1
|
He Y, Xu W, Xiao YT, Huang H, Gu D, Ren S. Targeting signaling pathways in prostate cancer: mechanisms and clinical trials. Signal Transduct Target Ther 2022; 7:198. [PMID: 35750683 PMCID: PMC9232569 DOI: 10.1038/s41392-022-01042-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) affects millions of men globally. Due to advances in understanding genomic landscapes and biological functions, the treatment of PCa continues to improve. Recently, various new classes of agents, which include next-generation androgen receptor (AR) signaling inhibitors (abiraterone, enzalutamide, apalutamide, and darolutamide), bone-targeting agents (radium-223 chloride, zoledronic acid), and poly(ADP-ribose) polymerase (PARP) inhibitors (olaparib, rucaparib, and talazoparib) have been developed to treat PCa. Agents targeting other signaling pathways, including cyclin-dependent kinase (CDK)4/6, Ak strain transforming (AKT), wingless-type protein (WNT), and epigenetic marks, have successively entered clinical trials. Furthermore, prostate-specific membrane antigen (PSMA) targeting agents such as 177Lu-PSMA-617 are promising theranostics that could improve both diagnostic accuracy and therapeutic efficacy. Advanced clinical studies with immune checkpoint inhibitors (ICIs) have shown limited benefits in PCa, whereas subgroups of PCa with mismatch repair (MMR) or CDK12 inactivation may benefit from ICIs treatment. In this review, we summarized the targeted agents of PCa in clinical trials and their underlying mechanisms, and further discussed their limitations and future directions.
Collapse
Affiliation(s)
- Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Weidong Xu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China
| | - Yu-Tian Xiao
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China.,Department of Urology, Shanghai Changhai Hospital, Shanghai, China
| | - Haojie Huang
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Di Gu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China.
| |
Collapse
|
2
|
Giridharan M, Rupani V, Banerjee S. Signaling Pathways and Targeted Therapies for Stem Cells in Prostate Cancer. ACS Pharmacol Transl Sci 2022; 5:193-206. [PMID: 35434534 PMCID: PMC9003388 DOI: 10.1021/acsptsci.2c00019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCa) is one of the most frequently occurring cancers among men, and the current statistics show that it is the second leading cause of cancer-related deaths among men. Over the years, research in PCa treatment and therapies has made many advances. Despite these efforts, the standardized therapies such as radiation, chemotherapy, hormonal therapy and surgery are not considered completely effective in treating advanced and metastatic PCa. In most situations, fast-dividing tumor cells are targeted, leaving behind relatively slowly dividing, chemoresistant cells known as cancer stem cells. Therefore, following the seemingly successful treatments, the lingering quiescent cancer stem cells are able to renew themselves, undergo differentiation into mature tumor cells, and sufficiently reinitiate the disease, leading to cancer relapse. Thus, prostate cancer stem cells (PCSCs) have been reported to play a vital role in controlling the dynamics of tumorigenesis, progression, and resistance to therapies in PCa. However, the complete knowledge on the mechanisms regulating the stemness of PCSCs is still unclear. Thus, studying the stemness of PCSCs will allow for the development of more effective cancer therapies due to the durable response, resulting in a reduction in recurrences of cancer. In this Review, we will specifically describe the molecular mechanisms responsible for regulating the stemness of PCSCs. Furthermore, current developments in stem cell-specific therapeutic approaches along with future prospects will also be discussed.
