1
|
Liao J, Liao J, Wang Y, Wang X, Chai X, Wang H, Xu L, Shan L, Xu X, Fu W, Pan P, Hou T, Sheng R, Li D. Discovery of N-(1,2,4-Thiadiazol-5-yl)benzo[ b]oxepine-4-carboxamide Derivatives as Novel Antiresistance Androgen Receptor Antagonists. J Med Chem 2025. [PMID: 39848621 DOI: 10.1021/acs.jmedchem.4c02649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
The ligand-binding pocket of the androgen receptor (AR) is the targeting site of all clinically used AR antagonists. However, various drug-resistant mutations emerged in the pocket. We previously reported a new targeting site at the dimer interface of AR (dimer interface pocket) and identified a novel antagonist M17-B15 that failed in oral administration. In this study, the head part of M17-B15 was substituted with divergent structures. Potent antagonist Z10 with benzo[b]oxepine was first identified. Subsequent structural optimization on the 2-oxopropyl moiety of Z10 generated the more powerful Y5 (IC50 = 0.04 μM). Out of the ordinary, Y5 demonstrated dual mechanisms of action, antagonized AR by disrupting AR dimerization, and induced AR degradation via the ubiquitin-proteasome pathway. Furthermore, Y5 exhibited excellent activity against variant drug-resistant AR mutants comparable to recently approved darolutamide. Furthermore, Y5 effectively suppressed the tumor growth of the LNCaP xenograft via oral administration, providing a potential novel therapeutic for drug-resistant prostate cancer.
Collapse
Affiliation(s)
- Jianing Liao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jinbiao Liao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xinyue Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xin Chai
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huating Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Luhu 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, Zhejiang 310022, China
| | - Xiaohong 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, Zhejiang 310022, China
| | - Weitao Fu
- Insitute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui 230601, China
| | - Peichen Pan
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Tingjun Hou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Rong Sheng
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321000, China
| | - Dan Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321000, China
| |
Collapse
|
2
|
Liao J, Liao J, Zhang M, Yu Y, Cai L, Le K, Fu W, Qin Y, Hou T, Li D, Sheng R. Identification of Oral Bioavailable Coumarin Derivatives as Potential AR Antagonists Targeting Prostate Cancer. J Med Chem 2024; 67:19395-19416. [PMID: 39492719 DOI: 10.1021/acs.jmedchem.4c01752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Androgen receptor (AR) is a crucial driver of prostate cancer (PCa), but acquired resistance to AR antagonists significantly undermines their clinical efficacy. We previously discovered coumarin derivative 1, which is capable of disrupting AR ligand-binding domain dimers, offering the potential for overcoming resistance. However, its poor oral bioavailability limited further development. In this study, comprehensive structure optimizations led to compound 4a (IC50 = 0.051 μM), which exhibited comparable AR antagonistic activity to enzalutamide (IC50 = 0.060 μM) and demonstrated excellent selectivity over other nuclear receptors in vitro. Especially, 4a showed superior efficacy against ARF876L/T877A and ARW741C mutants compared to darolutamide and enzalutamide. Moreover, 4a exhibited favorable pharmacokinetic profiles (F = 66.24%) in vivo and significant tumor growth inhibition in an LNCaP xenograft mouse model upon oral administration. These results highlight the potential of 4a as a promising oral AR antagonist for overcoming drug resistance in PCa.
