101
|
Gene Expression Alterations during Development of Castration-Resistant Prostate Cancer Are Detected in Circulating Tumor Cells. Cancers (Basel) 2019; 12:cancers12010039. [PMID: 31877738 PMCID: PMC7016678 DOI: 10.3390/cancers12010039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 12/19/2019] [Indexed: 11/24/2022] Open
Abstract
Development of castration-resistant prostate cancer (CRPC) is associated with alterations in gene expression involved in steroidogenesis and androgen signaling. This study investigates whether gene expression changes related to CRPC development can be identified in circulating tumor cells (CTCs). Gene expression in paired CTC samples from 29 patients, before androgen deprivation therapy (ADT) and at CRPC relapse, was compared using a panel including 47 genes related to prostate cancer progression on a qPCR platform. Fourteen genes displayed significantly changed gene expression in CTCs at CRPC relapse compared to before start of ADT. The genes with increased expression at CRPC relapse were related to steroidogenesis, AR-signaling, and anti-apoptosis. In contrast, expression of prostate markers was downregulated at CRPC. We also show that midkine (MDK) expression in CTCs from metastatic hormone-sensitive prostate cancer (mHSPC) was associated to short cancer-specific survival (CSS). In conclusion, this study shows that gene expression patterns in CTCs reflect the development of CRPC, and that MDK expression levels in CTCs are prognostic for cancer-specific survival in mHSPC. This study emphasizes the role of CTCs in exploring mechanisms of therapy resistance, as well as a promising biomarker for prognostic and treatment-predictive purposes in advanced mHSPC.
Collapse
|
102
|
DE Luca S, Amante E, Fiori C, Alleva G, Alladio E, Marini F, Garrou D, Manfredi M, Amparore D, Checcucci E, Pruner S, Salomone A, Scarpa RM, Vincenti M, Porpiglia F. Prospective evaluation of urinary steroids and prostate carcinoma-induced deviation: preliminary results. Minerva Urol Nephrol 2019; 73:98-106. [PMID: 31833333 DOI: 10.23736/s2724-6051.19.03529-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The serum prostate-specific antigen is the most widespread biomarker for prostate disease. Its low specificity for prostatic malignancies is a matter of concern and the reason why new biomarkers for screening purposes are needed. The correlation between altered production of the main steroids and prostate carcinoma (PCa) occurrence is historically known. The purpose of this study is to evaluate the modifications of a comprehensive urinary endogenous steroidal profile (USP) induced by PCa, by multivariate statistical methods. METHODS A total of 283 Italian subjects were included in the study, 139 controls and 144 PCa-affected patients. The USP, including 17 steroids and five urinary steroidal ratios, was quantitatively evaluated using gas chromatography coupled with single quadrupole mass spectrometry (GC-MS). The data were interpreted using a chemometric, multivariate approach (intrinsically more sensible to alterations with respect to traditional statistics) and a model for the discrimination of cancer-affected profiles was built. RESULTS Two multivariate classification models were calculated, the former including three steroids with the highest statistical significance (e.g. testosterone, etiocholanolone and 7β-OH-DHEA) and PSA values, the latter considering the three steroids' levels only. Both models yielded high sensitivity and specificity scores near to 70%, resulting significantly higher than PSA alone. CONCLUSIONS Three USP steroids resulted significantly altered in our PCa population. These preliminary results, combined with the simplicity and low-cost of the analysis, open to further investigation of the potential role of this restricted USP in PCa diagnosis.
Collapse
Affiliation(s)
- Stefano DE Luca
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Eleonora Amante
- Department of Chemistry, University of Turin, Turin, Italy - .,A. Bertinaria Anti-Doping Center, Orbassano, Turin, Italy
| | - Cristian Fiori
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Giorgio Alleva
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | | | | | - Diletta Garrou
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Matteo Manfredi
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Daniele Amparore
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Enrico Checcucci
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Serena Pruner
- Department of Chemistry, University of Turin, Turin, Italy
| | - Alberto Salomone
- Department of Chemistry, University of Turin, Turin, Italy.,A. Bertinaria Anti-Doping Center, Orbassano, Turin, Italy
| | - Roberto M Scarpa
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Marco Vincenti
- Department of Chemistry, University of Turin, Turin, Italy.,A. Bertinaria Anti-Doping Center, Orbassano, Turin, Italy
| | - Francesco Porpiglia
- Division of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| |
Collapse
|
103
|
Giatromanolaki A, Fasoulaki V, Kalamida D, Mitrakas A, Kakouratos C, Lialiaris T, Koukourakis MI. CYP17A1 and Androgen-Receptor Expression in Prostate Carcinoma Tissues and Cancer Cell Lines. Curr Urol 2019; 13:157-165. [PMID: 31933595 DOI: 10.1159/000499276] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
Background CYP17A1 is involved in the steroidogenesis of dehydroepiandrosterone and androstenedione. CYP17A is a target for the hormonal treatment of prostate cancer (PCa). Objectives To investigate the role of CYP17A1 as a driver of PCa growth. Materials and Methods We examined the expression of CYP17A1 and of androgen receptors (AR) in PCa specimens and in PCa cell lines. Results CYP17A1 was strongly expressed in the cytoplasm of PCa cells (median 50% of cancer cells, range 0-100%). The nuclear AR expression in cancer cells was directly related with CYP17A1 (p < 0.0001, r = 0.51). The hormone dependent 22Rv1 cell line expressed the CYP17A1 and AR protein and mRNA, in contrast to the PC3 and DU145 cell lines (p < 0.0001). Testosterone and dexamethasone induced nuclear expression of AR and this effect was abolished by abiraterone. CYP17A1 levels were not affected by the incubation with testosterone, while abiraterone significantly reduced its expression. Abiraterone reduced the growth rate and migration of testosterone stimulated 22Rv1 cells. Conclusions CYP17A1 is strongly expressed in half about of human prostate carcinomas, implying an intracellular androgen synthesis by cancer cells. Abiraterone effectively blocked nuclear accumulation of AR and suppressed CYP17A1 expression. CYP17A1 may function as a biomarker to select the best hormonal anticancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Theodoros Lialiaris
- Department of Genetics, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|
104
|
Josefsson A, Damber JE, Welén K. AR-V7 expression in circulating tumor cells as a potential prognostic marker in metastatic hormone-sensitive prostate cancer. Acta Oncol 2019; 58:1660-1664. [PMID: 31286815 DOI: 10.1080/0284186x.2019.1637540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Andreas Josefsson
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgical and Perioperative Sciences, Urology & Andrology, Umeå University, Umeå, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Jan-Erik Damber
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Welén
- Department of Urology, Sahlgrenska Cancer Center, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
105
|
Coates HW, Chua NK, Brown AJ. Consulting prostate cancer cohort data uncovers transcriptional control: Regulation of the MARCH6 gene. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1656-1668. [PMID: 31422115 DOI: 10.1016/j.bbalip.2019.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/28/2022]
Abstract
Cholesterol accumulation is a hallmark of prostate cancer (PCa) enabled by the upregulation of its synthesis, which presents a potential therapeutic target. This pathway is suppressed by the E3 ubiquitin ligase membrane-associated RING-CH-type finger 6 (MARCH6); however, little is known of MARCH6 regulation, particularly at the transcriptional level. Here, we consulted large transcriptomic PCa datasets to investigate transcription factors and DNA sequence elements that regulate the MARCH6 gene. Amongst 498 primary PCa tissues of The Cancer Genome Atlas, we identified a striking positive correlation between MARCH6 and androgen receptor (AR) gene expression (r = 0.81, p < 1 × 10-117) that held in other primary tumour datasets. Two putative androgen response elements were identified in the MARCH6 gene using motif prediction and mining of publicly accessible chromatin immunoprecipitation-sequencing data. However, MARCH6 expression was not androgen-responsive in luciferase reporter and qRT-PCR assays. Instead, we established that the MARCH6-AR correlation in primary PCa is due to common regulation by the transcription factor Sp1. We located a region 100 bp downstream of the MARCH6 transcriptional start site that contains three Sp1 binding sites and strongly upregulates promoter activity. The functionality of this region, and Sp1-mediated upregulation of MARCH6, was confirmed using pharmacological and genetic inhibition of Sp1. Moreover, modulation of Sp1 activity affected the stability of squalene monooxygenase, a cholesterol biosynthesis enzyme and MARCH6 substrate. We thus establish Sp1 as the first known regulator of the MARCH6 gene and demonstrate that interrogation of transcriptomic datasets can assist in the de novo inference of transcriptional regulation.
Collapse
Affiliation(s)
- Hudson W Coates
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Ngee Kiat Chua
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
106
|
Senapati D, Kumari S, Heemers HV. Androgen receptor co-regulation in prostate cancer. Asian J Urol 2019; 7:219-232. [PMID: 32742924 PMCID: PMC7385509 DOI: 10.1016/j.ajur.2019.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/30/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) progression relies on androgen receptor (AR) action. Preventing AR's ligand-activation is the frontline treatment for metastatic PCa. Androgen deprivation therapy (ADT) that inhibits AR ligand-binding initially induces remission but eventually fails, mainly because of adaptive PCa responses that restore AR action. The vast majority of castration-resistant PCa (CRPC) continues to rely on AR activity. Novel therapeutic strategies are being explored that involve targeting other critical AR domains such as those that mediate its constitutively active transactivation function, its DNA binding ability, or its interaction with co-operating transcriptional regulators. Considerable molecular and clinical variability has been found in AR's interaction with its ligands, DNA binding motifs, and its associated coregulators and transcription factors. Here, we review evidence that each of these levels of AR regulation can individually and differentially impact transcription by AR. In addition, we examine emerging insights suggesting that each can also impact the other, and that all three may collaborate to induce gene-specific AR target gene expression, likely via AR allosteric effects. For the purpose of this review, we refer to the modulating influence of these differential and/or interdependent contributions of ligands, cognate DNA-binding motifs and critical regulatory protein interactions on AR's transcriptional output, which may influence the efficiency of the novel PCa therapeutic approaches under consideration, as co-regulation of AR activity.
Collapse
Affiliation(s)
| | - Sangeeta Kumari
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Hannelore V Heemers
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, USA.,Department of Urology, Cleveland Clinic, Cleveland, OH, USA.,Department of Hematology/Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
107
|
Wolny-Rokicka E, Tukiendorf A, Wydmański J, Ostrowska M, Zembroń-Łacny A. Lipid Status During Combined Treatment in Prostate Cancer Patients. Am J Mens Health 2019; 13:1557988319876488. [PMID: 31559896 PMCID: PMC6767737 DOI: 10.1177/1557988319876488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The aim of this study was to provide a specific review of current medical literature regarding the lipid profile during prostate carcinoma (PCa) treatment. The main aim was to analyze the results presented by different authors and to find a commonality in the changes occurring during the treatment-hormonotherapy. The levels of total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein (LDL) cholesterol were measured before and after the follow-up treatment. The manuscripts reviewed came from the period between 2008 and 2016. The size of the studies ranged from 16 participants to 310. The mean age was from 65 to 74 years in all studies. The Q test was used to attain all lipid parameters and to specify heterogeneity (p < .0001). After 12 months of androgen deprivation therapy (ADT), the patients had a significantly higher level serum TC and TG.