Collapse
Affiliation(s)
- Madhuvanthi Giridharan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore-632104, Tamil Nadu, India
| | - Vasu Rupani
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore-632104, Tamil Nadu, India
| | - Satarupa Banerjee
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore-632104, Tamil Nadu, India
| |
Collapse
|
3
|
Omoboyowa DA, Balogun TA, Saibu OA, Chukwudozie OS, Alausa A, Olubode SO, Aborode AT, Batiha GE, Bodun DS, Musa SO. Structure-based discovery of selective CYP 17A 1 inhibitors for Castration-resistant prostate cancer treatment. Biol Methods Protoc 2021; 7:bpab026. [PMID: 35146123 PMCID: PMC8824735 DOI: 10.1093/biomethods/bpab026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/11/2021] [Accepted: 12/22/2021] [Indexed: 11/12/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy found in men and the second leading cause of cancer-related death worldwide. Castration-resistant PCa (CRPC) is defined by PCa cells that stop responding to hormone therapy. Cytochrome P450 17α-hydroxylase/17,20-lyase (CYP17A1) plays a critical role in the biosynthesis of androgens in humans. Androgen signaling cascade is a principal survival pathway for PCa cells and androgen-deprivation therapy (ADT) remains the key treatment for patients marked with locally advanced and metastatic PCa cells. Available synthetic drugs have been reported for toxicity, drug resistance, and decreasing efficacy. Thus, the design of novel selective inhibitors of CYP17A1 lyase would help circumvent associated side effects and improve pharmacological activities. Therefore, we employed structural bioinformatics techniques via molecular docking; molecular mechanics generalized born surface area (MM-GBSA), molecular dynamics (MD) simulation, and pharmacokinetic study to identify putative CYP17A1 lyase inhibitors. The results of the computational investigation showed that the Prunus dulcis compounds exhibited higher binding energy than the clinically approved abiraterone acetate. The stability of the ligand with the highest binding affinity (quercetin-3-o-rutinoside) was observed during MD simulation for 10 ns. Quercetin-3-o-rutinoside was observed to be stable within the active site of CYP17A1Lyase throughout the simulation period. The result of the pharmacokinetic study revealed that these compounds are promising therapeutic agents. Collectively, this study proposed that bioactive compounds from P. dulcis may be potential selective inhibitors of CYP17A1Lyase in CRPC treatments.
Collapse
Affiliation(s)
| | - Toheeb A Balogun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Oluwatosin A Saibu
- Department of Environmental Toxicology, University of Duisburg-Essen, North Rhine-Westphalia, Germany
| | - Onyeka S Chukwudozie
- Division of Biological Science, University of California San Diego, CA 92161, USA
| | - Abdullahi Alausa
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Samuel O Olubode
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | | | - Gaber E Batiha
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Damanhour University, Damanhour City, Egypt
| | - Damilola S Bodun
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Sekinat O Musa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| |
Collapse
|
4
|
Saranyutanon S, Srivastava SK, Pai S, Singh S, Singh AP. Therapies Targeted to Androgen Receptor Signaling Axis in Prostate Cancer: Progress, Challenges, and Hope. Cancers (Basel) 2019; 12:cancers12010051. [PMID: 31877956 PMCID: PMC7016833 DOI: 10.3390/cancers12010051] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the mostly commonly diagnosed non-cutaneous malignancy and the second leading cause of cancer-related death affecting men in the United States. Moreover, it disproportionately affects the men of African origin, who exhibit significantly greater incidence and mortality as compared to the men of European origin. Since androgens play an important role in the growth of normal prostate and prostate tumors, targeting of androgen signaling has remained a mainstay for the treatment of aggressive prostate cancer. Over the years, multiple approaches have been evaluated to effectively target the androgen signaling pathway that include direct targeting of the androgens, androgen receptor (AR), AR co-regulators or other alternate mechanisms that impact the outcome of androgen signaling. Several of these approaches are currently in clinical practice, while some are still pending further development and clinical evaluation. This remarkable progress has resulted from extensive laboratory, pre-clinical and clinical efforts, and mechanistic learnings from the therapeutic success and failures. In this review, we describe the importance of androgen signaling in prostate cancer biology and advances made over the years to effectively target this signaling pathway. We also discuss emerging data on the resistance pathways associated with the failure of various androgen signaling- targeted therapies and potential of this knowledge for translation into future therapies for prostate cancer.
Collapse
Affiliation(s)
- Sirin Saranyutanon
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Correspondence: (S.K.S.); (A.P.S.); Tel.: +1-251-445-9874 (S.K.S.); +1-251-445-9843 (A.P.S.)
| | - Sachin Pai
- Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA;
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL 36617, USA; (S.S.)
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
- Correspondence: (S.K.S.); (A.P.S.); Tel.: +1-251-445-9874 (S.K.S.); +1-251-445-9843 (A.P.S.)
