the highly conserved DNA binding domain (DBD), the only AR inhibitor that functions via this domain. Furthermore, computational modeling predicts that pyrvinium binds at the interface of the DBD dimer and the minor groove of the AR response element. Because PP acts through the DBD, PP is able to inhibit the constitutive activity of AR splice variants, which are thought to contribute to the growth of castration resistant prostate cancer (CRPC). PP also inhibits androgen-independent AR activation by HER2 kinase. The antiandrogen activity of pyrvinium manifests in the ability to inhibit the in vivo growth of CRPC xenografts that express AR splice variants. Interestingly, PP was most potent in cells with endogenous AR expression derived from prostate or bone. PP was able to inhibit several other hormone nuclear receptors (NRs) but not structurally unrelated transcription factors. PP inhibition of other NRs was similarly cell-type selective. Using dual-energy X-ray absorptiometry, we demonstrate that the cell-type specificity of PP manifests in tissue-selective inhibition of AR activity in mice, as PP decreases prostate weight and bone mineral density but does not affect lean body mass. Our results suggest that the noncompetitive AR inhibitor pyrvinium has significant potential to treat CRPC, including cancers driven by ligand-independent AR signaling.
Collapse
Affiliation(s)
- Minyoung Lim
- Department of Molecular Pharmacology, ‡Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center , Duarte, California 91010, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Pinto Á. Beyond abiraterone: new hormonal therapies for metastatic castration-resistant prostate cancer. Cancer Biol Ther 2014; 15:149-55. [PMID: 24100689 PMCID: PMC3928129 DOI: 10.4161/cbt.26724] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 09/10/2013] [Accepted: 10/06/2013] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer is a heterogeneous disease where the previous concept of "hormone resistance" has been changed by a new generation of hormonal therapies that have proven efficacy in the castration-resistant setting. The fact is that androgens play a crucial role in the whole clinical course of prostate cancer, even when a patient meets castration-resistance criteria. The development of abiraterone showed how important and clinically meaningful can be to achieve the lowest possible levels of testosterone, and androgen receptor overexpression, mutation, or enhanced crosstalk with other pathways, which can also be targeted with new agents tested in the last few years. New androgen biosynthesis inhibitors have been developed, such as orteronel (TAK-700), but also new antiandrogens (enzalutamide, ARN-509, ODM-201) or even agents with a dual mechanism of action (galeterone). In this review the development of new hormonal therapies following the arrival of abiraterone for the treatment of prostate cancer will be summarized.
Collapse
Affiliation(s)
- Álvaro Pinto
- Medical Oncology Department; University Hospital La Paz; IdiPAZ; Madrid, Spain
| |
Collapse
|
34
|
Ahmed A, Ali S, Sarkar FH. Advances in androgen receptor targeted therapy for prostate cancer. J Cell Physiol 2014; 229:271-6. [PMID: 24037862 DOI: 10.1002/jcp.24456] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/14/2013] [Indexed: 12/31/2022]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Current research findings suggest that the androgen receptor (AR) and its signaling pathway contribute significantly to the progression of metastatic PCa. The AR is a ligand activated transcription factor, where androgens such as testosterone (T) and dihydroxytestosterone (DHT) act as the activating ligands. However in many metastatic PCa, the AR functions promiscuously and is constitutively active through multiple mechanisms. Inhibition of enzymes that take part in androgen synthesis or synthesizing antiandrogens that can inhibit the AR are two popular methods of impeding the androgen receptor signaling axis; however, the inhibition of androgen-independent activated AR function has not yet been fully exploited. This article focuses on the development of emerging novel agents that act at different steps along the androgen-AR signaling pathway to help improve the poor prognosis of PCa patients.
