1
|
Dhawale SA, Bhosle P, Mahajan S, Patil G, Gawale S, Ghodke M, Tapadiya G, Ansari A. Dual targeting in prostate cancer with phytoconstituents as a potent lead: a computational approach for novel drug discovery. J Biomol Struct Dyn 2024; 42:8906-8919. [PMID: 37649379 DOI: 10.1080/07391102.2023.2251059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
Prostate Cancer (PCa) is an abnormal cell growth within the prostate. This condition is the second most widespread malignancy in elderly males and one of the most frequently diagnosed life-threatening conditions. The Androgen receptor signaling pathway played a crucial role in the initiation and spread to increase the risk of PCa. Hence, targeting the AR receptor signaling pathway is a key strategy for a therapeutic plan for PCa. Our study focuses on recognizing potential inhibitors for dual targeting in PCa by using the in-silico approach. In this study, we target the two enzymes that are CYP17A1 (3RUK) and 5α-reductase (3G1R) responsible for PCa, with the help of phytoconstituents. The natural plant contains various phytochemical types produced from secondary metabolites and used as a medical treatment. The in-silico investigation of phytoconstituents and enzymes was done by approaching molecular docking, ADMET analysis, and high-level molecular dynamic simulation used to assess the stability and binding affinities of the protein-ligand complex. Some phytoconstituents, such as Peonidin, Pelargonidin, Malvidin and Berberine show complex has good molecular interaction with protein. The reliability of the docking scores was examined using a molecular dynamic simulation, which revealed that the complex remained stable throughout the simulation, which ranged from 0 to 200 ns. The selected hits may be effective against CYP17A1 (3RUK) and 5α-reductase (3G1R) (PCa) using a computer-aided drug design (CADD) method, which further enables researchers for upcoming in-vivo and in-vitro research, according to our in-silico approach.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sachin A Dhawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Pallavi Bhosle
- Pharmacology, Shrinath College of Pharmacy, Aurangabad, India
| | | | - Geetanjali Patil
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Sachin Gawale
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Mangesh Ghodke
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | - Ganesh Tapadiya
- Department of Pharmaceutical Chemistry, Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad, India
| | | |
Collapse
|
2
|
Thomas S, Ricke WA, Li L. Toxicoproteomics of Mono(2-ethylhexyl) phthalate and Perfluorooctanesulfonic Acid in Models of Prostatic Diseases. Chem Res Toxicol 2023; 36:251-259. [PMID: 36749316 PMCID: PMC10041651 DOI: 10.1021/acs.chemrestox.2c00328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Benign and malignant prostatic diseases are common, costly, and burdensome; moreover, they share fundamental underlying molecular processes. Several ubiquitous contaminants may perturb these processes, possibly via peroxisome proliferator-activated receptor (PPAR) signaling, but the role of environmental exposures─particularly mixtures─in prostatic diseases is undefined. In the present study, nontumorigenic prostate stromal cells and metastatic prostate epithelial cells were exposed to ubiquitous exogenous PPAR ligands under different dosing paradigms, including a mixture, and effects were assessed via mass spectrometry-based global proteomics. In prostate stromal cells, environmentally relevant levels of mono(2-ethylhexyl) phthalate (MEHP), alone and in combination with perfluorooctanesulfonic acid, led to significant changes in proteins involved in key processes underlying prostatic diseases: oxidative stress defense, proteostasis, damage-associated molecular pattern signaling, and innate immune response signaling. A follow-up experiment in metastatic prostate epithelial cells showed that the occupationally relevant levels of MEHP perturbed similar processes, including lipid, cholesterol, steroid, and alcohol metabolism; apoptosis and coagulation regulation; wound response; and aging. This work shows that environmental exposures may contribute to prostatic diseases by perturbing key processes of a proposed adverse outcome pathway, including lipid metabolism, oxidative stress, and inflammation. Future in vivo research will investigate the role of contaminants in prostatic diseases and in preventative agents.
Collapse
Affiliation(s)
- Samuel Thomas
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - William A. Ricke
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53706, USA
- George M. O’Brien Research Center of Excellence, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Lingjun Li
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
3
|
Preclinical models of prostate cancer - modelling androgen dependency and castration resistance in vitro, ex vivo and in vivo. Nat Rev Urol 2023:10.1038/s41585-023-00726-1. [PMID: 36788359 DOI: 10.1038/s41585-023-00726-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
Prostate cancer is well known to be dependent on the androgen receptor (AR) for growth and survival. Thus, AR is the main pharmacological target to treat this disease. However, after an initially positive response to AR-targeting therapies, prostate cancer will eventually evolve to castration-resistant prostate cancer, which is often lethal. Tumour growth was initially thought to become androgen-independent following treatments; however, results from molecular studies have shown that most resistance mechanisms involve the reactivation of AR. Consequently, tumour cells become resistant to castration - the blockade of testicular androgens - and not independent of AR per se. However, confusion still remains on how to properly define preclinical models of prostate cancer, including cell lines. Most cell lines were isolated from patients for cell culture after evolution of the tumour to castration-resistant prostate cancer, but not all of these cell lines are described as castration resistant. Moreover, castration refers to the blockade of testosterone production by the testes; thus, even the concept of "castration" in vitro is questionable. To ensure maximal transfer of knowledge from scientific research to the clinic, understanding the limitations and advantages of preclinical models, as well as how these models recapitulate cancer cell androgen dependency and can be used to study castration resistance mechanisms, is essential.
Collapse
|
4
|
Ohlmann CH, Jäschke M, Jaehnig P, Krege S, Gschwend J, Rexer H, Junker K, Zillmann R, Rüssel C, Hellmis E, Suttmann H, Janssen M, Marin J, Hübner A, Mathers M, Gleißner J, Scheffler M, Feyerabend S, Telle J, Klier J, Stöckle M. LHRH sparing therapy in patients with chemotherapy-naïve, mCRPC treated with abiraterone acetate plus prednisone: results of the randomized phase II SPARE trial. Prostate Cancer Prostatic Dis 2022; 25:778-784. [PMID: 35430584 PMCID: PMC9705242 DOI: 10.1038/s41391-022-00533-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/06/2022] [Accepted: 03/24/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Although the benefit of androgen deprivation therapy (ADT) continuation in metastatic castration-resistant prostate cancer (mCRPC) remains controversial, clinical evidence is lacking. Recent results indicated that treatment with abiraterone acetate (AA) plus prednisone (P) further suppresses serum testosterone levels over ADT alone, suggesting that continuation of ADT in the treatment of mCRPC may not be necessary. METHODS In this exploratory phase 2 study, mCRPC patients were randomized with a 1:1 ratio to receive either continued ADT plus AA + P (Arm A) or AA + P alone (Arm B). The primary endpoint was the rate of radiographic progression-free survival (rPFS) at month 12. Secondary endpoints included PSA-response rate, objective response, time to PSA progression and safety. RESULTS A total of 68 patients were equally randomized between the two study arms. Median testosterone-levels remained below castrate-levels throughout treatment in all patients. According to the intention-to-treat analysis the rPFS rate was 0.84 in Arm A and 0.89 in Arm B. Moderate and severe treatment-emergent adverse events were reported for 72% of the patients in Arm A and for 85% of the patients in Arm B. CONCLUSIONS AA + P treatment without ADT may be effective in mCRPC patients and ADT may not be necessary in patients receiving AA + P.
Collapse
Affiliation(s)
- Carsten-Henning Ohlmann
- grid.411937.9Department of Urology, Saarland University Medical Center, Homburg/Saar, Germany ,Department of Urology, Johanniter-Kliniken Bonn, Bonn, Germany
| | - Michelle Jäschke
- grid.411937.9Department of Urology, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Susanne Krege
- grid.461714.10000 0001 0006 4176Department of Urology, Evangelische Kliniken Essen Mitte, Essen, Germany
| | - Jürgen Gschwend
- grid.6936.a0000000123222966Department of Urology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | | | - Kerstin Junker
- grid.411937.9Department of Urology, Saarland University Medical Center, Homburg/Saar, Germany
| | | | | | - Eva Hellmis
- Urologicum-Duisburg Fachärztesozietät, Duisburg, Germany
| | | | - Martin Janssen
- grid.411937.9Department of Urology, Saarland University Medical Center, Homburg/Saar, Germany ,grid.5949.10000 0001 2172 9288Department of Urology, University Münster, Münster, Germany
| | | | | | | | | | | | | | - Jens Telle
- Urologische Praxisgemeinschaft, Wolfsburg, Germany
| | - Jörg Klier
- Urologische Partnerschaft, Köln, Germany
| | - Michael Stöckle
- grid.411937.9Department of Urology, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
5
|
Sharpe MA, Baskin DS, Jenson AV, Baskin AM. Hijacking Sexual Immuno-Privilege in GBM-An Immuno-Evasion Strategy. Int J Mol Sci 2021; 22:10983. [PMID: 34681642 PMCID: PMC8536168 DOI: 10.3390/ijms222010983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/12/2023] Open
Abstract
Regulatory T-cells (Tregs) are immunosuppressive T-cells, which arrest immune responses to 'Self' tissues. Some immunosuppressive Tregs that recognize seminal epitopes suppress immune responses to the proteins in semen, in both men and women. We postulated that GBMs express reproductive-associated proteins to manipulate reproductive Tregs and to gain immune privilege. We analyzed four GBM transcriptome databases representing ≈900 tumors for hypoxia-responsive Tregs, steroidogenic pathways, and sperm/testicular and placenta-specific genes, stratifying tumors by expression. In silico analysis suggested that the presence of reproductive-associated Tregs in GBM tumors was associated with worse patient outcomes. These tumors have an androgenic signature, express male-specific antigens, and attract reproductive-associated Related Orphan Receptor C (RORC)-Treg immunosuppressive cells. GBM patient sera were interrogated for the presence of anti-sperm/testicular antibodies, along with age-matched controls, utilizing monkey testicle sections. GBM patient serum contained anti-sperm/testicular antibodies at levels > six-fold that of controls. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are associated with estrogenic tumors which appear to mimic placental tissue. We demonstrate that RORC-Tregs drive poor patient outcome, and Treg infiltration correlates strongly with androgen levels. Androgens support GBM expression of sperm/testicular proteins allowing Tregs from the patient's reproductive system to infiltrate the tumor. In contrast, estrogen appears responsible for MDSC/TAM immunosuppression.
Collapse
MESH Headings
- Androgens/metabolism
- Brain Neoplasms/immunology
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Databases, Factual
- Estrogens/metabolism
- Female
- Glioblastoma/immunology
- Glioblastoma/mortality
- Glioblastoma/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Microglia/immunology
- Microglia/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amanda V. Jenson
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - Alexandra M. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| |
Collapse
|
6
|
Cortés-Benítez F, Roy J, Perreault M, Maltais R, Poirier D. 16-Picolyl-androsterone derivative exhibits potent 17β-HSD3 inhibitory activity, improved metabolic stability and cytotoxic effect on various cancer cells: Synthesis, homology modeling and docking studies. J Steroid Biochem Mol Biol 2021; 210:105846. [PMID: 33609690 DOI: 10.1016/j.jsbmb.2021.105846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 11/18/2022]
Abstract
A new androsterone derivative bearing a 16β-picolyl group (compound 5; FCO-586-119) was synthetized in four steps from the lead compound 1 (RM-532-105). We measured its inhibitory activity on 17β-HSD3 using microsomal fraction of rat testes as well as transfected LNCaP[17β-HSD3] cells. We then assessed its metabolic stability as well as its cytotoxic effect against a panel of cancer cell lines. The addition of a picolyl moiety at C-16 of RM-532-105 steroid core improves the 17β-HSD3 inhibitory activity in the microsomal fraction of rat testes, but not in whole LNCaP[17β-HSD3] cells. Interestingly, this structural modification enhances 3-fold the metabolic stability in conjunction with a significant cytotoxic effect against pancreatic, ovarian, breast, lung, and prostate cancer cells. Because the inhibitory activity data against 17β-HSD3 suggested that both steroid derivatives are non-competitive inhibitors, we performed docking and molecular dynamics simulations using a homology model of this membrane-associated enzyme. The results of these simulations revealed that both RM-532-105 (1) and FCO-586-119 (5) can compete for the cofactor-binding site displaying better binding energy than NADP+.