| |
Collapse
|
5
|
Gumede NJ, Nxumalo W, Bisetty K, Escuder Gilabert L, Medina-Hernandez MJ, Sagrado S. Prospective computational design and in vitro bio-analytical tests of new chemical entities as potential selective CYP17A1 lyase inhibitors. Bioorg Chem 2019; 94:103462. [PMID: 31818479 DOI: 10.1016/j.bioorg.2019.103462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/27/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Abstract
The development and advancement of prostate cancer (PCa) into stage 4, where it metastasize, is a major problem mostly in elder males. The growth of PCa cells is stirred up by androgens and androgen receptor (AR). Therefore, therapeutic strategies such as blocking androgens synthesis and inhibiting AR binding have been explored in recent years. However, recently approved drugs (or in clinical phase) failed in improving the expected survival rates for this metastatic-castration resistant prostate cancer (mCRPC) patients. The selective CYP17A1 inhibition of 17,20-lyase route has emerged as a novel strategy. Such inhibition blocks the production of androgens everywhere they are found in the body. In this work, a three dimensional-quantitative structure activity relationship (3D-QSAR) pharmacophore model is developed on a diverse set of non-steroidal inhibitors of CYP17A1 enzyme. Highly active compounds are selected to define a six-point pharmacophore hypothesis with a unique geometrical arrangement fitting the following description: two hydrogen bond acceptors (A), two hydrogen bond donors (D) and two aromatic rings (R). The QSAR model showed adequate predictive statistics. The 3D-QSAR model is further used for database virtual screening of potential inhibitory hit structures. Density functional theory (DFT) optimization provides the electronic properties explaining the reactivity of the hits. Docking simulations discovers hydrogen bonding and hydrophobic interactions as responsible for the binding affinities of hits to the CYP17A1 Protein Data Bank structure. 13 hits from the database search (including five derivatives) are then synthesized in the laboratory as different scaffolds. Ultra high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) in vitro experiments reveals three new chemical entities (NCEs) with half maximal inhibitory concentration (IC50) values against the lyase route at mid-micromolar range with favorable selectivity to the lyase over the hydroxylase route (one of them with null hydroxylase inhibition). Thus, prospective computational design has enabled the design of potential lead lyase-selective inhibitors for further studies.
Collapse
Affiliation(s)
- N J Gumede
- Department of Chemistry, Mangosuthu University of Technology, PO Box 12363, Jacobs 4026, South Africa.
| | - W Nxumalo
- Department of Chemistry, University of Limpopo, Private Bag X 1106, Sovenga 0727, South Africa
| | - K Bisetty
- Department of Chemistry, Durban University of Technology, PO Box 1334, Durban 4000, South Africa
| | - L Escuder Gilabert
- Departamento de Química Analítica, Facultad de Farmacia, Universidad de Valencia, Avda. Vicent Andrés Estellés, s/n, E-46100 Burjassot, Valencia, Spain
| | - M J Medina-Hernandez
- Departamento de Química Analítica, Facultad de Farmacia, Universidad de Valencia, Avda. Vicent Andrés Estellés, s/n, E-46100 Burjassot, Valencia, Spain
| | - S Sagrado
- Departamento de Química Analítica, Facultad de Farmacia, Universidad de Valencia, Avda. Vicent Andrés Estellés, s/n, E-46100 Burjassot, Valencia, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Avda. Vicent Andrés Estellés, s/n, E-46100 Burjassot, Valencia, Spain
| |
Collapse
|
6
|
Nakazawa M, Kyprianou N. Epithelial-mesenchymal-transition regulators in prostate cancer: Androgens and beyond. J Steroid Biochem Mol Biol 2017; 166:84-90. [PMID: 27189666 DOI: 10.1016/j.jsbmb.2016.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/20/2016] [Accepted: 05/07/2016] [Indexed: 12/16/2022]
Abstract
Castration resistant prostate cancer (CRPC) remains one of the leading causes of cancer deaths among men. Conventional therapies targeting androgen signaling driven tumor growth have provided limited survival benefit in patients. Recent identification of the critical molecular and cellular events surrounding tumor progression, invasion, and metastasis to the bone as well as other sites provide new insights in targeting advanced disease. Epithelial mesenchymal transition (EMT) is a process via which epithelial cells undergo morphological changes to a motile mesenchymal phenotype, a phenomenon implicated in cancer metastasis but also therapeutic resistance. Therapeutic targeting of EMT has the potential to open a new avenue in the treatment paradigm of CRPC through the reversion of the invasive mesenchymal phenotype to the well differentiated tumor epithelial tumor phenotype. Overcoming therapeutic resistance in metastatic prostate cancer is an unmet need in today's clinical management of advanced disease. This review outlines our current understanding of the contribution of EMT and its reversal to MET in prostate cancer progression and therapeutic resistance, and the impact of selected targeting of mechanisms of resistance via EMT towards a therapeutic benefit in patients with CRPC.