Collapse
Affiliation(s)
- Alia Ahmed
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | | | | |
Collapse
|
35
|
Lebdai S, Branchereau J, Robert G, De La Taille A, Bouchaert P. [Corticotherapy in castration-resistant prostate cancer]. Prog Urol 2013; 23 Suppl 1:S23-33. [PMID: 24314736 DOI: 10.1016/s1166-7087(13)70043-5] [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] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Corticosteroids are commonly used in the treatment of prostate cancer resistant to castration (PCRC), partly due to the inhibitory effects on adrenal androgen production acting as a pituitary suppressant. METHODS A literature search was conducted in PubMed/MEDLINE database using the following key words: prostate cancer; castration resistance; metastasis; corticotherapy. RESULTS Corticosteroids exert direct anti-tumoral activities mediated by the glucocorticoids receptor and involving cellular/tissue functions as growth, apoptosis, inflammation, metastasis, differentiation and angiogenesis. As a pain relieving agents, corticosteroids significantly relieve PCRC clinical symptoms, especially those due to bone metastasis. In the comparative arm of phase II-III trials, corticosteroids administered daily produce a PSA decline. Among the adverse effects due to corticosteroids, bone loss and cardiovascular risk should be carefully monitored. In association with abiraterone acetate, corticosteroids increase overall survival in PCRC patients, and reduce the mineralocorticoid side effects of abiraterone. CONCLUSION Corticosteroids in monotherapy for PCRC have a limited efficacy. In association with abiraterone acetate it reduces the mineralocorticoid toxicity and enhances the androgenic suppression.
Collapse
Affiliation(s)
- S Lebdai
- Service d'urologie, CHU Angers, 4 rue Larrey, 49100 Angers, France
| | | | | | | | | |
Collapse
|
36
|
11β-hydroxyandrostenedione returns to the steroid arena: biosynthesis, metabolism and function. Molecules 2013; 18:13228-44. [PMID: 24165582 PMCID: PMC6270415 DOI: 10.3390/molecules181113228] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 01/22/2023] Open
Abstract
The biological significance of 11β-hydroxyandrostenedione (11OHA4) has eluded researchers for the past six decades. It is now known that 11OHA4 is biosynthesized in the androgen arm of the adrenal steroidogenesis pathway and subsequently metabolized by steroidogenic enzymes in vitro, serving as precursor to recognized and novel androgenic steroids. These in vitro findings extend beyond the adrenal, suggesting that 11OHA4 could be metabolized in steroid-responsive peripheral tissues, as is the case for androgen precursor metabolites of adrenal origin. The significance thereof becomes apparent when considering that the metabolism of 11OHA4 in LNCaP androgen dependent prostate cancer cells yields androgenic steroid metabolites. It is thus possible that 11OHA4 may be metabolized to yield ligands for steroid receptors in not only the prostate but also in other steroid-responsive tissues. Future investigations of 11OHA4 may therefore characterize it as a vital steroid with far-reaching physiological consequences. An overview of the research on 11OHA4 since its identification in 1953 will be presented, with specific focus on the most recent works that have advanced our understanding of its biological role, thereby underscoring its relevance in health and disease.