Collapse
Affiliation(s)
- Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada; Laboratory of Synthesis and Isolation of Bioactive Substances, Department of Biological Systems, Biological and Health Sciences Division, Metropolitan Autonomous University- Xochimilco (UAM-X), Mexico City 04960, Mexico
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - Martin Perreault
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU De Québec - Research Center, Québec City, Québec, G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, G1V 0A6, Canada.
| |
Collapse
|
7
|
Steroidogenesis in Peripheral and Transition Zones of Human Prostate Cancer Tissue. Int J Mol Sci 2021; 22:ijms22020487. [PMID: 33418978 PMCID: PMC7825320 DOI: 10.3390/ijms22020487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
The peripheral zone (PZ) and transition zone (TZ) represent about 70% of the human prostate gland with each zone having differential ability to develop prostate cancer. Androgens and their receptor are the primary driving cause of prostate cancer growth and eventually castration-resistant prostate cancer (CRPC). De novo steroidogenesis has been identified as a key mechanism that develops during CRPC. Currently, there is very limited information available on human prostate tissue steroidogenesis. The purpose of the present study was to investigate steroid metabolism in human prostate cancer tissues with comparison between PZ and TZ. Human prostate cancer tumors were procured from the patients who underwent radical prostatectomy without any neoadjuvant therapy. Human prostate homogenates were used to quantify steroid levels intrinsically present in the tissues as well as formed after incubation with 2 µg/mL of 17-hydroxypregnenolone (17-OH-pregnenolone) or progesterone. A Waters Acquity ultraperformance liquid chromatography coupled to a Quattro Premier XE tandem quadrupole mass spectrometer using a C18 column was used to measure thirteen steroids from the classical and backdoor steroidogenesis pathways. The intrinsic prostate tissue steroid levels were similar between PZ and TZ with dehydroepiandrosterone (DHEA), dihydrotestosterone (DHT), pregnenolone and 17-OH-pregnenolone levels higher than the other steroids measured. Interestingly, 5-pregnan-3,20-dione, 5-pregnan-3-ol-20-one, and 5-pregnan-17-ol-3,20-dione formation was significantly higher in both the zones of prostate tissues, whereas, androstenedione, testosterone, DHT, and progesterone levels were significantly lower after 60 min incubation compared to the 0 min control incubations. The incubations with progesterone had a similar outcome with 5-pregnan-3,20-dione and 5-pregnan-3-ol-20-one levels were elevated and the levels of DHT were lower in both PZ and TZ tissues. The net changes in steroid formation after the incubation were more observable with 17-OH-pregnenolone than with progesterone. In our knowledge, this is the first report of comprehensive analyses of intrinsic prostate tissue steroids and precursor-driven steroid metabolism using a sensitive liquid chromatography-mass spectrometry assay. In summary, the PZ and TZ of human prostate exhibited similar steroidogenic ability with distinction in the manner each zone utilizes the steroid precursors to divert the activity towards backdoor pathway through a complex matrix of steroidogenic mechanisms.
Collapse
|
8
|
Sviripa VM, Fiandalo MV, Begley KL, Wyrebek P, Kril LM, Balia AG, Parkin SR, Subramanian V, Chen X, Williams AH, Zhan CG, Liu C, Mohler JL, Watt DS. Pictet-Spengler condensations using 4-(2-aminoethyl)coumarins. NEW J CHEM 2020; 44:13415-13429. [PMID: 33795928 DOI: 10.1039/d0nj02664f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Androgen-deprivation therapy (ADT) is only a palliative measure, and prostate cancer invariably recurs in a lethal, castration-resistant form (CRPC). Prostate cancer resists ADT by metabolizing weak, adrenal androgens to growth-promoting 5α-dihydrotestosterone (DHT), the preferred ligand for the androgen receptor (AR). Developing small-molecule inhibitors for the final steps in androgen metabolic pathways that utilize 17-oxidoreductases required probes that possess fluorescent groups at C-3 and intact, naturally occurring functionality at C-17. Application of the Pictet-Spengler condensation to substituted 4-(2-aminoethyl)coumarins and 5α-androstane-3-ones furnished spirocyclic, fluorescent androgens at the desired C-3 position. Condensations required the presence of activating C-7 amino or N,N-dialkylamino groups in the 4-(2-aminoethyl)coumarin component of these condensation reactions. Successful Pictet-Spengler condensation, for example, of DHT with 9-(2-aminoethyl)-2,3,6,7-tetrahydro-1H,5H,11H-pyrano[2,3-f]pyrido[3,2,1-ij]quinolin-11-one led to a spirocyclic androgen, (3R,5S,10S,13S,17S)-17-hydroxy-10,13-dimethyl-1,2,2',3',4,5,6,7,8,8',9,9',10,11,12,12',13,13',14,15,16,17-docosahydro-7'H,11'H-spiro-[cyclopenta[a]phenanthrene-3,4'-pyrido[3,2,1-ij]pyrido[4',3':4,5]pyrano[2,3-f]quinolin]-5'(1'H)-one. Computational modeling supported the surrogacy of the C-3 fluorescent DHT analog as a tool to study 17-oxidoreductases for intracrine, androgen metabolism.
Collapse
Affiliation(s)
- Vitaliy M Sviripa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0093 USA
| | - Michael V Fiandalo
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
| | - Kristin L Begley
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Przemyslaw Wyrebek
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Liliia M Kril
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Andrii G Balia
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Sean R Parkin
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506 USA
| | | | - Xi Chen
- College of Chemistry and Material Science, South Central University for Nationalities, Wuhan 430074, People's Republic of China
| | - Alexander H Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA
| | - Chunming Liu
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0093 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| | - James L Mohler
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA.,Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
| | - David S Watt
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596 USA.,Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0093 USA.,Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY 40536-0509 USA
| |
Collapse
|
9
|
Wróbel TM, Rogova O, Andersen KL, Yadav R, Brixius-Anderko S, Scott EE, Olsen L, Jørgensen FS, Björkling F. Discovery of Novel Non-Steroidal Cytochrome P450 17A1 Inhibitors as Potential Prostate Cancer Agents. Int J Mol Sci 2020; 21:ijms21144868. [PMID: 32660148 PMCID: PMC7402352 DOI: 10.3390/ijms21144868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The current study presents the design, synthesis, and evaluation of novel cytochrome P450 17A1 (CYP17A1) ligands. CYP17A1 is a key enzyme in the steroidogenic pathway that produces androgens among other steroids, and it is implicated in prostate cancer. The obtained compounds are potent enzyme inhibitors (sub µM) with antiproliferative activity in prostate cancer cell lines. The binding mode of these compounds is also discussed.
Collapse
Affiliation(s)
- Tomasz M. Wróbel
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
- Correspondence: ; Tel.: +48-814-487-273
| | - Oksana Rogova
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| | - Kasper L. Andersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen, Denmark;
| | - Rahul Yadav
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
| | - Simone Brixius-Anderko
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
| | - Emily E. Scott
- Department of Medicinal Chemistry, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA; (R.Y.); (S.B.-A.); (E.E.S.)
- Department of Pharmacology, University of Michigan, 428 Church Street, Ann Arbor, MI 48109-1065, USA
| | - Lars Olsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
- Protein Engineering, Novozymes A/S, Krogshøjvej 36, DK-2880 Bagsvaerd, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (O.R.); (L.O.); (F.S.J.); (F.B.)
| |
Collapse
|
10
|
Penning TM, Detlefsen AJ. Intracrinology-revisited and prostate cancer. J Steroid Biochem Mol Biol 2020; 196:105499. [PMID: 31614208 PMCID: PMC6954292 DOI: 10.1016/j.jsbmb.2019.105499] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 01/22/2023]
Abstract
The formation of steroid hormones in peripheral target tissues is referred to as their intracrine formation. This process occurs in hormone dependent malignancies such as prostate and breast cancer in which the disease can be either castrate resistant or occur post-menopausally, respectively. In these instances, the major precursor steroid of androgens and estrogens is dehydroepiandrosterone (DHEA) and DHEA-SO4. This article reviews the major pathways by which adrenal steroids are converted to the potent male sex hormones, testosterone (T) and 5α-dihydrotestosterone (5α-DHT) and the discrete enzyme isoforms involved in castration resistant prostate cancer. Previous studies have mainly utilized radiotracers to investigate these pathways but have not used prevailing concentrations of precursors found in castrate male human serum. In addition, the full power of stable-isotope dilution liquid chromatography tandem mass spectrometry has not been applied routinely. Furthermore, it is clear that adaptive responses occur in the transporters and enzyme isoforms involved in response to androgen deprivation therapy that need to be considered.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, 421 Curie Blvd, 1350 BRBII/IIII, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-6084, United States.
| | - Andrea J Detlefsen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania School Philadelphia, PA, United States
| |
Collapse
|
11
|
Boutin S, Roy J, Maltais R, Poirier D. Formation of 5α-dihydrotestosterone from 5α-androstane-3α,17β-diol in prostate cancer LAPC-4 cells - Identifying inhibitors of non-classical pathways producing the most potent androgen. Bioorg Med Chem Lett 2020; 30:126783. [PMID: 31753699 DOI: 10.1016/j.bmcl.2019.126783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022]
Abstract
5α-Dihydrotestosterone (5α-DHT) possesses a great affinity for the androgen receptor (AR), and its binding to AR promotes the proliferation of prostate cancer (PC) cells in androgen-dependent PC. Primarily synthesized from testosterone (T) in testis, 5α-DHT could also be produced from 5α-androstane-3α,17β-diol (3α-diol), an almost inactive androgen, following non-classical pathways. We reported the chemical synthesis of non-commercially available [4-14C]-3α-diol from [4-14C]-T, and the development of a biological assay to identify inhibitors of the 5α-DHT formation from radiolabeled 3α-diol in LAPC-4 cell PC model. We measured the inhibitory potency of 5α-androstane derivatives against the formation of 5α-DHT, and inhibition curves were obtained for the most potent compounds (IC50 = 1.2-14.1 μM). The most potent inhibitor 25 (IC50 = 1.2 μM) possesses a 4-(4-CF3-3-CH3O-benzyl)piperazinyl methyl side chain at C3β and 17β-OH/17α-CCH functionalities at C17 of a 5α-androstane core.
Collapse
Affiliation(s)
- Sophie Boutin
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
12
|
Gonthier K, Poluri RTK, Audet-Walsh É. Functional genomic studies reveal the androgen receptor as a master regulator of cellular energy metabolism in prostate cancer. J Steroid Biochem Mol Biol 2019; 191:105367. [PMID: 31051242 DOI: 10.1016/j.jsbmb.2019.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022]
Abstract
Sex-steroid hormones have been investigated for decades for their oncogenic properties in hormone-dependent cancers. The increasing body of knowledge on the biological actions of androgens in prostate cancer has led to the development of several targeted therapies that still represent the standard of care for cancer patients to this day. In the prostate, androgens promote cellular differentiation and proper tissue development. These hormones also promote the aberrant proliferation and survival of prostate cancer cells. Over the past few years, sequencing technologies for functional genomic analyses have rapidly expanded, revealing novel functions of sex-steroid hormone receptors other than their classic roles. In this article, we will focus on transcriptomic- and genomic-based evidence that demonstrates the importance of the androgen receptor signaling in the regulation of prostate cancer cell metabolism. This is significant because the reprogramming of cell metabolism is a hallmark of cancer. In fact, it is clear now that the androgen receptor contributes to the reprogramming of specific cellular metabolic pathways that promote tumor growth and disease progression, including aerobic glycolysis, mitochondrial respiration, fatty acid ß-oxidation, and de novo lipid synthesis. Overall, beyond regulating development, differentiation, and proliferation, the androgen receptor is also a master regulator of cellular energy metabolism.