Collapse
Affiliation(s)
- Mary Nakazawa
- Departments of Urology, Biochemistry, Pathology and Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States, United States
| | - Natasha Kyprianou
- Departments of Urology, Biochemistry, Pathology and Toxicology & Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, United States, United States.
| |
Collapse
|
7
|
Abstract
Prostate cancer is the second leading cause of cancer deaths in the USA. The challenge in managing castration-resistant prostate cancer (CRPC) stems not from the lack of therapeutic options but from the limited duration of clinical and survival benefit offered by treatments in this setting due to primary and acquired resistance. The remarkable molecular heterogeneity and tumor adaptability in advanced prostate cancer necessitate optimization of such treatment strategies. While the future of CRPC management will involve newer targeted therapies in deliberately biomarker-selected patients, interventions using current approaches may exhibit improved clinical benefit if employed in the context of optimal sequencing and combinations. This review outlines our current understanding of mechanisms of therapeutic resistance in progression to and after the development of castration resistance, highlighting targetable and reversible mechanisms of resistance.
Collapse
Affiliation(s)
- Mary Nakazawa
- Departments of Urology, Molecular and Cellular Biochemistry, Pathology and Toxicology and Cancer Biology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536, USA
| | - Channing Paller
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Natasha Kyprianou
- Departments of Urology, Molecular and Cellular Biochemistry, Pathology and Toxicology and Cancer Biology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY, 40536, USA.
| |
Collapse
|
8
|
Oskarsson A, Ohlsson Andersson Å. Suppressed Sex Hormone Biosynthesis by Alkylresorcinols: A Possible Link to Chemoprevention. Nutr Cancer 2016; 68:978-87. [DOI: 10.1080/01635581.2016.1190022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
9
|
Dellis A, Papatsoris AG. Phase I and II therapies targeting the androgen receptor for the treatment of castration resistant prostate cancer. Expert Opin Investig Drugs 2016; 25:697-707. [PMID: 26954621 DOI: 10.1517/13543784.2016.1162784] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Prostate cancer is the most common cancer in elderly males. Regardless of the initial hormonal treatment in metastatic disease, a significant proportion of patients develop castration resistant prostate cancer (CRPC). A better understanding of the molecular mechanisms behind castration resistance has led to the approval of oral medications such as abiraterone acetate and enzalutamide. Relevant research is accelerated with numerous agents being tested for the management of CRPC. AREAS COVERED The authors present Phase I and II studies targeting the androgen receptor for the treatment of CRPC. Three groups of agents are identified according to the mechanism of action. These include the CYP-17 modulators (Orteronel, Galeterone, VT-464 and CFG-920), novel antiandrogens (Apatorsen, ARN-509, ODM-201, EZN-4176, AZD-3514) and bipolar androgen therapy. EXPERT OPINION Further understanding of the mechanisms leading to castration resistance in prostate cancer can reveal potential targets for the development of novel anti-cancer agents. Except for the development of novel antiandrogens and CYP-17 modulators, bipolar androgen therapy is an interesting therapeutic approach. The combinations of the novel agents tested in Phase I and II studies with established agents is another field of interest. The real challenge is to distinguish a novel anti-cancer agent with acceptable tolerability and the best outcome.
Collapse
Affiliation(s)
- Athanasios Dellis
- a University Department of Urology , Sismanoglio Hospital , Athens , Greece
| | - Athanasios G Papatsoris
- a University Department of Urology , Sismanoglio Hospital , Athens , Greece.,b Department of Urology, Addenbrooke's Hospital , Cambridge University Hospitals NHS , Cambridge , UK
| |
Collapse
|
10
|
Huang A, Jayaraman L, Fura A, Vite GD, Trainor GL, Gottardis MM, Spires T, Spires VM, Rizzo CA, Obermeier MT, Elzinga PA, Todderud G, Fan Y, Newitt JA, Beyer SM, Zhu Y, Warrack BM, Goodenough AK, Tebben AJ, Doweyko AM, Gold DL, Balog A. Discovery of the Selective CYP17A1 Lyase Inhibitor BMS-351 for the Treatment of Prostate Cancer. ACS Med Chem Lett 2016; 7:40-5. [PMID: 26819663 PMCID: PMC4716597 DOI: 10.1021/acsmedchemlett.5b00310] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/02/2015] [Indexed: 11/28/2022] Open
Abstract
Efforts to identify a potent, reversible, nonsteroidal CYP17A1 lyase inhibitor with good selectivity over CYP17A1 hydroxylase and CYPs 11B1 and 21A2 for the treatment of castration-resistant prostate cancer (CRPC) culminated in the discovery of BMS-351 (compound 18), a pyridyl biaryl benzimidazole with an excellent in vivo profile. Biological evaluation of BMS-351 at a dose of 1.5 mg in castrated cynomolgus monkeys revealed a remarkable reduction in testosterone levels with minimal glucocorticoid and mineralcorticoid perturbation. Based on a favorable profile, BMS-351 was selected as a candidate for further preclinical evaluation.