Collapse
Affiliation(s)
- Edyta Wolny-Rokicka
- Department of Radiotherapy, Multidisciplinary Hospital, Gorzow Wielkopolski, Poland.,Faculty of Medicine and Health Sciences, University of Zielona Góra, Zielona Góra, Poland
| | | | - Jerzy Wydmański
- Department of Radiotherapy, Centre of Oncology, Maria Skłodowska-Curie Memorial Institute, Branch in Gliwice, Poland
| | - Małgorzata Ostrowska
- Department of Nephrology, Medical University Hospital of Karol Marcinkowski, Zielona Góra, Poland
| | | |
Collapse
|
108
|
Iguchi T, Tamada S, Kato M, Yasuda S, Machida Y, Ohmachi T, Ishii K, Iwata H, Yamamoto S, Kanamaru T, Morimoto K, Hase T, Tashiro K, Harimoto K, Deguchi T, Adachi T, Iwamoto K, Takegaki Y, Nakatani T. Enzalutamide versus flutamide for castration-resistant prostate cancer after combined androgen blockade therapy with bicalutamide: the OCUU-CRPC study. Int J Clin Oncol 2019; 25:486-494. [PMID: 31564004 DOI: 10.1007/s10147-019-01554-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/20/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Before the androgen target therapy era, flutamide was widely used for castration-resistant prostate cancer in Japan. Enzalutamide is currently the recommended treatment; however, the efficacy and safety of enzalutamide and flutamide after combined androgen blockade therapy with bicalutamide, has not been compared. METHODS Patients with castration-resistant prostate cancer who received combined androgen blockade therapy with bicalutamide were randomly assigned to receive either enzalutamide or flutamide. The primary endpoint for efficacy was the 3-month prostate-specific antigen response rate. This trial is registered with ClinicalTrials.gov (NCT02346578) and the University hospital Medical Information Network (UMIN000016301). RESULTS Overall, 103 patients were enrolled. The 3- (80.8% vs. 35.3%; p < 0.001) and 6-month (73.1% vs. 31.4%; p < 0.001) prostate-specific antigen response rates were higher in the enzalutamide than in the flutamide group. The 3-month disease progression rates (radiographic or prostate-specific antigen progression) were 6.4% and 38.8% in the enzalutamide and flutamide groups, respectively [hazard ratio (HR): 0.16; 95% confidence interval (CI): 0.05-0.47; p < 0.001]; the 6-month rates were 11.4% and 51.1%, respectively (HR 0.22; 95% CI 0.09-0.50; p < 0.001). Enzalutamide provided superior prostate-specific antigen progression-free survival compared with flutamide (HR 0.29; 95% CI 0.15-0.54; p < 0.001). Median time to prostate-specific antigen progression-free survival was not reached and was 6.6 months in the enzalutamide and flutamide groups, respectively. CONCLUSIONS As an alternative anti-androgen therapy in patients with castration-resistant prostate cancer who fail bicalutamide-combined androgen blockade therapy, enzalutamide provides superior clinical outcomes compared with flutamide. Enzalutamide should be preferred over flutamide in these patients.
Collapse
Affiliation(s)
- Taro Iguchi
- Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Satoshi Tamada
- Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Minoru Kato
- Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Sayaka Yasuda
- Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yuichi Machida
- Department of Urology, Yao Municipal Hospital, Yao, Japan
| | - Tetsuji Ohmachi
- Department of Urology, Bell-land General Hospital, Sakai, Japan
| | - Keiichi Ishii
- Department of Urology, Osaka City General Hospital, Osaka, Japan
| | - Hiroyuki Iwata
- Department of Urology, Itami City Hospital, Itami, Japan
| | - Shinji Yamamoto
- Department of Urology, Ikuwakai Memorial Hospital, Osaka, Japan
| | | | - Kazuya Morimoto
- Department of Urology, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | - Taro Hase
- Department of Urology, Suita Municipal Hospital, Suita, Japan
| | | | | | | | - Takahisa Adachi
- Department of Urology, Osaka City Juso Hospital, Osaka, Japan
| | - Katsuki Iwamoto
- Department of Urology, Ishikiriseiki Hospital, Higashiosaka, Japan
| | | | - Tatsuya Nakatani
- Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
109
|
Smith KW, Thompson PD, Rodriguez EP, Mackay L, Cobice DF. Effects of vitamin D as a regulator of androgen intracrinology in LNCAP prostate cancer cells. Biochem Biophys Res Commun 2019; 519:579-584. [PMID: 31537382 DOI: 10.1016/j.bbrc.2019.09.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/14/2019] [Indexed: 12/27/2022]
Abstract
Prostate cancer is initially treated via androgen deprivation therapy (ADT), a highly successful treatment in the initial pursuit of tumor regression, but commonly restricted by the eventual emergence of a more lethal 'castrate resistant' form of the disease. Intracrine pathways that utilize dehydroepiandrosterone (DHEA) or other circulatory precursor steroids, are thought to generate relevant levels of growth-stimulating androgens such as testosterone (T) and dihydrotestosterone (DHT). In this study, we explored the capacity of the active vitamin D hormone to interact and elicit changes upon this prostatic intracrine pathway at a metabolic level. We used androgen dependent LNCaP cells cultured under steroid-depleted conditions and assessed the impact of vitamin D-based compounds upon intracrine pathways that convert exogenously added DHEA to relevant metabolites, through Mass Spectrometry (MS). Changes in relevant metabolism-related gene targets were also assessed. Our findings confirm that exposure to vitamin D based compounds, within LNCaP cells, elicits measurable and significant reduction in the intracrine conversion of DHEA to T, DHT and other intermediate metabolites within the androgenic pathway. The aassessment and validation of the biological model and analytical platforms were performed by pharmacological manipulations of the SRD5α and HSD-17β enzymes. The data provides further confirmation for how a vitamin D-based regime may be used to counter intracrine mechanisms contributing to the emergence of castrate-resistant tumors.
Collapse
Affiliation(s)
- Karl W Smith
- Mass Spectrometry Centre, Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK; The Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Paul D Thompson
- The Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Edna Patricia Rodriguez
- The Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Logan Mackay
- Scottish Instrumentation and Research Centre for Advanced Mass Spectrometry (SIRCAMS), EastChem School of Chemistry, University of Edinburgh, Scotland, UK
| | - Diego F Cobice
- Mass Spectrometry Centre, Biomedical Sciences Research Institute (BMSRI), School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
110
|
Abstract
Ketoconazole is a nonselective steroid 17α-hydroxylase/17,20 lyase (CYP17A1) inhibitor that has been used, off-label, as a second-line therapy for castration-resistant prostate cancer (CRPC). The drug has shown clinical efficacy without survival benefit. Despite not improving survival, ketoconazole has beneficial characteristics, such as its low cost, a relatively favourable toxicity profile compared with chemotherapy, and its efficacy both before and after chemotherapy. The approval of several new, highly effective treatments, including abiraterone acetate, enzalutamide, and apalutamide, warrants re-evaluation of the role of ketoconazole and other classic agents in achieving the optimal timing and sequencing of available agents to prolong survival and maintain patients' quality of life. In the current CRPC treatment landscape, we believe that ketoconazole can be considered in patients with nonmetastatic CRPC and in those with metastatic CRPC who do not respond to, tolerate, or have access to chemotherapy and other standard therapeutic options.
Collapse
Affiliation(s)
- Vaibhav Patel
- Division of Hematology and Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Bobby Liaw
- Division of Hematology and Oncology, Department of Medicine, The Mount Sinai Hospital, Mount Sinai Beth Israel, New York, NY, USA
| | - William Oh
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
111
|
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: 1.4] [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
|
112
|
Zhao J, Zhang M, Liu J, Liu Z, Shen P, Nie L, Guo W, Cai D, Liu J, Armstrong CM, Sun G, Chen J, Zhu S, Dai J, Zhang H, Zhao P, Zhang X, Yin X, Zhu X, Ni Y, Chen N, Zeng H. AKR1C3 expression in primary lesion rebiopsy at the time of metastatic castration-resistant prostate cancer is strongly associated with poor efficacy of abiraterone as a first-line therapy. Prostate 2019; 79:1553-1562. [PMID: 31294486 DOI: 10.1002/pros.23875] [Citation(s) in RCA: 6] [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: 04/28/2019] [Accepted: 06/17/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Previous studies had demonstrated that aldo-keto reductase family 1 member C3 (AKR1C3), a crucial enzyme in the steroidogenic pathway, played an important role in abiraterone (ABI)-resistance in metastatic castration-resistant prostate cancer (mCRPC) by increasing intratumoral androgen synthesis. However, its value in predicting treatment response in patients with mCRPC is unknown. METHOD AND MATERIALS Data of 163 patients with metastatic prostate cancer between 2016 and 2018 were retrospectively analyzed. All patients received androgen deprivation therapy plus bicalutamide after initial diagnosis. After mCRPC, either ABI or docetaxel (DOC) treatment was used. No patient had the experience of therapy to the primary tumor. AKR1C3 protein was detected by immunohistochemical staining from rebiopsy (re-Bx) of primary prostate lesions at mCRPC. Kaplan-Meier curves and Cox regression were used to analyze the association between AKR1C3 and treatment outcomes. RESULTS AKR1C3 was positive in 58 of 163 (35.6%) cases. AKR1C3 was associated with significantly shorter median prostate-specific antigen progression-free survival (mPSA-PFS, 5.6 mo vs 10.7 mo; P < .001), median radiographic progression-free survival (mrPFS, 11.1 mo vs 18.0 mo; P = .018), and numerically shorter median overall survival (mOS, 20.4 mo vs 26.4 mo; P = .157). Notably, AKR1C3-positive patients treated with ABI, but not DOC, had shorter mPSA-PFS and mrPFS compared with AKR1C3-negative men, (mPSA-PFS, 5.7 mo vs. 11.2 mo; P < .001; mrPFS, 12.4 mo vs 23.3 mo; P = .048). However, AKR1C3 expression had no correlation to PSA response or OS. Multivariate Cox regression indicated that AKR1C3 was independently accompanied with rapid PSA progression (hazard ratio [HR], 3.64; 95% confidence interval [CI], 2.10-6.31; P < 0.001) and radiological progression (HR, 2.08; 95% CI, 1.05-4.11; P = .036) in the ABI-treated subgroup. CONCLUSION This study demonstrated that AKR1C3 detection in tissues from prostate re-Bx at mCRPC was associated with early resistance to ABI but not DOC. These results will help to make optimal personalized treatment decisions for patients with mCRPC, facilitate physicians predicting the effectiveness of ABI.