Collapse
Affiliation(s)
- Kevin Gonthier
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada
| | - Raghavendra Tejo Karthik Poluri
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada
| | - Étienne Audet-Walsh
- Department of Molecular Medicine, Axe Endocrinologie - Néphrologie du Centre de recherche du CHU de Québec, Canada; Centre de recherche sur le cancer - Université Laval, Canada.
| |
Collapse
|
13
|
Knuuttila M, Hämäläinen E, Poutanen M. Applying mass spectrometric methods to study androgen biosynthesis and metabolism in prostate cancer. J Mol Endocrinol 2019; 62:R255-R267. [PMID: 30917337 DOI: 10.1530/jme-18-0150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022]
Abstract
Recent development of gas chromatography and liquid chromatography-tandem mass spectrometry (GC-MS/MS, LC-MS/MS) has provided novel tools to define sex steroid concentrations. These new methods overcome several of the problems associated with immunoassays for sex steroids. With the novel MS-based applications we are now able to measure small concentrations of the steroid hormones reliably and with high accuracy in both body fluids and tissue homogenates. The sensitivity of the tandem mass spectrometry assays allows us also for the first time to reliably measure picomolar or even femtomolar concentrations of estrogens and androgens. Furthermore, due to a high sensitivity and specificity of MS technology, we are also able to measure low concentrations of steroid hormones of interest in the presence of pharmacological concentration of other steroids and structurally closely related compounds. Both of these features are essential for multiple preclinical models for prostate cancer. The MS assays are also valuable for the simultaneous measurement of multiple steroids and their metabolites in small sample volumes in serum and tissue biopsies of prostate cancer patients before and after drug interventions. As a result, novel information about steroid hormone synthesis and metabolic pathways in prostate cancer has been obtained. In our recent studies, we have extensively applied a GC-MS/MS method to study androgen biosynthesis and metabolism in VCaP prostate cancer xenografts in mice. In the present review, we shortly summarize some of the benefits of the GC-MS/MS and novel LC-MS/MS assays, and provide examples of their use in defining novel mechanisms of androgen action in prostate cancer.
Collapse
Affiliation(s)
- Matias Knuuttila
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry and HUSLAB, Helsinki University and Helsinki University Hospital, HUSLAB, Helsinki, Finland
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Fiandalo MV, Gewirth DT, Mohler JL. Potential impact of combined inhibition of 3α-oxidoreductases and 5α-reductases on prostate cancer. Asian J Urol 2018; 6:50-56. [PMID: 30775248 PMCID: PMC6363635 DOI: 10.1016/j.ajur.2018.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) growth and progression rely on the interaction between the androgen receptor (AR) and the testicular ligands, testosterone and dihydrotestosterone (DHT). Almost all men with advanced PCa receive androgen deprivation therapy (ADT). ADT lowers circulating testosterone levels, which impairs AR activation and leads to PCa regression. However, ADT is palliative and PCa recurs as castration-recurrent/resistant PCa (CRPC). One mechanism for PCa recurrence relies on intratumoral synthesis of DHT, which can be synthesized using the frontdoor or primary or secondary backdoor pathway. Androgen metabolism inhibitors, such as those targeting 5α-reductase, aldo-keto-reductase family member 3 (AKR1C3), or cytochrome P450 17A1 (CYP17A1) have either failed or produced only modest clinical outcomes. The goal of this review is to describe the therapeutic potential of combined inhibition of 5α-reductase and 3α-oxidoreductase enzymes that facilitate the terminal steps of the frontdoor and primary and secondary backdoor pathways for DHT synthesis. Inhibition of the terminal steps of the androgen metabolism pathways may be a way to overcome the shortcomings of existing androgen metabolism inhibitors and thereby delay PCa recurrence during ADT or enhance the response of CRPC to androgen axis manipulation.
Collapse
Affiliation(s)
- Michael V Fiandalo
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
15
|
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
|
16
|
Cook SF, Fiandalo MV, Watt DS, Wu Y, Mohler JL, Bies RR. Mathematical modeling of intracrine androgen metabolism in prostate cancer: Methodological aspects. Prostate 2018; 78:1069-1076. [PMID: 29938815 DOI: 10.1002/pros.23665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 06/07/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Progression of castration-recurrent/resistant prostate cancer (CRPC) relies in part on dihydrotestosterone derived from intratumoral androgen metabolism. Mathematical modeling provides a valuable tool for studies of androgen metabolism in CRPC. This modeling approach integrates existing knowledge about complex biologic systems and provides a means of interrogating the effects of various interventions. We sought to model a single reaction in the androgen biosynthesis network, namely the oxidation of androsterone (AND) to androstanedione (5α-dione) by four 3α-oxidoreductase enzymes, as an initial effort to establish the feasibility of our modeling approach. METHODS Models were constructed for two cell culture systems, a non-prostate cancer cell line (CV-1) and a prostate cancer cell line (LAPC-4), using the SimBiology app (version 5.3) in MATLAB (version 8.6). The models included components for substrate (AND), product (5α-dione), each of the four enzymes, and each of the four enzyme-substrate complexes. Each enzymatic reaction consisted of a reversible enzyme-substrate binding step and an irreversible catalysis step. Rates of change for each component were described using ordinary differential equations. RESULTS Mathematical models were developed with model parameter values derived from literature sources or from existing experimental data, which included gene expression measurements and substrate and product concentrations determined using liquid chromatography-tandem mass spectrometry. The models for both cell lines adequately described substrate and product concentrations observed after 12 h treatment with AND. CONCLUSIONS This modeling approach represents an adaptable, extensible and mechanistic framework that reflects androgen metabolism. The models can be expanded systematically to describe the complex androgen metabolic pathways important for study of novel therapies for CRPC.
Collapse
Affiliation(s)
- Sarah F Cook
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York
| | | | - David S Watt
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky
- Lucille Parker Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Yue Wu
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - James L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Robert R Bies
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York
- Computational and Data-Enabled Science and Engineering Program, University at Buffalo, State University of New York, Buffalo, New York
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
17
|
Penning TM. Dehydroepiandrosterone (DHEA)-SO 4 Depot and Castration-Resistant Prostate Cancer. VITAMINS AND HORMONES 2018; 108:309-331. [PMID: 30029732 DOI: 10.1016/bs.vh.2018.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dehydroepiandrosterone (DHEA)-SO4 of adrenal origin is the major C19 steroid in the serum. It is a precursor of intratumoral androgen biosynthesis in patients with advanced prostate cancer following chemical or surgical castration. DHEA is a product of the P450c17 (17α-hydroxylase-17,20-lyase) enzyme. Despite inhibition of P450c17 with new agents, e.g., Abiraterone acetate, Orterenol, and Galeterone, the level of enzyme inhibition rarely exceeds 90% leaving behind a significant depot for androgen biosynthesis within the tumor. For DHEA-SO4 to be utilized there is uptake by organic anion transporter polypeptides, deconjugation catalyzed by steroid sulfatase, and adaptive upregulation of prostate steroidogenic enzymes that will convert DHEA into either testosterone or dihydrotestosterone. The depot of DHEA-SO4 that remains after P450c17 inhibition and the adaptive responses that occur within the tumor to promote DHEA utilization contribute to mechanisms of drug resistance observed with P450c17 inhibitors. Knowledge of these mechanisms identify new targets for therapeutics that could be used to surmount drug resistance in prostate cancer.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Ohlmann CH, Jäschke M, Jaehnig P, Krege S, Gschwend J, Rexer H, Stöckle M. Abiraterone acetate plus LHRH therapy versus abiraterone acetate while sparing LHRH therapy in patients with progressive, metastatic and chemotherapy-naïve, castration-resistant prostate cancer (SPARE): study protocol for a randomized controlled trial. Trials 2017; 18:457. [PMID: 28978327 PMCID: PMC5628432 DOI: 10.1186/s13063-017-2195-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/14/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The value of continuation of luteinizing hormone-releasing hormone (LHRH) therapy in castration-resistant prostate cancer (CRPC) remains controversial and clear evidence is lacking. Argumentation for cessation of LHRH therapy is the prolonged suppression of testosterone levels after the withdrawal of LHRH analogues and the fact that disease progression occurs despite castration levels of testosterone. Especially upon treatment with the life-prolonging cytochrome P450 17-alpha-hydroxylase (Cyp17)-inhibitor, abiraterone, which has the ability to further suppress testosterone serum levels over LHRH therapy alone, continuation of LHRH therapy seems to be negligible. However, the proven increase of luteinizing hormone levels after LHRH withdrawal, which is even further increased by abiraterone, may counteract the effects of abiraterone by the induction of enzymes of steroidogenesis. Therefore, cessation of LHRH therapy when starting treatment with abiraterone in CRPC may display an unpredictable hazard to the patients. This study will explore the role of continuation of LHRH therapy when starting treatment with abiraterone in patients with asymptomatic or mildly symptomatic, chemotherapy-naïve CPRC. METHODS/DESIGN The trial will assess radiographic progression-free survival after 12 months of treatment with abiraterone/prednisone in patients who will be randomized to receive continuing LHRH therapy versus LHRH withdrawal at the time of starting abiraterone therapy. DISCUSSION This multicenter, prospective, randomized, exploratory phase-II trial will bring about new data regarding the efficacy and safety of abiraterone/prednisone treatment with or without continuation of LHRH therapy. In addition, further insight into the complex hormonal changes under treatment will be gained and the results of this trial may give rise to a larger phase-III trial to examine the possibility of withdrawing LHRH therapy in patients with CRPC. TRIAL REGISTRATION ClinicalTrials.gov, ID: NCT02077634 . Registered on 9 December 2013.
Collapse
Affiliation(s)
- Carsten-Henning Ohlmann
- Department of Urology, Saarland University, Kirrbergerstrasse, 66421 Homburg/Saar, Germany
- Working Group on Urological Oncology (AUO), Germany Cancer Society, Berlin, Germany
| | - Michelle Jäschke
- Department of Urology, Saarland University, Kirrbergerstrasse, 66421 Homburg/Saar, Germany
| | | | - Susane Krege
- Department of Urology, Klinikum Essen Mitte, Essen, Germany
- Working Group on Urological Oncology (AUO), Germany Cancer Society, Berlin, Germany
| | - Jürgen Gschwend
- Department of Urology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
- Working Group on Urological Oncology (AUO), Germany Cancer Society, Berlin, Germany
| | - Heidrun Rexer
- Working Group on Urological Oncology (AUO), Germany Cancer Society, Berlin, Germany
| | - Michael Stöckle
- Department of Urology, Saarland University, Kirrbergerstrasse, 66421 Homburg/Saar, Germany
| |
Collapse
|
19
|
Abstract
INTRODUCTION AKR1C3 is a drug target in hormonal and hormonal independent malignancies and acts as a major peripheral 17β-hydroxysteroid dehydrogenase to yield the potent androgens testosterone and dihydrotestosterone, and as a prostaglandin (PG) F synthase to produce proliferative ligands for the PG FP receptor. AKR1C3 inhibitors may have distinct advantages over existing therapeutics for the treatment of castration resistant prostate cancer, breast cancer and acute myeloid leukemia. Area covered: This article reviews the patent literature on AKR1C3 inhibitors using SciFinder which identified inhibitors in the following chemical classes: N-phenylsulfonyl-indoles, N-(benzimidazoylylcarbonyl)- N-(indoylylcarbonyl)- and N-(pyridinepyrrolyl)- piperidines, N-benzimidazoles and N-benzindoles, repurposed nonsteroidal antiinflammatory drugs (indole acetic acids, N-phenylanthranilates and aryl propionic acids), isoquinolines, and nitrogen and sulfur substituted estrenes. The article evaluates inhibitor AKR potency, specificity, efficacy in cell-based and xenograft models and clinical utility. The advantage of bifunctional compounds that either competitively inhibit AKR1C3 and block its androgen receptor (AR) coactivator function or act as AKR1C3 inhibitors and direct acting AR antagonists are discussed. Expert opinion: A large number of potent and selective inhibitors of AKR1C3 have been described however, preclinical optimization, is required before their benefit in human disease can be assessed.