Collapse
Affiliation(s)
- Audris Huang
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Lata Jayaraman
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Aberra Fura
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Gregory D. Vite
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - George L. Trainor
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Marco M. Gottardis
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Thomas
E. Spires
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Vanessa M. Spires
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Cheryl A. Rizzo
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Mary T. Obermeier
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Paul A. Elzinga
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Gordon Todderud
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Yi Fan
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - John A. Newitt
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Sophie M. Beyer
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Yongxin Zhu
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Bethanne M. Warrack
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Angela K. Goodenough
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Andrew J. Tebben
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Arthur M. Doweyko
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - David L. Gold
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| | - Aaron Balog
- Bristol-Myers
Squibb Research
and Development, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
11
|
Nakazawa M, Lu C, Chen Y, Paller CJ, Carducci MA, Eisenberger MA, Luo J, Antonarakis ES. Serial blood-based analysis of AR-V7 in men with advanced prostate cancer. Ann Oncol 2015; 26:1859-1865. [PMID: 26117829 DOI: 10.1093/annonc/mdv282] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We previously showed that pretreatment detection of androgen receptor splice variant-7 (AR-V7) in circulating tumor cells (CTCs) from men with castration-resistant prostate cancer is associated with resistance to abiraterone and enzalutamide, but not to taxane chemotherapies. Here, we conducted serial measurements of AR-V7 and evaluated patterns of longitudinal AR-V7 dynamics over the course of multiple sequential therapies. PATIENTS AND METHODS Metastatic prostate cancer patients treated at Johns Hopkins with AR-directed therapies or taxane chemotherapies underwent serial liquid biopsies for CTC-based AR-V7 analysis at baseline, during therapy, and at progression. We used a CTC enrichment platform followed by multiplexed reverse-transcription polymerase chain reaction analysis to detect full-length androgen receptor and AR-V7 transcripts. Patients selected for inclusion in this report were those who provided ≥4 CTC samples, at least one of which was AR-V7 positive, over the course of ≥2 consecutive therapies. RESULTS We identified 14 patients who received a total of 37 therapies and contributed 70 CTC samples for AR-V7 analysis during a median follow-up period of 11 months. Three patients remained AR-V7 positive during the entire course of therapy. The remainder underwent transitions in AR-V7 status: there were eight instances of 'conversions' from AR-V7-negative to -positive status (during treatment with first-line androgen deprivation therapy, abiraterone, enzalutamide, and docetaxel), and six instances of 'reversions' from AR-V7-positive to -negative status (during treatment with docetaxel and cabazitaxel). CONCLUSIONS AR-V7 is a dynamic marker, and transitions in AR-V7 status may reflect selective pressures on the tumor exerted by therapeutic interventions. While 'conversions' to AR-V7-positive status were observed with both AR-directed therapies and taxane chemotherapies, 'reversions' to AR-V7-negative status only occurred during taxane therapies. Serial blood-based AR-V7 testing is feasible in routine clinical practice, and may provide insights into temporal changes in tumor evolution.
Collapse
Affiliation(s)
- M Nakazawa
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Lu
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Y Chen
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C J Paller
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - M A Carducci
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - M A Eisenberger
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - J Luo
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - E S Antonarakis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
12
|
Gomez L, Kovac JR, Lamb DJ. CYP17A1 inhibitors in castration-resistant prostate cancer. Steroids 2015; 95:80-7. [PMID: 25560485 PMCID: PMC4323677 DOI: 10.1016/j.steroids.2014.12.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/14/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
The majority of prostate cancer (PCa) cases are diagnosed as a localized disease. Definitive treatment, active surveillance or watchful waiting are employed as therapeutic paradigms. The current standard of care for the treatment of metastatic PCa is either medical or surgical castration. Once PCa progresses in spite of castrate androgen levels it is termed 'castration-resistant prostate cancer' (CRPC). Patients may even exhibit rising PSA levels with possible bone, lymph node or solid organ metastases. In 2010, the only agent approved for the treatment of CRPC was docetaxel, a chemotherapeutic agent. It is now known that cells from patients with CRPC express androgen receptors (AR) and remain continuously influenced by androgens. As such, treatments with novel hormonal agents that specifically target the biochemical conversion of cholesterol to testosterone have come to the forefront. The use of cytochrome P450c17 (CYP17A1) inhibitor underlies one of the most recent advances in the treatment of CRPC. Abiraterone acetate (AA) was the first CYP17A1 inhibitor approved in the United States. This review will discuss CRPC in general with a specific focus on AA and novel CYP17A1 inhibitors. AA clinical trials will be reviewed along with other novel adjunct treatments that may enhance the effectiveness of abiraterone therapy. Furthermore, the most recently identified CYP17A1 inhibitors Orteronel, Galeterone, VT-464, and CFG920 will also be explored.