Collapse
|
37
|
Ripert T, Crouzet S, Ploussard G, De La Taille A, Robert G. Mécanismes de résistance aux in hibiteurs de CYP17A1 dans le cancer de la prostate résistant à la castration. Prog Urol 2013; 23 Suppl 1:S16-22. [DOI: 10.1016/s1166-7087(13)70042-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Murez T, Basset V, Audenet F, Lebret T, Branchereau J. Régulation hormonale qualitative et quantitative dans le cancer de la prostate résistant à la castration. Prog Urol 2013; 23 Suppl 1:S1-8. [DOI: 10.1016/s1166-7087(13)70040-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Kosaka T, Miyajima A, Nagata H, Maeda T, Kikuchi E, Oya M. Human castration resistant prostate cancer rather prefer to decreased 5α-reductase activity. Sci Rep 2013; 3:1268. [PMID: 23429215 PMCID: PMC3572449 DOI: 10.1038/srep01268] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 01/28/2013] [Indexed: 01/22/2023] Open
Abstract
Physiologically relevant steroid 5α-reductase (SRD5A) activity that is essential for dihydrotestosterone (DHT) biosynthesis in human castration-resistant prostate cancer (CRPC) has not been fully characterized yet. In this study to ascertain the potential SRD5A activity, we cultured two human CRPC cell lines, C4-2 and C4-2AT6, with the steroid precursor: 13C-[2,3,4]-androstenedione (13C-Adione), and analyzed the sequential biosynthesis of 13C-[2,3,4]-testosterone (13C-T) and 13C-[2,3,4]-DHT (13C-DHT) by liquid chromatography/mass spectrometry (LC/MS/MS). The 13C-DHT/13C-T concentration ratio detected by LC/MS/MS in C4-2AT6 cells appeared to reflect the SRD5A activity. The ratio in C4-2AT6 was significantly lower than that in C4-2. An increased concentration of DHT did not have a positive effect on cell proliferation, rather it exhibited inhibitory effects. 5α-reductase inhibitors did not have any inhibitory effect at clinically achievable concentrations. These results indicate that CRPC cells may have an unknown regulation system to protect themselves from an androgenic suppressive effect mediated by SRD5A activity.
Collapse
Affiliation(s)
- Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The palliative goal of the treatment of metastatic prostate cancer is to prolong survival and decrease cancer-related complications. Androgen ablation therapy is widely accepted as the initial treatment of choice; when the disease becomes resistant to castration-resistant prostate cancer (CRPC), docetaxel-based chemotherapy aids in prolonging overall survival and controlling disease-related symptoms. Until a few years ago, no drug had showed efficacy in docetaxel-resistant patients. Recently, cabazitaxel, a taxane family compound, has been shown to help prolong survival in patients previously treated with docetaxel, even if a high grade of myelotoxicity has been reported. Moreover, a better understanding of the biology of CRPC has demonstrated that prostate cancer proliferation is largely mediated through the androgen receptor, which could be reactivated by androgens produced by the adrenal glands. Abiraterone acetate is an orally active acetate salt of the steroidal compound abiraterone with antiandrogen activity. Abiraterone inhibits the enzymatic activity of steroid 17α-monooxygenase, a member of the cytochrome P450 family that catalyzes the 17α-hydroxylation of steroid intermediates involved in testosterone synthesis from the adrenal glands. This review focuses on abiraterone acetate, the first compound that, through the inhibition of adrenal gland production of testosterone, increases the overall survival in CRPC patients. The role of possible predictive biomarkers and future perspectives are also discussed.
Collapse
|
41
|
Hendrix LN, Hamilton DA, Kyprianou N. Emerging therapeutics targeting castration-resistant prostate cancer: the AR-mageddon of tumor epithelial-mesenchymal transition. Expert Rev Endocrinol Metab 2013; 8:403-416. [PMID: 30736155 DOI: 10.1586/17446651.2013.811914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Advanced prostate cancer will claim nearly 30,000 lives among men in the USA in the year 2013. Most of these will be castration-resistant prostate cancers that are not responsive to traditional therapeutic modalities, and there is no available regimen that fully eradicates metastatic disease. This poses a significant clinical challenge for practitioners and has stimulated the development of novel agents that target these castration-resistant tumor cells. Development of metastatic prostate cancer is orchestrated by multiple signaling pathways that regulate cell survival, apoptosis, anoikis, epithelial-mesenchymal transition (EMT), invasion, the androgen signaling axis and angiogenesis. Disruption of the mechanisms underlying these processes is critical for development of agents that can target otherwise resistant tumor cells. Insights into the mechanisms by which rounds of EMT/mesenchymal-epithelial transition conversions facilitate the progression of localized prostate carcinomas to advanced metastatic and castration-resistant disease emerge as attractive targets for drug development. In this review, the authors discuss the current understanding of therapeutic resistance in castration-resistant prostate cancer with focus on the androgen receptor signaling axis and EMT. Novel therapeutic approaches targeting critical players of both pathways as well as the results from ongoing clinical trials will be discussed in this review.
