1
|
Lee S, Kim HR, Woo Y, Kim J, Kim HW, Park JY, Suh B, Choi Y, Ahn J, Ryu JH, Roe JS, Song J, Lee SH. UBX-390: A Novel Androgen Receptor Degrader for Therapeutic Intervention in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400398. [PMID: 38958553 PMCID: PMC11434238 DOI: 10.1002/advs.202400398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The androgen receptor (AR) is an attractive target for treating prostate cancer, considering its role in the development and progression of localized and metastatic prostate cancer. The high global mortality burden of prostate cancer, despite medical treatments such as androgen deprivation or AR antagonist therapy, highlights the need to explore alternative strategies. One strategy involves the use of heterobifunctional degraders, also known as proteolysis-targeting chimeras, which are novel small-molecule therapeutics that inhibit amplified or mutated targets. Here, the study reports a novel cereblon-based AR degrader, UBX-390, and demonstrates its superior activity over established AR degraders, such as ARV-110 or ARCC-4, in prostate cancer cells under short- and long-term treatment conditions. UBX-390 suppresses chromatin binding and gene expression of AR and demonstrates substantial efficacy in the degradation of AR mutants in patients with treatment-resistant prostate cancer. UBX-390 is presented as an optimized AR degrader with remarkable potential for treating castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Soohyun Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yaejin Woo
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jiyoung Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Han Wool Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Ji Youn Park
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Beomseon Suh
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Yuri Choi
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jungmin Ahn
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Je Ho Ryu
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Song Hee Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| |
Collapse
|
2
|
Ahmad I, Sood S, Al-Daqqaq Z, Bagit A, Maliyar K, Mufti A, Yeung J. Cutaneous Eruptions Associated With Androgen Receptor Inhibitors for Castration-Resistant Prostate Cancer: A Meta-Analysis of Randomized Controlled Trials. J Cutan Med Surg 2024; 28:395-396. [PMID: 38468198 DOI: 10.1177/12034754241239278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Affiliation(s)
- Ihtisham Ahmad
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Siddhartha Sood
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zizo Al-Daqqaq
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ahmed Bagit
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Khalad Maliyar
- Department of Medicine, Division of Dermatology, University of Toronto, Toronto, ON, Canada
| | - Asfandyar Mufti
- Department of Medicine, Division of Dermatology, University of Toronto, Toronto, ON, Canada
- Department of Dermatology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Jensen Yeung
- Department of Medicine, Division of Dermatology, University of Toronto, Toronto, ON, Canada
- Department of Dermatology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Dermatology, Women's College Hospital, Toronto, ON, Canada
- Probity Medical Research, Waterloo, ON, Canada
| |
Collapse
|
3
|
Kumar V, Sari AN, Gupta D, Ishida Y, Terao K, Kaul SC, Vrati S, Sundar D, Wadhwa R. Anti-COVID-19 Potential of Withaferin-A and Caffeic Acid Phenethyl Ester. Curr Top Med Chem 2024; 24:830-842. [PMID: 38279743 DOI: 10.2174/0115680266280720231221100004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/15/2023] [Accepted: 12/05/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND The recent COVID-19 (coronavirus disease 2019) pandemic triggered research on the development of new vaccines/drugs, repurposing of clinically approved drugs, and assessment of natural anti-COVID-19 compounds. Based on the gender difference in the severity of the disease, such as a higher number of men hospitalized and in intense care units, variations in sex hormones have been predicted to play a role in disease susceptibility. Cell surface receptors (Angiotensin-Converting Enzyme 2; ACE2 and a connected transmembrane protease serine 2- TMPSS2) are upregulated by androgens. Conversely, androgen antagonists have also been shown to lower ACE2 levels, implying their usefulness in COVID-19 management. OBJECTIVES In this study, we performed computational and cell-based assays to investigate the anti- COVID-19 potential of Withaferin-A and Caffeic acid phenethyl ester, natural compounds from Withania somnifera and honeybee propolis, respectively. METHODS Structure-based computational approach was adopted to predict binding stability, interactions, and dynamics of the two test compounds to three target proteins (androgen receptor, ACE2, and TMPRSS2). Further, in vitro, cell-based experimental approaches were used to investigate the effect of compounds on target protein expression and SARS-CoV-2 replication. RESULTS Computation and experimental analyses revealed that (i) CAPE, but not Wi-A, can act as androgen antagonist and hence inhibit the transcriptional activation function of androgen receptor, (ii) while both Wi-A and CAPE could interact with ACE2 and TMPRSS2, Wi-A showed higher binding affinity, and (iii) combination of Wi-A and CAPE (Wi-ACAPE) caused strong downregulation of ACE2 and TMPRSS2 expression and inhibition of virus infection. CONCLUSION Wi-A and CAPE possess multimodal anti-COVID-19 potential, and their combination (Wi-ACAPE) is expected to provide better activity and hence warrant further attention in the laboratory and clinic.
Collapse
Affiliation(s)
- Vipul Kumar
- DAILAB, Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, 110 016, India
| | - Anissa Nofita Sari
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305 8565, Japan
| | - Dharmender Gupta
- Regional Centre for Biotechnology (RCB), Faridabad, 121 001, India
| | - Yoshiyuki Ishida
- CycloChem Bio Co., Ltd., 7-4-5 Minatojima-minamimachi, Kobe, 6500047, Japan
| | - Keiji Terao
- CycloChem Bio Co., Ltd., 7-4-5 Minatojima-minamimachi, Kobe, 6500047, Japan
| | - Sunil C Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305 8565, Japan
| | - Sudhanshu Vrati
- Regional Centre for Biotechnology (RCB), Faridabad, 121 001, India
| | - Durai Sundar
- DAILAB, Department of Biochemical Engineering & Biotechnology, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, 110 016, India
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305 8565, Japan
| |
Collapse
|
4
|
Buck SAJ, Meertens M, van Ooijen FMF, Oomen-de Hoop E, de Jonge E, Coenen MJH, Bergman AM, Koolen SLW, de Wit R, Huitema ADR, van Schaik RHN, Mathijssen RHJ. A common germline variant in CYP11B1 is associated with adverse clinical outcome of treatment with abiraterone or enzalutamide. Biomed Pharmacother 2023; 169:115890. [PMID: 37988848 DOI: 10.1016/j.biopha.2023.115890] [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: 08/25/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
Extragonadal androgens play a pivotal role in prostate cancer disease progression on androgen receptor signaling inhibitors (ARSi), including abiraterone and enzalutamide. We aimed to investigate if germline variants in genes involved in extragonadal androgen synthesis contribute to resistance to ARSi and may predict clinical outcomes on ARSi. We included ARSi naive metastatic prostate cancer patients treated with abiraterone or enzalutamide and determined 18 germline variants in six genes involved in extragonadal androgen synthesis. Variants were tested in univariate and multivariable analysis for the relation with overall survival (OS) and time to progression (TTP) by Cox regression, and PSA response by logistic regression. A total of 275 patients were included. From the investigated genes CYP17A1, HSD3B1, CYP11B1, AKR1C3, SRD5A1 and SRD5A2, only rs4736349 in CYP11B1 in homozygous form (TT), present in 54 patients (20%), was related with a significantly worse OS (HR = 1.71, 95% CI 1.09 - 2.68, p = 0.019) and TTP (HR = 1.50, 95% CI 1.08 - 2.09, p = 0.016), and was related with a significantly less frequent PSA response (OR = 0.48, 95% CI 0.24 - 0.96, p = 0.038) on abiraterone or enzalutamide in a multivariable analysis. The frequent germline variant rs4736349 in CYP11B1 is, as homozygote, an independent negative prognostic factor for treatment with abiraterone or enzalutamide in ARSi naive metastatic prostate cancer patients. Our findings warrant prospective investigation of this potentially important predictive biomarker.
Collapse
Affiliation(s)
- Stefan A J Buck
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.
| | - Marinda Meertens
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | | | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Evert de Jonge
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke J H Coenen
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andries M Bergman
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands; Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Morozov VM, Riva A, Sarwar S, Kim WJ, Li J, Zhou L, Licht J, Daaka Y, Ishov A. HIRA-mediated loading of histone variant H3.3 controls androgen-induced transcription by regulation of AR/BRD4 complex assembly at enhancers. Nucleic Acids Res 2023; 51:10194-10217. [PMID: 37638746 PMCID: PMC10602887 DOI: 10.1093/nar/gkad700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/21/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Incorporation of histone variant H3.3 comprises active territories of chromatin. Exploring the function of H3.3 in prostate cancer (PC), we found that knockout (KO) of H3.3 chaperone HIRA suppresses PC growth in vitro and in xenograft settings, deregulates androgen-induced gene expression and alters androgen receptor (AR) binding within enhancers of target genes. H3.3 affects transcription in multiple ways, including activation of p300 by phosphorylated H3.3 at Ser-31 (H3.3S31Ph), which results in H3K27 acetylation (H3K27Ac) at enhancers. In turn, H3K27Ac recruits bromodomain protein BRD4 for enhancer-promoter interaction and transcription activation. We observed that HIRA KO reduces H3.3 incorporation, diminishes H3.3S31Ph and H3K27Ac, modifies recruitment of BRD4. These results suggest that H3.3-enriched enhancer chromatin serves as a platform for H3K27Ac-mediated BRD4 recruitment, which interacts with and retains AR at enhancers, resulting in transcription reprogramming. In addition, HIRA KO deregulates glucocorticoid- (GR) driven transcription of genes co-regulated by AR and GR, suggesting a common H3.3/HIRA-dependent mechanism of nuclear receptors function. Expression of HIRA complex proteins is increased in PC compared with normal prostate tissue, especially in high-risk PC groups, and is associated with a negative prognosis. Collectively, our results demonstrate function of HIRA-dependent H3.3 pathway in regulation of nuclear receptors activity.