Collapse
Affiliation(s)
- Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiandong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenhao Guo
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Diming Cai
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Cameron M Armstrong
- Department of Urology and Comprehensive Cancer Center, University of California Davis, Sacramento, California
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Haoran Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xudong Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
113
|
Cucchiara V, Yang JC, Liu C, Adomat HH, Tomlinson Guns ES, Gleave ME, Gao AC, Evans CP. GnRH Antagonists Have Direct Inhibitory Effects On Castration-Resistant Prostate Cancer Via Intracrine Androgen and AR-V7 Expression. Mol Cancer Ther 2019; 18:1811-1821. [DOI: 10.1158/1535-7163.mct-18-1337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/28/2019] [Accepted: 07/23/2019] [Indexed: 11/16/2022]
|
114
|
Affiliation(s)
- Ian F Tannock
- From the Division of Medical Oncology, Princess Margaret Cancer Centre and the University of Toronto, Toronto
| |
Collapse
|
115
|
Gordon JA, Noble JW, Midha A, Derakhshan F, Wang G, Adomat HH, Tomlinson Guns ES, Lin YY, Ren S, Collins CC, Nelson PS, Morrissey C, Wasan KM, Cox ME. Upregulation of Scavenger Receptor B1 Is Required for Steroidogenic and Nonsteroidogenic Cholesterol Metabolism in Prostate Cancer. Cancer Res 2019; 79:3320-3331. [PMID: 31064850 PMCID: PMC6606386 DOI: 10.1158/0008-5472.can-18-2529] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/25/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Aberrant cholesterol metabolism is increasingly appreciated to be essential for prostate cancer initiation and progression. Transcript expression of the high-density lipoprotein-cholesterol receptor scavenger receptor B1 (SR-B1) is elevated in primary prostate cancer. Hypothesizing that SR-B1 expression may help facilitate malignant transformation, we document increased SR-B1 protein and transcript expression in prostate cancer relative to normal prostate epithelium that persists in lethal castration-resistant prostate cancer (CRPC) metastasis. As intratumoral steroid synthesis from the precursor cholesterol can drive androgen receptor (AR) pathway activity in CRPC, we screened androgenic benign and cancer cell lines for sensitivity to SR-B1 antagonism. Benign cells were insensitive to SR-B1 antagonism, and cancer line sensitivity inversely correlated with expression levels of full-length and splice variant AR. In androgen-responsive CRPC cell model C4-2, SR-B1 antagonism suppressed cholesterol uptake, de novo steroidogenesis, and AR activity. SR-B1 antagonism also suppressed growth and viability and induced endoplasmic reticulum stress and autophagy. The inability of exogenous steroids to reverse these effects indicates that AR pathway activation is insufficient to overcome cytotoxic stress caused by a decrease in the availability of cholesterol. Furthermore, SR-B1 antagonism decreased cholesterol uptake, growth, and viability of the AR-null CRPC cell model PC-3, and the small-molecule SR-B1 antagonist block lipid transport-1 decreased xenograft growth rate despite poor pharmacologic properties. Overall, our findings show that SR-B1 is upregulated in primary and castration-resistant disease and is essential for cholesterol uptake needed to drive both steroidogenic and nonsteroidogenic biogenic pathways, thus implicating SR-B1 as a novel and potentially actionable target in CRPC. SIGNIFICANCE: These findings highlight SR-B1 as a potential target in primary and castration-resistant prostate cancer that is essential for cholesterol uptake needed to drive steroidogenic and nonsteroidogenic biogenic pathways.
Collapse
MESH Headings
- Androgens/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Bone Neoplasms/metabolism
- Bone Neoplasms/secondary
- Bone Neoplasms/surgery
- Cell Proliferation
- Cholesterol/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Liver Neoplasms/surgery
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lung Neoplasms/surgery
- Male
- Mice
- Mice, Nude
- Orchiectomy
- Prognosis
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/surgery
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jacob A Gordon
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Jake W Noble
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Ankur Midha
- Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Fatemeh Derakhshan
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Gang Wang
- Department of Pathology, British Columbia Cancer Agency, Vancouver, Canada
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Emma S Tomlinson Guns
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Shancheng Ren
- Department of Urology, Second Military Medical University, Shanghai, China
| | - Collin C Collins
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Kishor M Wasan
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada
| | - Michael E Cox
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- Department of Urologic Sciences, University of British Columbia, Canada
| |
Collapse
|
116
|
Shen MM, Rubin MA. Prostate Cancer Research at the Crossroads. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a036277. [PMID: 30348836 DOI: 10.1101/cshperspect.a036277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Michael M Shen
- Departments of Medicine, Genetics and Development, Urology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | - Mark A Rubin
- Englander Institute for Precision Medicine, Department of Pathology and Laboratory Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021.,Department of BioMedical Research, University of Bern and Inselspital, 3008 Bern, Switzerland
| |
Collapse
|
117
|
Blatt EB, Raj GV. Molecular mechanisms of enzalutamide resistance in prostate cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:189-197. [PMID: 35582713 PMCID: PMC8992629 DOI: 10.20517/cdr.2019.25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/06/2019] [Accepted: 05/10/2019] [Indexed: 11/12/2022]
Abstract
An estimated 30,000 men in the United States will die of metastatic prostate cancer (PCa) each year due to the development of therapy resistance, most notably resistance to second-generation antiandrogen enzalutamide. The vast majority of PCa is driven by the androgen receptor (AR). Enzalutamide is an AR antagonist, which extends patient survival and is widely used in the clinic for the treatment of castration-resistant prostate cancer (CRPC); however, many patients will have primary or develop acquired resistance and continue to progress. Characterization of the molecular mechanisms of enzalutamide resistance provides insight into potentially efficacious therapies for enzalutamide-resistant CRPC (ER-CRPC). Understanding these mechanisms is critical for the identification of biomarkers predictive of therapy resistance and the development of therapeutic strategies to target ER-CRPC.
Collapse
Affiliation(s)
- Eliot B. Blatt
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ganesh V. Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
118
|
Moll JM, Kumagai J, van Royen ME, Teubel WJ, van Soest RJ, French PJ, Homma Y, Jenster G, de Wit R, van Weerden WM. A bypass mechanism of abiraterone-resistant prostate cancer: Accumulating CYP17A1 substrates activate androgen receptor signaling. Prostate 2019; 79:937-948. [PMID: 31017696 PMCID: PMC6593470 DOI: 10.1002/pros.23799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/08/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Intratumoral steroidogenesis and its potential relevance in castration-resistant prostate cancer (CRPC) and in cytochrome P450, family 17, subfamily A, polypeptide 1 (CYP17A1)-inhibitor treated hormone-naïve and patients with CRPC are not well established. In this study, we tested if substrates for de novo steroidogenesis accumulating during CYP17A1 inhibition may drive cell growth in relevant preclinical models. METHODS PCa cell lines and their respective CRPC sublines were used to model CRPC in vitro. Precursor steroids pregnenolone (Preg) and progesterone (Prog) served as substrate for de novo steroid synthesis. TAK700 (orteronel), abiraterone, and small interfering RNA (siRNA) against CYP17A1 were used to block CYP17A1 enzyme activity. The antiandrogen RD162 was used to assess androgen receptor (AR) involvement. Cell growth was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. AR-target gene expression was quantified by reverse transcription polymerase chain reaction (RT-PCR). Nuclear import studies using cells with green fluorescent protein (GFP)-tagged AR were performed to assess the potential of precursor steroids to directly activate AR. RESULTS Preg and Prog stimulated cell proliferation and AR target gene expression in VCaP, DuCaP, LNCaP, and their respective CRPC sublines. The antiandrogen RD162, but not CYP17A1 inhibition with TAK700, abiraterone or siRNA, was able to block Preg- and Prog-induced proliferation. In contrast to TAK700, abiraterone also affected dihydrotestosterone-induced cell growth, indicating direct AR binding. Furthermore, Prog-induced AR translocation was not affected by treatment with TAK700 or abiraterone, while it was effectively blocked by the AR antagonist enzalutamide, further demonstrating the direct AR activation by Prog. CONCLUSION Activation of the AR by clinically relevant levels of Preg and Prog accumulating in abiraterone-treated patients may act as a driver for CRPC. These data provide a scientific rationale for combining CYP17A1 inhibitors with antiandrogens, particularly in patients with overexpressed or mutated-AR.
Collapse
Affiliation(s)
- Jan M. Moll
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Jinpei Kumagai
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of UrologyUniversity of TokyoTokyoJapan
| | - Martin E. van Royen
- Department of PathologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of Erasmus Optical Imaging CentreErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of Cancer Treatment Screening FacilityErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Wilma J. Teubel
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Robert J. van Soest
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Pim J. French
- Department of Cancer Treatment Screening FacilityErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of NeurologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Yukio Homma
- Department of UrologyUniversity of TokyoTokyoJapan
| | - Guido Jenster
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Ronald de Wit
- Department of Medical OncologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Wytske M. van Weerden
- Department of UrologyErasmus University Medical Center, Erasmus MC Cancer InstituteRotterdamThe Netherlands
| |
Collapse
|
119
|
Androgen decline and survival during docetaxel therapy in metastatic castration resistant prostate cancer (mCRPC). Prostate Cancer Prostatic Dis 2019; 23:66-73. [PMID: 31053766 PMCID: PMC6825875 DOI: 10.1038/s41391-019-0152-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/21/2019] [Accepted: 03/20/2019] [Indexed: 11/18/2022]
Abstract
Background: Multiple androgens drive prostate cancer progression and higher pre-treatment levels of androgens, even within the castrate range, have been previously shown to be associated with an improved overall survival (OS) in mCRPC. Docetaxel impairs microtubules, has androgen receptor (AR) inhibitory effects and is used in both the castration resistant and sensitive settings, where androgen dynamics may impact outcome. The present analysis evaluates the association of decline in serum androgen levels (Testosterone (T), Androstenedione (A) and DHEA in docetaxel-treated mCRPC patients with OS. Methods: Data from 1,050 men treated on CALGB 90401 with docetaxel, prednisone and either bevacizumab or placebo were evaluated. Eligibility required progressive mCRPC and no prior chemotherapy. Pre-treatment, 6 week and progression serum assays for T, A and DHEA were performed via tandem Liquid Chromatography-Mass Spectrometry (LC-MS/MS). Changes in T, A and DHEA levels from baseline to 6 weeks were calculated as the ratio of 6-week over baseline. The proportional hazards model was used to assess the prognostic significance of changes in T, A, and DHEA from baseline to 6 weeks in predicting OS adjusting for known prognostic factors. Results: Median baseline values for T, A, and, DHEA were 1.0, 13.5 and 8.1, ng/dL respectively while 6 week levels were 0.64, 7.0 and 6.8, ng/dL respectively. Median OS for low testosterone decline is 20.9 months vs 26.3 months for high testosterone decline. In multivariable analysis including known prognostic variables, change in testosterone levels was independently associated with greater OS; the hazard ratio for death with each unit increase in the 6-week/baseline ratio is 1.02 (95% CI=1.01–1.03, p=0.001). Decline in A and DHEA were not significant predictors of OS. In multivariable analysis change in the serum changes did not predict PFS however the ratio of T at 6-weeks over baseline was prognostic of ≥50% decline in PSA with an odds ratio of 0.93 (95% CI=0.85–0.98, p-value=0.039). Conclusions: Declines in testosterone during docetaxel treatment is associated with a longer survival, consistent with a favorable prognostic significance of higher serum androgens in the CRPC.
Collapse
|
120
|
Miyazaki Y, Teramoto Y, Shibuya S, Goto T, Okasho K, Mizuno K, Uegaki M, Yoshikawa T, Akamatsu S, Kobayashi T, Ogawa O, Inoue T. Consecutive Prostate Cancer Specimens Revealed Increased Aldo⁻Keto Reductase Family 1 Member C3 Expression with Progression to Castration-Resistant Prostate Cancer. J Clin Med 2019; 8:E601. [PMID: 31052459 PMCID: PMC6571723 DOI: 10.3390/jcm8050601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 01/03/2023] Open
Abstract
Aldo-keto reductase family 1 member C3 (AKR1C3) is an enzyme in the steroidogenesis pathway, especially in formation of testosterone and dihydrotestosterone, and is believed to have a key role in promoting prostate cancer (PCa) progression, particularly in castration-resistant prostate cancer (CRPC). This study aims to compare the expression level of AKR1C3 between benign prostatic epithelium and cancer cells, and among hormone-naïve prostate cancer (HNPC) and CRPC from the same patients, to understand the role of AKR1C3 in PCa progression. Correlation of AKR1C3 immunohistochemical expression between benign and cancerous epithelia in 134 patient specimens was analyzed. Additionally, correlation between AKR1C3 expression and prostate-specific antigen (PSA) progression-free survival (PFS) after radical prostatectomy was analyzed. Furthermore, we evaluated the consecutive prostate samples derived from 11 patients both in the hormone-naïve and castration-resistant states. AKR1C3 immunostaining of cancer epithelium was significantly stronger than that of the benign epithelia in patients with localized HNPC (p < 0.0001). High AKR1C3 expression was an independent factor of poor PSA PFS (p = 0.032). Moreover, AKR1C3 immunostaining was significantly stronger in CRPC tissues than in HNPC tissues in the same patients (p = 0.0234). Our findings demonstrate that AKR1C3 is crucial in PCa progression.