Collapse
Affiliation(s)
- Lauren N Hendrix
- a Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - David A Hamilton
- a Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Natasha Kyprianou
- b Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- c Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
- d Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
42
|
The androgen receptor, androgen synthesis, and new designer antiandrogens for metastatic castration-resistant prostate cancer: teaching old dogs new tricks. Am J Ther 2013; 20:128-31. [PMID: 23466618 DOI: 10.1097/mjt.0b013e3182857f8e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
[Novel agents for the therapy of castration-resistant prostate cancer: overview of pivotal studies and new strategies to come]. Prog Urol 2013; 23:1-7. [PMID: 23287477 DOI: 10.1016/j.purol.2012.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 05/14/2012] [Accepted: 07/11/2012] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Recently, new agents have been developed in the treatment of prostate cancer. Our aim was to review phase III studies that involved novel agents in the treatment of castration resistant prostate cancer. METHODS PubMed databases were searched for original articles published with the search terms: prostate cancer, castration resistant, metastatic, targeted therapy, biologic agents, immunotherapy and clinical trials. Proceedings from 2008 of conferences of the American Society of Clinical Oncology, American Urological Association, and the European Association of Urology were also searched. We included phase III studies that involved: abiraterone, MDV 3100, cabazitaxel, sipuleucel-T, radium-223, and denosumab. RESULTS Abiraterone and MDV 3100 are two new hormotherapies that showed an increased overall survival of 15 and 18 months respectively before after docetaxel based chemotherapy in randomized trials. Cabazitaxel became the standard second line chemotherapy after docetaxel. Sipuleucel-T has emerged as the first approved vaccine in prostate cancer. It showed a 22 % reduction of mortality and a prolonged survival time of 4.1 months compared to placebo. A radium-223 based metabolic radiotherapy has showed a better overall survival, delayed and reduced skeletal-related events in placebo controlled randomized trials. Denosumab also delayed the first skeletal-related event in a zoledronic acid controlled trial (20.7 versus 17.1 months, P=0.0002). Moreover, Denosumab delays bone metastases by 4.1 months compared to placebo. CONCLUSION The novel agents that emerged in the treatment of prostate cancer showed an efficacy in placebo controlled trials. They added new tools in the armamentarium of therapies of castration resistant prostate cancer.
Collapse
|
44
|
Gauthier S, Martel C, Labrie F. Steroid derivatives as pure antagonists of the androgen receptor. J Steroid Biochem Mol Biol 2012; 132:93-104. [PMID: 22449547 DOI: 10.1016/j.jsbmb.2012.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND While the androgens of testicular origin (representing about 50% of total androgens in men over 50 years) can be completely eliminated by surgical or medical castration with GnRH (gonadotropin-releasing hormone) agonists or antagonists, the antiandrogens currently available as blockers of androgen binding to the androgen receptor (AR), namely bicalutamide (BICA), flutamide (FLU) and nilutamide have too weak affinity to completely neutralize the other 50% of androgens made locally from dehydroepiandrosterone (DHEA) in the prostate cancer tissue by the mechanisms of intracrinology. MATERIALS AND METHODS Series of steroid derivatives having pure and potent antagonistic activity on the human and rodent AR were synthesized. Assays of AR binding and activity in carcinoma mouse Shionogi and human LNCaP cells as well as in vivo bioavailability measurements and in vivo prostate weight assays in the rat were used. RESULTS The chosen lead steroidal compound, namely EM-5854, has a 3.7-fold higher affinity than BICA for the human AR while EM-5855, an important metabolite of EM-5854, has a 94-fold higher affinity for the human AR compared to BICA. EM-5854 and EM-5855 are 14 times more potent than BICA in inhibiting androgen (R1881)-stimulated prostatic specific antigen (PSA) secretion in human prostatic carcinoma LNCaP cells in vitro. MDV3100 has a potency comparable to bicalutamide in these assays. Depending upon the oral formulation, EM-5854 is 5- to 10-times more potent than BICA to inhibit dihydrotestosterone (DHT)-stimulated ventral prostatic weight in vivo in the rat while MDV3100 has lower activity than BICA in this in vivo model. These data are supported by respective 40-fold and 105-fold higher potencies of EM-5854 and EM-5855 compared to BICA to inhibit cell proliferation in the androgen-sensitive Shionogi carcinoma cell model. CONCLUSIONS Although the present preclinical results data need evaluation in clinical trials in men, combination of the data obtained in vitro in human LNCaP cells as indicator of potency in the human prostate and the data on metabolism evaluated in vivo on ventral prostate weight in the rat, could suggest the possibility of a 70- to 140-fold higher potency of EM-5854 compared to bicalutamide (Casodex) for the treatment of prostate cancer in men.