Collapse
Affiliation(s)
- Lissette Gomez
- Scott Department of Urology and The Center for Reproductive Medicine, and the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Jason R Kovac
- Urology of Indiana, Male Reproductive Endocrinology and Surgery, Carmel, IN, United States
| | - Dolores J Lamb
- Scott Department of Urology and The Center for Reproductive Medicine, and the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
13
|
Tilli TM, Ferreira LB, Gimba ERP. Osteopontin-c mediates the upregulation of androgen responsive genes in LNCaP cells through PI3K/Akt and androgen receptor signaling. Oncol Lett 2015; 9:1845-1850. [PMID: 25789054 PMCID: PMC4356391 DOI: 10.3892/ol.2015.2939] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 11/27/2014] [Indexed: 11/09/2022] Open
Abstract
Androgen receptor (AR) signaling is a key pathway modulating prostate cancer (PCa) progression. Several steps in this pathway have been investigated in order to propose novel treatment strategies for advanced PCa. Total osteopontin (OPN) has been described as a biomarker for PCa, in addition to its role in activating the progression of this tumor. Based on the known effects of the OPNc splice variant on PCa progression, the present study investigated whether this isoform can also modulate AR signaling. In order to test this, an in vitro model was used in which LNCaP cells were cultured in the presence of conditioned medium (CM) secreted by PCa cells overexpressing OPNc (OPNc-CM). The activation of AR signaling was evaluated by measuring the expression levels of AR-responsive genes (ARGs) using quantitative polymerase chain reaction and specific oligonucleotides. The data demonstrated that all nine tested ARGs (Fgf8, TMPRSS2, Greb1, Cdk2, Ndrg1, Cdk1, Pmepa1, Psa and Ar) are significantly upregulated in response to OPNc-CM compared with LNCaP cells cultured in CM secreted by control cells transfected with empty expression vector. The specific involvement of OPNc was demonstrated by depleting OPNc from OPNc-CM using an anti-OPNc neutralizing antibody. In addition, by using a phosphoinositide 3-kinase (PI3K)-specific inhibitor and AR antagonists, such as flutamide and bicalutamide, it was also observed that upregulation of ARGs in response to OPNc-CM involves PI3K signaling and depends on the AR. In conclusion, these data indicated that OPNc is able to activate AR signaling through the PI3K pathway and the AR. These data further corroborate our previous data, revealing the OPNc splice variant to be a key molecule that is able to modulate key signaling pathways involved in PCa progression.
Collapse
Affiliation(s)
- Tatiana Martins Tilli
- Molecular Carcinogenesis Program, Research Coordination, National Institute of Cancer, Rio de Janeiro 22743-051, Brazil
| | - Luciana Bueno Ferreira
- Institute of Molecular Pathology and Immunology, University of Porto, Porto 4200-465, Portugal
| | - Etel Rodrigues Pereira Gimba
- Molecular Carcinogenesis Program, Research Coordination, National Institute of Cancer, Rio de Janeiro 22743-051, Brazil ; Natural Sciences Department, Health and Humanities Institute, Fluminense Federal University, Rio das Ostras, Rio de Janeiro 28895-532, Brazil
| |
Collapse
|
14
|
Androgen receptor: structure, role in prostate cancer and drug discovery. Acta Pharmacol Sin 2015; 36:3-23. [PMID: 24909511 PMCID: PMC4571323 DOI: 10.1038/aps.2014.18] [Citation(s) in RCA: 572] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/05/2014] [Indexed: 12/15/2022] Open
Abstract
Androgens and androgen receptors (AR) play a pivotal role in expression of the male phenotype. Several diseases, such as androgen insensitivity syndrome (AIS) and prostate cancer, are associated with alterations in AR functions. Indeed, androgen blockade by drugs that prevent the production of androgens and/or block the action of the AR inhibits prostate cancer growth. However, resistance to these drugs often occurs after 2–3 years as the patients develop castration-resistant prostate cancer (CRPC). In CRPC, a functional AR remains a key regulator. Early studies focused on the functional domains of the AR and its crucial role in the pathology. The elucidation of the structures of the AR DNA binding domain (DBD) and ligand binding domain (LBD) provides a new framework for understanding the functions of this receptor and leads to the development of rational drug design for the treatment of prostate cancer. An overview of androgen receptor structure and activity, its actions in prostate cancer, and how structural information and high-throughput screening have been or can be used for drug discovery are provided herein.