Collapse
Affiliation(s)
- Jinbiao Liao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jianing Liao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Minkui Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yanzhen Yu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lvtao Cai
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321000 Zhejiang, China
| | - Kaixin Le
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321000 Zhejiang, China
| | - Weitao Fu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yiyang Qin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Tingjun Hou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dan Li
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321000 Zhejiang, China
| | - Rong Sheng
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321000 Zhejiang, China
| |
Collapse
|
3
|
Ochiai K, Yonezawa R, Fujii S. Structural Development of Androgen Receptor Antagonists Using Phenylferrocene Framework as a Hydrophobic Pharmacophore. ChemMedChem 2024; 19:e202400040. [PMID: 38291942 DOI: 10.1002/cmdc.202400040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
We previously identified nitrophenylferrocenes and cyanophenylferrocenes as promising lead structures of novel androgen receptor (AR) antagonists, based on the structural similarity between ferrocene and the steroidal skeleton. In the present research, we explored the structure-activity relationship (SAR) of phenylferrocene derivatives. Introduction of a hydrophobic substituent such as a chlorine atom at the 2-position or 3-position of phenylferrocene derivatives significantly increased the antagonistic activity toward wild-type AR, and among the synthesized compounds, 3-chloro-4-cyanophenylferrocene (29) exhibited the most potent anti-proliferative activity toward the androgen-dependent growth of SC-3 cells expressing wild-type AR (IC50 14 nM). Like conventional antiandrogens such as hydroxyflutamide, the major active metabolite of flutamide, compound 29 exhibited agonistic activity toward T877A-AR, a mutant AR expressed in human prostate cancer cell line LNCaP. Notably, however, the 2-chloro isomer 27 showed potent antagonistic activity toward wild-type AR (IC50 49 nM) and also exhibited antagonistic activity toward T877A-AR. Our SAR data should prove helpful for the development of new-generation AR antagonists based on phenylferrocene as candidate agents to treat drug-resistant prostate cancer.
Collapse
Affiliation(s)
- Kotaro Ochiai
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Ryo Yonezawa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| |
Collapse
|
4
|
Kumar GG, Kilari EK, Nelli G, Salleh N. Oral administration of Turnera diffusa willd. ex Schult. extract ameliorates steroidogenesis and spermatogenesis impairment in the testes of rats with type-2 diabetes mellitus. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116638. [PMID: 37187362 DOI: 10.1016/j.jep.2023.116638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/01/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Turnera diffusa Willd. ex Schult. (T. diffusa) has traditionally been used to treat male reproductive dysfunction and have aphrodisiac properties. AIMS OF THE STUDY This study aims to investigate the ability of T. diffusa to ameliorate the impairment in testicular steroidogenesis and spermatogenesis in DM that might help to improve testicular function, and subsequently restore male fertility. MATERIALS AND METHODS DM-induced adult male rats were given 100 mg/kg/day and 200 mg/kg/day T. diffusa leaf extract orally for 28 consecutive days. Rats were then sacrificed; sperm and testes were harvested and sperm parameter analysis were performed. Histo-morphological changes in the testes were observed. Biochemical assays were performed to measure testosterone and testicular oxidative stress levels. Immunohistochemistry and double immunofluorescence were used to monitor oxidative stress and inflammation levels in testes as well as Sertoli and steroidogenic marker proteins' expression. RESULTS Treatment with T. diffusa restores sperm count, motility, and viability near normal and reduces sperm morphological abnormalities and sperm DNA fragmentation in diabetic rats. T. diffusa treatment also reduces testicular NOX-2 and lipid peroxidation levels, increases testicular antioxidant enzymes (SOD, CAT, and GPx) activities, ameliorates testicular inflammation via downregulating NF-ΚB, p-Ikkβ and TNF-α and upregulating IκBα expression. In diabetic rats, T. diffusa treatment increases testicular steroidogenic proteins (StAR, CYP11A1, SHBG, and ARA54, 3 and 17β-HSD) and plasma testosterone levels. Furthermore, in diabetic rats treated with T. diffusa, Sertoli cell marker proteins including Connexin 43, N-cadherin, and occludin levels in the testes were elevated. CONCLUSION T. diffusa treatment could help to ameliorate the detrimental effects of DM on the testes, thus this plant has potential to be used to restore male fertility.