Collapse
Affiliation(s)
- Viacheslav M Morozov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alberto Riva
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, USA
| | - Sadia Sarwar
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Wan-Ju Kim
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jianping Li
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lei Zhou
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Yehia Daaka
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Alexander M Ishov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| |
Collapse
|
6
|
Elshazly AM, Gewirtz DA. Making the Case for Autophagy Inhibition as a Therapeutic Strategy in Combination with Androgen-Targeted Therapies in Prostate Cancer. Cancers (Basel) 2023; 15:5029. [PMID: 37894395 PMCID: PMC10605431 DOI: 10.3390/cancers15205029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Androgen receptor targeting remains the primary therapeutic strategy in prostate cancer, encompassing androgen biosynthesis inhibitors and androgen receptor antagonists. While both androgen-receptor-positive and "castration-resistant" prostate cancer are responsive to these approaches, the development of resistance is an almost inevitable outcome leading to the castration-resistant form of the disease. Given that "cytoprotective" autophagy is considered to be a predominant mechanism of resistance to various chemotherapeutic agents as well as to radiation in the cancer literature, the purpose of this review is to evaluate whether autophagy plays a central role in limiting the utility of androgen deprivation therapies in prostate cancer. Unlike most of our previous reports, where multiple functional forms of autophagy were identified, making it difficult if not impossible to propose autophagy inhibition as a therapeutic strategy, the cytoprotective form of autophagy appears to predominate in the case of androgen deprivation therapies. This opens a potential pathway for improving the outcomes for prostate cancer patients once effective and reliable pharmacological autophagy inhibitors have been developed.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
7
|
Venugopal S, Kaur B, Verma A, Wadhwa P, Magan M, Hudda S, Kakoty V. Recent advances of benzimidazole as anticancer agents. Chem Biol Drug Des 2023; 102:357-376. [PMID: 37009821 DOI: 10.1111/cbdd.14236] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/20/2023] [Accepted: 03/14/2023] [Indexed: 04/04/2023]
Abstract
Cancer is the second leading cause of death globally, with 9.6 million deaths yearly. As a life-threatening disease, it necessitates the emergence of new therapies. Resistance to current chemotherapies drives scientists to develop new medications that will eventually be accessible. Because heterocycles are so common in biological substances, compounds play a big part in the variety of medications that have been developed. The "Master Key" is the benzimidazole nucleus, which consists of a six-membered benzene ring fused with a five-membered imidazole/imidazoline ring, which is an azapyrrole. One of the five-membered aromatic nitrogen heterocycles identified in American therapies that have been approved by the Food and Drug Administration (FDA). Our results show that benzimidazole's broad therapeutic spectrum is due to its structural isosteres with purine, which improves hydrogen bonding, electrostatic interactions with topoisomerase complexes, intercalation with DNA, and other functions. It also enhances protein and nucleic acid inhibition, tubulin microtubule degeneration, apoptosis, DNA fragmentation, and other functions. Additionally, readers for designing the more recent benzimidazole analogues as prospective cancer treatments.
Collapse
Affiliation(s)
- Sneha Venugopal
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Balwinder Kaur
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Anil Verma
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Pankaj Wadhwa
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Muskan Magan
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Sharwan Hudda
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| | - Violina Kakoty
- Department of Pharmaceutical Sciences, School of Pharmacy, Lovely Professional University, Punjab, India
| |
Collapse
|
8
|
Morozov VM, Riva A, Sarwar S, Kim W, Li J, Zhou L, Licht JD, Daaka Y, Ishov AM. HIRA-mediated loading of histone variant H3.3 controls androgen-induced transcription by regulation of AR/BRD4 complex assembly at enhancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.536256. [PMID: 37214820 PMCID: PMC10197601 DOI: 10.1101/2023.05.08.536256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Incorporation of histone variant H3.3 comprises active territories of chromatin. Exploring the function of H3.3 in prostate cancer (PC), we found that knockout (KO) of H3.3 chaperone HIRA suppresses PC growth in vitro and in xenograft settings, deregulates androgen-induced gene expression and alters androgen receptor (AR) binding within enhancers of target genes. H3.3 affects transcription in multiple ways, including activation of p300 by phosphorylated H3.3 at Ser-31 (H3.3S31Ph), which results in H3K27 acetylation (H3K27Ac) at enhancers. In turn, H3K27Ac recruits bromodomain protein BRD4 for enhancer-promoter interaction and transcription activation. We observed that HIRA KO reduces H3.3 incorporation, diminishes H3.3S31Ph and H3K27Ac, modifies recruitment of BRD4. These results suggest that H3.3-enriched enhancer chromatin serves as a platform for H3K27Ac-mediated BRD4 recruitment, which interacts with and retains AR at enhancers, resulting in transcription reprogramming. AR KO reduced levels of H3.3 at enhancers, indicating feedback mechanism. In addition, HIRA KO deregulates glucocorticoid-driven transcription, suggesting a common H3.3/HIRA-dependent mechanism of nuclear receptors function. Expression of HIRA complex proteins is increased in PC compared with normal prostate tissue, especially in high-risk PC groups, and is associated with a negative prognosis. Collectively, our results demonstrate function of HIRA-dependent H3.3 pathway in regulation of nuclear receptors activity. Key points *H3.3 at enhancers promotes acetylation of H3K27Ac and retention of AR/BRD4 complex for transcription regulation*Knockout of H3.3 chaperone HIRA suppresses PC cells growth and deregulates androgen-induced transcription*H3.3/HIRA pathway regulates both AR and GR, suggesting a common HIRA/H3.3 mechanism of nuclear receptors function.
Collapse
|
9
|
Fleming B, Edison P, Kenny L. Cognitive impairment after cancer treatment: mechanisms, clinical characterization, and management. BMJ 2023; 380:e071726. [PMID: 36921926 DOI: 10.1136/bmj-2022-071726] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is a debilitating side effect experienced by patients with cancer treated with systemically administered anticancer therapies. With around 19.3 million new cases of cancer worldwide in 2020 and the five year survival rate growing from 50% in 1970 to 67% in 2013, an urgent need exists to understand enduring side effects with severe implications for quality of life. Whereas cognitive impairment associated with chemotherapy is recognized in patients with breast cancer, researchers have started to identify cognitive impairment associated with other treatments such as immune, endocrine, and targeted therapies only recently. The underlying mechanisms are diverse and therapy specific, so further evaluation is needed to develop effective therapeutic interventions. Drug and non-drug management strategies are emerging that target mechanistic pathways or the cognitive deficits themselves, but they need to be rigorously evaluated. Clinically, consistent use of objective diagnostic tools is necessary for accurate diagnosis and clinical characterization of cognitive impairment in patients treated with anticancer therapies. This should be supplemented with clinical guidelines that could be implemented in daily practice. This review summarizes the recent advances in the mechanisms, clinical characterization, and novel management strategies of cognitive impairment associated with treatment of non-central nervous system cancers.
Collapse
Affiliation(s)
- Ben Fleming
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
10
|
Wang YW, Lan L, Wang M, Zhang JY, Gao YH, Shi L, Sun LP. PROTACS: A technology with a gold rush-like atmosphere. Eur J Med Chem 2023; 247:115037. [PMID: 36566716 DOI: 10.1016/j.ejmech.2022.115037] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/03/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Abnormally expressed or malfunctioning proteins may affect or even damage cells, leading to the onset of diseases. Proteolysis targeting chimera (PROTAC) technology has been proven to be a fresh therapeutic strategy, superior to conventional small molecule inhibitors for the treatment of diseases caused by pathogenic proteins. Unlike conventional small molecule inhibitors that are occupancy-driven, PROTACs are heterobifunctional small molecules with catalytic properties. They combine with E3 ligases and target proteins to form a ternary complex, rendering the target protein ubiquitous and subsequently degraded by the proteasome. This paper focuses first on significant events in the development of PROTAC technology from 2001 to 2022, followed by a brief overview of various PROTACs categorized by target proteins. In addition, the applications of PROTACs in the treatment of diseases and fundamental biology are also under discussion.
Collapse
Affiliation(s)
- Yu-Wei Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Li Lan
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Min Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Jin-Yang Zhang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yu-Hui Gao
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Lei Shi
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Li-Ping Sun
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
11
|
Gerald T, Raj G. Testosterone and the Androgen Receptor. Urol Clin North Am 2022; 49:603-614. [DOI: 10.1016/j.ucl.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
12
|
Khan A, Mao Y, Tahreem S, Wei DQ, Wang Y. Structural and molecular insights into the mechanism of resistance to enzalutamide by the clinical mutants in androgen receptor (AR) in castration-resistant prostate cancer (CRPC) patients. Int J Biol Macromol 2022; 218:856-865. [PMID: 35905763 DOI: 10.1016/j.ijbiomac.2022.07.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
Abstract
Androgen receptor (AR) is a key contributing element in the prostate cancer (PCa) instigation, progression and it is among the vastly discovered target for prostate cancer. Numerous mechanisms trigger the expansion of CRPC among which the aberrant AR gene is considered as the prime factor. Recently three essential substitutions H875Y, F877L, and T878A are reported to cause resistance to Enzalutamide. However, no detailed study is available to explore the key events that contribute to the resistance. Hence, considering the applicability of structural bioinformatics and molecular simulation-based methods in the current study, we assessed the impact of these mutations on the binding of Enzalutamide. Using a long-run simulation approach the binding stability, residues flexibility, hydrogen bonding, and protein compactness for each complex were determined to reveal the dynamic variations induced by these mutations. We discovered that the binding mode of Enzalutamide is altered by these mutations which misstarget the key residues required for the antagonistic activity. Molecular simulation of each complex revealed that the wild type H11 and H12 are more flexible moving outside and provides more volume for the ligand optimization. In the mutant complexes, the H12 remained tighter pushing out enzalutamide from the key residues which then essentially misstarget the correct orientation for the antagonist activity. The binding free energy (BFE) for the wild type was computed to be -59.92 ± 0.18 kcal/mol, for H875Y the BFE was -55.92 ± 0.18 kcal/mol, -54.82 ± 0.15 kcal/mol for F877L and -53.87 ± 0.18 kcal/mol for T878A, which further demonstrate that these mutations have destabilized the binding of enzalutamide. The proteins' motion and FEL further validated the aforementioned findings where the wild type reported different dynamic features than the mutant complexes. In conclusion, this study provides a structural basis for the resistance to Enzalutamide, which can be used to design novel effective drugs using structure-based and rationale approaches.