Collapse
Affiliation(s)
- Yu Miyazaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Yuki Teramoto
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | - Shinsuke Shibuya
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | - Takayuki Goto
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Kosuke Okasho
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Kei Mizuno
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Masayuki Uegaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takeshi Yoshikawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| |
Collapse
|
121
|
Kaiser A, Haskins C, Siddiqui MM, Hussain A, D’Adamo C. The evolving role of diet in prostate cancer risk and progression. Curr Opin Oncol 2019; 31:222-229. [PMID: 30893147 PMCID: PMC7379157 DOI: 10.1097/cco.0000000000000519] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This overview examines the rationale for dietary interventions for prostate cancer by summarizing the current evidence base and biological mechanisms for the involvement of diet in disease incidence and progression. RECENT FINDINGS Recent data have further solidified the association between insulin resistance and prostate cancer with the homeostatic model assessment of insulin resistance. Data also show that periprostatic adipocytes promote extracapsular extension of prostate cancer through chemokines, thereby providing a mechanistic explanation for the association observed between obesity and high-grade cancer. Regarding therapeutics, hyperinsulinemia may be the cause of resistance to phosphatidylinositol-3 kinase inhibitors in the treatment of prostate cancer, leading to new investigations combining these drugs with ketogenic diets. SUMMARY Given the recently available data regarding insulin resistance and adipokine influence on prostate cancer, dietary strategies targeting metabolic syndrome, diabetes, and obesity should be further explored. In macronutrient-focused therapies, low carbohydrate/ketogenic diets should be favored in such interventions because of their superior impact on weight loss and metabolic parameters and encouraging clinical data. Micronutrients, including the carotenoid lycopene which is found in highest concentrations in tomatoes, may also play a role in prostate cancer prevention and prognosis through complementary metabolic mechanisms. The interplay between genetics, diet, and prostate cancer is an area of emerging focus that might help optimize therapeutic dietary response in the future through personalization.
Collapse
Affiliation(s)
- Adeel Kaiser
- Department of Radiation Oncology, Univ. of Maryland School of Medicine, Baltimore, MD USA
| | - Christopher Haskins
- Department of Radiation Oncology, Univ. of Maryland School of Medicine, Baltimore, MD USA
| | - Mohummad M. Siddiqui
- Division of of Urology, Department of Surgery, Univ. of Maryland School of Medicine, Baltimore, MD USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD USA
| | - Arif Hussain
- Department of Medicine and University of Maryland Greenebaum Comprehensive Cancer Center, Univ. of Maryland School of Medicine, Baltimore, MD USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD USA
| | - Christopher D’Adamo
- Department of Family and Community Medicine, Univ. of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
122
|
An actionable sterol-regulated feedback loop modulates statin sensitivity in prostate cancer. Mol Metab 2019; 25:119-130. [PMID: 31023626 PMCID: PMC6600047 DOI: 10.1016/j.molmet.2019.04.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The statin family of cholesterol-lowering drugs has been shown to induce tumor-specific apoptosis by inhibiting the rate-limiting enzyme of the mevalonate (MVA) pathway, HMG-CoA reductase (HMGCR). Accumulating evidence suggests that statin use may delay prostate cancer (PCa) progression in a subset of patients; however, the determinants of statin drug sensitivity in PCa remain unclear. Our goal was to identify molecular features of statin-sensitive PCa and opportunities to potentiate statin-induced PCa cell death. METHODS Deregulation of HMGCR expression in PCa was evaluated by immunohistochemistry. The response of PCa cell lines to fluvastatin-mediated HMGCR inhibition was assessed using cell viability and apoptosis assays. Activation of the sterol-regulated feedback loop of the MVA pathway, which was hypothesized to modulate statin sensitivity in PCa, was also evaluated. Inhibition of this statin-induced feedback loop was performed using RNA interference or small molecule inhibitors. The achievable levels of fluvastatin in mouse prostate tissue were measured using liquid chromatography-mass spectrometry. RESULTS High HMGCR expression in PCa was associated with poor prognosis; however, not all PCa cell lines underwent apoptosis in response to treatment with physiologically-achievable concentrations of fluvastatin. Rather, most cell lines initiated a feedback response mediated by sterol regulatory element-binding protein 2 (SREBP2), which led to the further upregulation of HMGCR and other lipid metabolism genes. Overcoming this feedback mechanism by knocking down or inhibiting SREBP2 potentiated fluvastatin-induced PCa cell death. Notably, we demonstrated that this feedback loop is pharmacologically-actionable, as the drug dipyridamole can be used to block fluvastatin-induced SREBP activation and augment apoptosis in statin-insensitive PCa cells. CONCLUSION Our study implicates statin-induced SREBP2 activation as a PCa vulnerability that can be exploited for therapeutic purposes using clinically-approved agents.
Collapse
|
123
|
Dong J, Zhang J, Li Z, Asnake S, Zhang D, Olsson PE, Zhao G. Design, synthesis, and biological evaluation of 2-(5-methyl-1H-pyrazol-1-yl) acetamide derivatives as androgen receptor antagonists. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
124
|
Tousignant KD, Rockstroh A, Taherian Fard A, Lehman ML, Wang C, McPherson SJ, Philp LK, Bartonicek N, Dinger ME, Nelson CC, Sadowski MC. Lipid Uptake Is an Androgen-Enhanced Lipid Supply Pathway Associated with Prostate Cancer Disease Progression and Bone Metastasis. Mol Cancer Res 2019; 17:1166-1179. [PMID: 30808729 DOI: 10.1158/1541-7786.mcr-18-1147] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/03/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
De novo lipogenesis is a well-described androgen receptor (AR)-regulated metabolic pathway that supports prostate cancer tumor growth by providing fuel, membrane material, and steroid hormone precursor. In contrast, our current understanding of lipid supply from uptake of exogenous lipids and its regulation by AR is limited, and exogenous lipids may play a much more significant role in prostate cancer and disease progression than previously thought. By applying advanced automated quantitative fluorescence microscopy, we provide the most comprehensive functional analysis of lipid uptake in cancer cells to date and demonstrate that treatment of AR-positive prostate cancer cell lines with androgens results in significantly increased cellular uptake of fatty acids, cholesterol, and low-density lipoprotein particles. Consistent with a direct, regulatory role of AR in this process, androgen-enhanced lipid uptake can be blocked by the AR-antagonist enzalutamide, but is independent of proliferation and cell-cycle progression. This work for the first time comprehensively delineates the lipid transporter landscape in prostate cancer cell lines and patient samples by analysis of transcriptomics and proteomics data, including the plasma membrane proteome. We show that androgen exposure or deprivation regulates the expression of multiple lipid transporters in prostate cancer cell lines and tumor xenografts and that mRNA and protein expression of lipid transporters is enhanced in bone metastatic disease when compared with primary, localized prostate cancer. Our findings provide a strong rationale to investigate lipid uptake as a therapeutic cotarget in the fight against advanced prostate cancer in combination with inhibitors of lipogenesis to delay disease progression and metastasis. IMPLICATIONS: Prostate cancer exhibits metabolic plasticity in acquiring lipids from uptake and lipogenesis at different disease stages, indicating potential therapeutic benefit by cotargeting lipid supply.
Collapse
Affiliation(s)
- Kaylyn D Tousignant
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Atefeh Taherian Fard
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Melanie L Lehman
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Chenwei Wang
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Stephen J McPherson
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lisa K Philp
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nenad Bartonicek
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Martin C Sadowski
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
125
|
Anderson-Carter I, Posielski N, Liou JI, Khemees TA, Downs TM, Abel EJ, Jarrard DF, Richards KA. The impact of statins in combination with androgen deprivation therapyin patients with advanced prostate cancer: A large observational study. Urol Oncol 2019; 37:130-137. [PMID: 30528885 PMCID: PMC6919968 DOI: 10.1016/j.urolonc.2018.11.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Statins are thought to possess antineoplastic properties related to their effect on cell proliferation and steroidogenesis. Progression to castrate resistant prostate cancer (CaP) includes de-regulation of androgen synthesis suggesting a role for statins in this setting. Our goal was to assess the role of statin use on oncologic outcomes in patients with advanced CaP being treated with androgen deprivation therapy (ADT). METHODS The national VA database was used to identify all men diagnosed with CaP who were treated with ADT for at least 6 months between 2000 and 2008 with follow-up through May 2016. Our cohort was stratified based on statin use of at least 6 months duration during the same time. Multivariable Cox proportional hazards analyses with inverse propensity score weighted (IPSW) adjustment were calculated to assess for primary outcomes of CaP-specific survival (CSS), overall survival (OS) and skeletal related events (SREs). RESULTS A total of 87,346 patients on ADT were included in the study cohort, 53,360 patients used statins and 33,986 did not. Statin users were younger in age (median 73 vs. 76, P < 0.001), more likely to have a higher Charlson comorbidity index (CCI) >3 (3.1% vs. 2.5%, P < 0.001) and more likely to have a high grade (Gleason score 8-10) cancer (12.3% vs. 10.9%, P < 0.001). Statin users had longer OS (median 6.5 vs. 4.0 years P < 0.001) and decreased death from CaP (5-year CSS 94.0% vs. 87.3%, P < 0.001). Statin use was also associated with longer time to a SRE (median 5.9 vs. 3.7 years, P < 0.001). On multivariable Cox proportional hazards analysis with inverse propensity score weighted, statin use was an independent predictor of improved OS (hazard ratio [HR] 0.66, confidence interval [CI] 0.63-0.68; P < 0.001), CSS (HR 0.56, 95% CI 0.53-0.60; P < 0.001), and SREs (HR 0.64, 95%CI 0.59-0.71; P < 0.001) when controlling for age, race, Charlson comorbidity index, prostate-specific antigen, and Gleason score. CONCLUSION The use of statins in men on ADT for CaP is associated with improved CSS and OS. Statins are inexpensive, well-tolerated medications that offer a promising adjunct to ADT, but require further prospective studies.