Collapse
Affiliation(s)
- Gowri Gopa Kumar
- Department of Physiology, Faculty of Medicine, University Malaya, 50603, Kuala Lumpur, Malaysia
| | - Eswar Kumar Kilari
- Pharmacology Division, A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530 003, India
| | - Giribabu Nelli
- Department of Physiology, Faculty of Medicine, University Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
Schnöller LE, Piehlmaier D, Weber P, Brix N, Fleischmann DF, Nieto AE, Selmansberger M, Heider T, Hess J, Niyazi M, Belka C, Lauber K, Unger K, Orth M. Systematic in vitro analysis of therapy resistance in glioblastoma cell lines by integration of clonogenic survival data with multi-level molecular data. Radiat Oncol 2023; 18:51. [PMID: 36906590 PMCID: PMC10007763 DOI: 10.1186/s13014-023-02241-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
Despite intensive basic scientific, translational, and clinical efforts in the last decades, glioblastoma remains a devastating disease with a highly dismal prognosis. Apart from the implementation of temozolomide into the clinical routine, novel treatment approaches have largely failed, emphasizing the need for systematic examination of glioblastoma therapy resistance in order to identify major drivers and thus, potential vulnerabilities for therapeutic intervention. Recently, we provided proof-of-concept for the systematic identification of combined modality radiochemotherapy treatment vulnerabilities via integration of clonogenic survival data upon radio(chemo)therapy with low-density transcriptomic profiling data in a panel of established human glioblastoma cell lines. Here, we expand this approach to multiple molecular levels, including genomic copy number, spectral karyotyping, DNA methylation, and transcriptome data. Correlation of transcriptome data with inherent therapy resistance on the single gene level yielded several candidates that were so far underappreciated in this context and for which clinically approved drugs are readily available, such as the androgen receptor (AR). Gene set enrichment analyses confirmed these results, and identified additional gene sets, including reactive oxygen species detoxification, mammalian target of rapamycin complex 1 (MTORC1) signaling, and ferroptosis/autophagy-related regulatory circuits to be associated with inherent therapy resistance in glioblastoma cells. To identify pharmacologically accessible genes within those gene sets, leading edge analyses were performed yielding candidates with functions in thioredoxin/peroxiredoxin metabolism, glutathione synthesis, chaperoning of proteins, prolyl hydroxylation, proteasome function, and DNA synthesis/repair. Our study thus confirms previously nominated targets for mechanism-based multi-modal glioblastoma therapy, provides proof-of-concept for this workflow of multi-level data integration, and identifies novel candidates for which pharmacological inhibitors are readily available and whose targeting in combination with radio(chemo)therapy deserves further examination. In addition, our study also reveals that the presented workflow requires mRNA expression data, rather than genomic copy number or DNA methylation data, since no stringent correlation between these data levels could be observed. Finally, the data sets generated in the present study, including functional and multi-level molecular data of commonly used glioblastoma cell lines, represent a valuable toolbox for other researchers in the field of glioblastoma therapy resistance.
Collapse
Affiliation(s)
- Leon Emanuel Schnöller
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Daniel Piehlmaier
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Peter Weber
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Nikko Brix
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Daniel Felix Fleischmann
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Edward Nieto
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Martin Selmansberger
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Theresa Heider
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Bavarian Cancer Research Center (BKFZ), Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Bavarian Cancer Research Center (BKFZ), Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium (DKTK), Munich, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany. .,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
6
|
Yavuz M, Sabour Takanlou L, Biray Avcı Ç, Demircan T. A Selective Androgen Receptor Modulator, S4, Displays Robust Anti-cancer Activity on Hepatocellular Cancer Cells by Negatively Regulating PI3K/AKT/mTOR Signaling Pathway. Gene 2023; 869:147390. [PMID: 36990257 DOI: 10.1016/j.gene.2023.147390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is a major global health problem that often correlates with poor prognosis. Due to the insufficient therapy options with limited benefits, it is crucial to identify new therapeutic approaches to overcome HCC. One of the vital signaling pathways in organ homeostasis and male sexual development is Androgen Receptor (AR) signaling. Its activity affects several genes that contribute to cancer characteristics and have essential roles in cell cycle progression, proliferation, angiogenesis, and metastasis. AR signaling has been shown to be misregulated in many cancers, including HCC, suggesting that it might contribute to hepatocarcinogenesis. Targeting AR signaling using anti-androgens, AR inhibitors, or AR-degrading molecules is a powerful and promising strategy to defeat HCC. In this study, AR signaling was targeted by a novel Selective Androgen Receptor Modulator (SARM), S4, in HCC cells to evaluate its potential anti-cancer effect. To date, S4 activity in cancer has not been demonstrated, and our data unrevealed that S4 significantly impaired HCC growth, migration, proliferation, and induced apoptosis through inhibiting PI3K/AKT/mTOR signaling. Since PI3K/AKT/mTOR signaling is frequently activated in HCC and contributes to its aggressiveness and poor prognosis, its negative regulation by the downregulation of critical components via S4 was a prominent finding. Further studies are necessary to investigate the S4 action mechanism and anti-tumorigenic capacity in in-vivo.