Collapse
|
15
|
Androgen receptor splice variants in the era of enzalutamide and abiraterone. Discov Oncol 2014; 5:265-73. [PMID: 25048254 DOI: 10.1007/s12672-014-0190-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/10/2014] [Indexed: 01/24/2023] Open
Abstract
The FDA approvals of enzalutamide and abiraterone have rapidly changed the clinical landscape of prostate cancer treatment. Both drugs were designed to further suppress androgen receptor (AR) signaling, which is restored following first-line androgen deprivation therapies. Resistance to enzalutamide and abiraterone, however, is again marked by a return of AR signaling, indicating a remarkable "addiction" of prostate cancer cells to the AR pathway. Several mechanisms of castration resistance have been uncovered in the past decades, featuring a wide spectrum of molecular alterations that may explain sustained AR signaling in castration-resistant prostate cancers (CRPC). Among these, the androgen receptor splice variants (AR-Vs), particularly variant 7 (AR-V7), have been implicated in resistance to enzalutamide and abiraterone in preclinical studies, and they cannot be targeted by currently available AR-directed drugs. Drug development for AR-V-associated CRPC may therefore be necessary to augment the preexisting treatment repertoire. In this mini-review, we will discuss general mechanisms of resistance to AR-directed therapies, with a focus on the role of androgen receptor splice variants in the new era of treating advanced prostate cancer with enzalutamide and abiraterone.
Collapse
|
16
|
Oskarsson A, Spatafora C, Tringali C, Andersson ÅO. Inhibition of CYP17A1 activity by resveratrol, piceatannol, and synthetic resveratrol analogs. Prostate 2014; 74:839-51. [PMID: 24610083 DOI: 10.1002/pros.22801] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 02/13/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND Resveratrol (RSV) and resveratrol analogs have a potential use in prostate cancer chemoprevention due to effects on for example, cell growth, apoptosis, angiogenesis, and metastasis. However, inhibition of CYP17A1, a key enzyme in the androgen biosynthesis and a target for prostate cancer therapy, has not been explored as a possible mechanism behind the effects on prostate cancer. METHODS Human adrenocortical carcinoma cells, H295R, were treated with RSV, piceatannol (PIC), 3,5,4'-triacetylresveratrol (RSVTA), 3,5-diacetylresveratrol (RSVDA), and 3,5,4'-trimethylresveratrol (RSVTM) for 24 hr at concentrations of 1, 5, 10, 25, and 50 µM. Steroid secretion, enzyme activities, and gene expression of key steps in steroidogenesis were investigated. RESULTS Secretion of dihydroepiandrosterone (DHEA), testosterone, and cortisol were drastically decreased by all test compounds at concentrations that did not affect cell viability. Progesterone and aldosterone secretion were increased. This steroid secretion pattern can be explained by the demonstrated inhibition of CYP17A1 enzyme activity. The most efficient CYP17A1 inhibitors were the synthetic analogs RSVTA, RSVDA, and RSVTM. Inhibition by RSVTM was more selective on the 17,20-lyase activity than hydroxylase activity of CYP17A1. Treatment of cells with all compounds, except RSVTM, caused increased estradiol levels, which could be explained by the demonstrated inhibition of estrogen sulfate conjugation, catalyzed by SULT1E1. CONCLUSIONS Our results on CYP17A1 inhibition of RSV and RSV analogs suggest a novel mechanism for chemoprevention of prostate cancer by resveratrol and the analogs. Especially RSVTM, which has a preferential inhibition on the 17,20-lyase activity of CYP17A1, may be a promising candidate for prostate cancer chemoprevention.