Collapse
Affiliation(s)
- Sylvain Gauthier
- Endoresearch Inc., 2989, de la Promenade, Quebec City, QC, Canada
| | | | | |
Collapse
|
45
|
Green SM, Mostaghel EA, Nelson PS. Androgen action and metabolism in prostate cancer. Mol Cell Endocrinol 2012; 360:3-13. [PMID: 22453214 PMCID: PMC4124858 DOI: 10.1016/j.mce.2011.09.046] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Revised: 09/26/2011] [Accepted: 09/26/2011] [Indexed: 11/21/2022]
Abstract
The transcriptional programs regulated through the activity of the androgen receptor (AR) modulate normal prostate development and the maintenance of prostatic functions at maturity. AR signaling also controls key survival and growth functions operative in prostate cancer. Inhibiting the AR program remains the key target in the treatment of advanced prostate cancer, and suppressing AR also holds great potential for preventing the development or progression of early stage prostate cancer. In this review, we detail molecular mechanisms of AR activity, cellular components contributing to the maintenance of AR signaling despite AR-ligand suppression, and discuss treatment strategies designed to target components of resistance to AR-directed therapeutics.
Collapse
Affiliation(s)
- Sean M. Green
- Divisions of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Elahe A Mostaghel
- Clinical Research, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Peter S. Nelson
- Divisions of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA
- Corresponding Author Fred Hutchinson Cancer Research Center 1100 Fairview Ave NE, MS D4-100 Seattle, WA 98109 phone 206-667-3377 fax 206-667-2917
| |
Collapse
|
46
|
Mitani T, Harada N, Nakano Y, Inui H, Yamaji R. Coordinated action of hypoxia-inducible factor-1α and β-catenin in androgen receptor signaling. J Biol Chem 2012; 287:33594-606. [PMID: 22865883 DOI: 10.1074/jbc.m112.388298] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The androgen receptor (AR) acts as a ligand-dependent transcriptional factor and plays a critical role in the development and progression of androgen-dependent and castration-resistant prostate cancer. Castration results in hypoxia in prostate cancer cells, and hypoxia enhances transcriptional activity of AR through hypoxia-inducible factor (HIF)-1α at low serum androgen levels mimicking the castration-resistant stage. However, HIF-1α is necessary but not sufficient for hypoxia-activated AR transactivation, and the molecular mechanism that regulates AR function in castration-resistant prostate cancer remains unclear. Here, we report that β-catenin is required for HIF-1α-mediated AR transactivation in hypoxic LNCaP prostate cancer cells under low androgen conditions. HIF-1α and β-catenin coordinately enhanced AR N-terminal and C-terminal interaction. β-Catenin accumulated in the nucleus in the HIF-1α protein-positive cells of LNCaP xenografts in castrated mice. In LNCaP cells, when HIF-1α was knocked down or was exogenously expressed in the cytoplasm, hypoxia-induced nuclear localization of β-catenin was inhibited. β-Catenin formed a complex with HIF-1α both in the nucleus and in the cytoplasm. Hypoxia increased the amount of a complex composed of AR and β-catenin, and knockdown of HIF-1α attenuated the recruitment of AR and β-catenin to the androgen response elements (AREs) of androgen-responsive genes. Furthermore, together with β-catenin, HIF-1α bound to the AREs in the presence of androgen. These results demonstrate that (i) HIF-1α and β-catenin coordinately enhance AR transactivation by accelerating N-terminal and C-terminal interaction; (ii) HIF-1α promotes nuclear translocation of β-catenin in hypoxia; and (iii) AR, HIF-1α, and β-catenin form a ternary complex on AREs.