Collapse
|
7
|
Xu P, Yang JC, Ning S, Chen B, Nip C, Wei Q, Liu L, Johnson OT, Gao AC, Gestwicki JE, Evans CP, Liu C. Allosteric inhibition of HSP70 in collaboration with STUB1 augments enzalutamide efficacy in antiandrogen resistant prostate tumor and patient-derived models. Pharmacol Res 2023; 189:106692. [PMID: 36773708 PMCID: PMC10162009 DOI: 10.1016/j.phrs.2023.106692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Ubiquitin proteasome activity is suppressed in enzalutamide resistant prostate cancer cells, and the heat shock protein 70/STIP1 homology and U-box-containing protein 1 (HSP70/STUB1) machinery are involved in androgen receptor (AR) and AR variant protein stabilization. Targeting HSP70 could be a viable strategy to overcome resistance to androgen receptor signaling inhibitor (ARSI) in advanced prostate cancer. Here, we showed that a novel HSP70 allosteric inhibitor, JG98, significantly suppressed drug-resistant C4-2B MDVR and CWR22Rv1 cell growth, and enhanced enzalutamide treatment. JG98 also suppressed cell growth in conditional reprogramed cell cultures (CRCs) and organoids derived from advanced prostate cancer patient samples. Mechanistically, JG98 degraded AR/AR-V7 expression in resistant cells and promoted STUB1 nuclear translocation to bind AR-V7. Knockdown of the E3 ligase STUB1 significantly diminished the anticancer effects and partially restored AR-V7 inhibitory effects of JG98. JG231, a more potent analog developed from JG98, effectively suppressed the growth of the drug-resistant prostate cancer cells, CRCs, and organoids. Notably, the combination of JG231 and enzalutamide synergistically inhibited AR/AR-V7 expression and suppressed CWR22Rv1 xenograft tumor growth. Inhibition of HSP70 using novel small-molecule inhibitors coordinates with STUB1 to regulate AR/AR-V7 protein stabilization and ARSI resistance.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA; Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA.
| |
Collapse
|
8
|
Alemany M. The Roles of Androgens in Humans: Biology, Metabolic Regulation and Health. Int J Mol Sci 2022; 23:11952. [PMID: 36233256 PMCID: PMC9569951 DOI: 10.3390/ijms231911952] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Androgens are an important and diverse group of steroid hormone molecular species. They play varied functional roles, such as the control of metabolic energy fate and partition, the maintenance of skeletal and body protein and integrity and the development of brain capabilities and behavioral setup (including those factors defining maleness). In addition, androgens are the precursors of estrogens, with which they share an extensive control of the reproductive mechanisms (in both sexes). In this review, the types of androgens, their functions and signaling are tabulated and described, including some less-known functions. The close interrelationship between corticosteroids and androgens is also analyzed, centered in the adrenal cortex, together with the main feedback control systems of the hypothalamic-hypophysis-gonads axis, and its modulation by the metabolic environment, sex, age and health. Testosterone (T) is singled out because of its high synthesis rate and turnover, but also because age-related hypogonadism is a key signal for the biologically planned early obsolescence of men, and the delayed onset of a faster rate of functional losses in women after menopause. The close collaboration of T with estradiol (E2) active in the maintenance of body metabolic systems is also presented Their parallel insufficiency has been directly related to the ravages of senescence and the metabolic syndrome constellation of disorders. The clinical use of T to correct hypoandrogenism helps maintain the functionality of core metabolism, limiting excess fat deposition, sarcopenia and cognoscitive frailty (part of these effects are due to the E2 generated from T). The effectiveness of using lipophilic T esters for T replacement treatments is analyzed in depth, and the main problems derived from their application are discussed.