Collapse
Affiliation(s)
- Abbas Khan
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yuanshen Mao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, PR China
| | - Sana Tahreem
- Sharif Medical and Dental College Lahore, Pakistan
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China; State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint Laboratory of International Laboratory of Metabolic and Developmental Sciences, Ministry of Education and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yanjing Wang
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
13
|
Choi YH, Kim J, Shin JY, Kang NG, Lee S. Antiandrogenic activity of Riboflavin 5'-phosphate (FMN) in 22Rv1 and LNCaP human prostate cancer cell lines. Eur J Pharmacol 2022; 917:174743. [PMID: 34998793 DOI: 10.1016/j.ejphar.2022.174743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/29/2021] [Accepted: 01/03/2022] [Indexed: 11/03/2022]
Abstract
The androgen receptor is a hormone activated transcription factor that regulates the development and maintenance of male characteristics and represents one of the most well-established drug targets, being implicated not only in prostate cancer but also in many non-cancerous human diseases including androgenetic alopecia, acne vulgaris, and hirsutism. In this study, the antiandrogenic effects of FMN were investigated in 22Rv1 and LNCaP prostate cancer cells. FMN inhibited dihydrotestosterone (DHT)-induced protein expression of androgen receptor in 22Rv1cells. In another prostate cancer LNCaP cells, FMN decreased the protein level of DHT-induced prostate specific antigen (PSA). In addition, FMN downregulated DHT-induced mRNA expression of androgen regulated genes in both cell lines, showing less prominent inhibition in 22Rv1cells where androgen receptor had been significantly decreased by FMN. FMN was found to bind androgen receptor, demonstrating that it acted as a competitive androgen receptor antagonist. FMN increased the phosphorylation of Akt in 22Rv1 cells and this increment was abrogated by PI3K inhibitor wortmannin, resulting in a rescued androgen receptor protein level which was decreased by FMN. Additionally, FMN was found to increase the mRNA and protein level of E3 ligase mouse double minute 2. Our data suggest that the androgen receptor signaling is regulated through PI3K-Akt-MDM2 pathway in 22Rv1 cells. Together, our results indicate that FMN facilitated the degradation of androgen receptor in 22Rv1 cells and inhibited the expression of androgen regulated genes by competing the binding of DHT to androgen receptor in LNCaP cells, demonstrating its therapeutic potential as an antiandrogen.
Collapse
Affiliation(s)
- Yun-Ho Choi
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul, 07795, Republic of Korea
| | - Jaeyoon Kim
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul, 07795, Republic of Korea
| | - Jae Young Shin
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul, 07795, Republic of Korea
| | - Nae-Gyu Kang
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul, 07795, Republic of Korea
| | - Sanghwa Lee
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul, 07795, Republic of Korea.
| |
Collapse
|
14
|
Harris AE, Metzler VM, Lothion-Roy J, Varun D, Woodcock CL, Haigh DB, Endeley C, Haque M, Toss MS, Alsaleem M, Persson JL, Gudas LJ, Rakha E, Robinson BD, Khani F, Martin LM, Moyer JE, Brownlie J, Madhusudan S, Allegrucci C, James VH, Rutland CS, Fray RG, Ntekim A, de Brot S, Mongan NP, Jeyapalan JN. Exploring anti-androgen therapies in hormone dependent prostate cancer and new therapeutic routes for castration resistant prostate cancer. Front Endocrinol (Lausanne) 2022; 13:1006101. [PMID: 36263323 PMCID: PMC9575553 DOI: 10.3389/fendo.2022.1006101] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Androgen deprivation therapies (ADTs) are important treatments which inhibit androgen-induced prostate cancer (PCa) progression by either preventing androgen biosynthesis (e.g. abiraterone) or by antagonizing androgen receptor (AR) function (e.g. bicalutamide, enzalutamide, darolutamide). A major limitation of current ADTs is they often remain effective for limited durations after which patients commonly progress to a lethal and incurable form of PCa, called castration-resistant prostate cancer (CRPC) where the AR continues to orchestrate pro-oncogenic signalling. Indeed, the increasing numbers of ADT-related treatment-emergent neuroendocrine-like prostate cancers (NePC), which lack AR and are thus insensitive to ADT, represents a major therapeutic challenge. There is therefore an urgent need to better understand the mechanisms of AR action in hormone dependent disease and the progression to CRPC, to enable the development of new approaches to prevent, reverse or delay ADT-resistance. Interestingly the AR regulates distinct transcriptional networks in hormone dependent and CRPC, and this appears to be related to the aberrant function of key AR-epigenetic coregulator enzymes including the lysine demethylase 1 (LSD1/KDM1A). In this review we summarize the current best status of anti-androgen clinical trials, the potential for novel combination therapies and we explore recent advances in the development of novel epigenetic targeted therapies that may be relevant to prevent or reverse disease progression in patients with advanced CRPC.
Collapse
Affiliation(s)
- Anna E. Harris
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Veronika M. Metzler
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Jennifer Lothion-Roy
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Dhruvika Varun
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Corinne L. Woodcock
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Daisy B. Haigh
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Chantelle Endeley
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Maria Haque
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Michael S. Toss
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Mansour Alsaleem
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- Department of Applied Medical Science, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Jenny L. Persson
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Department of Biomedical Sciences, Malmö Universitet, Malmö, Sweden
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Emad Rakha
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Brian D. Robinson
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
| | - Francesca Khani
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
| | - Laura M. Martin
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Jenna E. Moyer
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Juliette Brownlie
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Srinivasan Madhusudan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Cinzia Allegrucci
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Victoria H. James
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Catrin S. Rutland
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Rupert G. Fray
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Atara Ntekim
- Department of Oncology, University Hospital Ibadan, Ibadan, Nigeria
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| | - Simone de Brot
- Comparative Pathology Platform (COMPATH), Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Nigel P. Mongan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| | - Jennie N. Jeyapalan
- University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom
- *Correspondence: Jennie N. Jeyapalan, ; Nigel P. Mongan, ; ; Atara Ntekim,
| |
Collapse
|
15
|
Pang JP, Shen C, Zhou WF, Wang YX, Shan LH, Chai X, Shao Y, Hu XP, Zhu F, Zhu DY, Xiao L, Xu L, Xu XH, Li D, Hou TJ. Discovery of novel antagonists targeting the DNA binding domain of androgen receptor by integrated docking-based virtual screening and bioassays. Acta Pharmacol Sin 2022; 43:229-239. [PMID: 33767381 PMCID: PMC8724294 DOI: 10.1038/s41401-021-00632-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/24/2021] [Indexed: 02/01/2023] Open
Abstract
Androgen receptor (AR), a ligand-activated transcription factor, is a master regulator in the development and progress of prostate cancer (PCa). A major challenge for the clinically used AR antagonists is the rapid emergence of resistance induced by the mutations at AR ligand binding domain (LBD), and therefore the discovery of novel anti-AR therapeutics that can combat mutation-induced resistance is quite demanding. Therein, blocking the interaction between AR and DNA represents an innovative strategy. However, the hits confirmed targeting on it so far are all structurally based on a sole chemical scaffold. In this study, an integrated docking-based virtual screening (VS) strategy based on the crystal structure of the DNA binding domain (DBD) of AR was conducted to search for novel AR antagonists with new scaffolds and 2-(2-butyl-1,3-dioxoisoindoline-5-carboxamido)-4,5-dimethoxybenzoicacid (Cpd39) was identified as a potential hit, which was competent to block the binding of AR DBD to DNA and showed decent potency against AR transcriptional activity. Furthermore, Cpd39 was safe and capable of effectively inhibiting the proliferation of PCa cell lines (i.e., LNCaP, PC3, DU145, and 22RV1) and reducing the expression of the genes regulated by not only the full-length AR but also the splice variant AR-V7. The novel AR DBD-ARE blocker Cpd39 could serve as a starting point for the development of new therapeutics for castration-resistant PCa.
Collapse
Affiliation(s)
- Jin-Ping Pang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chao Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Fang Zhou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yun-Xia Wang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lu-Hu Shan
- Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xin Chai
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Shao
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xue-Ping Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng Zhu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dan-Yan Zhu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Li Xiao
- School of Life Science, Huzhou University, Huzhou, 313000, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Xiao-Hong Xu
- Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Dan Li
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Lab of CAD & CG, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Carlsson SV, Murata K, Danila DC, Lilja H. PSA: role in screening and monitoring patients with prostate cancer. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00001-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
17
|
Hua C, Chen J, Li S, Zhou J, Fu J, Sun W, Wang W. KDM6 Demethylases and Their Roles in Human Cancers. Front Oncol 2021; 11:779918. [PMID: 34950587 PMCID: PMC8688854 DOI: 10.3389/fonc.2021.779918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/17/2021] [Indexed: 12/31/2022] Open
Abstract
Cancer therapy is moving beyond traditional chemotherapy to include epigenetic approaches. KDM6 demethylases are dynamic regulation of gene expression by histone demethylation in response to diverse stimuli, and thus their dysregulation has been observed in various cancers. In this review, we first briefly introduce structural features of KDM6 subfamily, and then discuss the regulation of KDM6, which involves the coordinated control between cellular metabolism (intrinsic regulators) and tumor microenvironment (extrinsic stimuli). We further describe the aberrant functions of KDM6 in human cancers, acting as either a tumor suppressor or an oncoprotein in a context-dependent manner. Finally, we propose potential therapy of KDM6 enzymes based on their structural features, epigenetics, and immunomodulatory mechanisms, providing novel insights for prevention and treatment of cancers.
Collapse
Affiliation(s)
- Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | | | - Shuting Li
- Wenzhou Medical University, Wenzhou, China
| | | | - Jiahong Fu
- Wenzhou Medical University, Wenzhou, China
| | - Weijian Sun
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenqian Wang
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Leblanc E, Ban F, Cavga AD, Lawn S, Huang CCF, Mohan S, Chang MEK, Flory MR, Ghaidi F, Lingadahalli S, Chen G, Yu IPL, Morin H, Lallous N, Gleave ME, Mohammed H, Young RN, Rennie PS, Lack NA, Cherkasov A. Development of 2-(5,6,7-Trifluoro-1 H-Indol-3-yl)-quinoline-5-carboxamide as a Potent, Selective, and Orally Available Inhibitor of Human Androgen Receptor Targeting Its Binding Function-3 for the Treatment of Castration-Resistant Prostate Cancer. J Med Chem 2021; 64:14968-14982. [PMID: 34661404 DOI: 10.1021/acs.jmedchem.1c00681] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) patients undergoing androgen deprivation therapy almost invariably develop castration-resistant prostate cancer (CRPC). Targeting the androgen receptor (AR) Binding Function-3 (BF3) site offers a promising option to treat CRPC. However, BF3 inhibitors have been limited by poor potency or inadequate metabolic stability. Through extensive medicinal chemistry, molecular modeling, and biochemistry, we identified 2-(5,6,7-trifluoro-1H-Indol-3-yl)-quinoline-5-carboxamide (VPC-13789), a potent AR BF3 antagonist with markedly improved pharmacokinetic properties. We demonstrate that VPC-13789 suppresses AR-mediated transcription, chromatin binding, and recruitment of coregulatory proteins. This novel AR antagonist selectively reduces the growth of both androgen-dependent and enzalutamide-resistant PCa cell lines. Having demonstrated in vitro efficacy, we developed an orally bioavailable prodrug that reduced PSA production and tumor volume in animal models of CRPC with no observed toxicity. VPC-13789 is a potent, selective, and orally bioavailable antiandrogen with a distinct mode of action that has a potential as novel CRPC therapeutics.