Collapse
Affiliation(s)
| | - Natasza Posielski
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
| | - Jinn-ing Liou
- The University of Wisconsin-Madison, Department of Medicine, Madison, WI
| | - Tariq A. Khemees
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
| | - Tracy M. Downs
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
| | - E. Jason Abel
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
| | - David F. Jarrard
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
| | - Kyle A. Richards
- The University of Wisconsin-Madison, Department of Urology, Madison, WI
- William S. Middleton Memorial Veterans Hospital, Department of Surgery, Section of Urology, Madison, WI
| |
Collapse
|
126
|
Shiota M, Narita S, Akamatsu S, Fujimoto N, Sumiyoshi T, Fujiwara M, Uchiumi T, Habuchi T, Ogawa O, Eto M. Association of Missense Polymorphism in HSD3B1 With Outcomes Among Men With Prostate Cancer Treated With Androgen-Deprivation Therapy or Abiraterone. JAMA Netw Open 2019; 2:e190115. [PMID: 30794306 PMCID: PMC6484618 DOI: 10.1001/jamanetworkopen.2019.0115] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IMPORTANCE Recently, genetic polymorphism in HSD3B1 encoding 3β-hydroxysteroid dehydrogenase-1 has been shown to be associated with oncological outcome when treated with androgen-deprivation therapy (ADT) for prostate cancer. Upfront abiraterone combined with ADT has proved survival benefit. However, its effect on oncological outcome among different ethnicities and in abiraterone treatment remain unclear. OBJECTIVE To investigate the significance of missense polymorphism in HSD3B1 gene among men treated with primary ADT or abiraterone. DESIGN, SETTING, AND PARTICIPANTS This prognostic study included Japanese patients with metastatic hormone-sensitive prostate cancer between June 1993 and July 2005 and with castration-resistant prostate cancer between September 2014 and February 2018. Genome DNA was obtained from patient whole blood samples, and genotyping on HSD3B1 (rs1047303, 1245C) was performed by Sanger sequencing. EXPOSURES Primary ADT for metastatic hormone-sensitive prostate cancer and abiraterone for castration-resistant prostate cancer. MAIN OUTCOMES AND MEASURES The association of genotype in HSD3B1 with clinicopathological parameters and oncological outcome, including prostate-specific antigen response, progression-free survival, treatment failure-free survival, and overall survival was examined. RESULTS Of 203 men, 104 were in the primary ADT cohort (median [interquartile range] age, 72 [67-76] years) and 99 men were in the abiraterone group (median [interquartile range] age, 74 [67-80] years). Most patients carried metastatic lesions in each cohort. Among the cohort of primary ADT, men carrying heterozygous and homozygous variant types in HSD3B1 gene showed higher progression risk (hazard ratio [HR], 2.34; 95% CI, 1.08-4.49; P = .03) but not any-caused death risk (HR, 1.36; 95% CI, 0.52-2.92; P = .50), compared with men carrying homozygous wild type. In contrast, among the abiraterone cohort, men carrying variant type in HSD3B1 gene showed lower progression risk (HR, 0.32; 95% CI, 0.12-0.69; P = .006) and lower all-cause mortality risk (HR, 0.40; 95% CI, 0.13-0.94; P = .04) compared with men carrying homozygous wild type. CONCLUSIONS AND RELEVANCE This study showed that HSD3B1 genetic variant is distinctly associated with oncological outcome between primary ADT and abiraterone in Japanese men, suggesting universal significance among different ethnicities in primary ADT, as well as promise as a predictive biomarker of ADT and abiraterone.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shintaro Narita
- Department of Urology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naohiro Fujimoto
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takayuki Sumiyoshi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Maki Fujiwara
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Habuchi
- Department of Urology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
127
|
Pre- and post-radical prostatectomy testosterone levels in prostate cancer patients. Int J Impot Res 2019; 31:145-149. [PMID: 30659293 DOI: 10.1038/s41443-019-0116-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/30/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
Radical prostatectomy is one of the treatment of choices for localized prostate cancer. Published data show that radical prostatectomy is associated with both an increase and decrease in testosterone levels. This study aimed to document the changes in pre- and postoperative serum testosterone levels after radical prostatectomy along with the associations between serum testosterone levels and prostate cancer profiles in Thai population. Localized and locally advanced prostate cancer patients who elected to have radical prostatectomy without prior androgen deprivation therapy were included in the study. Patients' demographic data, pre- and postoperative serum testosterone levels, sex hormone binding globulin, albumin, prostate-specific antigen, and final pathologic reports were collected. Eighty-five prostate cancer patients were included in this study. Mean age was 67.32 years. Mean pre- and postoperative serum testosterone levels were 424.95 ng/dL and 371.94 ng/dL, respectively (p-value < 0.001). There was a greater testosterone reduction in patients with a final pathologic report of Gleason 4 + 3 and above compared with those with a Gleason 3 + 3 and 3 + 4 (p-value = 0.001). No significant association between preoperative testosterone levels and final Gleason scores was observed. This study documented significant postoperative testosterone reductions in prostate cancer patients after a radical prostatectomy. Patients with high Gleason grades had greater testosterone reductions. These findings may have clinical implications for the prediction of postoperative hypogonadal states in prostate cancer patients.
Collapse
|
128
|
Wnt/Beta-Catenin Signaling and Prostate Cancer Therapy Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:351-378. [PMID: 31900917 DOI: 10.1007/978-3-030-32656-2_16] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastatic or locally advanced prostate cancer (PCa) is typically treated with androgen deprivation therapy (ADT). Initially, PCa responds to the treatment and regresses. However, PCa almost always develops resistance to androgen deprivation and progresses to castrate-resistant prostate cancer (CRPCa), a currently incurable form of PCa. Wnt/β-Catenin signaling is frequently activated in late stage PCa and contributes to the development of therapy resistance. Although activating mutations in the Wnt/β-Catenin pathway are not common in primary PCa, this signaling cascade can be activated through other mechanisms in late stage PCa, including cross talk with other signaling pathways, growth factors and cytokines produced by the damaged tumor microenvironment, release of the co-activator β-Catenin from sequestration after inhibition of androgen receptor (AR) signaling, altered expression of Wnt ligands and factors that modulate the Wnt signaling, and therapy-induced cellular senescence. Research from genetically engineered mouse models indicates that activation of Wnt/β-Catenin signaling in the prostate is oncogenic, enables castrate-resistant PCa growth, induces an epithelial-to-mesenchymal transition (EMT), promotes neuroendocrine (NE) differentiation, and confers stem cell-like features to PCa cells. These important roles of Wnt/β-Catenin signaling in PCa progression underscore the need for the development of drugs targeting this pathway to treat therapy-resistant PCa.
Collapse
|
129
|
Li Y, Zhang Q, Lovnicki J, Chen R, Fazli L, Wang Y, Gleave M, Huang J, Dong X. SRRM4 gene expression correlates with neuroendocrine prostate cancer. Prostate 2019; 79:96-104. [PMID: 30155992 DOI: 10.1002/pros.23715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/14/2018] [Indexed: 12/29/2022]
Abstract
Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of castrate-resistant prostate cancer characterized by poor patient outcome. Whole transcriptome sequencing analyses identified a NEPC-specific RNA splicing program that is predominantly controlled by the SRRM4 gene, suggesting that SRRM4 drives NEPC development. However, whether SRRM4 expression in patients may aid pathologists in diagnosing NEPC and predicting patient survival remains to be determined. In this study, we have applied RNA in situ hybridization and immunohistochemistry assays to measure the expressions of SRRM4, NEPC markers (SYP, CD56, and CHGA), and adenocarcinoma (AdPC) markers (AR, PSA) in a series of tissue microarrays constructed from castrate-resistant prostate tumors, treatment-naïve tumors collected from radical prostatectomy, and tumors treated with neoadjuvant hormonal therapy (NHT) for 0-12 months. Three pathologists also independently evaluated tumor histology and NEPC marker status. Here, we report that SRRM4 in castrate-resistant tumors is highly expressed in NEPC, strongly correlated with SYP, CD56, and CHGA expressions (Pearson correlation r = 0.883, 0.675, and 0.881; P < 0.0001) and negatively correlated with AR and PSA expressions (Pearson correlation r = -0.544 and -0.310; P < 0.05). Overall survival is 12.3 months for patients with SRRM4 positive tumors, comparing to 23 months for patients with SRRM4 negative tumors. In treatment-naïve AdPC, low SRRM4 expression is detected in ∼16% tumor cores. It correlates with SYP and CHGA expressions, but not Gleason scores. AdPC treated with >7 month NHT has significantly higher SRRM4 expression. Based on these findings, we conclude that SRRM4 expression in castrate-resistant tumors is highly correlated with NEPC and poor patient survival. It may serve as a diagnosis and prognosis biomarker of NEPC.
Collapse
Affiliation(s)
- Yinan Li
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qingfu Zhang
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina
- China Medical University, Shenyang, China
| | - Jessica Lovnicki
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ruiqi Chen
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Xuesen Dong
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
130
|
Storbeck KH, Mostaghel EA. Canonical and Noncanonical Androgen Metabolism and Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:239-277. [PMID: 31900912 DOI: 10.1007/978-3-030-32656-2_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Androgens are critical drivers of prostate cancer. In this chapter we first discuss the canonical pathways of androgen metabolism and their alterations in prostate cancer progression, including the classical, backdoor and 5α-dione pathways, the role of pre-receptor DHT metabolism, and recent findings on oncogenic splicing of steroidogenic enzymes. Next, we discuss the activity and metabolism of non-canonical 11-oxygenated androgens that can activate wild-type AR and are less susceptible to glucuronidation and inactivation than the canonical androgens, thereby serving as an under-recognized reservoir of active ligands. We then discuss an emerging literature on the potential non-canonical role of androgen metabolizing enzymes in driving prostate cancer. We conclude by discussing the potential implications of these findings for prostate cancer progression, particularly in context of new agents such as abiraterone and enzalutamide, which target the AR-axis for prostate cancer therapy, including mechanisms of response and resistance and implications of these findings for future therapy.
Collapse
Affiliation(s)
- Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Geriatric Research, Education and Clinical Center S-182, VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
131
|
He Y, Hooker E, Yu EJ, Cunha GR, Liao L, Xu J, Earl A, Wu H, Gonzalgo ML, Sun Z. Androgen signaling is essential for development of prostate cancer initiated from prostatic basal cells. Oncogene 2018; 38:2337-2350. [PMID: 30510232 PMCID: PMC6440846 DOI: 10.1038/s41388-018-0583-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/03/2018] [Accepted: 10/17/2018] [Indexed: 12/15/2022]
Abstract
Emerging evidence has shown that both prostatic basal and luminal cells are able to initiate oncogenic transformation. However, despite the diversity of tumor-initiating cells, most prostate cancer cells express the androgen receptor (AR) and depend on androgens for their growth and expansion, implicating an essential role of androgen signaling in prostate tumorigenesis. Prostatic basal cells express p63 and are able to differentiate into luminal, neuroendocrine, and basal cells. Here, we directly assessed the essential role of androgen signaling in prostatic p63-expressing cell initiated oncogenic transformation and tumor formation. Using novel and relevant mouse models, we demonstrated that, with stabilized β-catenin expression, prostatic p63-expressing cells possess the ability to initiate oncogenic transformation and, in the presence of androgens, they further transdifferentiate into luminal-like tumor cells and develop adenocarcinomas. Castration prior to activating stabilized β-catenin sensitizes p63-expressing cells and increases their sensitivity to androgens, resulting in aggressive and fast growing tumor phenotypes. These findings are consistent with what have been observed in human prostate cancers, demonstrating an essential role for androgen signaling in prostate cancer initiation and progression. This study also provides fresh insight into developing new therapeutic strategies for better treating prostate cancer patients.