Collapse
Affiliation(s)
- Trevor M Penning
- a Center of Excellence in Environmental Toxicology & Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
20
|
Zeng CM, Chang LL, Ying MD, Cao J, He QJ, Zhu H, Yang B. Aldo-Keto Reductase AKR1C1-AKR1C4: Functions, Regulation, and Intervention for Anti-cancer Therapy. Front Pharmacol 2017; 8:119. [PMID: 28352233 PMCID: PMC5349110 DOI: 10.3389/fphar.2017.00119] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/27/2017] [Indexed: 12/31/2022] Open
Abstract
Aldo-keto reductases comprise of AKR1C1-AKR1C4, four enzymes that catalyze NADPH dependent reductions and have been implicated in biosynthesis, intermediary metabolism, and detoxification. Recent studies have provided evidences of strong correlation between the expression levels of these family members and the malignant transformation as well as the resistance to cancer therapy. Mechanistically, most studies focus on the catalytic-dependent function of AKR1C isoforms, like their impeccable roles in prostate cancer, breast cancer, and drug resistance due to the broad substrates specificity. However, accumulating clues showed that catalytic-independent functions also played critical roles in regulating biological events. This review summarizes the catalytic-dependent and -independent roles of AKR1Cs, as well as the small molecule inhibitors targeting these family members.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang UniversityHangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang UniversityHangzhou, China
| |
Collapse
|
21
|
Kenmogne LC, Roy J, Maltais R, Rouleau M, Neveu B, Pouliot F, Poirier D. Investigation of the In Vitro and In Vivo efficiency of RM-532-105, a 17β-hydroxysteroid dehydrogenase type 3 inhibitor, in LAPC-4 prostate cancer cell and tumor models. PLoS One 2017; 12:e0171871. [PMID: 28182747 PMCID: PMC5300232 DOI: 10.1371/journal.pone.0171871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/26/2017] [Indexed: 11/27/2022] Open
Abstract
In the fight against androgen-sensitive prostate cancer, the enzyme 17β-hydroxysteroid dehydrogenase type 3 (17β-HSD3) is an attractive therapeutic target considering its key role in the formation of androgenic steroids. In this study, we attempted to assess the in vivo efficacy of the compound RM-532-105, an androsterone derivative developed as an inhibitor of 17β-HSD3, in the prostate cancer model of androgen-sensitive LAPC-4 cells xenografted in nude mice. RM-532-105 did not inhibit the tumor growth induced by 4-androstene-3,17-dione (4-dione); rather, the levels of the androgens testosterone (T) and dihydrotestosterone (DHT) increased within the tumors. In plasma, however, DHT levels increased but T levels did not. In troubleshooting experiments, the non-androgenic potential of RM-532-105 was confirmed by two different assays (LAPC-4 proliferation and androgen receptor transcriptional activity assays). The enzyme 5α-reductase was also revealed to be the predominant enzyme metabolizing 4-dione in LAPC-4 cells, yielding 5α-androstane-3,17-dione and not T. Other 17β-HSDs than 17β-HSD3 seem responsible in the androgen synthesis. From experiments with LAPC-4 cells, we fortuitously came across the interesting finding that 17β-HSD3 inhibitor RM-532-105 is concentrated inside tumors.
Collapse
Affiliation(s)
- Lucie Carolle Kenmogne
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), Québec, Québec, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), Québec, Québec, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), Québec, Québec, Canada
| | - Mélanie Rouleau
- CHU de Québec - Research Center, Axe Cancer, Québec, Québec, Canada
| | - Bertrand Neveu
- CHU de Québec - Research Center, Axe Cancer, Québec, Québec, Canada
| | - Frédéric Pouliot
- CHU de Québec - Research Center, Axe Cancer, Québec, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Center (CHUL, T4), Québec, Québec, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
- * E-mail:
| |
Collapse
|
22
|
Cortés-Benítez F, Roy J, Maltais R, Poirier D. Impact of androstane A- and D-ring inversion on 17β-hydroxysteroid dehydrogenase type 3 inhibitory activity, androgenic effect and metabolic stability. Bioorg Med Chem 2017; 25:2065-2073. [PMID: 28254377 DOI: 10.1016/j.bmc.2017.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/31/2017] [Accepted: 02/06/2017] [Indexed: 10/20/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 3 (17β-HSD3) is a major player in human endocrinology, being one of the most important enzymes involved in testosterone production. To capitalize on the discovery of RM-532-105, a steroidal 17β-HSD3 inhibitor, we explored the effect of its backbone configuration on inhibitory activity, androgenic profile, and metabolic stability. Two modifications that greatly alter the natural shape of steroids, i.e. inversion of the methyl on carbon 13 (13α-CH3 instead of 13β-CH3) and inversion of the hydrogen on carbon 5 (5β-H instead of 5α-H), were tested after the syntheses in 6 steps of 2 isomeric forms (5α/13α-RM-532-105 (6a) and 5β/13β-RM-532-105 (6b), respectively) of the 17β-HSD3 inhibitor RM-532-105 (5α/13β-configurations). For compound 6b, a cis/trans junction of the A/B rings did not significantly alter the inhibitory activity on 17β-HSD3 (IC50=0.15μM) as well as the liver microsomal stability (16.6% of 6b remaining after 1h incubation) compared to RM-532-105 (IC50=0.11μM and 14.1% remaining). In contrast, a trans/cis junction of C/D rings reduced the inhibitory activity on 17β-HSD3 (IC50=1.09μM) but increased the metabolic stability with 29.4% of compound 6a remaining after incubation. The structural modifications represented by compounds 6a and 6b did not change the non-androgenicity profile of an androsterone derivative such as RM-532-105, but slightly increased its cytotoxic activity.
Collapse
Affiliation(s)
- Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec - Research Center, Québec, Québec, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
23
|
Capogrosso P, Ventimiglia E, Moschini M, Boeri L, Farina E, Finocchio N, Gandaglia G, Fossati N, Briganti A, Montorsi F, Salonia A. Testosterone Levels Correlate With Grade Group 5 Prostate Cancer: Another Step Toward Personalized Medicine. Prostate 2017; 77:234-241. [PMID: 27775173 DOI: 10.1002/pros.23266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/20/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Controversial results have shown a significant association with either low or high total testosterone (tT) levels and high risk prostate cancer (PCa). We tested the relationship between circulating tT and grade group 5 (G5) PCa at radical prostatectomy (RP) in patients with preoperative low- to intermediate-risk disease. METHODS Serum sex hormones were assessed the day before RP in a cohort of 846 patients with low- to intermediate-risk PCa. Patients were segregated using the new 5-tiered Gleason grade groups. Restricted cubic spline functions and logistic regression analyses tested the association between sex hormones and G5 PCa. Differences in potential predictive accuracy (PA) were assessed for tT and prostate-specific antigen (PSA) levels. RESULTS Overall, 27 men (3.2%) had G5 PCa at RP, and this group had higher PSA values than patients with G1-G4 PCa (P = 0.02). The groups did not differ in terms of preoperative mean hormonal values. Both low and high circulating tT values depicted a nonlinear U-shaped correlation with G5 PCa at RP. The lowest and highest (10th and 90th percentiles) tT values and biopsy PCa grade emerged as multivariable independent predictors of G5 PCa at RP (all P < 0.05). PA for G5 PCa did not differ between tT (area under the curve [AUC] 0.631) and PSA (AUC 0.636). CONCLUSIONS Circulating tT was a significant predictor of G5 PCa at RP in patients with preoperative low- to intermediate-risk disease. Preoperative tT and PSA values showed similar PA for the most aggressive disease, confirming a potential role for circulating androgens in preoperative risk assessment of PCa patients. Prostate 77:234-241, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paolo Capogrosso
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Eugenio Ventimiglia
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Marco Moschini
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Luca Boeri
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Elena Farina
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Nadia Finocchio
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Giorgio Gandaglia
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Nicola Fossati
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Alberto Briganti
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Franscesco Montorsi
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI-Urological Research Institute, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
24
|
Narayanan R, Dalton JT. Androgen Receptor: A Complex Therapeutic Target for Breast Cancer. Cancers (Basel) 2016; 8:cancers8120108. [PMID: 27918430 PMCID: PMC5187506 DOI: 10.3390/cancers8120108] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 12/29/2022] Open
Abstract
Molecular and histopathological profiling have classified breast cancer into multiple sub-types empowering precision treatment. Although estrogen receptor (ER) and human epidermal growth factor receptor (HER2) are the mainstay therapeutic targets in breast cancer, the androgen receptor (AR) is evolving as a molecular target for cancers that have developed resistance to conventional treatments. The high expression of AR in breast cancer and recent discovery and development of new nonsteroidal drugs targeting the AR provide a strong rationale for exploring it again as a therapeutic target in this disease. Ironically, both nonsteroidal agonists and antagonists for the AR are undergoing clinical trials, making AR a complicated target to understand in breast cancer. This review provides a detailed account of AR’s therapeutic role in breast cancer.
Collapse
Affiliation(s)
- Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - James T Dalton
- College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
25
|
Penning TM. Mechanisms of drug resistance that target the androgen axis in castration resistant prostate cancer (CRPC). J Steroid Biochem Mol Biol 2015; 153:105-13. [PMID: 26032458 PMCID: PMC4568163 DOI: 10.1016/j.jsbmb.2015.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/23/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
Castrate resistant prostate cancer (CRPC) is the fatal-form of prostate cancer and remains androgen dependent. The reactivation of the androgen axis occurs due to adaptive intratumoral androgen biosynthesis which can be driven by adrenal androgens and/or by changes in the androgen receptor (AR) including AR gene amplification. These mechanisms are targeted with P450c17 inhibitors e.g., abiraterone acetate and AR super-antagonists e.g., enzalutamide, respectively. Clinical experience indicates that with either agent an initial response is followed by drug resistance and the patient clinically progresses on these agents. This article reviews the mechanisms of intrinsic and acquired drug resistance that target the androgen axis and how this might be surmounted.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA.
| |
Collapse
|
26
|
Roy J, Fournier MA, Maltais R, Kenmogne LC, Poirier D. Reprint of "In vitro and in vivo evaluation of a 3β-androsterone derivative as inhibitor of 17β-hydroxysteroid dehydrogenase type 3". J Steroid Biochem Mol Biol 2015; 153:170-8. [PMID: 26291835 DOI: 10.1016/j.jsbmb.2015.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/29/2013] [Accepted: 12/31/2013] [Indexed: 10/23/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 3 (17β-HSD3 or HSD17B3) catalyzes the last step in the biosynthesis of the potent androgen testosterone (T), by stereoselectively reducing the C17 ketone of 4-androstene-3,17-dione (4-dione), with NADPH as cofactor. Since T plays an important role in androgen-sensitive diseases, this enzyme is thus an interesting therapeutic target. In an attempt to design compounds to lower the level of T, we synthesized androsterone derivatives substituted at position 3 as inhibitors of 17β-HSD3, and selected one of the most potent compounds for additional studies. In an enzymatic assay in homogenized and whole HEK-293 cells overexpressing 17β-HSD3, the inhibitor RM-532-105 efficiently inhibited the conversion of natural substrate 4-dione (50nM) into T with an IC50 of 26nM and 5nM, respectively. Moreover, the inhibitor RM-532-105 (10mg/kg) reached a plasma concentration of 250ng/mL at 7h (AUC 24h: 3485ngh/mL) after subcutaneous (s.c.) injection in the rat. In order to mimic the human situation in which 4-dione is converted to T in the testis, we used intact rats. Treatment for 7 days with 17β-HSD3 inhibitor RM-532-105 by s.c. injection or oral gavage exerted no effect on the testis, prostate and seminal vesicle weight and no modification in the levels of plasma steroids. However, after this treatment, the concentration of inhibitor in plasma increased depending on the dose. We thereafter determined the concentration of inhibitor in the testis and we discovered that the compound was slightly present. In fact, at 10mg/kg, the inhibitor RM-532-105 seems to have difficulty penetrating inside the testis and was found to be concentrated in the testicular capsule, and therefore unable to inhibit the 17β-HSD3 located inside the testis. However, with a higher dose of 50mg/kg injected s.c. in rats, RM-532-105 significantly decreased the level of T and dihydrotestosterone measured in plasma at 2h.