Collapse
Affiliation(s)
- Eric Leblanc
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Fuqiang Ban
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Ayse Derya Cavga
- School of Medicine, Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey
| | - Sam Lawn
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Chia-Chi Flora Huang
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Sankar Mohan
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Matthew E K Chang
- Knight Cancer Institute, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Mark R Flory
- Knight Cancer Institute, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Fariba Ghaidi
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Shreyas Lingadahalli
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Gang Chen
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Ivan Pak Lok Yu
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Hélène Morin
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Nada Lallous
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Hisham Mohammed
- Knight Cancer Institute, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Robert N Young
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Paul S Rennie
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Nathan A Lack
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada.,School of Medicine, Koç University, Rumelifeneri Yolu, Istanbul 34450, Turkey
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|
19
|
Influence of Androgen Receptor Antagonist MDV3100 Therapy on Rats With Benign Prostatic Hyperplasia. Int Neurourol J 2021; 25:219-228. [PMID: 34610715 PMCID: PMC8497737 DOI: 10.5213/inj.2142004.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/15/2021] [Indexed: 11/08/2022] Open
Abstract
Purpose To probe the effect and mechanism of androgen receptor antagonist MDV3100 on benign prostatic hyperplasia (BPH) of rats Methods BPH rat model was induced by testosterone propionate. Then antagomir-miR-21-3p or agomir-miR-21-3p was injected into rats before MDV3100 treatment. The prostate index was measured by weighing the wet weight of the rat prostate. The structural morphology of rat prostate was observed after hematoxylin & eosin staining. Immunohistochemistry was applied to evaluate the expression levels of Ki-6 and inflammatory cytokines (interleukin [IL]-6, IL-18, and tumor necrosis factor-α) in rat prostate tissues. Quantitative reverse transcription polymerase chain reaction was utilized for assessment of miR-21-3p expression, and Western blot for the performance of the phosphorylation levels of IKKα and p65. Results Injection of testosterone propionate caused increased prostate gland hyperplasia, heightened miR-21-3p level, and activated nuclear factor-kappa B (NF-κB) signaling pathway. Additionally, BPH was accompanied by inflammatory response, as evidenced by enhanced expressions of Ki-67 and inflammatory cytokines. MDV3100 exposure ameliorated BPH and suppressed miR-21-3p expression. Overexpression of miR-21-3p intensified BPH and inflammation level, while knockdown of miR-21-3p relieved BPH. The coeffect of miR-21-3p upregulation and MDV3100 subjection led to higher inflammatory response, elevated phosphorylation levels of IKKα and p65 than MDV3100 treatment alone. Conclusions Androgen receptor antagonist MDV3100 alleviates BPH and inflammatory response through miR-21-3p downregulation and NF-κB signaling pathway blockade.
Collapse
|
20
|
Xiang W, Zhao L, Han X, Qin C, Miao B, McEachern D, Wang Y, Metwally H, Kirchhoff PD, Wang L, Matvekas A, He M, Wen B, Sun D, Wang S. Discovery of ARD-2585 as an Exceptionally Potent and Orally Active PROTAC Degrader of Androgen Receptor for the Treatment of Advanced Prostate Cancer. J Med Chem 2021; 64:13487-13509. [PMID: 34473519 PMCID: PMC8855934 DOI: 10.1021/acs.jmedchem.1c00900] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We report herein the discovery of exceptionally potent and orally bioavailable PROTAC AR degraders with ARD-2585 being the most promising compound. ARD-2585 achieves DC50 values of ≤0.1 nM in the VCaP cell line with AR gene amplification and in the LNCaP cell line carrying an AR mutation. It potently inhibits cell growth with IC50 values of 1.5 and 16.2 nM in the VCaP and LNCaP cell lines, respectively, and achieves excellent pharmacokinetics and 51% of oral bioavailability in mice. It is more efficacious than enzalutamide in inhibition of VCaP tumor growth and does not cause any sign of toxicity in mice. ARD-2585 is a promising AR degrader for extensive investigations for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Weiguo Xiang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lijie Zhao
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xin Han
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chong Qin
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bukeyan Miao
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul D Kirchhoff
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aleksas Matvekas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Miao He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
21
|
Konoshenko MY, Bryzgunova OE, Laktionov PP. miRNAs and androgen deprivation therapy for prostate cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188625. [PMID: 34534639 DOI: 10.1016/j.bbcan.2021.188625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Androgen deprivation therapy (ADT) is mainly used for the treatment of advanced, metastatic or recurrent prostate cancer (PCa). However, patients progress to ADT resistance and castration-resistant prostate cancer (CRPC) with a poor prognosis. Reliable validated markers of ADT resistance with proven clinical utility are necessary for timely correction of the therapy as well as for improvement of patient quality of life. MiRNAs involved in the ADT response and CRPC development via multiple mechanisms may act as biomarkers for patient outcomes. Available data on miRNAs associated with the ADT response (resistance and sensitivity) are summarized and analyzed in the manuscript, including analyses using bioinformatics resources. Molecular targets of miRNAs, as well as reciprocal relations between miRNAs and their targets, were studied using different databases. Special attention was dedicated to the mechanisms of ADT resistance and CRPC development, including testosterone, PI3K-AKT, VEGF pathways and associated genes. Several different approaches can be used to search for miRNAs associated with the ADT response, each of which focuses on the associated set of miRNAs - potential markers of ADT. The intersection of these approaches and combined analysis allowed us to select the most promising miRNA markers of the ADT response. Meta-analysis of the current data indicated that the selected 5 miRNAs (miRNAs - 125b, miR-21, miR-23b, miR-27b and miR-221) and 14 genes are involved in the regulation of key processes of CRPC development and represent the most promising predictors of the ADT response, further demonstrating their potential in combination therapy for advanced PCa.
Collapse
Affiliation(s)
- Maria Yu Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia.
| | - Olga E Bryzgunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Pavel P Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
22
|
Spratt DE, Shore N, Sartor O, Rathkopf D, Olivier K. Treating the patient and not just the cancer: therapeutic burden in prostate cancer. Prostate Cancer Prostatic Dis 2021; 24:647-661. [PMID: 33603236 PMCID: PMC8384628 DOI: 10.1038/s41391-021-00328-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prostate cancer (PC) is a leading cause of death in older men. Androgen deprivation therapy (ADT) is considered the standard-of-care for men with locally advanced disease. However, continuous androgen ablation is associated with acute and long-term adverse effects and most patients will eventually develop castration-resistant PC (CRPC). The recent approval of three, second-generation androgen receptor inhibitors (ARIs), apalutamide, enzalutamide, and darolutamide, has transformed the treatment landscape of PC. Treatment with these second-generation ARIs have produced positive trends in metastasis-free survival, progression-free survival, and overall survival. For patients with non-metastatic CRPC, who are mainly asymptomatic from their disease, maintaining quality of life is a major objective when prescribing therapy. Polypharmacy for age-related comorbidities also is common in this population and may increase the potential for drug-drug interactions (DDIs). METHOD This review summarizes the multiple factors that may contribute to the therapeutic burden of patients with CRPC, including the interplay between age, comorbidities, concomitant medications, the use of ARIs, and financial distress. CONCLUSIONS As the treatment landscape in PC continues to rapidly evolve, consideration must be given to the balance between therapeutic benefits and potential treatment-emergent adverse events that may be further complicated by DDIs with concomitant medications. Patient-centered communication is a crucial aspect of alleviating this burden, and healthcare professionals (HCPs) may benefit from training in effective patient communication. HCPs should closely and frequently monitor patient treatment responses, in order to better understand symptom onset and exacerbation. Patients also should be encouraged to participate in exercise programs, and health information and support groups, which may assist them in preventing or mitigating certain determinants of the therapeutic burden associated with PC and its management.
Collapse
Affiliation(s)
| | - Neal Shore
- Carolina Urologic Research Center, Atlantic Urology Clinics, Myrtle Beach, SC, USA
| | - Oliver Sartor
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kara Olivier
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
23
|
Singh R, Alsayadi YMMA, Singh V, Chawla P, Rawal RK. Prospects of Treating Prostate Cancer through Apalutamide: A Mini-Review. Anticancer Agents Med Chem 2021; 22:1056-1067. [PMID: 34431470 DOI: 10.2174/1871520621666210824113736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Prostate cancer is considered the second most diagnosed cancer, and one of the most common causes of death from cancer in men. Apalutamide is an effective, safe, and well-tolerated agent used for the treatment of men with non-metastatic castration-resistant prostate cancer (nmCRPC) and metastatic hormone-naive prostate cancer (mHNPC). Androgen receptor signaling is a leading factor that drives these prostate tumors. USFDA has approved apalutamide on 14 February 2018 as an agent that targets androgen receptor signaling through inhibition causing significant improvement in metastasis-free survival in patients with prostate cancer. <P> Objective: In this review, various aspects related to apalutamide have been summarized which involve the mechanism of action, chemistry, synthesis, pharmacokinetics, pharmacodynamics, adverse reactions, and safety parameters. <P> Methods: The literature was thoroughly searched in the relevant databases to identify studies published in this field during recent years. Special attention has been given to apalutamide clinical trials phases and its promising future as one of the first-line agents for the treatment of patients with advanced prostate cancer. <P> Results: Ongoing trials are progressing for apalutamide monotherapy and also for its combinations in other disease settings. The expected results of such trials will shape the future scenario of prostate cancer therapy. <P> Conclusion: This review article has highlighted different aspects of Apalutamide like its mechanism of action, adverse effects, pharmacokinetics, pharmacodynamics, clinical trials among others. The contents of this article should make an excellent read for prospective researchers in this field.