Collapse
Affiliation(s)
- Takakazu Mitani
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | | | | | | | | |
Collapse
|
47
|
Chrvala CA. The changing landscape of treatment options for metastatic castrate-resistant prostate cancer: challenges and solutions for physicians and patients. P & T : A PEER-REVIEWED JOURNAL FOR FORMULARY MANAGEMENT 2012; 37:453-463. [PMID: 23091338 PMCID: PMC3474424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Accepted: 06/13/2012] [Indexed: 06/01/2023]
Abstract
Four recently approved drugs (cabazitaxel, sipuleucel-T, abiraterone, and denosumab), along with emerging therapies, bone-building therapies, hormonal treatments, and immunotherapies, have all demonstrated promise in advanced prostate cancer. It appears that the best outcomes will be achieved from the sequential use of multiple agents.
Collapse
|
48
|
Venkateswaran S, Margel D, Yap S, Hersey K, Yip P, Fleshner NE. Comparison of serum testosterone levels in prostate cancer patients receiving LHRH agonist therapy with or without the removal of the prostate. Can Urol Assoc J 2012; 6:183-6. [PMID: 22664629 DOI: 10.5489/cuaj.11278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION : The prostate secretes enzymes and nutrients to promote sperm motility. Recent reports suggest that the prostate may also secrete testosterone, which is believed to be a fuel for prostate tumour growth. The aim of this study was to determine if a difference in serum testosterone levels exists between men on luteinizing hormone releasing-hormone (LHRH) agonists who have undergone radical prostatectomy, radiation or hormone therapy as primary prostate cancer treatment. METHODS : Serum testosterone levels were evaluated in 165 consecutive prostate cancer patients using LHRH analogues for >3 months. We excluded patients receiving either radiation or chemotherapy at time of time of testosterone measurement. Patients were classified based on primary treatment: (1) radical prostatectomy; (2) radiation; or (3) primary hormone therapy. We used one-way ANOVA to compare testosterone levels. Pearson correlation was used to correlate testosterone with prostate-specific antigen (PSA) and time on LHRH agonists. Multivariable linear regression was used to predict serum testosterone levels. RESULTS : The median (interquartile range) serum testosterone levels were 1.4 (1-1.9), 1.3 (1-1.625) and 1.25 (0.9-1.525) nmol/L for radical prostatectomy, radiation and primary hormone therapy groups, respectively. There was no statistically significant difference in testosterone levels between the groups (p = 0.3). No correlation was found between testosterone and PSA levels or time on LHRH (r = 0.02 and r = 0.01), respectively. Multivariable linear regression showed that none of the clinical variables were predictors of serum testosterone levels. CONCLUSION : Our study suggests that primary treatment does not affect serum testosterone levels among men using LHRH analogues.