Collapse
Affiliation(s)
- Jenny Roy
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Michelle-Audrey Fournier
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Lucie Carolle Kenmogne
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada.
| |
Collapse
|
27
|
Stojkovic R, Ivankovic S, Ivankovic D, Attias L, Mantovani A, Fucic A. Testosterone-induced micronuclei and increased nuclear division rate in L929 cell line expressing the androgen receptor. Toxicol In Vitro 2015; 29:1021-5. [DOI: 10.1016/j.tiv.2015.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/24/2015] [Accepted: 04/13/2015] [Indexed: 02/01/2023]
|
28
|
Powell K, Semaan L, Conley-LaComb MK, Asangani I, Wu YM, Ginsburg KB, Williams J, Squire JA, Maddipati KR, Cher ML, Chinni SR. ERG/AKR1C3/AR Constitutes a Feed-Forward Loop for AR Signaling in Prostate Cancer Cells. Clin Cancer Res 2015; 21:2569-79. [PMID: 25754347 PMCID: PMC4976600 DOI: 10.1158/1078-0432.ccr-14-2352] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/19/2015] [Indexed: 01/26/2023]
Abstract
PURPOSE Intratumoral androgen synthesis in prostate cancer contributes to the development of castration-resistant prostate cancer (CRPC). Several enzymes responsible for androgen biosynthesis have been shown to be overexpressed in CRPC, thus contributing to CRPC in a castrated environment. The TMPRSS2-ERG transcription factor has been shown to be present in primary prostate cancer tumors as well as CRPC tumors. We hypothesize that TMPRSS2-ERG fusions regulate androgen biosynthetic enzyme (ABE) gene expression and the production of androgens, which contributes to the development of CRPC. EXPERIMENTAL DESIGN We used a panel of assays, including lentivirus transduction, gene expression, chromatin immunoprecipitation and sequencing, liquid chromatography-mass spectrometric quantitation, immunocytochemistry, immunohistochemistry, and bioinformatics analysis of gene microarray databases, to determine ERG regulation of androgen synthesis. RESULTS We found that ERG regulated the expression of the ABE AKR1C3 in prostate cancer cells via direct binding to the AKR1C3 gene. Knockdown of ERG resulted in reduced AKR1C3 expression, which caused a reduction in both DHT synthesis and PSA expression in VCaP prostate cancer cells treated with 5α-androstanedione (5α-Adione), a DHT precursor metabolite. Immunohistochemical staining revealed that ERG was coexpressed with AKR1C3 in prostate cancer tissue samples. CONCLUSIONS These data suggest that AKR1C3 catalyzes the biochemical reduction of 5α-Adione to DHT in prostate cancer cells, and that ERG regulates this step through upregulation of AKR1C3 expression. Elucidation of ERG regulation of ABEs in CRPC may help to stratify TMPRSS2-ERG fusion-positive prostate cancer patients in the clinic for anti-androgen receptor-driven therapies; and AKR1C3 may serve as a valuable therapeutic target in the treatment of CRPC.
Collapse
Affiliation(s)
- Katelyn Powell
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan
| | - Louie Semaan
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan
| | | | - Irfan Asangani
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Kevin B Ginsburg
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan
| | - Julia Williams
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jeremy A Squire
- Department of Pathology and Forensic Medicine, University of Sao Paulo at Ribeirão Preto, Brazil
| | - Krishna R Maddipati
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Michael L Cher
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan. Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan. Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Sreenivasa R Chinni
- Department of Urology, Wayne State University School of Medicine, Detroit, Michigan. Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan. Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
29
|
Han CS, Patel R, Kim IY. Pharmacokinetics, pharmacodynamics and clinical efficacy of abiraterone acetate for treating metastatic castration-resistant prostate cancer. Expert Opin Drug Metab Toxicol 2015; 11:967-75. [DOI: 10.1517/17425255.2015.1041918] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Livingstone DEW, Barat P, Di Rollo EM, Rees GA, Weldin BA, Rog-Zielinska EA, MacFarlane DP, Walker BR, Andrew R. 5α-Reductase type 1 deficiency or inhibition predisposes to insulin resistance, hepatic steatosis, and liver fibrosis in rodents. Diabetes 2015; 64:447-58. [PMID: 25239636 DOI: 10.2337/db14-0249] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
5α-Reductase type 1 (5αR1) catalyses A-ring reduction of androgens and glucocorticoids in liver, potentially influencing hepatic manifestations of the metabolic syndrome. Male mice, homozygous for a disrupted 5αR1 allele (5αR1 knockout [KO] mice), were studied after metabolic (high-fat diet) and fibrotic (carbon tetrachloride [CCl4]) challenge. The effect of the 5α-reductase inhibitor finasteride on metabolism was investigated in male obese Zucker rats. While eating a high-fat diet, male 5αR1-KO mice demonstrated greater mean weight gain (21.6 ± 1.4 vs 16.2 ± 2.4 g), hyperinsulinemia (insulin area under the curve during glucose tolerance test 609 ± 103 vs. 313 ± 66 ng ⋅ mL(-1) ⋅ min), and hepatic steatosis (liver triglycerides 136.1 ± 17.0 vs. 89.3 ± 12.1 μmol ⋅ g(-1)). mRNA transcript profiles in liver were consistent with decreased fatty acid β-oxidation and increased triglyceride storage. 5αR1-KO male mice were more susceptible to fibrosis after CCl4 administration (37% increase in collagen staining). The nonselective 5α-reductase inhibitor finasteride induced hyperinsulinemia and hepatic steatosis (10.6 ± 1.2 vs. 7.0 ± 1.0 μmol ⋅ g(-1)) in obese male Zucker rats, both intact and castrated. 5αR1 deficiency induces insulin resistance and hepatic steatosis, consistent with the intrahepatic accumulation of glucocorticoids, and predisposes to hepatic fibrosis. Hepatic steatosis is independent of androgens in rats. Variations in 5αR1 activity in obesity and with nonselective 5α-reductase inhibition in men with prostate disease may have important consequences for the onset and progression of metabolic liver disease.
Collapse
Affiliation(s)
- Dawn E W Livingstone
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K.
| | - Pascal Barat
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Emma M Di Rollo
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Georgina A Rees
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Benjamin A Weldin
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Eva A Rog-Zielinska
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - David P MacFarlane
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Brian R Walker
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Ruth Andrew
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
31
|
Boberg J, Johansson HKL, Hadrup N, Dreisig K, Berthelsen L, Almstrup K, Vinggaard AM, Hass U. Perinatal exposure to mixtures of anti-androgenic chemicals causes proliferative lesions in rat prostate. Prostate 2015; 75:126-40. [PMID: 25327291 DOI: 10.1002/pros.22897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 08/21/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND Elevated levels of endogenous or exogenous estrogens during fetal life can induce permanent disturbances in prostate growth and predispose to precancerous lesions. Recent studies have indicated that also early anti-androgen exposure may affect prostate cancer risk. METHODS We examined the influence of perinatal exposure to mixtures of anti-androgenic and estrogenic chemicals on prostate development. Wistar rats were exposed from gestation day 7 to postnatal day 22 to a mixture of 8 anti-androgenic compounds (AAMix), a mixture of four estrogenic compounds (EMix), or paracetamol or a mixture of all 13 compounds (TotalMix) in mixture ratios reflecting human exposure levels. RESULTS Ventral prostate weights were reduced by the TotalMix and AAMix in pre-pubertal rats. Histological changes in prostate appeared with increasing age and indicated a shift from the normal age-dependent epithelial atrophy towards hyperplasia. These lesions showed similarities to pre-cancerous lesions in humans. Increased proliferation was observed already in pre-puberty and it was hypothesized that this could be associated with reduced ERβ signaling, but no clear conclusions could be made from gene expression studies on ERβ-related pathways. The influences of the estrogenic chemicals and paracetamol on prostate morphology were minor, but in young adulthood the estrogen mixture reduced ventral prostate mRNA levels of Igf1 and paracetamol reduced the mRNA level ofPbpc3. CONCLUSIONS Mixtures of endocrine disrupters relevant for human exposure was found to elicit persistent effects on the rat prostate following perinatal exposure, suggesting that human perinatal exposure to environmental chemicals may increase the risk of prostate cancer later in life.
Collapse
Affiliation(s)
- Julie Boberg
- Division of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Quintela ML, Mateos LL, Estévez SV, Calvo OF, Herranz UA, Afonso FJA, Santomé L, Aparicio LA. Enzalutamide: a new prostate cancer targeted therapy against the androgen receptor. Cancer Treat Rev 2015; 41:247-53. [PMID: 25638257 DOI: 10.1016/j.ctrv.2014.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 10/24/2022]
Abstract
Enzalutamide (MDV3100), an androgen receptor-signalling inhibitor, represents the most recent compound added to the therapeutic armamentarium for the treatment of metastatic castration-resistant prostate cancer (mCRPC) who progressed to docetaxel. The anti-tumour activity and safety of enzalutamide has been demonstrated in a phase III clinical trial, showing a benefit in overall survival, which was the primary endpoint. There are no head-to-head studies comparing the different treatment options in this subset of patients. In this article, most relevant data published in the literature have been reviewed, with special attention to the therapeutic alternatives currently available for postdocexatel mCRPC patients, emphasising the mechanisms of action of the different drugs, efficacy and quality of life-related aspects.