Collapse
Affiliation(s)
- Yongfeng He
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA.,Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Erika Hooker
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA.,Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Eun-Jeong Yu
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA.,Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Gerald R Cunha
- Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrew Earl
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA
| | - Huiqing Wu
- Department of Pathology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA
| | - Michael L Gonzalgo
- Department of Urology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Zijie Sun
- Department of Cancer Biology, Beckman Research Institute and Cancer Center, City of Hope, Duarte, CA, 91010, USA. .,Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
132
|
Huhtaniemi R, Oksala R, Knuuttila M, Mehmood A, Aho E, Laajala TD, Nicorici D, Aittokallio T, Laiho A, Elo L, Ohlsson C, Kallio P, Mäkelä S, Mustonen MV, Sipilä P, Poutanen M. Adrenals Contribute to Growth of Castration-Resistant VCaP Prostate Cancer Xenografts. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2890-2901. [DOI: 10.1016/j.ajpath.2018.07.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/24/2018] [Accepted: 07/30/2018] [Indexed: 01/01/2023]
|
133
|
Shiota M, Kashiwagi E, Murakami T, Takeuchi A, Imada K, Inokuchi J, Tatsugami K, Eto M. Serum testosterone level as possible predictive marker in androgen receptor axis-targeting agents and taxane chemotherapies for castration-resistant prostate cancer. Urol Oncol 2018; 37:180.e19-180.e24. [PMID: 30446464 DOI: 10.1016/j.urolonc.2018.10.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/23/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Currently, several therapeutic options for castration-resistant prostate cancer (CRPC) are available, for which predictive biomarkers have not been established. Therefore, we aimed to reveal the association between pretreatment serum testosterone level and antitumor outcomes when treated with androgen receptor axis-targeting agents and taxane chemotherapies for CRPC. PATIENTS AND METHODS The present study included Japanese patients with metastatic prostate cancer whose serum testosterone levels during androgen-deprivation therapy were available. The antitumor outcomes when treated with enzalutamide, abiraterone, docetaxel, and cabazitaxel with clinicopathological parameters including serum testosterone levels during androgen-deprivation therapy, as well as prognoses including progression-free survival and overall survival, were examined. RESULTS Progression-free survival among men with higher serum testosterone level was superior to that among men with lower serum testosterone level when treated with enzalutamide. On the contrary, progression-free survival and overall survival among men with higher serum testosterone level were significantly inferior to those among men with lower serum testosterone level when treated with docetaxel and cabazitaxel, respectively. CONCLUSIONS The present study indicated distinct prognostic values of serum testosterone level when treated with androgen receptor axis-targeting agent and taxane chemotherapy for CRPC, suggesting that serum testosterone level may be useful predictive biomarker to navigate the appropriate therapy in patients with CRPC.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Eiji Kashiwagi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiko Murakami
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ario Takeuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenjiro Imada
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junichi Inokuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsunori Tatsugami
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
134
|
Hillebrand AC, Pizzolato LS, Branchini G, Brum IS. Androgenic modulation of AR-Vs. Endocrine 2018; 62:477-486. [PMID: 30027434 DOI: 10.1007/s12020-018-1682-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE The importance of androgen receptor variants (AR-Vs) is recognized in prostate cancer. AR-Vs have been the focus of many studies. Expression of AR-Vs has been proposed as a biomarker for resistance to androgen deprivation therapy for metastatic disease. Herein, we show dynamic changes in AR-Vs expression in response to androgen modulation. METHODS The C4-2B cell line was exposed to low (10-13 M) and high (10-8 M) androgen (dihydrotestosterone, DHT) levels, with or without flutamide. mRNA and protein expression levels were assessed by qPCR and immunohistochemistry, respectively. RESULTS We demonstrated that high levels of DHT downregulate AR-FL and AR-Vs. Even though AR-Vs did not present ligand-binding domain, thus were not capable of binding to DHT, they present dynamic changes under androgen treatment. Treatment with flutamide alone or in association with low levels of DHT stimulates growth of prostatic cells. CONCLUSIONS Importantly, we provide evidence that AR-Vs respond differently to androgenic modulation. These findings have implications for a better understanding of the role of AR-Vs in prostate carcinogenesis.
Collapse
Affiliation(s)
- Ana Caroline Hillebrand
- Laboratory of Molecular Endocrine and Tumoral Biology, Department of Physiology, Institute of Basic Sciences of Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Lolita Schneider Pizzolato
- Laboratory of Molecular Endocrine and Tumoral Biology, Department of Physiology, Institute of Basic Sciences of Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Gisele Branchini
- Department of Basic Sciences of Health, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Ilma Simoni Brum
- Laboratory of Molecular Endocrine and Tumoral Biology, Department of Physiology, Institute of Basic Sciences of Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil.
- Laboratory of Molecular Obstetrics and Gynecology, Experimental Research Center, Department of Obstetrics and Gynecology, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-903, Brazil.
| |
Collapse
|
135
|
Prekovic S, van den Broeck T, Linder S, van Royen ME, Houtsmuller AB, Handle F, Joniau S, Zwart W, Claessens F. Molecular underpinnings of enzalutamide resistance. Endocr Relat Cancer 2018; 25:R545–R557. [PMID: 30306781 DOI: 10.1530/erc-17-0136] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Prostate cancer (PCa) is among the most common adult malignancies, and the second leading cause of cancer-related death in men. As PCa is hormone dependent, blockade of the androgen receptor (AR) signaling is an effective therapeutic strategy for men with advanced metastatic disease. The discovery of enzalutamide, a compound that effectively blocks the AR axis and its clinical application has led to a significant improvement in survival time. However, the effect of enzalutamide is not permanent, and resistance to treatment ultimately leads to development of lethal disease, for which there currently is no cure. This review will focus on the molecular underpinnings of enzalutamide resistance, bridging the gap between the preclinical and clinical research on novel therapeutic strategies for combating this lethal stage of prostate cancer.
Collapse
Affiliation(s)
- S Prekovic
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T van den Broeck
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - S Linder
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M E van Royen
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
- Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, The Netherlands
| | - A B Houtsmuller
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
- Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, The Netherlands
| | - F Handle
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - S Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - W Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - F Claessens
- Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
136
|
Linder S, van der Poel HG, Bergman AM, Zwart W, Prekovic S. Enzalutamide therapy for advanced prostate cancer: efficacy, resistance and beyond. Endocr Relat Cancer 2018; 26:R31-R52. [PMID: 30382692 PMCID: PMC6215909 DOI: 10.1530/erc-18-0289] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
Abstract
The androgen receptor drives the growth of metastatic castration-resistant prostate cancer. This has led to the development of multiple novel drugs targeting this hormone-regulated transcription factor, such as enzalutamide – a potent androgen receptor antagonist. Despite the plethora of possible treatment options, the absolute survival benefit of each treatment separately is limited to a few months. Therefore, current research efforts are directed to determine the optimal sequence of therapies, discover novel drugs effective in metastatic castration-resistant prostate cancer and define patient subpopulations that ultimately benefit from these treatments. Molecular studies provide evidence on which pathways mediate treatment resistance and may lead to improved treatment for metastatic castration-resistant prostate cancer. This review provides, firstly a concise overview of the clinical development, use and effectiveness of enzalutamide in the treatment of advanced prostate cancer, secondly it describes translational research addressing enzalutamide response vs resistance and lastly highlights novel potential treatment strategies in the enzalutamide-resistant setting.
Collapse
Affiliation(s)
- Simon Linder
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Division of UrologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andries M Bergman
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Laboratory of Chemical Biology and Institute for Complex Molecular SystemsDepartment of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Stefan Prekovic
- Division of OncogenomicsOncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Correspondence should be addressed to S Prekovic:
| |
Collapse
|
137
|
Marret G, Doucet L, Hennequin C, Fizazi K, Culine S. Abiraterone in metastatic castration-resistant prostate cancer: Efficacy and safety in unselected patients. Cancer Treat Res Commun 2018; 17:37-42. [PMID: 30347333 DOI: 10.1016/j.ctarc.2018.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/24/2018] [Accepted: 10/08/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Abiraterone acetate (AA), an androgen biosynthesis inhibitor, is now a standard of care for men with metastatic, castration-sensitive and castration-resistant prostate cancer (mCRPC). Data exploring real-world toxicity and outcomes are scarce. METHODS Retrospective study on unselected patients with mCRPC on AA plus steroids. RESULTS 93 patients were included in the study. Median duration of treatment by AA was 7.5 months (95% CI 5.7-12) among the 58 patients pretreated with chemotherapy, versus 12.7 months ( 95% CI 8.2-35.9) among the 33 chemo-naive patients. Median survivals would reach 13.4 months (95% CI 10.2-19.1) and 36.4 months (95% CI 24.7-41.5) respectively. Rates of hypokalemia, peripheral edema, hypertension, cardiac failure, and overall survival assessments in patients with and without prior chemotherapy were similar to that previously reported in phase 3 randomized trials. The median survival time without adverse event of special interest was 7.5 months for hypokalemia and hypertension, and 5.3 months for liver-function test abnormalities (it was not reached for cardiac disorders). CONCLUSION Our findings provide further evidence for the survival benefits of AA with a low frequency of additional adverse events among unselected patients. In patients who have not developed hypokalemia or a transaminase increase within 7.5 and 5.3 months respectively, a lighter systematic monitoring may be considered.
Collapse
Affiliation(s)
- Grégoire Marret
- Department of Cancer Medicine, Hôpital Saint Louis, Paris, France.
| | - Ludovic Doucet
- Department of Cancer Medicine, Hôpital Saint Louis, Paris, France.
| | | | - Karim Fizazi
- Department of Cancer Medicine, Gustave Roussy, University of Paris Saclay, Villejuif, France.
| | - Stéphane Culine
- Department of Cancer Medicine, Hôpital Saint Louis, Paris, France.
| |
Collapse
|
138
|
Sundaresan L, Kumar P, Chatterjee S. Mechanistic insights into the differential effects of thalidomide and lenalidomide in metastatic prostate cancer. Future Oncol 2018; 14:2383-2401. [DOI: 10.2217/fon-2018-0090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aim: To understand why thalidomide and lenalidomide exhibit different responses in metastatic prostate cancer (mPCa) treatment. Methods: We analyzed the perturbation signatures of thalidomide, lenalidomide, flutamide treated mPCa cell line from Library of Integrated Network-based Cellular Signatures database and transcriptome of docetaxel-treated mPCa patients. Results: Flutamide and docetaxel downregulated ‘Steroid Biosynthesis’, ‘Cell cycle’ and PCa specific transcription factor networks. Thalidomide inhibited ‘Cell cycle’ and ‘E2F network’, possibly accounting for its synergistic effects with docetaxel. Conversely, lenalidomide promoted ‘Cell cycle’ and ‘Cholesterol biosynthesis’. Conclusion: Hence, we propose that lenalidomide upregulates cholesterol synthesis followed by enhanced rate of cell cycle, thereby nurturing a hyperproliferative tumor microenvironment. In summary, this study offers a possible explanation for the differential outcomes in the treatment of mPCa with thalidomide and lenalidomide.
Collapse
Affiliation(s)
- Lakshmikirupa Sundaresan
- AU-KBC Research Center, Anna University, Chennai, India
- Department of Biotechnology, Anna University, Chennai, India
| | - Pavitra Kumar
- AU-KBC Research Center, Anna University, Chennai, India
| | - Suvro Chatterjee
- AU-KBC Research Center, Anna University, Chennai, India
- Department of Biotechnology, Anna University, Chennai, India
| |
Collapse
|
139
|
Deb S, Pham S, Ming DS, Chin MY, Adomat H, Hurtado-Coll A, Gleave ME, Guns EST. Characterization of Precursor-Dependent Steroidogenesis in Human Prostate Cancer Models. Cancers (Basel) 2018; 10:cancers10100343. [PMID: 30241348 PMCID: PMC6210088 DOI: 10.3390/cancers10100343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 11/17/2022] Open
Abstract
Castration-resistant prostate tumors acquire the independent capacity to generate androgens by upregulating steroidogenic enzymes or using steroid precursors produced by the adrenal glands for continued growth and sustainability. The formation of steroids was measured by liquid chromatography-mass spectrometry in LNCaP and 22Rv1 prostate cancer cells, and in human prostate tissues, following incubation with steroid precursors (22-OH-cholesterol, pregnenolone, 17-OH-pregnenolone, progesterone, 17-OH-progesterone). Pregnenolone, progesterone, 17-OH-pregnenolone, and 17-OH-progesterone increased C21 steroid (5-pregnan-3,20-dione, 5-pregnan-3,17-diol-20-one, 5-pregnan-3-ol-20-one) formation in the backdoor pathway, and demonstrated a trend of stimulating dihydroepiandrosterone or its precursors in the backdoor pathway in LNCaP and 22Rv1 cells. The precursors differentially affected steroidogenic enzyme messenger RNA (mRNA) expressions in the cell lines. The steroidogenesis following incubation of human prostate tissue with 17-OH-pregnenolone and progesterone produced trends similar to those observed in cell lines. Interestingly, the formation of C21 steroids from classical pathway was not stimulated but backdoor pathway steroids (e.g., 5-pregnan-3,20-dione, 5-pregnan-3-ol-20-one) were elevated following incubations with prostate tissues. Overall, C21 steroids were predominantly formed in the classical as well as backdoor pathways, and steroid precursors induced a diversion of steroidogenesis to the backdoor pathway in both cell lines and human prostate tissue, and influenced adaptive steroidogenesis to form C21 steroids.