Collapse
Affiliation(s)
- Seetha Venkateswaran
- Department of Surgery, Division of Urology, Princess Margaret Hospital, University Health Network, Toronto, ON
| | | | | | | | | | | |
Collapse
|
49
|
Levina E, Chen M, Carkner R, Shtutman M, Buttyan R. Paracrine Hedgehog increases the steroidogenic potential of prostate stromal cells in a Gli-dependent manner. Prostate 2012; 72:817-24. [PMID: 22025366 DOI: 10.1002/pros.21500] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 09/19/2011] [Indexed: 01/03/2023]
Abstract
Acquired intratumoral steroidogenesis is involved in progression of prostate cancer to castration resistant disease (CRPC) and a target for improved therapeutics. Recent work has shown that prostate cancer cells can acquire steroidogenic activity as they progress to a therapeutic-resistant state. However, benign prostate stromal cells (PrSCs) also have steroidogenic potential though they are often overlooked as a source of intratumoral androgens. Here, we present preliminary studies showing that the steroidogenic activity of primary human PrSCs is significantly increased by exposure to a Hedgehog agonist (SAG) or by transduction of PrSCs with lentiviruses that expresses active Gli2 (Gli2ΔN), a transcription factor that is triggered by Hh signaling. Comparative gene expression profiling on Chips, that was confirmed by quantitative real-time PCR, revealed that hedgehog agonist treatment induced in these cells expressions of hedgehog target genes (Gli1, Ptch1, and SCUBE1) plus a specific cadre of genes involved in cholesterol/steroid biosynthesis, metabolism, and transport. Genes involved downstream in steroid hormone generation, including CYP17A1 and CYP19A1 were also induced. Both the hedgehog agonist and the Gli2-expressing lentivirus significantly increased the output of testosterone (T) from PrSCs that were supplemented with dihydroepiandrosterone (DHEA), an adrenal precursor of T. Finally, knockdown of Gli2 by siRNA suppressed the ability of SAG to induce this response. Collectively, our data indicate that hedgehog/Gli signaling may be a factor in acquired intratumoral steroidogenesis of a prostate tumor through its actions on stromal cells in the tumor microenvironment and an influence for the development of CRPC.
Collapse
Affiliation(s)
- Elina Levina
- South Carolina College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, Columbia, South Carolina, USA
| | | | | | | | | |
Collapse
|
50
|
New and emerging agents for the treatment of castration-resistant prostate cancer. Urol Oncol 2012; 29:S1-8. [PMID: 22074657 DOI: 10.1016/j.urolonc.2011.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/24/2011] [Accepted: 08/24/2011] [Indexed: 11/20/2022]
Abstract
Most men with recurrent prostate cancer (CaP) initially respond to androgen deprivation therapy but eventually develop metastatic castration-resistant prostate cancer (CRPC). Over the last decade, new therapeutic targets have been identified in CRPC and several new drugs have reached advanced stages of clinical development. In 2010, the Food and Drug Administration (FDA) approved sipuleucel-T and cabazitaxel, and in 2011, abiraterone for patients with metastatic CRPC based on phase 3 trials showing improved survival. Although not yet available for clinical use, a press release in June 2011 announced that radium 223 also demonstrated a survival advantage in men with metastatic CRPC. Emerging therapies in advanced stages of clinical development in CRPC include the hormonal therapies MDV3100 and TAK 700, and the immunotherapy ipilimumab. Results are also pending on phase 3 studies comparing docetaxel plus prednisone with docetaxel given with the novel agents aflibercept, dasatinib, lenalidomide, and custirsen. In addition to these new and emerging therapeutic agents, denosumab was approved for the prevention of skeletal complications in patients with bone metastases due to solid tumor malignancies, providing an alternative to zoledronic acid. While the addition of these new treatment options is a great advance for men with metastatic CRPC, there are many new questions arising regarding sequencing of these treatments with each other, with previously existing therapies, and with the emerging agents now in clinical trials. Furthermore, there are concerns that on-going phase 3 trials may be contaminated if patients go off study treatment to start 1 of the newly approved agents or take the agent subsequently. These realities make clinical trial design more challenging than ever.
Collapse
|