Collapse
Affiliation(s)
- Martín Lázaro Quintela
- Oncología Médica, Complexo Hospitalario Universitario de Vigo, C/Pizarro 22, 36204 Vigo, Spain.
| | - Luis León Mateos
- Servicio de Oncología Médica, Complexo Hospitalario Universitario de Pontevedra, Spain.
| | - Sergio Vázquez Estévez
- Servicio de Oncología Médica, Hospital Universitario Lucus Augusti, C/Doutor Ulises Romero, s/n, 27003 Lugo, Spain.
| | - Ovidio Fernández Calvo
- Servicio Oncologia Medica, Complejo Hospitalario Universitario Ourense, C/Ramón Puga 54.32005, Spain.
| | - Urbano Anido Herranz
- Oncología Médica, Complejo Hospitalario Universitario de Santiago de Compostela, c/Choupana s/n, 15706 Santiago de Compostela, A Coruña, Spain.
| | | | - Lucía Santomé
- Servicio de Oncología Médica, Hospital Povisa, C/Salamanca 5, 36211 Vigo, Spain.
| | | |
Collapse
|
33
|
Fokidis HB, Adomat HH, Kharmate G, Hosseini-Beheshti E, Guns ES, Soma KK. Regulation of local steroidogenesis in the brain and in prostate cancer: lessons learned from interdisciplinary collaboration. Front Neuroendocrinol 2015; 36:108-29. [PMID: 25223867 DOI: 10.1016/j.yfrne.2014.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/16/2022]
Abstract
Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Department of Biology, Rollins College, Winter Park, FL 37289, USA; Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | | | - Emma S Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; Department of Urological Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Brain Research Centre, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
34
|
Upreti R, Naredo G, Faqehi AMM, Hughes KA, Stewart LH, Walker BR, Homer NZM, Andrew R. Simultaneous pharmacokinetic and pharmacodynamic analysis of 5α-reductase inhibitors and androgens by liquid chromatography tandem mass spectrometry. Talanta 2015; 131:728-35. [PMID: 25281165 PMCID: PMC4196769 DOI: 10.1016/j.talanta.2014.07.087] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/25/2014] [Accepted: 07/30/2014] [Indexed: 11/29/2022]
Abstract
Benign prostatic hyperplasia and prostate cancer can be treated with the 5α-reductase inhibitors, finasteride and dutasteride, when pharmacodynamic biomarkers are useful in assessing response. A novel method was developed to measure the substrates and products of 5α-reductases (testosterone, 5α-dihydrotestosterone (DHT), androstenedione) and finasteride and dutasteride simultaneously by liquid chromatography tandem mass spectrometry, using an ABSciex QTRAP(®) 5500, with a Waters Acquity™ UPLC. Analytes were extracted from serum (500 µL) via solid-phase extraction (Oasis(®) HLB), with (13)C3-labelled androgens and d9-finasteride included as internal standards. Analytes were separated on a Kinetex C18 column (150 × 3 mm, 2.6 µm), using a gradient run of 19 min. Temporal resolution of analytes from naturally occurring isomers and mass +2 isotopomers was ensured. Protonated molecular ions were detected in atmospheric pressure chemical ionisation mode and source conditions optimised for DHT, the least abundant analyte. Multiple reaction monitoring was performed as follows: testosterone (m/z 289 → 97), DHT (m/z 291 → 255), androstenedione (m/z 287 → 97), dutasteride (m/z 529 → 461), finasteride (m/z 373 → 317). Validation parameters (intra- and inter-assay precision and accuracy, linearity, limits of quantitation) were within acceptable ranges and biological extracts were stable for 28 days. Finally the method was employed in men treated with finasteride or dutasteride; levels of DHT were lowered by both drugs and furthermore the substrate concentrations increased.
Collapse
Affiliation(s)
- Rita Upreti
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Gregorio Naredo
- Mass Spectrometry Core, Wellcome Trust Clinical Research Facility, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Abdullah M M Faqehi
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Katherine A Hughes
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Laurence H Stewart
- Department of Urology, NHS Lothian, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, United Kingdom.
| | - Brian R Walker
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom; Mass Spectrometry Core, Wellcome Trust Clinical Research Facility, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Natalie Z M Homer
- Mass Spectrometry Core, Wellcome Trust Clinical Research Facility, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | - Ruth Andrew
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom; Mass Spectrometry Core, Wellcome Trust Clinical Research Facility, Queen׳s Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
35
|
Mohler JL. Concept and viability of androgen annihilation for advanced prostate cancer. Cancer 2014; 120:2628-37. [PMID: 24771515 PMCID: PMC4140964 DOI: 10.1002/cncr.28675] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/17/2013] [Accepted: 01/23/2014] [Indexed: 12/22/2022]
Abstract
There remains no standard of care for patients with a rising prostate-specific antigen level after radical prostatectomy or radiotherapy but who have no radiographic metastases, even though this is the second largest group of patients with prostate cancer (CaP) in the United States. Androgen deprivation therapy (ADT) may cure some men with advanced CaP based on single-institution series and a randomized clinical trial of immediate versus delayed ADT for men found to have pelvic lymph node metastasis at the time of radical prostatectomy. ADT may be more effective when initiated for minimal disease burden, which can be detected using PSA after radical prostatectomy or radiotherapy, and if more complete disruption of the androgen axis using newer agents decreases the chance that androgen-sensitive cells survive to adapt to a low-androgen environment. Androgens may be "annihilated" simultaneously using a luteinizing hormone-releasing hormone antagonist or agonist to inhibit testicular production of testosterone, a P45017A1 (CYP17A1) inhibitor to diminish metabolism of testosterone via the adrenal pathway and dihydrotestosterone (DHT) via the backdoor pathway, a 5α-reductase (SRD5A) inhibitor to diminish testosterone reduction to DHT and backdoor metabolism of progesterone substrates to DHT, and a newer antiandrogen to compete better with DHT for the androgen receptor ligand-binding domain. Early initiation of androgen annihilation for induction as part of planned intermittent ADT should be safe, may reduce tumor burden below a threshold that allows eradication by the immune system, and may cure many men who have failed definitive local therapy.
Collapse
Affiliation(s)
- James L Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York; Department of Urology, State University of New York at Buffalo School of Medicine and Biotechnology, Buffalo, New York
| |
Collapse
|
36
|
Kuo KF, Hunter-Merrill R, Gulati R, Hall SP, Gambol TE, Higano CS, Yu EY. Relationships between times to testosterone and prostate-specific antigen rises during the first off-treatment interval of intermittent androgen deprivation are prognostic for castration resistance in men with nonmetastatic prostate cancer. Clin Genitourin Cancer 2014; 13:10-6. [PMID: 25242417 DOI: 10.1016/j.clgc.2014.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/03/2014] [Accepted: 08/05/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Intermittent androgen deprivation (IAD) represents an alternative to continuous AD with quality-of-life benefit and no evidence of inferior overall survival for nonmetastatic prostate cancer. Early markers of prognosis for men treated with IAD have not been described. PATIENTS AND METHODS Men with nonmetastatic prostate cancer were treated with 9 months of leuprolide and flutamide followed by a variable off-treatment interval; AD was resumed when prostate specific antigen (PSA) reached a prespecified value (1 ng/mL, radical prostatectomy; 4 ng/mL, intact prostate). Cycles were repeated until castration resistance (marking the advent of castration-resistant prostate cancer [CRPC]), defined as 2 PSA rises with testosterone (T) ≤ 50 ng/dL. Kinetics and relationships of PSA and T levels were evaluated, with a focus on times to rise in each level, during the first off-treatment interval. Associations with CRPC and prostate cancer mortality were estimated using Cox proportional hazards models controlling for age and Gleason score. RESULTS Each 30-day increase in time to PSA rise was associated with a 21% reduction in the risk of developing CRPC (95% CI, 3%-36%; P = .02). Longer time (≥ 60 days) to PSA rise after rise to T > 50 ng/dL was associated with a 71% reduction in the risk of developing CRPC (95% CI, 92% reduction to 2% inflation; P = .05). Time to first T > 50 ng/dL and PSA doubling time were not prognostic for progression to CRPC. No time interval was prognostic for prostate cancer mortality. CONCLUSION During the first off-treatment interval of IAD, longer times to PSA rise overall and after T > 50 ng/dL were associated with reduced risk of developing CRPC.
Collapse
Affiliation(s)
- Kevin F Kuo
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Rachel Hunter-Merrill
- Fred Hutchinson Cancer Research Center, Biostatistics and Biomathematics Program, Public Health Sciences Division, Seattle, WA
| | - Roman Gulati
- Fred Hutchinson Cancer Research Center, Biostatistics and Biomathematics Program, Public Health Sciences Division, Seattle, WA
| | - Suzanne P Hall
- University of Washington/Fred Hutchinson Cancer Research Center, Department of Medicine, Division of Oncology, Seattle, WA
| | - Teresa E Gambol
- University of Washington/Fred Hutchinson Cancer Research Center, Department of Medicine, Division of Oncology, Seattle, WA
| | - Celestia S Higano
- University of Washington/Fred Hutchinson Cancer Research Center, Department of Medicine, Division of Oncology, Seattle, WA
| | - Evan Y Yu
- University of Washington/Fred Hutchinson Cancer Research Center, Department of Medicine, Division of Oncology, Seattle, WA.
| |
Collapse
|
37
|
Hoy SM. Abiraterone acetate: a review of its use in patients with metastatic castration-resistant prostate cancer. Drugs 2014; 73:2077-91. [PMID: 24271422 DOI: 10.1007/s40265-013-0150-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Abiraterone acetate (Zytiga(®)) is an orally administered, selective inhibitor of the 17α-hydroxylase and C17,20-lyase enzymatic activities of cytochrome P450 (CYP) 17. CYP17 is required for androgen biosynthesis, with androgen receptor signalling crucial in the progression from primary to metastatic prostate cancer. Abiraterone acetate is approved in the European Union and the US, in combination with prednisone or prednisolone, for the treatment of men with metastatic castration-resistant prostate cancer (CRPC). When administered in combination with prednisone in a placebo-controlled, multinational phase III study, abiraterone acetate significantly prolonged overall survival and radiographic progression-free survival (rPFS) in men with metastatic CRPC who had previously received docetaxel. In men with metastatic CRPC who had not previously received chemotherapy participating in a placebo-controlled, multinational phase III study, there was a strong trend towards an overall survival benefit, a significant prolongation in rPFS and significant delays in clinical decline, the need for chemotherapy and the onset of pain observed. Given the nature of the therapy, the overall tolerability profile of abiraterone acetate, in combination with prednisone, was acceptable in men with metastatic CRPC. Abiraterone acetate is associated with hypokalaemia, hypertension, and fluid retention or oedema, secondary to its mechanism of action, and with cardiac adverse events and hepatotoxicity; however, in the phase III studies the incidences of the most frequently reported grade 3 or 4 adverse events of special interest were relatively low. Although the final overall survival data in men with metastatic CRPC who have not previously received chemotherapy are awaited, current evidence indicates that abiraterone acetate is a useful option for the treatment of metastatic CRPC.
Collapse
Affiliation(s)
- Sheridan M Hoy
- Adis, 41 Centorian Drive, Private Bag 65901, Mairangi Bay, North Shore, 0754, Auckland, New Zealand,
| |
Collapse
|
38
|
Abstract
Prostate cancer is the second leading cause of death in adult males in the USA. Recent advances have revealed that the fatal form of this cancer, known as castration-resistant prostate cancer (CRPC), remains hormonally driven despite castrate levels of circulating androgens. CRPC arises as the tumor undergoes adaptation to low levels of androgens by either synthesizing its own androgens (intratumoral androgens) or altering the androgen receptor (AR). This article reviews the major routes to testosterone and dihydrotestosterone synthesis in CRPC cells and examines the enzyme targets and progress in the development of isoform-specific inhibitors that could block intratumoral androgen biosynthesis. Because redundancy exists in these pathways, it is likely that inhibition of a single pathway will lead to upregulation of another so that drug resistance would be anticipated. Drugs that target multiple pathways or bifunctional agents that block intratumoral androgen biosynthesis and antagonize the AR offer the most promise. Optimal use of enzyme inhibitors or AR antagonists to ensure maximal benefits to CRPC patients will also require application of precision molecular medicine to determine whether a tumor in a particular patient will be responsive to these treatments either alone or in combination.