Collapse
Affiliation(s)
- Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| | - Steven Pham
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Dong-Sheng Ming
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Mei Yieng Chin
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Hans Adomat
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Antonio Hurtado-Coll
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | - Martin E Gleave
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
140
|
Fucic A, Aghajanyan A, Culig Z, Le Novere N. Systems Oncology: Bridging Pancreatic and Castrate Resistant Prostate Cancer. Pathol Oncol Res 2018; 25:1269-1277. [PMID: 30220022 DOI: 10.1007/s12253-018-0467-8] [Citation(s) in RCA: 2] [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: 12/08/2017] [Accepted: 09/03/2018] [Indexed: 12/31/2022]
Abstract
Large investments by pharmaceutical companies in the development of new antineoplastic drugs have not been resulting in adequate advances of new therapies. Despite the introduction of new methods, technologies, translational medicine and bioinformatics, the usage of collected knowledge is unsatisfactory. In this paper, using examples of pancreatic ductal adenocarcinoma (PaC) and castrate-resistant prostate cancer (CRPC), we proposed a concept showing that, in order to improve applicability of current knowledge in oncology, the re-clustering of clinical and scientific data is crucial. Such an approach, based on systems oncology, would include bridging of data on biomarkers and pathways between different cancer types. Proposed concept would introduce a new matrix, which enables combining of already approved therapies between cancer types. Paper provides a (a) detailed analysis of similarities in mechanisms of etiology and progression between PaC and CRPC, (b) diabetes as common hallmark of both cancer types and (c) knowledge gaps and directions of future investigations. Proposed horizontal and vertical matrix in cancer profiling has potency to improve current antineoplastic therapy efficacy. Systems biology map using Systems Biology Graphical Notation Language is used for summarizing complex interactions and similarities of mechanisms in biology of PaC and CRPC.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Ksaverska c 2, 10000, Zagreb, Croatia.
| | - A Aghajanyan
- Institute of Medicine, Peoples' Friendship University of Russia, Moscow, Russian Federation
| | - Z Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
141
|
Poulose N, Mills IG, Steele RE. The impact of transcription on metabolism in prostate and breast cancers. Endocr Relat Cancer 2018; 25:R435-R452. [PMID: 29760165 DOI: 10.1530/erc-18-0048] [Citation(s) in RCA: 7] [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: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
Abstract
Metabolic dysregulation is regarded as an important driver in cancer development and progression. The impact of transcriptional changes on metabolism has been intensively studied in hormone-dependent cancers, and in particular, in prostate and breast cancer. These cancers have strong similarities in the function of important transcriptional drivers, such as the oestrogen and androgen receptors, at the level of dietary risk and epidemiology, genetics and therapeutically. In this review, we will focus on the function of these nuclear hormone receptors and their downstream impact on metabolism, with a particular focus on lipid metabolism. We go on to discuss how lipid metabolism remains dysregulated as the cancers progress. We conclude by discussing the opportunities that this presents for drug repurposing, imaging and the development and testing of new therapeutics and treatment combinations.
Collapse
Affiliation(s)
- Ninu Poulose
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
| | - Ian G Mills
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
- Nuffield Department of Surgical SciencesJohn Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Rebecca E Steele
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
| |
Collapse
|
142
|
Staal J, Beyaert R. Inflammation and NF-κB Signaling in Prostate Cancer: Mechanisms and Clinical Implications. Cells 2018; 7:E122. [PMID: 30158439 PMCID: PMC6162478 DOI: 10.3390/cells7090122] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer is a highly prevalent form of cancer that is usually slow-developing and benign. Due to its high prevalence, it is, however, still the second most common cause of death by cancer in men in the West. The higher prevalence of prostate cancer in the West might be due to elevated inflammation from metabolic syndrome or associated comorbidities. NF-κB activation and many other signals associated with inflammation are known to contribute to prostate cancer malignancy. Inflammatory signals have also been associated with the development of castration resistance and resistance against other androgen depletion strategies, which is a major therapeutic challenge. Here, we review the role of inflammation and its link with androgen signaling in prostate cancer. We further describe the role of NF-κB in prostate cancer cell survival and proliferation, major NF-κB signaling pathways in prostate cancer, and the crosstalk between NF-κB and androgen receptor signaling. Several NF-κB-induced risk factors in prostate cancer and their potential for therapeutic targeting in the clinic are described. A better understanding of the inflammatory mechanisms that control the development of prostate cancer and resistance to androgen-deprivation therapy will eventually lead to novel treatment options for patients.
Collapse
Affiliation(s)
- Jens Staal
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, 9052 Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
143
|
Sowalsky AG, Ye H, Bhasin M, Van Allen EM, Loda M, Lis RT, Montaser-Kouhsari L, Calagua C, Ma F, Russo JW, Schaefer RJ, Voznesensky OS, Zhang Z, Bubley GJ, Montgomery B, Mostaghel EA, Nelson PS, Taplin ME, Balk SP. Neoadjuvant-Intensive Androgen Deprivation Therapy Selects for Prostate Tumor Foci with Diverse Subclonal Oncogenic Alterations. Cancer Res 2018; 78:4716-4730. [PMID: 29921690 PMCID: PMC6095796 DOI: 10.1158/0008-5472.can-18-0610] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/02/2018] [Accepted: 06/05/2018] [Indexed: 01/06/2023]
Abstract
Primary prostate cancer can have extensive microheterogeneity, but its contribution to the later emergence of metastatic castration-resistant prostate cancer (mCRPC) remains unclear. In this study, we microdissected residual prostate cancer foci in radical prostatectomies from 18 men treated with neoadjuvant-intensive androgen deprivation therapy (leuprolide, abiraterone acetate, and prednisone) and analyzed them for resistance mechanisms. Transcriptome profiling showed reduced but persistent androgen receptor (AR) activity in residual tumors, with no increase in neuroendocrine differentiation. Proliferation correlated negatively with AR activity but positively with decreased RB1 expression, and whole-exome sequencing (WES) further showed enrichment for RB1 genomic loss. In 15 cases where 2 or 3 tumor foci were microdissected, WES confirmed a common clonal origin but identified multiple oncogenic alterations unique to each focus. These findings show that subclones with oncogenic alterations found in mCRPC are present in primary prostate cancer and are selected for by neoadjuvant-intense androgen deprivation therapy. In particular, this study indicates that subclonal RB1 loss may be more common than previously appreciated in intermediate- to high-risk primary prostate cancer and may be an early event, independent of neuroendocrine differentiation, in the development of mCRPC. Comprehensive molecular analyses of primary prostate cancer may detect aggressive subclones and possibly inform adjuvant strategies to prevent recurrence.Significance: Neoadjuvant androgen deprivation therapy for prostate cancer selects for tumor foci with subclonal genomic alterations, which may comprise the origin of metastatic castration-resistant prostate cancer. Cancer Res; 78(16); 4716-30. ©2018 AACR.
Collapse
Affiliation(s)
- Adam G Sowalsky
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, NIH, Bethesda, Maryland
| | - Huihui Ye
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Manoj Bhasin
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Eliezer M Van Allen
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Massimo Loda
- Department of Molecular Oncologic Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, Masschusetts
| | - Rosina T Lis
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Carla Calagua
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Fen Ma
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Joshua W Russo
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rachel J Schaefer
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Olga S Voznesensky
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Zhenwei Zhang
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Glenn J Bubley
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Bruce Montgomery
- Medical Oncology Division, University of Washington, Seattle, Washington
| | - Elahe A Mostaghel
- Medical Oncology Division, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Medical Oncology Division, University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mary-Ellen Taplin
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Steven P Balk
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| |
Collapse
|
144
|
Kallio HML, Hieta R, Latonen L, Brofeldt A, Annala M, Kivinummi K, Tammela TL, Nykter M, Isaacs WB, Lilja HG, Bova GS, Visakorpi T. Constitutively active androgen receptor splice variants AR-V3, AR-V7 and AR-V9 are co-expressed in castration-resistant prostate cancer metastases. Br J Cancer 2018; 119:347-356. [PMID: 29988112 PMCID: PMC6070921 DOI: 10.1038/s41416-018-0172-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND A significant subset of prostate cancer (PC) patients with a castration-resistant form of the disease (CRPC) show primary resistance to androgen receptor (AR)-targeting drugs developed against CRPC. As one explanation could be the expression of constitutively active androgen receptor splice variants (AR-Vs), our current objectives were to study AR-Vs and other AR aberrations to better understand the emergence of CRPC. METHODS We analysed specimens from different stages of prostate cancer by next-generation sequencing and immunohistochemistry. RESULTS AR mutations and copy number variations were detected only in CRPC specimens. Genomic structural rearrangements of AR were observed in 5/30 metastatic CRPC patients, but they were not associated with expression of previously known AR-Vs. The predominant AR-Vs detected were AR-V3, AR-V7 and AR-V9, with the expression levels being significantly higher in CRPC cases compared to prostatectomy samples. Out of 25 CRPC metastases that expressed any AR variant, 17 cases harboured expression of all three of these AR-Vs. AR-V7 protein expression was highly heterogeneous and higher in CRPC compared to hormone-naïve tumours. CONCLUSIONS AR-V3, AR-V7 and AR-V9 are co-expressed in CRPC metastases highlighting the fact that inhibiting AR function via regions common to all AR-Vs is likely to provide additional benefit to patients with CRPC.