Collapse
Affiliation(s)
- Agneta Oskarsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | | |
Collapse
|
17
|
Ahmed A, Ali S, Sarkar FH. Advances in androgen receptor targeted therapy for prostate cancer. J Cell Physiol 2014; 229:271-6. [PMID: 24037862 DOI: 10.1002/jcp.24456] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/14/2013] [Indexed: 12/31/2022]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Current research findings suggest that the androgen receptor (AR) and its signaling pathway contribute significantly to the progression of metastatic PCa. The AR is a ligand activated transcription factor, where androgens such as testosterone (T) and dihydroxytestosterone (DHT) act as the activating ligands. However in many metastatic PCa, the AR functions promiscuously and is constitutively active through multiple mechanisms. Inhibition of enzymes that take part in androgen synthesis or synthesizing antiandrogens that can inhibit the AR are two popular methods of impeding the androgen receptor signaling axis; however, the inhibition of androgen-independent activated AR function has not yet been fully exploited. This article focuses on the development of emerging novel agents that act at different steps along the androgen-AR signaling pathway to help improve the poor prognosis of PCa patients.
Collapse
Affiliation(s)
- Alia Ahmed
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | | | | |
Collapse
|
18
|
Goyal J, Antonarakis ES. Clinical Evaluation of Abiraterone in the Treatment of Metastatic Prostate Cancer. ACTA ACUST UNITED AC 2013; 2013:1-14. [PMID: 24482578 PMCID: PMC3904375 DOI: 10.4137/cmu.s8337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Treatment of castration-resistant prostate cancer remains an area of unmet medical need. Evidence suggests that this entity continues to be driven by androgens and androgen receptor (AR) signaling. Abiraterone acetate, a pregnenolone derivative, is an oral selective and irreversible inhibitor of the key steroidogenic enzyme CYP17. It possesses dual 17-α hydroxylase and C17,20-lyase blocking activity, the result of which is decreased gonadal and extra-gonadal androgen synthesis. Abiraterone was first approved by the US Food and Drug Administration (FDA) in 2011 following the demonstration of superior survival compared with placebo in the post-docetaxel population. Since that time, more evidence has been generated from preclinical studies and clinical trials which have considerably enhanced our understanding of this complex disease. In this paper, we review the development of abiraterone acetate, its pharmacological characteristics, and its effects on the androgen-AR signaling axis, along with the combined experience from clinical trials. We also discuss some of the ongoing trials using this agent, as well as potential mechanisms of abiraterone resistance, novel bio-marker development, and future directions using AR-directed therapies.
Collapse
Affiliation(s)
- Jatinder Goyal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Emmanuel S Antonarakis
- Assistant Professor of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore MD
| |
Collapse
|
19
|
Lallous N, Dalal K, Cherkasov A, Rennie PS. Targeting alternative sites on the androgen receptor to treat castration-resistant prostate cancer. Int J Mol Sci 2013; 14:12496-519. [PMID: 23771019 PMCID: PMC3709796 DOI: 10.3390/ijms140612496] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 01/22/2023] Open
Abstract
Recurrent, metastatic prostate cancer continues to be a leading cause of cancer-death in men. The androgen receptor (AR) is a modular, ligand-inducible transcription factor that regulates the expression of genes that can drive the progression of this disease, and as a consequence, this receptor is a key therapeutic target for controlling prostate cancer. The current drugs designed to directly inhibit the AR are called anti-androgens, and all act by competing with androgens for binding to the androgen/ligand binding site. Unfortunately, with the inevitable progression of the cancer to castration resistance, many of these drugs become ineffective. However, there are numerous other regulatory sites on this protein that have not been exploited therapeutically. The regulation of AR activity involves a cascade of complex interactions with numerous chaperones, co-factors and co-regulatory proteins, leading ultimately to direct binding of AR dimers to specific DNA androgen response elements within the promoter and enhancers of androgen-regulated genes. As part of the family of nuclear receptors, the AR is organized into modular structural and functional domains with specialized roles in facilitating their inter-molecular interactions. These regions of the AR present attractive, yet largely unexploited, drug target sites for reducing or eliminating androgen signaling in prostate cancers. The design of small molecule inhibitors targeting these specific AR domains is only now being realized and is the culmination of decades of work, including crystallographic and biochemistry approaches to map the shape and accessibility of the AR surfaces and cavities. Here, we review the structure of the AR protein and describe recent advancements in inhibiting its activity with small molecules specifically designed to target areas distinct from the receptor’s androgen binding site. It is anticipated that these new classes of anti-AR drugs will provide an additional arsenal to treat castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Nada Lallous
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | | | | | | |
Collapse
|