Collapse
Affiliation(s)
- Trevor M Penning
- Perelman School of MedicineCenter of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| |
Collapse
|
39
|
Upreti R, Hughes KA, Livingstone DEW, Gray CD, Minns FC, Macfarlane DP, Marshall I, Stewart LH, Walker BR, Andrew R. 5α-reductase type 1 modulates insulin sensitivity in men. J Clin Endocrinol Metab 2014; 99:E1397-406. [PMID: 24823464 PMCID: PMC4207930 DOI: 10.1210/jc.2014-1395] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
CONTEXT 5α-Reductase (5αR) types 1 and 2 catalyze the A-ring reduction of steroids, including androgens and glucocorticoids. 5α-R inhibitors lower dihydrotestosterone in benign prostatic hyperplasia; finasteride inhibits 5αR2, and dutasteride inhibits both 5αR2 and 5αR1. In rodents, loss of 5αR1 promotes fatty liver. OBJECTIVE Our objective was to test the hypothesis that inhibition of 5αR1 causes metabolic dysfunction in humans. DESIGN, SETTING, AND PARTICIPANTS This double-blind randomized controlled parallel group study at a clinical research facility included 46 men (20-85 years) studied before and after intervention. INTERVENTION Oral dutasteride (0.5 mg daily; n = 16), finasteride (5 mg daily; n = 16), or control (tamsulosin; 0.4 mg daily; n = 14) was administered for 3 months. MAIN OUTCOME MEASURE Glucose disposal was measured during a stepwise hyperinsulinemic-euglycemic clamp. Data are mean (SEM). RESULTS Dutasteride and finasteride had similar effects on steroid profiles, with reduced urinary androgen and glucocorticoid metabolites and reduced circulating DHT but no change in plasma or salivary cortisol. Dutasteride, but not finasteride, reduced stimulation of glucose disposal by high-dose insulin (dutasteride by -5.7 [3.2] μmol/kg fat-free mass/min, versus finasteride +7.2 [3.0], and tamsulosin +7.0 [2.0]). Dutasteride also reduced suppression of nonesterified fatty acids by insulin and increased body fat (by 1.6% [0.6%]). Glucose production and glycerol turnover were unchanged. Consistent with metabolic effects of dutasteride being mediated in peripheral tissues, mRNA for 5αR1 but not 5αR2 was detected in human adipose tissue. CONCLUSION Dual inhibition of 5αRs, but not inhibition of 5αR2 alone, modulates insulin sensitivity in human peripheral tissues rather than liver. This may have important implications for patients prescribed dutasteride for prostatic disease.
Collapse
Affiliation(s)
- Rita Upreti
- University/British Heart Foundation Centre for Cardiovascular Science (R.U., K.A.H., D.E.W.L., D.P.M., I.M., B.R.W., R.A.) and Clinical Research Imaging Centre (C.D.G.), University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom; and Radiology (F.C.M.) and Urology (L.H.S.) Departments, National Health Service Lothian University Hospitals Division, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Knuuttila M, Yatkin E, Kallio J, Savolainen S, Laajala TD, Aittokallio T, Oksala R, Häkkinen M, Keski-Rahkonen P, Auriola S, Poutanen M, Mäkelä S. Castration induces up-regulation of intratumoral androgen biosynthesis and androgen receptor expression in an orthotopic VCaP human prostate cancer xenograft model. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2163-73. [PMID: 24949550 DOI: 10.1016/j.ajpath.2014.04.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/03/2014] [Accepted: 04/16/2014] [Indexed: 11/17/2022]
Abstract
Androgens are key factors involved in the development and progression of prostate cancer (PCa), and PCa growth can be suppressed by androgen deprivation therapy. In a considerable proportion of men receiving androgen deprivation therapy, however, PCa progresses to castration-resistant PCa (CRPC), making the development of efficient therapies challenging. We used an orthotopic VCaP human PCa xenograft model to study cellular and molecular changes in tumors after androgen deprivation therapy (castration). Tumor growth was monitored through weekly serum prostate-specific antigen measurements, and mice with recurrent tumors after castration were randomized to treatment groups. Serum prostate-specific antigen concentrations showed significant correlation with tumor volume. Castration-resistant tumors retained concentrations of intratumoral androgen (androstenedione, testosterone, and 5α-dihydrotestosterone) at levels similar to tumors growing in intact hosts. Accordingly, castration induced up-regulation of enzymes involved in androgen synthesis (CYP17A1, AKR1C3, and HSD17B6), as well as expression of full-length androgen receptor (AR) and AR splice variants (AR-V1 and AR-V7). Furthermore, AR target gene expression was maintained in castration-resistant xenografts. The AR antagonists enzalutamide (MDV3100) and ARN-509 suppressed PSA production of castration-resistant tumors, confirming the androgen dependency of these tumors. Taken together, the findings demonstrate that our VCaP xenograft model exhibits the key characteristics of clinical CRPC and thus provides a valuable tool for identifying druggable targets and for testing therapeutic strategies targeting AR signaling in CRPC.
Collapse
Affiliation(s)
- Matias Knuuttila
- Department of Physiology, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Emrah Yatkin
- Department of Physiology, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Functional Foods Forum, University of Turku, Turku, Finland
| | - Jenny Kallio
- Department of Physiology, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Saija Savolainen
- Department of Physiology, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Teemu D Laajala
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Tero Aittokallio
- Department of Mathematics and Statistics, University of Turku, Turku, Finland; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Riikka Oksala
- Department of Oncology and Critical Care Research, Orion Pharma, Turku, Finland
| | - Merja Häkkinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Matti Poutanen
- Department of Physiology, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Sari Mäkelä
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Functional Foods Forum, University of Turku, Turku, Finland.
| |
Collapse
|
41
|
Roy J, Fournier MA, Maltais R, Kenmogne LC, Poirier D. In vitro and in vivo evaluation of a 3β-androsterone derivative as inhibitor of 17β-hydroxysteroid dehydrogenase type 3. J Steroid Biochem Mol Biol 2014; 141:44-51. [PMID: 24434282 DOI: 10.1016/j.jsbmb.2013.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/29/2013] [Accepted: 12/31/2013] [Indexed: 11/17/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 3 (17β-HSD3 or HSD17B3) catalyzes the last step in the biosynthesis of the potent androgen testosterone (T), by stereoselectively reducing the C17 ketone of 4-androstene-3,17-dione (4-dione), with NADPH as cofactor. Since T plays an important role in androgen-sensitive diseases, this enzyme is thus an interesting therapeutic target. In an attempt to design compounds to lower the level of T, we synthesized androsterone derivatives substituted at position 3 as inhibitors of 17β-HSD3, and selected one of the most potent compounds for additional studies. In an enzymatic assay in homogenized and whole HEK-293 cells overexpressing 17β-HSD3, the inhibitor RM-532-105 efficiently inhibited the conversion of natural substrate 4-dione (50nM) into T with an IC50 of 26nM and 5nM, respectively. Moreover, the inhibitor RM-532-105 (10mg/kg) reached a plasma concentration of 250ng/mL at 7h (AUC 24h: 3485ngh/mL) after subcutaneous (s.c.) injection in the rat. In order to mimic the human situation in which 4-dione is converted to T in the testis, we used intact rats. Treatment for 7 days with 17β-HSD3 inhibitor RM-532-105 by s.c. injection or oral gavage exerted no effect on the testis, prostate and seminal vesicle weight and no modification in the levels of plasma steroids. However, after this treatment, the concentration of inhibitor in plasma increased depending on the dose. We thereafter determined the concentration of inhibitor in the testis and we discovered that the compound was slightly present. In fact, at 10mg/kg, the inhibitor RM-532-105 seems to have difficulty penetrating inside the testis and was found to be concentrated in the testicular capsule, and therefore unable to inhibit the 17β-HSD3 located inside the testis. However, with a higher dose of 50mg/kg injected s.c. in rats, RM-532-105 significantly decreased the level of T and dihydrotestosterone measured in plasma at 2h.
Collapse
Affiliation(s)
- Jenny Roy
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Michelle-Audrey Fournier
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Lucie Carolle Kenmogne
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL, T4) - Research Center and Laval University (Faculty of Medicine), Québec, Québec G1V 4G2, Canada.
| |
Collapse
|
42
|
Sharad S, Ravindranath L, Haffner MC, Li H, Yan W, Sesterhenn IA, Chen Y, Ali A, Srinivasan A, McLeod DG, Yegnasubramanian S, Srivastava S, Dobi A, Petrovics G. Methylation of the PMEPA1 gene, a negative regulator of the androgen receptor in prostate cancer. Epigenetics 2014; 9:918-27. [PMID: 24694733 PMCID: PMC4065188 DOI: 10.4161/epi.28710] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The prostate transmembrane protein androgen induced 1 (PMEPA1) gene is highly expressed in prostate epithelial cells and is a direct transcriptional target for the androgen receptor (AR). AR protein levels are controlled by the AR-PMEPA1 negative feedback loop through NEDD4-E3 ligase. Reduced expression of PMEPA1 observed in prostate tumors, suggests that loss of PMEPA1 may play critical roles in prostate tumorigenesis. This study focuses on epigenetic mechanisms of reduced PMEPA1 expression in the cancer of the prostate (CaP). Benign (n = 77) and matched malignant (n = 77) prostate epithelial cells were laser capture micro-dissected from optimum cutting temperature embedded frozen prostate sections from 42 Caucasian American (CA) and 35 African American (AA) cases. Purified DNA specimens were analyzed for CpG methylation of the PMEPA1 gene. PMEPA1 mRNA expression levels were evaluated by qRT-PCR. Analysis of PMEPA1 methylation and mRNA expression in the same tumor cell populations indicated a significant inverse correlation between mRNA expression and methylation in CaP (P = 0.0115). We noted higher frequency of CpG methylation within the evaluated first intronic region of the PMEPA1 gene in prostate tumors of CA men as compared with AA. In CaP cell lines, PMEPA1 expression was induced and AR protein levels were diminished in response to treatment with the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (decitabine). Cell culture-based studies demonstrated that decitabine restores PMEPA1 expression in AR-positive CaP cell lines. This report reveals the potential role of PMEPA1 gene methylation in the regulation of AR stability. Thus, downregulation of PMEPA1 may result in increased AR protein levels and function in CaP cells, contributing to prostate tumorigenesis.
Collapse
Affiliation(s)
- Shashwat Sharad
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Lakshmi Ravindranath
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Michael C Haffner
- Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University; Baltimore, MD USA
| | - Hua Li
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Wusheng Yan
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | | | - Yongmei Chen
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Amina Ali
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA; Urology Service; Walter Reed National Military Medical Center; Bethesda, MD USA
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - David G McLeod
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA; Urology Service; Walter Reed National Military Medical Center; Bethesda, MD USA
| | | | - Shiv Srivastava
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Albert Dobi
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research; Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| |
Collapse
|
43
|
Titus MA, Li Y, Kozyreva OG, Maher V, Godoy A, Smith GJ, Mohler JL. 5α-reductase type 3 enzyme in benign and malignant prostate. Prostate 2014; 74:235-49. [PMID: 24150795 PMCID: PMC3992828 DOI: 10.1002/pros.22745] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 09/23/2013] [Indexed: 11/09/2022]
Abstract
BACKGROUND Currently available 5α-reductase inhibitors are not completely effective for treatment of benign prostate enlargement, prevention of prostate cancer (CaP), or treatment of advanced castration-recurrent (CR) CaP. We tested the hypothesis that a novel 5α-reductase, 5α-reductase-3, contributes to residual androgen metabolism, especially in CR-CaP. METHODS A new protein with potential 5α-reducing activity was expressed in CHO-K1 cellsandTOP10 E. coli for characterization. Protein lysates and total mRNA were isolated from preclinical and clinical tissues. Androgen metabolism was assessed using androgen precursors and thin layer chromatography or liquid chromatography tandem mass spectrometry. RESULTS The relative mRNA expression for the three 5α-reductase enzymes in clinical samples of CR-CaP was 5α-reductase-3 ≫ 5α-reductase-1> 5α-reductase-2. Recombinant 5α-reductase-3 protein incubations converted testosterone, 4-androstene-3,17-dione (androstenedione) and 4-pregnene-3,20-dione (progesterone) to dihydrotestosterone, 5α-androstan-3,17-dione, and 5α-pregnan-3,20-dione, respectively. 5α-Reduced androgen metabolites were measurable in lysates from androgen-stimulated (AS) CWR22 and CR-CWR22 tumors and clinical specimens of AS-CaP and CR-CaP pre-incubated with dutasteride (a bi-specific inhibitor of 5α-reductase-1 and 2). CONCLUSION Human prostate tissues contain a third 5α-reductase that was inhibited poorly by dutasteride at high androgen substrate concentration in vitro, and it may promote DHT formation in vivo, through alternative androgen metabolism pathways when testosterone levels are low.