Collapse
Affiliation(s)
- Ranapartap Singh
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy, GT Road, Moga-142001, Punjab. India
| | - Yunes M M A Alsayadi
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy, GT Road, Moga-142001, Punjab. India
| | - VikramJeet Singh
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy, GT Road, Moga-142001, Punjab. India
| | - Pooja Chawla
- Department of Pharmaceutical Chemistry & Analysis, ISF College of Pharmacy, GT Road, Moga-142001, Punjab. India
| | - Ravindra Kumar Rawal
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana-133207, Haryana. India
| |
Collapse
|
24
|
Nicola C, Dubois M, Campart C, Al Sagheer T, Desrues L, Schapman D, Galas L, Lange M, Joly F, Castel H. The Prostate Cancer Therapy Enzalutamide Compared with Abiraterone Acetate/Prednisone Impacts Motivation for Exploration, Spatial Learning and Alters Dopaminergic Transmission in Aged Castrated Mice. Cancers (Basel) 2021; 13:cancers13143518. [PMID: 34298734 PMCID: PMC8304001 DOI: 10.3390/cancers13143518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Cognitive side effects and fatigue after cancer treatment now constitute a major challenge in oncology. Abiraterone acetate plus prednisone (AAP) and enzalutamide (ENZ) are next-generation therapies improving metastatic castration-resistant prostate cancer (mCRPC) patient survival, but also associated with neurological disturbances. We developed a behavioral 17 months-aged and castrated mouse model receiving AAP or ENZ for 5 days per week for six weeks. We establish that ENZ impacts locomotor and explorative behaviors, and strength capacity likely by preventing binding of central synthetized androgens to androgen receptors expressed by dopamine neurons of the Substantia Nigra and the Ventral Tegmentum. ENZ also reduces the cognitive score, associated with less neuronal activity in dorsal hippocampal areas. This demonstrates ENZ-specific consequences on motivation to exploration and cognition, being of particular importance for future management of elderly prostate cancer patients and their quality of life. Abstract Cognitive side effects after cancer treatment threatening quality of life (QoL) constitute a major challenge in oncology. Abiraterone acetate plus prednisone (AAP) and enzalutamide (ENZ) are examples of next-generation therapy (NGT) administered to metastatic castration-resistant prostate cancer (mCRPC) patients. NGT significantly improved mCRPC overall survival but neurological side effects such as fatigue and cognitive impairment were reported. We developed a behavioral 17 months-aged and castrated mouse model receiving per os AAP or ENZ for 5 days per week for six consecutive weeks. ENZ exposure reduced spontaneous activity and exploratory behavior associated with a decreased tyrosine hydroxylase (TH)-dopaminergic activity in the substantia nigra pars compacta and the ventral tegmental area. A decrease in TH+-DA afferent fibers and Phospho-DARPP32-related dopaminergic neuronal activities in the striatum and the ventral hippocampus highlighted ENZ-induced dopaminergic regulation within the nigrostriatal and mesolimbocortical pathways. ENZ and AAP treatments did not substantially modify spatial learning and memory performances, but ENZ led to a thygmotaxis behavior impacting the cognitive score, and reduced c-fos-related activity of NeuN+-neurons in the dorsal hippocampus. The consequences of the mCRPC treatment ENZ on aged castrated mouse motivation to exploration and cognition should make reconsider management strategy of elderly prostate cancer patients.
Collapse
Affiliation(s)
- Celeste Nicola
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
| | - Martine Dubois
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
| | - Cynthia Campart
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
| | - Tareq Al Sagheer
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
| | - Laurence Desrues
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
| | - Damien Schapman
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Normandie University, UNIROUEN, INSERM, PRIMACEN, 76000 Rouen, France
| | - Ludovic Galas
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Normandie University, UNIROUEN, INSERM, PRIMACEN, 76000 Rouen, France
| | - Marie Lange
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
- Centre François Baclesse, Clinical Research Department, 14000 Caen, France
- Normandie University, UNICAEN, INSERM, U1086 ANTICIPE, 14000 Caen, France
| | - Florence Joly
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
- Centre François Baclesse, Clinical Research Department, 14000 Caen, France
- Normandie University, UNICAEN, INSERM, U1086 ANTICIPE, 14000 Caen, France
- University Hospital of Caen, 14000 Caen, France
| | - Hélène Castel
- Normandie University, UNIROUEN, INSERM, U1239 DC2N, 76000 Rouen, France; (C.N.); (M.D.); (C.C.); (T.A.S.); (L.D.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France; (D.S.); (L.G.)
- Cancer and Cognition Platform, Ligue Nationale contre le Cancer, 14000 Caen, France; (M.L.); (F.J.)
- Normandie University, UNIROUEN, INSERM, DC2N, Team Astrocyte and Vascular Niche, Place Emile Blondel, CEDEX, 76821 Mont-Saint-Aignan, France
- Correspondence: ; Tel.: +33-2-35-14-66-23
| |
Collapse
|
25
|
Nie X, Liang Z, Li K, Yu H, Huang Y, Ye L, Yang Y. Novel organoid model in drug screening: Past, present, and future. LIVER RESEARCH 2021. [DOI: 10.1016/j.livres.2021.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Law CSW, Yeong KY. Benzimidazoles in Drug Discovery: A Patent Review. ChemMedChem 2021; 16:1861-1877. [PMID: 33646618 DOI: 10.1002/cmdc.202100004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 01/10/2023]
Abstract
Benzimidazole is a heterocyclic ring system that has been widely studied in the pharmaceutical field. For the past decade, numerous benzimidazole derivatives have been synthesized and evaluated for their wide range of pharmacological activities, which are beneficial for drug development. This article presents the biological effects of benzimidazole derivatives in each invention from 2015 to 2020. Two patent databases, Google Patents and Lens, were used to locate relevant granted patent applications. Specifically, this review delineates the role of patented benzimidazoles from a disease-centric perspective and examines the mechanisms of action of these compounds in related diseases. Most of the benzimidazoles have shown good activities against various target proteins. Whilst several of them have progressed into clinical trials, most patents presented novel therapeutic approaches for respective target diseases. Hence, their potential in being developed into clinical drugs are also discussed.
Collapse
Affiliation(s)
- Christine S W Law
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan Bandar Sunway, 47500, Selangor, Malaysia
| | - Keng Y Yeong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan Bandar Sunway, 47500, Selangor, Malaysia.,Tropical Medicine and Biology (TMB) multidisciplinary platform, Monash University Malaysia, Jalan Lagoon Selatan Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
27
|
Kvízová J, Pavlíčková V, Kmoníčková E, Ruml T, Rimpelová S. Quo Vadis Advanced Prostate Cancer Therapy? Novel Treatment Perspectives and Possible Future Directions. Molecules 2021; 26:2228. [PMID: 33921501 PMCID: PMC8069564 DOI: 10.3390/molecules26082228] [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: 03/21/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Prostate cancer is a very common disease, which is, unfortunately, often the cause of many male deaths. This is underlined by the fact that the early stages of prostate cancer are often asymptomatic. Therefore, the disease is usually detected and diagnosed at late advanced or even metastasized stages, which are already difficult to treat. Hence, it is important to pursue research and development not only in terms of novel diagnostic methods but also of therapeutic ones, as well as to increase the effectiveness of the treatment by combinational medicinal approach. Therefore, in this review article, we focus on recent approaches and novel potential tools for the treatment of advanced prostate cancer; these include not only androgen deprivation therapy, antiandrogen therapy, photodynamic therapy, photothermal therapy, immunotherapy, multimodal therapy, but also poly(ADP-ribose) polymerase, Akt and cyclin-dependent kinase inhibitors.
Collapse
Affiliation(s)
- Jana Kvízová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
- Bioinova, s.r.o., Vídeňská 1083, 140 20 Praha, Czech Republic
| | - Vladimíra Pavlíčková
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| | - Eva Kmoníčková
- Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic;
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| |
Collapse
|
28
|
Goka ET, Mesa Lopez DT, Lippman ME. Hormone-Dependent Prostate Cancers are Dependent on Rac Signaling for Growth and Survival. Mol Cancer Ther 2021; 20:1052-1061. [PMID: 33722851 DOI: 10.1158/1535-7163.mct-20-0695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/20/2020] [Accepted: 03/01/2021] [Indexed: 11/16/2022]
Abstract
Prostate cancer remains a common cause of cancer mortality in men. Initially, cancers are dependent of androgens for growth and survival. First line therapies reduce levels of circulating androgens or target the androgen receptor (AR) directly. Although most patients show durable responses, many patients eventually progress to castration-resistant prostate cancer (CRPC) creating a need for alternative treatment options. The Rac1 signaling pathway has previously been implicated as a driver of cancer initiation and disease progression. We investigated the role of HACE1, the E3 ubiquitin ligase for Rac1, in prostate cancer and found that HACE1 is commonly lost resulting in hyperactive Rac signaling leading to enhanced cellular proliferation, motility and viability. Importantly, we show that a Rac inhibitor can attenuate the growth and survival of prostate cancer cells. Rac signaling was also found to be critical in prostate cancers that express the AR. Rac inhibition in androgen dependent cells resulted in reduction of AR target gene expression suggesting that targeting Rac1 may be an alternative method for blocking the AR signaling axis. Finally, when used in combination with AR antagonists, Rac inhibition enhanced the suppression of AR target gene expression. Therefore, targeting Rac in prostate cancer has the potential to enhance the efficacy of approved AR therapies.