Collapse
Affiliation(s)
- Heini M L Kallio
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.
| | - Reija Hieta
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Leena Latonen
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Anniina Brofeldt
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Kati Kivinummi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Teuvo L Tammela
- Department of Urology, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - William B Isaacs
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans G Lilja
- Departments of Surgery (Urology), Laboratory Medicine and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Translational Medicine, Lund University, Malmö, Sweden.,Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - G Steven Bova
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
145
|
Wang W, Chen ZX, Guo DY, Tao YX. Regulation of prostate cancer by hormone-responsive G protein-coupled receptors. Pharmacol Ther 2018; 191:135-147. [PMID: 29909235 DOI: 10.1016/j.pharmthera.2018.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/01/2018] [Indexed: 11/27/2022]
Abstract
Regulation of prostate cancer by androgen and androgen receptor (AR), and blockade of AR signaling by AR antagonists and steroidogenic enzyme inhibitors have been extensively studied. G protein-coupled receptors (GPCRs) are a family of membrane receptors that regulate almost all physiological processes. Nearly 40% of FDA-approved drugs in the market target GPCRs. A variety of GPCRs that mediate reproductive function have been demonstrated to be involved in the regulation of prostate cancer. These GPCRs include gonadotropin-releasing hormone receptor, luteinizing hormone receptor, follicle-stimulating hormone receptor, relaxin receptor, ghrelin receptor, and kisspeptin receptor. We highlight here GPCR regulation of prostate cancer by these GPCRs. Further therapeutic approaches targeting these GPCRs for the treatment of prostate cancer are summarized.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Zhao-Xia Chen
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA.
| |
Collapse
|
146
|
Verma K, Gupta N, Zang T, Wangtrakluldee P, Srivastava SK, Penning TM, Trippier PC. AKR1C3 Inhibitor KV-37 Exhibits Antineoplastic Effects and Potentiates Enzalutamide in Combination Therapy in Prostate Adenocarcinoma Cells. Mol Cancer Ther 2018; 17:1833-1845. [PMID: 29891491 DOI: 10.1158/1535-7163.mct-17-1023] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/01/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
Abstract
Aldo-keto reductase 1C3 (AKR1C3), also known as type 5 17 β-hydroxysteroid dehydrogenase, is responsible for intratumoral androgen biosynthesis, contributing to the development of castration-resistant prostate cancer (CRPC) and eventual chemotherapeutic failure. Significant upregulation of AKR1C3 is observed in CRPC patient samples and derived CRPC cell lines. As AKR1C3 is a downstream steroidogenic enzyme synthesizing intratumoral testosterone (T) and 5α-dihydrotestosterone (DHT), the enzyme represents a promising therapeutic target to manage CRPC and combat the emergence of resistance to clinically employed androgen deprivation therapy. Herein, we demonstrate the antineoplastic activity of a potent, isoform-selective and hydrolytically stable AKR1C3 inhibitor (E)-3-(4-(3-methylbut-2-en-1-yl)-3-(3-phenylpropanamido)phenyl)acrylic acid (KV-37), which reduces prostate cancer cell growth in vitro and in vivo and sensitizes CRPC cell lines (22Rv1 and LNCaP1C3) toward the antitumor effects of enzalutamide. Crucially, KV-37 does not induce toxicity in nonmalignant WPMY-1 prostate cells nor does it induce weight loss in mouse xenografts. Moreover, KV-37 reduces androgen receptor (AR) transactivation and prostate-specific antigen expression levels in CRPC cell lines indicative of a therapeutic effect in prostate cancer. Combination studies of KV-37 with enzalutamide reveal a very high degree of synergistic drug interaction that induces significant reduction in prostate cancer cell viability via apoptosis, resulting in >200-fold potentiation of enzalutamide action in drug-resistant 22Rv1 cells. These results demonstrate a promising therapeutic strategy for the treatment of drug-resistant CRPC that invariably develops in prostate cancer patients following initial treatment with AR antagonists such as enzalutamide. Mol Cancer Ther; 17(9); 1833-45. ©2018 AACR.
Collapse
Affiliation(s)
- Kshitij Verma
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Nehal Gupta
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Tianzhu Zang
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Phumvadee Wangtrakluldee
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sanjay K Srivastava
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas.,Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, Texas
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas. .,Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| |
Collapse
|
147
|
Cioni B, Zwart W, Bergman AM. Androgen receptor moonlighting in the prostate cancer microenvironment. Endocr Relat Cancer 2018; 25:R331-R349. [PMID: 29618577 DOI: 10.1530/erc-18-0042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023]
Abstract
Androgen receptor (AR) signaling is vital for the normal development of the prostate and is critically involved in prostate cancer (PCa). AR is not only found in epithelial prostate cells but is also expressed in various cells in the PCa-associated stroma, which constitute the tumor microenvironment (TME). In the TME, AR is expressed in fibroblasts, macrophages, lymphocytes and neutrophils. AR expression in the TME was shown to be decreased in higher-grade and metastatic PCa, suggesting that stromal AR plays a protective role against PCa progression. With that, the functionality of AR in stromal cells appears to deviate from the receptor's classical function as described in PCa cells. However, the biological action of AR in these cells and its effect on cancer progression remains to be fully understood. Here, we systematically review the pathological, genomic and biological literature on AR actions in various subsets of prostate stromal cells and aim to better understand the consequences of AR signaling in the TME in relation to PCa development and progression.
Collapse
Affiliation(s)
- B Cioni
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - W Zwart
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode InstituteThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A M Bergman
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
148
|
Lee HJ, Li J, Vickman RE, Li J, Liu R, Durkes AC, Elzey BD, Yue S, Liu X, Ratliff TL, Cheng JX. Cholesterol Esterification Inhibition Suppresses Prostate Cancer Metastasis by Impairing the Wnt/β-catenin Pathway. Mol Cancer Res 2018; 16:974-985. [PMID: 29545473 PMCID: PMC5984676 DOI: 10.1158/1541-7786.mcr-17-0665] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/11/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
Dysregulation of cholesterol is a common characteristic of human cancers including prostate cancer. This study observed an aberrant accumulation of cholesteryl ester in metastatic lesions using Raman spectroscopic analysis of lipid droplets in human prostate cancer patient tissues. Inhibition of cholesterol esterification in prostate cancer cells significantly suppresses the development and growth of metastatic cancer lesions in both orthotopic and intracardiac injection mouse models. Gene expression profiling reveals that cholesteryl ester depletion suppresses the metastatic potential through upregulation of multiple regulators that negatively impact metastasis. In addition, Wnt/β-catenin, a vital pathway for metastasis, is downregulated upon cholesteryl ester depletion. Mechanistically, inhibition of cholesterol esterification significantly blocks secretion of Wnt3a through reduction of monounsaturated fatty acid levels, which limits Wnt3a acylation. These results collectively validate cholesterol esterification as a novel metabolic target for treating metastatic prostate cancer. Mol Cancer Res; 16(6); 974-85. ©2018 AACR.
Collapse
Affiliation(s)
- Hyeon Jeong Lee
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, Indiana
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Renee E Vickman
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, Indiana
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Junjie Li
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Rui Liu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Abigail C Durkes
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Shuhua Yue
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Xiaoqi Liu
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, Indiana
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Timothy L Ratliff
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, Indiana
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Ji-Xin Cheng
- Interdisciplinary Life Science Program, Purdue University, West Lafayette, Indiana.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
- Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Photonics Center, Boston University, Boston, Massachusetts
| |
Collapse
|
149
|
Allott EH, Masko EM, Freedland AR, Macias E, Pelton K, Solomon KR, Mostaghel EA, Thomas GV, Pizzo SV, Freeman MR, Freedland SJ. Serum cholesterol levels and tumor growth in a PTEN-null transgenic mouse model of prostate cancer. Prostate Cancer Prostatic Dis 2018; 21:196-203. [PMID: 29795142 PMCID: PMC6026483 DOI: 10.1038/s41391-018-0045-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/28/2022]
Abstract
Background Some, but not all, epidemiologic evidence supports a role for cholesterol, the precursor for steroid hormone synthesis, in prostate cancer. Using a PTEN-null transgenic mouse model of prostate cancer, we tested the effect of modifying serum cholesterol levels on prostate tumor development and growth. We hypothesized that serum cholesterol reduction would lower tumor androgens and slow prostate cancer growth. Methods PTENloxP/loxP-Cre+ mice consuming ad libitum high fat, high cholesterol diets (40% fat, 1.25% cholesterol) were randomized after weaning to receive the cholesterol uptake inhibitor, ezetimibe (30 mg/kg/day), or no intervention, and sacrificed at 2, 3 or 4 months of age. Serum cholesterol and testosterone were measured by ELISA and intraprostatic androgens by mass spectrometry. Prostate histology was graded, and proliferation and apoptosis in tumor epithelium and stroma was assessed by Ki67 and TUNEL, respectively. Results Ezetimibe-treated mice had lower serum cholesterol at 4 months (p=0.031). Serum cholesterol was positively correlated with prostate weight (p=0.033) and tumor epithelial proliferation (p=0.069), and negatively correlated with tumor epithelial apoptosis (p=0.004). Serum cholesterol was unrelated to body weight (p=0.195). Tumor stromal cell proliferation was reduced in the ezetimibe group (p=0.010). Increased serum cholesterol at 4 months was associated with elevated intraprostatic DHEA, testosterone and androstenedione (p=0.043, p=0.074, p=0.031, respectively). However, cholesterol reduction did not significantly affect adenocarcinoma development at 2, 3 or 4 months of age (0%, 78%, 100% in ezetimibe-treated vs. 0%, 80%, 100% in mice not receiving ezetimibe). Conclusions Though serum cholesterol reduction did not significantly affect the rate of adenocarcinoma development in the PTEN-null transgenic mouse model of prostate cancer, it lowered intraprostatic androgens and slowed tumor growth. These findings support a role for serum cholesterol in promoting prostate cancer growth, potentially via enhanced tumor androgen signaling, and may provide new insight into cholesterol-lowering interventions for prostate cancer treatment.
Collapse
Affiliation(s)
- Emma H Allott
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth M Masko
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Alexis R Freedland
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Everardo Macias
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kristine Pelton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keith R Solomon
- The Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA.,Applied Photophysics, Beverly, MA, USA
| | - Elahe A Mostaghel
- Division of Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - George V Thomas
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.,Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Michael R Freeman
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Freedland
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Division of Urology, Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
150
|
Lee AR, Che N, Lovnicki JM, Dong X. Development of Neuroendocrine Prostate Cancers by the Ser/Arg Repetitive Matrix 4-Mediated RNA Splicing Network. Front Oncol 2018; 8:93. [PMID: 29666783 PMCID: PMC5891588 DOI: 10.3389/fonc.2018.00093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/16/2018] [Indexed: 12/19/2022] Open
Abstract
While the use of next-generation androgen receptor pathway inhibition (ARPI) therapy has significantly increased the survival of patients with metastatic prostate adenocarcinoma (AdPC), several groups have reported a treatment-resistant mechanism, whereby cancer cells can become androgen receptor (AR) indifferent and gain a neuroendocrine (NE)-like phenotype. This subtype of castration-resistant prostate cancer has been termed "treatment-induced castration-resistant neuroendocrine prostate cancer" (CRPC-NE). Recent reports indicate that the overall genomic landscapes of castration-resistant tumors with AdPC phenotypes and CRPC-NE are not significantly altered. However, CRPC-NE tumors have been found to contain a NE-specific pattern throughout their epigenome and splicing transcriptome, which are significantly modified. The molecular mechanisms by which CRPC-NE develops remain unclear, but several factors have been implicated in the progression of the disease. Recently, Ser/Arg repetitive matrix 4 (SRRM4), a neuronal-specific RNA splicing factor that is upregulated in CRPC-NE tumors, has been shown to establish a CRPC-NE-unique splicing transcriptome, to induce a NE-like morphology in AdPC cells, and, most importantly, to transform AdPC cells into CRPC-NE xenografts under ARPI. Moreover, the SRRM4-targeted splicing genes are highly enriched in various neuronal processes, suggesting their roles in facilitating a CRPC-NE program. This article will address the importance of SRRM4-mediated alternative RNA splicing in reprogramming translated proteins to facilitate NE differentiation, survival, and proliferation of cells to establish CRPC-NE tumors. In addition, we will discuss the potential roles of SRRM4 in conjunction with other known pathways and factors important for CRPC-NE development, such as the AR pathway, TP53 and RB1 genes, the FOXA family of proteins, and environmental factors. This study aims to explore the multifaceted functions of SRRM4 and SRRM4-mediated splicing in driving a CRPC-NE program as a coping mechanism for therapy resistance, as well as define future SRRM4-targeted therapeutic approaches for treating CRPC-NE or mitigating its development.
Collapse
Affiliation(s)
- Ahn R Lee
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Che
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jessica M Lovnicki
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|