Collapse
Affiliation(s)
- Mark A. Titus
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
- Correspondence to: Mark A. Titus, PhD, Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas, MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721.
| | - Yun Li
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Olga G. Kozyreva
- Lineberger Comprehensive Cancer Center University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Varun Maher
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Alejandro Godoy
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Gary J. Smith
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - James L. Mohler
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York
- Departmentof Urology, University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, New York
- Lineberger Comprehensive Cancer Center University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Departmentof Surgery, Division of Urology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
44
|
Pezaro C, Omlin A, Bianchini D, de Bono J. New Therapies in Hormone Relapsed Disease. Prostate Cancer 2014. [DOI: 10.1002/9781118347379.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
45
|
Abstract
As the first in class steroid 17α-hydroxylase/C17,20-lyase (CYP17) inhibitor, abiraterone acetate (of which the active metabolite is abiraterone) has been shown to improve overall survival in patients with castration-resistant prostate cancer (CRPC)--in those who are chemotherapy-naive and those previously treated with docetaxel. Furthermore, the clinical success of abiraterone demonstrated that CRPC, which has previously been regarded as an androgen-independent disease, is still driven, at least in part, by androgens. More importantly, abiraterone is a 'promiscuous' drug that interacts with a number of targets, which dictate its clinical benefits and adverse effects profile. Besides CYP17 inhibition, abiraterone acts as an antagonist to the androgen receptor and inhibits 3β-hydroxysteroid dehydrogenase--two effects that potentially contribute to its antitumour effects. However, the inhibition of the 17α-hydroxylase activity of CYP17, CYP11B1 and a panel of hepatic CYP enzymes leads to adverse effects and toxicities that include secondary mineralocorticoid excess. Abiraterone is also associated with increased incidence of cardiac disorders. Under such circumstances, development of new CYP17 inhibitors as an additional line of defence is urgently needed. To achieve enhanced clinical benefits, new strategies are being explored that include selective inhibition of the C17,20-lyase activity of CYP17 and multi-targeting strategies that affect androgen synthesis and signalling at different points. Some of these strategies-including the drugs orteronel, VT-464 and galeterone--are supported by preclinical data and are being explored in the clinic.
Collapse
|
46
|
Stanczyk FZ, Azen CG, Pike MC. Effect of finasteride on serum levels of androstenedione, testosterone and their 5α-reduced metabolites in men at risk for prostate cancer. J Steroid Biochem Mol Biol 2013; 138:10-6. [PMID: 23474436 DOI: 10.1016/j.jsbmb.2013.02.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 02/24/2013] [Accepted: 02/27/2013] [Indexed: 11/17/2022]
Abstract
Studies show that treatment of men with 5α-reductase inhibitors such as finasteride is effective for the primary prevention of prostate cancer. Although it is known that finasteride treatment suppresses serum levels of dihydrotestosterone (DHT) and its distal metabolite, 5α-androstane-3α,17β-diol glucuronide (3α-diol G), and increases serum testosterone (T) levels, little is known about its effect on other precursors and metabolites of DHT, as well as on the relationship of these androgens to prostate specific antigen (PSA), a marker of prostatic intraepithelial neoplasia. The present study provides new data on the effect of finasteride on precursors and metabolites of DHT. Fifty-three men, ages 57-79 years, with elevated PSA levels (>4ng/ml), were randomized to treatment with finasteride (5mg/day) or observation (controls) for 12 months. Blood samples were obtained at baseline, 1, 3, 6 and 12 months for measurement of PSA, androstenedione (A), T, DHT, 3α-diol G, androsterone glucuronide (ADT G) and DHT sulfate (DHT S) in serum by validated, highly specific radioimmunoassays. Statistical analysis was carried out using mixed model ANOVA and t-tests. In the control group, PSA and androgen levels were unchanged throughout the 12 months of treatment. In the finasteride group, PSA, DHT, DHT S, 3α-diol G and ADT G decreased from baseline to 1 month by 23.2%, 78.7%, 71.0%, 75.7% and 43.0%, respectively. The change in PSA decreased further to 46.1% and 55.1% at 3 and 12 months of treatment, respectively, whereas the decrease in androgens observed at 1 month did not change by more than 6.9% for DHT, DHT S and 3α-diol G in the subsequent months of sampling. However, the decline in ADT G was only 22.2% at month 3, and remained essentially at this level after that time. In contrast, T and A increased significantly from baseline, and the increase in A of approximately 34.5% was about 1.9 times the increase in T (approximately 18.3%). The present data suggest that either 3α-diol G or DHT S may serve as a potential diagnostic marker of intraprostatic 5α-reductase activity during treatment of patients with 5α-reductase inhibitors.
Collapse
Affiliation(s)
- Frank Z Stanczyk
- Department of Obstetrics and Gynecology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, United States; Department of Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, United States.
| | | | | |
Collapse
|
47
|
Chhipa RR, Halim D, Cheng J, Zhang HY, Mohler JL, Ip C, Wu Y. The direct inhibitory effect of dutasteride or finasteride on androgen receptor activity is cell line specific. Prostate 2013; 73:1483-94. [PMID: 23813737 PMCID: PMC3992475 DOI: 10.1002/pros.22696] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/15/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Finasteride and dutasteride were developed originally as 5α-reductase inhibitors to block the conversion of testosterone to dihydrotestosterone (DHT). These drugs may possess off-target effects on the androgen receptor (AR) due to their structural similarity to DHT. METHODS A total of four human prostate cancer cell models were examined: LNCaP (T877A mutant AR), 22Rv1 (H874Y mutant AR), LAPC4 (wild-type AR), and VCaP (wild-type AR). Cells were cultured in 10% charcoal-stripped fetal bovine serum, either with or without DHT added to the medium. AR activity was evaluated using the ARE-luciferase assay or the expression of AR regulated genes. RESULTS Dutasteride was more potent than finasteride in interfering with DHT-stimulated AR signaling. Disruption of AR function was accompanied by decreased cell growth. Cells that rely on DHT for protection against death were particularly vulnerable to dutasteride. Different prostate cancer cell models exhibited different sensitivities to dutasteride and finasteride. LNCaP was most sensitive, LAPC4 and VCaP were intermediate, while 22Rv1 was least sensitive. Regardless of the AR genotype, if AR was transfected into drug-sensitive cells, AR was inhibited by drug treatment; and if AR was transfected into drug-resistant cells, AR was not inhibited. CONCLUSIONS The direct inhibitory effect of dutasteride or finasteride on AR signaling is cell line specific. Mutations in the ligand binding domain of AR do not appear to play a significant role in influencing the AR antagonistic effect of these drugs. Subcellular constituent is an important factor in determining the drug effect on AR function.
Collapse
Affiliation(s)
- Rishi Raj Chhipa
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
| | - Danny Halim
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
- Health Research Unit Faculty of Medicine Universitas Padjadjaran Bandung 40161, Indonesia
| | - Jinrong Cheng
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
| | - Huan Yi Zhang
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
| | - James L. Mohler
- Department of Urology Roswell Park Cancer Institute Buffalo, NY 14263
- Department of Urology University at Buffalo School of Medicine and Biotechnology Buffalo, NY 14263
| | - Clement Ip
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
| | - Yue Wu
- Department of Cancer Prevention and Control Roswell Park Cancer Institute Buffalo, NY 14263
- Corresponding Author: Department of Cancer Prevention and Control Roswell Park Cancer Institute Elm & Carlton Streets Buffalo, NY 14263 Phone: 716-845-1704; Fax: 716-845-8100
| |
Collapse
|
48
|
Adeniji AO, Chen M, Penning TM. AKR1C3 as a target in castrate resistant prostate cancer. J Steroid Biochem Mol Biol 2013; 137:136-49. [PMID: 23748150 PMCID: PMC3805777 DOI: 10.1016/j.jsbmb.2013.05.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/04/2013] [Accepted: 05/08/2013] [Indexed: 01/27/2023]
Abstract
Aberrant androgen receptor (AR) activation is the major driver of castrate resistant prostate cancer (CRPC). CRPC is ultimately fatal and more therapeutic agents are needed to treat this disease. Compounds that target the androgen axis by inhibiting androgen biosynthesis and or AR signaling are potential candidates for use in CRPC treatment and are currently being pursued aggressively. Aldo-keto reductase 1C3 (AKR1C3) plays a pivotal role in androgen biosynthesis within the prostate. It catalyzes the 17-ketoreduction of weak androgen precursors to give testosterone and 5α-dihydrotestosterone. AKR1C3 expression and activity has been implicated in the development of CRPC, making it a rational target. Selective inhibition of AKR1C3 will be important, however, due to the presence of closely related isoforms, AKR1C1 and AKR1C2 that are also involved in androgen inactivation. We examine the evidence that supports the vital role of AKR1C3 in CRPC and recent developments in the discovery of potent and selective AKR1C3 inhibitors. This article is part of a Special Issue entitled 'CSR 2013'.
Collapse
Affiliation(s)
- Adegoke O. Adeniji
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6061
| | - Mo Chen
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6061
| | - Trevor M. Penning
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6061
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6061
| |
Collapse
|
49
|
Yepuru M, Wu Z, Kulkarni A, Yin F, Barrett CM, Kim J, Steiner MS, Miller DD, Dalton JT, Narayanan R. Steroidogenic enzyme AKR1C3 is a novel androgen receptor-selective coactivator that promotes prostate cancer growth. Clin Cancer Res 2013; 19:5613-25. [PMID: 23995860 DOI: 10.1158/1078-0432.ccr-13-1151] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Castration-resistant prostate cancer (CRPC) may occur by several mechanisms including the upregulation of androgen receptor (AR), coactivators, and steroidogenic enzymes, including aldo keto reductase 1C3 (AKR1C3). AKR1C3 converts weaker 17-keto androgenic precursors to more potent 17-hydroxy androgens and is consistently the major upregulated gene in CRPC. The studies in the manuscript were undertaken to examine the role of AKR1C3 in AR function and CRPC. EXPERIMENTAL DESIGN LNCaP cells stably transfected with AKR1C3 and VCaP cells endogenously expressing AKR1C3 were used to understand the effect of AKR1C3 on prostate cancer cell and tumor growth in nude mice. Chromatin immunoprecipitation, confocal microscopy, and co-immunoprecipitation studies were used to understand the recruitment of AKR1C3, intracellular localization of AKR1C3 and its interaction with AR in cells, tumor xenograft, and in Gleason sum 7 CRPC tissues. Cells were transiently transfected for AR transactivation. Novel small-molecule AKR1C3-selective inhibitors were synthesized and characterized in androgen-dependent prostate cancer and CRPC models. RESULTS We identified unique AR-selective coactivator- and prostate cancer growth-promoting roles for AKR1C3. AKR1C3 overexpression promotes the growth of both androgen-dependent prostate cancer and CRPC xenografts, with concomitant reactivation of androgen signaling. AKR1C3 interacted with AR in prostate cancer cells, xenografts, and in human CRPC samples and was recruited to the promoter of an androgen-responsive gene. The coactivator and growth-promoting functions of AKR1C3 were inhibited by an AKR1C3-selective competitive inhibitor. CONCLUSIONS AKR1C3 is a novel AR-selective enzymatic coactivator and may represent the first of more than 200 known nuclear hormone receptor coactivators that can be pharmacologically targeted.
Collapse
Affiliation(s)
- Muralimohan Yepuru
- Authors' Affiliations: Preclinical Research and Development, GTx Inc.; and Department of Pathology, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
|