Collapse
Affiliation(s)
| | | | - Marc E Lippman
- Department of Oncology, Georgetown University, Washington, District of Columbia.
| |
Collapse
|
29
|
Wang L, Xu M, Kao CY, Tsai SY, Tsai MJ. Small molecule JQ1 promotes prostate cancer invasion via BET-independent inactivation of FOXA1. J Clin Invest 2020; 130:1782-1792. [PMID: 31874106 DOI: 10.1172/jci126327] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Recent findings have shown that inhibitors targeting bromodomain and extraterminal domain (BET) proteins, such as the small molecule JQ1, are potent growth inhibitors of many cancers and hold promise for cancer therapy. However, some reports have also revealed that JQ1 can activate additional oncogenic pathways and may affect epithelial-to-mesenchymal transition (EMT). Therefore, it is important to address the potential unexpected effect of JQ1 treatment, such as cell invasion and metastasis. Here, we showed that in prostate cancer, JQ1 inhibited cancer cell growth but promoted invasion and metastasis in a BET protein-independent manner. Multiple invasion pathways including EMT, bone morphogenetic protein (BMP) signaling, chemokine signaling, and focal adhesion were activated by JQ1 to promote invasion. Notably, JQ1 induced upregulation of invasion genes through inhibition of Forkhead box protein A1 (FOXA1), an invasion suppressor in prostate cancer. JQ1 directly interacted with FOXA1 and inactivated FOXA1 binding to its interacting repressors TLE3, HDAC7, and NFIC, thereby blocking FOXA1-repressive function and activating the invasion genes. Our findings indicate that JQ1 has an unexpected effect of promoting invasion in prostate cancer. Thus, the ill effect of JQ1 or its derived therapeutic agents cannot be ignored during cancer treatment, especially in FOXA1-related cancers.
Collapse
Affiliation(s)
- Leiming Wang
- Department of Molecular and Cellular Biology, and
| | - Mafei Xu
- Department of Molecular and Cellular Biology, and
| | | | - Sophia Y Tsai
- Department of Molecular and Cellular Biology, and.,Department of Medicine and Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, and.,Department of Medicine and Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
31
|
Abstract
Prostate Cancer is now the second biggest cause of cancer mortality in the UK. Media coverage has been rising, with some attributing to a rise in the cases diagnosed and treated in the NHS down to the “Fry and Turnbull effect”. Our understanding of prostate cancer has increased tremendously in the past decades, with advances in molecular biology and genomics driving the way to new treatments and diagnostics. This Special Edition of Translational Andrology and Urology 2019: Prostate Cancer Biology and Genomics aims to review the current state of prostate cancer genomics, proteomics, diagnostics and treatment.
Collapse
Affiliation(s)
- Hayley Whitaker
- Division of Surgical and Interventional Sciences, Faculty of Medicine, University College London, London, UK
| | - Joseph O Tam
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Martin J Connor
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Alistair Grey
- Division of Surgical and Interventional Sciences, Faculty of Medicine, University College London, London, UK.,Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Bart's and Royal London Hospitals, London, UK
| |
Collapse
|
32
|
Wang L, Cheng CM, Qin J, Xu M, Kao CY, Shi J, You E, Gong W, Rosa LP, Chase P, Scampavia L, Madoux F, Spicer T, Hodder P, Xu HE, Tsai SY, Tsai MJ. Small-molecule inhibitor targeting orphan nuclear receptor COUP-TFII for prostate cancer treatment. SCIENCE ADVANCES 2020; 6:eaaz8031. [PMID: 32494682 PMCID: PMC7190335 DOI: 10.1126/sciadv.aaz8031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/04/2020] [Indexed: 06/01/2023]
Abstract
The orphan nuclear receptor COUP-TFII is expressed at a low level in adult tissues, but its expression is increased and shown to promote progression of multiple diseases, including prostate cancer, heart failure, and muscular dystrophy. Suppression of COUP-TFII slows disease progression, making it an intriguing therapeutic target. Here, we identified a potent and specific COUP-TFII inhibitor through high-throughput screening. The inhibitor specifically suppressed COUP-TFII activity to regulate its target genes. Mechanistically, the inhibitor directly bound to the COUP-TFII ligand-binding domain and disrupted COUP-TFII interaction with transcription regulators, including FOXA1, thus repressing COUP-TFII activity on target gene regulation. Through blocking COUP-TFII's oncogenic activity in prostate cancer, the inhibitor efficiently exerted a potent antitumor effect in xenograft mouse models and patient-derived xenograft models. Our study identified a potent and specific COUP-TFII inhibitor that may be useful for the treatment of prostate cancer and possibly other diseases.
Collapse
Affiliation(s)
- Leiming Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chiang-Min Cheng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Mafei Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chung-Yang Kao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jingjing Shi
- CAS Key Laboratory of Receptor Research, VARI-SIMM Center, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Erli You
- CAS Key Laboratory of Receptor Research, VARI-SIMM Center, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wanchun Gong
- CAS Key Laboratory of Receptor Research, VARI-SIMM Center, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Laura Pedro Rosa
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - Peter Chase
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - Louis Scampavia
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - Franck Madoux
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - Timothy Spicer
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - Peter Hodder
- Scripps Research, Molecular Screening Center, Jupiter, FL 33458, USA
| | - H. Eric Xu
- CAS Key Laboratory of Receptor Research, VARI-SIMM Center, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sophia Y. Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
33
|
Pharmacokinetic Drug–Drug Interaction of Apalutamide, Part 2: Investigating Interaction Potential Using a Physiologically Based Pharmacokinetic Model. Clin Pharmacokinet 2020; 59:1149-1160. [DOI: 10.1007/s40262-020-00881-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Burslem GM, Crews CM. Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020; 181:102-114. [PMID: 31955850 PMCID: PMC7319047 DOI: 10.1016/j.cell.2019.11.031] [Citation(s) in RCA: 561] [Impact Index Per Article: 140.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022]
Abstract
New biological tools provide new techniques to probe fundamental biological processes. Here we describe the burgeoning field of proteolysis-targeting chimeras (PROTACs), which are capable of modulating protein concentrations at a post-translational level by co-opting the ubiquitin-proteasome system. We describe the PROTAC technology and its application to drug discovery and provide examples where PROTACs have enabled novel biological insights. Furthermore, we provide a workflow for PROTAC development and use and discuss the benefits and issues associated with PROTACs. Finally, we compare PROTAC-mediated protein-level modulation with other technologies, such as RNAi and genome editing.
Collapse
Affiliation(s)
- George M Burslem
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Craig M Crews
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA; Departments of Chemistry and Pharmacology, Yale University, New Haven, CT, USA.
| |
Collapse
|
35
|
Hu JR, Duncan MS, Morgans AK, Brown JD, Meijers WC, Freiberg MS, Salem JE, Beckman JA, Moslehi JJ. Cardiovascular Effects of Androgen Deprivation Therapy in Prostate Cancer: Contemporary Meta-Analyses. Arterioscler Thromb Vasc Biol 2020; 40:e55-e64. [PMID: 31969015 PMCID: PMC7047549 DOI: 10.1161/atvbaha.119.313046] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Androgen deprivation therapy is a cornerstone of prostate cancer treatment. Pharmacological androgen deprivation includes gonadotropin-releasing hormone agonism and antagonism, androgen receptor inhibition, and CYP17 (cytochrome P450 17A1) inhibition. Studies in the past decade have raised concerns about the potential for androgen deprivation therapy to increase the risk of adverse cardiovascular events such as myocardial infarction, stroke, and cardiovascular mortality, possibly by exacerbating cardiovascular risk factors. In this review, we summarize existing data on the cardiovascular effects of androgen deprivation therapy. Among the therapies, abiraterone stands out for increasing risk of cardiac events in meta-analyses of both randomized controlled trials and observational studies. We find a divergence between observational studies, which show consistent positive associations between androgen deprivation therapy use and cardiovascular disease, and randomized controlled trials, which do not show these associations reproducibly.
Collapse
Affiliation(s)
- Jiun-Ruey Hu
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Meredith S Duncan
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN.,Department of Epidemiology (M.S.D., M.S.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Alicia K Morgans
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL (A.K.M.)
| | - Jonathan D Brown
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Wouter C Meijers
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Matthew S Freiberg
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN.,Department of Epidemiology (M.S.D., M.S.F.), Vanderbilt University Medical Center, Nashville, TN.,Nashville Veteran Affairs (VA) Medical Center, TN (M.S.F.)
| | - Joe-Elie Salem
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN.,Department of Pharmacology, AP-HP, Pitié-Salpêtrière Hospital, CIC-1421, INSERM, Sorbonne Universités, UNICO-GRECO Cardio-Oncology Program, Paris, France (J.-E.S.)
| | - Joshua A Beckman
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN
| | - Javid J Moslehi
- From the Division of Cardiovascular Medicine (J.-R.H., M.S.D., J.D.B., W.C.M., M.S.F., J.-E.S., J.A.B., J.J.M.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
36
|
Korotchkina L, Kazyulkin D, Komarov PG, Polinsky A, Andrianova EL, Joshi S, Gupta M, Vujcic S, Kononov E, Toshkov I, Tian Y, Krasnov P, Chernov MV, Veith J, Antoch MP, Middlemiss S, Somers K, Lock RB, Norris MD, Henderson MJ, Haber M, Chernova OB, Gudkov AV. OT-82, a novel anticancer drug candidate that targets the strong dependence of hematological malignancies on NAD biosynthesis. Leukemia 2020; 34:1828-1839. [PMID: 31896781 DOI: 10.1038/s41375-019-0692-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/23/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023]
Abstract
Effective treatment of some types of cancer can be achieved by modulating cell lineage-specific rather than tumor-specific targets. We conducted a systematic search for novel agents selectively toxic to cells of hematopoietic origin. Chemical library screenings followed by hit-to-lead optimization identified OT-82, a small molecule with strong efficacy against hematopoietic malignancies including acute myeloblastic and lymphoblastic adult and pediatric leukemias, erythroleukemia, multiple myeloma, and Burkitt's lymphoma in vitro and in mouse xenograft models. OT-82 was also more toxic towards patients-derived leukemic cells versus healthy bone marrow-derived hematopoietic precursors. OT-82 was shown to induce cell death by inhibiting nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the salvage pathway of NAD synthesis. In mice, optimization of OT-82 dosing and dietary niacin further expanded the compound's therapeutic index. In toxicological studies conducted in mice and nonhuman primates, OT-82 showed no cardiac, neurological or retinal toxicities observed with other NAMPT inhibitors and had no effect on mouse aging or longevity. Hematopoietic and lymphoid organs were identified as the primary targets for dose limiting toxicity of OT-82 in both species. These results reveal strong dependence of neoplastic cells of hematopoietic origin on NAMPT and introduce OT-82 as a promising candidate for the treatment of hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jean Veith
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | | | | | - Murray D Norris
- Children's Cancer Institute, Sydney, NSW, Australia.,University of New South Wales Centre for Childhood Cancer Research, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
37
|
Anti-androgen hormonal therapy for cancer and other diseases. Eur J Pharmacol 2020; 866:172783. [DOI: 10.1016/j.ejphar.2019.172783] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 01/27/2023]
|
38
|
Bernemann C, Krabbe LM, Schrader AJ. Considerations for AR-V7 testing in clinical routine practice. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S378. [PMID: 32016096 DOI: 10.21037/atm.2019.12.136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Christof Bernemann
- Department of Urology, University of Muenster Medical Center, Muenster, Germany
| | - Laura-Maria Krabbe
- Department of Urology, University of Muenster Medical Center, Muenster, Germany
| | - Andres Jan Schrader
- Department of Urology, University of Muenster Medical Center, Muenster, Germany
| |
Collapse
|
39
|
Pertusati F, Ferla S, Bassetto M, Brancale A, Khandil S, Westwell AD, McGuigan C. A new series of bicalutamide, enzalutamide and enobosarm derivatives carrying pentafluorosulfanyl (SF5) and pentafluoroethyl (C2F5) substituents: Improved antiproliferative agents against prostate cancer. Eur J Med Chem 2019; 180:1-14. [DOI: 10.1016/j.ejmech.2019.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 01/31/2023]
|
40
|
Cuneo MJ, Mittag T. The ubiquitin ligase adaptor SPOP in cancer. FEBS J 2019; 286:3946-3958. [PMID: 31495053 DOI: 10.1111/febs.15056] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/20/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
The dysregulation of ubiquitin-mediated proteasomal degradation has emerged as an important mechanism of pathogenesis in several cancers. The speckle-type POZ protein (SPOP) functions as a substrate adaptor for the cullin3-RING ubiquitin ligase and controls the cellular persistence of a diverse array of protein substrates in hormone signalling, epigenetic control and cell cycle regulation, to name a few. Mutations in SPOP and the resulting dysregulation of this proteostatic pathway play causative roles in the pathogenesis of prostate and endometrial cancers, whereas overexpression and mislocalization are associated with kidney cancer. Understanding the molecular mechanism of the normal function of SPOP as well as the cause of SPOP-mediated oncogenesis is thus critical for eventual therapeutic targeting of SPOP and other related pathways. Here, we will review SPOP structure, function and the molecular mechanism of how this function is achieved. We will then review how mutations and protein mislocalization contribute to cancer pathogenesis and will provide a perspective on how SPOP may be targeted therapeutically.
Collapse
Affiliation(s)
- Matthew J Cuneo
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tanja Mittag
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
41
|
Ghafoor S, Burger IA, Vargas AH. Multimodality Imaging of Prostate Cancer. J Nucl Med 2019; 60:1350-1358. [PMID: 31481573 DOI: 10.2967/jnumed.119.228320] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/29/2019] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is a very heterogeneous disease, and contemporary management is focused on identification and treatment of the prognostically adverse high-risk tumors while minimizing overtreatment of indolent, low-risk tumors. In recent years, imaging has gained increasing importance in the detection, staging, posttreatment assessment, and detection of recurrence of prostate cancer. Several imaging modalities including conventional and functional methods are used in different clinical scenarios with their very own advantages and limitations. This continuing medical education article provides an overview of available imaging modalities currently in use for prostate cancer followed by a more specific section on the value of these different imaging modalities in distinct clinical scenarios, ranging from initial diagnosis to advanced, metastatic castration-resistant prostate cancer. In addition to established imaging indications, we will highlight some potential future applications of contemporary imaging modalities in prostate cancer.
Collapse
Affiliation(s)
- Soleen Ghafoor
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Irene A Burger
- Department of Nuclear Medicine, Baden Cantonal Hospital, Baden, Switzerland
| | - Alberto H Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| |
Collapse
|
42
|
Okafor CD, Colucci JK, Ortlund EA. Ligand-Induced Allosteric Effects Governing SR Signaling. NUCLEAR RECEPTOR RESEARCH 2019. [DOI: 10.32527/2019/101382] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
43
|
Tenkorang MAA, Duong P, Cunningham RL. NADPH Oxidase Mediates Membrane Androgen Receptor-Induced Neurodegeneration. Endocrinology 2019; 160:947-963. [PMID: 30811529 PMCID: PMC6435014 DOI: 10.1210/en.2018-01079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Abstract
Oxidative stress (OS) is a common characteristic of several neurodegenerative disorders, including Parkinson disease (PD). PD is more prevalent in men than in women, indicating the possible involvement of androgens. Androgens can have either neuroprotective or neurodamaging effects, depending on the presence of OS. Specifically, in an OS environment, androgens via a membrane-associated androgen receptor (mAR) exacerbate OS-induced damage. To investigate the role of androgens on OS signaling and neurodegeneration, the effects of testosterone and androgen receptor activation on the major OS signaling cascades, the reduced form of NAD phosphate (NADPH) oxidase (NOX)1 and NOX2 and the Gαq/inositol trisphosphate receptor (InsP3R), were examined. To create an OS environment, an immortalized neuronal cell line was exposed to H2O2 prior to cell-permeable/cell-impermeable androgens. Different inhibitors were used to examine the role of G proteins, mAR, InsP3R, and NOX1/2 on OS generation and cell viability. Both testosterone and DHT/3-O-carboxymethyloxime (DHT)-BSA increased H2O2-induced OS and cell death, indicating the involvement of an mAR. Furthermore, classical AR antagonists did not block testosterone's negative effects in an OS environment. Because there are no known antagonists specific for mARs, an AR protein degrader, ASC-J9, was used to block mAR action. ASC-J9 blocked testosterone's negative effects. To determine OS-related signaling mediated by mAR, this study examined NOX1, NOX2, Gαq. NOX1, NOX2, and the Gαq complex with mAR. Only NOX inhibition blocked testosterone-induced cell loss and OS. No effects of blocking either Gαq or G protein activation were observed on testosterone's negative effects. These results indicate that androgen-induced OS is via the mAR-NOX complex and not the mAR-Gαq complex.
Collapse
Affiliation(s)
- Mavis A A Tenkorang
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
| | - Phong Duong
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, Texas
- Correspondence: Rebecca L. Cunningham, PhD, Department of Physiology and Anatomy, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, Texas 76107. E-mail:
| |
Collapse
|
44
|
Barrado M, Blanco-Luquin I, Navarrete PA, Visus I, Guerrero-Setas D, Escors D, Kochan G, Arias F. Radiopotentiation of enzalutamide over human prostate cancer cells as assessed by real-time cell monitoring. Rep Pract Oncol Radiother 2019; 24:221-226. [PMID: 30858765 DOI: 10.1016/j.rpor.2019.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/12/2018] [Accepted: 02/07/2019] [Indexed: 11/29/2022] Open
Abstract
Aim To evaluate the radiopotentiation of enzalutamide in human prostate cancer cells. Background While radiotherapy is the first line of treatment for prostate cancer, androgen blockade therapies are demonstrating significant survival benefit as monotherapies. As androgen blockade can cause cell death by apoptosis, it is likely that androgen blockade will potentiate the cytotoxic activities of radiotherapy. Materials and methods Here, we tested the potential synergistic effects of these two treatments over two human metastatic prostate cancer cells by real-time cell analysis (RTCA), androgen-sensitive LNCaP cells (Lymph Node Carcinoma of the Prostate) and androgen-independent PC-3. Both cell lines were highly resistant to high doses of radiotherapy. Results A pre-treatment of LNCaP cells with IC50 concentrations of enzalutamide significantly sensitized them to radiotherapy through enhanced apoptosis. In contrast, enzalutamide resistant PC-3 cells were not sensitized to radiotherapy by androgen blockade. Conclusions These results provide evidence that the enzalutamide/radiotherapy combination could maximize therapeutic responses in patients with enzalutamide-sensitive prostate cancer.
Collapse
Affiliation(s)
- Marta Barrado
- Biomedical Research Center of Navarra-Navarrabiomed, Fundación Miguel Servet, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain.,Department of Radiation Oncology, Hospital of Navarre, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - Idoia Blanco-Luquin
- Biomedical Research Center of Navarra-Navarrabiomed, Fundación Miguel Servet, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - Paola Andrea Navarrete
- Department of Radiation Oncology, Hospital of Navarre, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - Ignacio Visus
- Department of Radiation Oncology, Hospital of Navarre, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - David Guerrero-Setas
- Biomedical Research Center of Navarra-Navarrabiomed, Fundación Miguel Servet, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - David Escors
- Biomedical Research Center of Navarra-Navarrabiomed, Fundación Miguel Servet, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain.,Department of Infection and Immunity, Rayne Institute, University College London, 5 University Street, WC1E 6JJ London, United Kingdom
| | - Grazyna Kochan
- Biomedical Research Center of Navarra-Navarrabiomed, Fundación Miguel Servet, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| | - Fernando Arias
- Department of Radiation Oncology, Hospital of Navarre, IdISNA, Irunlarrea 3, 31008 Pamplona, Navarre, Spain
| |
Collapse
|
45
|
Carugo A, Draetta GF. Academic Discovery of Anticancer Drugs: Historic and Future Perspectives. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2019. [DOI: 10.1146/annurev-cancerbio-030518-055645] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The identification and prosecution of meritorious anticancer drug targets and the discovery of clinical candidates represent an extraordinarily time- and resource-intensive process, and the current failure rate of late-stage drugs is a critical issue that must be addressed. Relationships between academia and industry in drug discovery and development have continued to change over time as a result of technical and financial challenges and, most importantly, to the objective of translating impactful scientific discoveries into clinical opportunities. This Golden Age of anticancer drug discovery features an increased appreciation for the high-risk, high-innovation research conducted in the nonprofit sector, with the goals of infusing commercial drug development with intellectual capital and curating portfolios that are financially tenable and clinically meaningful. In this review, we discuss the history of academic-industry interactions in the context of antidrug discovery and offer a view of where these interactions are likely headed as we continue to reach new horizons in our understanding of the immense complexities of cancer biology.
Collapse
Affiliation(s)
- Alessandro Carugo
- Center for Co-Clinical Trials and Institute for Applied Cancer Science, MD Anderson Cancer Center, Houston, Texas 77030, USA
- Moon Shots Program™, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Giulio F. Draetta
- Moon Shots Program™, MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
46
|
Liu Q, Wang G, Li Q, Jiang W, Kim JS, Wang R, Zhu S, Wang X, Yan L, Yi Y, Zhang L, Meng Q, Li C, Zhao D, Qiao Y, Li Y, Gursel DB, Chinnaiyan AM, Chen K, Cao Q. Polycomb group proteins EZH2 and EED directly regulate androgen receptor in advanced prostate cancer. Int J Cancer 2019; 145:415-426. [PMID: 30628724 DOI: 10.1002/ijc.32118] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/19/2018] [Indexed: 01/01/2023]
Abstract
Polycomb group proteins are important epigenetic regulators for cell proliferation and differentiation, organ development, as well as initiation and progression of lethal diseases, including cancer. Upregulated Polycomb group proteins, including Enhancer of zeste homolog 2 (EZH2), promote proliferation, migration, invasion and metastasis of cancer cells, as well as self-renewal of cancer stem cells. In our study, we report that EZH2 and embryonic ectoderm development (EED) indicate respective direct interaction with androgen receptor (AR). In the context of AR-positive prostate cancer, EZH2 and EED regulate AR expression levels and AR downstream targets. More importantly, we demonstrate that targeting EZH2 with the small-molecule inhibitor astemizole in cancer significantly represses the EZH2 and AR expression as well as the neoplastic capacities. These results collectively suggest that pharmacologically targeting EZH2 might be a promising strategy for advanced prostate cancer.
Collapse
Affiliation(s)
- Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Urology, The Second Xiangya Hospital of Central South University, Hunan, China
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY
| | - Qiaqia Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Weihua Jiang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX
| | - Jung-Sun Kim
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX
| | - Rui Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX
| | - Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX
| | - Xiaoju Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Lin Yan
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yang Yi
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lili Zhang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX
| | - Qingshu Meng
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Chao Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Dongyu Zhao
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Yong Li
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Demirkan B Gursel
- Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL.,Pathology Department, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI.,Department of Pathology, University of Michigan, Ann Arbor, MI.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI.,Department of Urology, University of Michigan, Ann Arbor, MI.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI
| | - Kaifu Chen
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY.,Institutes for Academic Medicine, Houston Methodist Hospital, Houston, TX
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
47
|
Dellis AE, Papatsoris AG. Perspectives on the current and emerging chemical androgen receptor antagonists for the treatment of prostate cancer. Expert Opin Pharmacother 2018; 20:163-172. [PMID: 30462924 DOI: 10.1080/14656566.2018.1548611] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Prostate cancer is the most common cancer in men. Regardless of the initial treatment of localized disease, almost all patients develop castration resistant prostate cancer (CRPC). A better understanding of the molecular mechanisms behind castration resistance has led to the approval of novel oral androgen receptor (AR) antagonists, such as enzalutamide and apalutamide. Indeed, research has accelerated with numerous agents being studied for the management of CRPC. Areas covered: Herein, the authors present currently used and emerging AR antagonists for the treatment of CRPC. Emerging agents include darolutamide, EZN-4176, AZD-3514, and AZD-5312, apatorsen, galeterone, ODM-2014, TRC-253, BMS-641988, and proxalutamide. Expert opinion: Further understanding of the mechanisms leading to castration resistance in prostate cancer can reveal potential targets for the development of novel AR antagonists. Current novel agents are associated with modest clinical and survival benefit, while acquired resistance and safety issues are under continuous evaluation. The combination of AR antagonists used and ideal sequencing strategies are key tasks ahead, along with the investigation of molecular biomarkers for future personalized targeted therapies. In the future, the challenge will be to determine an AR antagonist with the best combination of outcome and tolerability.
Collapse
Affiliation(s)
- Athanasios E Dellis
- a 2nd Department of Surgery, Aretaieion Academic Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece.,b 1st Department of Urology, Laikon General Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Athanasios G Papatsoris
- c 2nd Department of Urology, Sismanogleion General Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
48
|
Koshkin VS, Small EJ. Apalutamide in the treatment of castrate-resistant prostate cancer: evidence from clinical trials. Ther Adv Urol 2018; 10:445-454. [PMID: 30574205 PMCID: PMC6295778 DOI: 10.1177/1756287218811450] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/16/2018] [Indexed: 11/28/2022] Open
Abstract
Apalutamide (ARN-509) is a second-generation androgen receptor (AR) antagonist that was developed to inhibit AR-mediated prostate cancer cell proliferation. Following the initial promising clinical efficacy results in phase I and II clinical trials of patients with metastatic castrate-resistant prostate cancer (CRPC), apalutamide has been investigated in several phase III trials. Particular interest has focused on the development of effective therapy for the prevention of disease progression in patients with nonmetastatic (nm or M0) CRPC, especially patients who have a rapid prostate-specific antigen (PSA) doubling time that is indicative of shorter bone metastasis-free survival and associated with significant morbidity and mortality. The results from the phase III SPARTAN trial were recently published and reported a significant benefit of apalutamide relative to placebo in patients with nmCRPC and a high risk of metastatic progression. The study noted marked improvement in the primary endpoint of metastasis-free survival as well as several relevant secondary clinical endpoints, including time to symptomatic progression. These results led to the United States Food and Drug Administration (US FDA) approval of apalutamide in the nmCRPC setting in February 2018. This review summarizes the clinical development of apalutamide, culminating with the pivotal SPARTAN trial as well as other phase III trials which may further expand potential indications for this agent in the near future.
Collapse
Affiliation(s)
- Vadim S Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, 550 16th Street, Box 3211, San Francisco, CA 94158, USA
| | - Eric J Small
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Luo J, Tian J, Chou F, Lin C, Xing EZ, Zuo L, Niu Y, Yeh S, Chang C. Targeting the androgen receptor (AR) with AR degradation enhancer ASC-J9® led to increase docetaxel sensitivity via suppressing the p21 expression. Cancer Lett 2018; 444:35-44. [PMID: 30248372 DOI: 10.1016/j.canlet.2018.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022]
Abstract
Chemotherapy with docetaxel remains the effective therapy to suppress castration resistant prostate cancer (CRPC) in some patients. However, most chemotherapy with docetaxel eventually fails with the development of docetaxel resistance after 18-weeks of treatment. Here we found docetaxel treatment might have an adverse effect of increasing the androgen receptor (AR) protein level in the CRPC cells, and combining docetaxel with anti-AR therapy using AR-shRNA or the AR degradation enhancer ASC-J9® may increase docetaxel sensitivity to better suppress the CRPC cell growth. Mechanism dissection found docetaxel might have the adverse effect of increasing the AR protein stability via suppressing the AR ubiquitination due to the increased AR phosphorylation. The consequence of such increased AR protein may then lead to increase p21 expression via transcriptional regulation. Preclinical studies with in vitro cells lines also demonstrated that targeting AR with ASC-J9® led to suppressing the AR-increased p21 expression to improve the docetaxel sensitivity in the CRPC cells that already developed docetaxel resistance. Together, these results suggest that a combined therapy of docetaxel and ASC-J9® is a novel therapy to better suppress CRPC in patients that already developed docetaxel resistance.
Collapse
Affiliation(s)
- Jie Luo
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Biology Department, University of Rochester, Rochester, NY, USA
| | - Jing Tian
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - FuJu Chou
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Changyi Lin
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Emily Zixin Xing
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Li Zuo
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Department of Urology, Changzhou Second People's Affiliated Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, 300211, China
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, 14642, USA; Sex Hormone Research Center, China Medical University and Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
50
|
Schultz NM, Shore ND, Chowdhury S, Klotz LH, Concepcion RS, Penson DF, Karsh LI, Yang H, Brown BA, Barlev A, Flanders SC. Number-needed-to-treat analysis of clinical progression in patients with metastatic castration-resistant prostate cancer in the STRIVE and TERRAIN trials. BMC Urol 2018; 18:77. [PMID: 30189902 PMCID: PMC6128000 DOI: 10.1186/s12894-018-0387-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/23/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND This analysis estimated the number needed to treat with enzalutamide versus bicalutamide to achieve one additional patient with chemotherapy-naïve metastatic castration-resistant prostate cancer who would obtain clinical benefit regarding progression-free survival, radiographic progression-free survival, or no prostate-specific antigen progression at 1 and 2 years following treatment initiation. METHODS Clinical event rates were obtained from the STRIVE (NCT01664923) and TERRAIN (NCT01288911) trials, and the number needed to treat was the inverse of the absolute rate difference between the event rates of enzalutamide and bicalutamide. The 95% Confidence Interval of the number needed to treat was derived from the 95% Confidence Interval of the event rate difference. RESULTS Using STRIVE data (patients with metastatic disease: n = 128 enzalutamide; n = 129 bicalutamide) comparing enzalutamide with bicalutamide at 1 and 2 years, the numbers needed to treat to achieve one additional patient with chemotherapy-naïve metastatic castration-resistant prostate cancer with progression-free survival were 2.0 and 2.8, respectively; with radiographic progression-free survival, 2.6 and 3.0, respectively; and without prostate-specific antigen progression, 1.8 and 2.4, respectively. Using TERRAIN data (n = 184 enzalutamide; n = 191 bicalutamide) comparing enzalutamide with bicalutamide at 1 and 2 years, the numbers needed to treat to achieve one additional patient with progression-free survival were 4.3 and 3.7, respectively; with radiographic progression-free survival, 10.0 and 2.8, respectively; and without prostate-specific antigen progression, 2.1 and 3.2, respectively. CONCLUSIONS The combined data from TERRAIN and STRIVE demonstrated that treating chemotherapy-naïve metastatic castration-resistant prostate cancer with enzalutamide leads to more patients without clinical progression at 1 and 2 years than with bicalutamide. TRIAL REGISTRATION STRIVE (NCT01664923; registration date: August 10, 2012) and TERRAIN (NCT01288911; registration date: February 1, 2011).
Collapse
Affiliation(s)
- Neil M. Schultz
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062 USA
| | - Neal D. Shore
- Carolina Urologic Research Center, Myrtle Beach, SC USA
| | | | - Laurence H. Klotz
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON Canada
| | | | | | | | | | - Bruce A. Brown
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062 USA
| | - Arie Barlev
- Medivation, Inc., San Francisco, CA USA
- Pfizer, Inc., New York, NY USA
| | | |
Collapse
|