Collapse
Affiliation(s)
- Marià Alemany
- Facultat de Biologia, Universitat de Barcelona, Av. Diagonal, 635, 08028 Barcelona, Catalonia, Spain;
- Institut de Biomedicina, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
9
|
The Crucial Role of AR-V7 in Enzalutamide-Resistance of Castration-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14194877. [PMID: 36230800 PMCID: PMC9563243 DOI: 10.3390/cancers14194877] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Androgen receptor splice variant 7 (AR-V7) has always been considered a key driver for triggering enzalutamide resistance of castration-resistant prostate cancer (CRPC). In recent years, both the homeostasis of AR-V7 protein and AR-V7’s relationship with LncRNAs have gained great attention with in-depth studies. Starting from protein stability and LncRNA, the paper discusses and summarizes the mechanisms and drugs that affect the CRPC patients’ sensitivity to enzalutamide by regulating the protein or transcriptional stability of AR-V7, hoping to provide therapeutic ideas for subsequent research to break through the CRPC therapeutic bottleneck. Abstract Prostate cancer (PCa) has the second highest incidence of malignancies occurring in men worldwide. The first-line therapy of PCa is androgen deprivation therapy (ADT). Nonetheless, most patients progress to castration-resistant prostate cancer (CRPC) after being treated by ADT. As a second-generation androgen receptor (AR) antagonist, enzalutamide (ENZ) is the current mainstay of new endocrine therapies for CRPC in clinical use. However, almost all patients develop resistance during AR antagonist therapy due to various mechanisms. At present, ENZ resistance (ENZR) has become challenging in the clinical treatment of CRPC. AR splice variant 7 (AR-V7) refers to a ligand-independent and constitutively active variant of the AR and is considered a key driver of ENZR in CRPC. In this review, we summarize the mechanisms and biological behaviors of AR-V7 in ENZR of CRPC to contribute novel insights for CRPC therapy.
Collapse
|
10
|
Chen C, Chai X, Hu X, Lou S, Li D, Hou T, Cui S. Discovery of 2-(1-(3-Chloro-4-cyanophenyl)-1 H-pyrazol-3-yl)acetamides as Potent, Selective, and Orally Available Antagonists Targeting the Androgen Receptor. J Med Chem 2022; 65:13074-13093. [PMID: 36154033 DOI: 10.1021/acs.jmedchem.2c00912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The androgen receptor (AR) antagonists are efficient therapeutics for the treatment of prostate cancer (PCa). All the approved AR antagonists to date are targeted to the ligand-binding pocket (LBP) of AR and have suffered from various drug resistances, whereas AR antagonist targeting non-LBP site of AR is conceived as a promising strategy. Through the scaffold hopping of AR LBP antagonists, the 2-chloro-4-(1H-pyrazol-1-yl)benzonitrile was designed as a new core structure for AR antagonists. A total of 46 compounds were synthesized and biologically evaluated to disclose compounds 2f, 2k, and 4c, exhibiting potent AR antagonistic activities (IC50 up to 69 nM), force against antiandrogen resistance, and untraditional targeting site of probably AR binding function 3. Therein, 4c exhibited effective tumor growth inhibition in LNCaP xenograft study upon oral administration. This work provides a novel chemical scaffold for AR antagonists and offers new perspective for the development of PCa therapy.
Collapse
Affiliation(s)
- Changwei Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xin Chai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueping Hu
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao 266237, China
| | - Shengying Lou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dan Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sunliang Cui
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
11
|
Handelsman DJ. History of androgens and androgen action. Best Pract Res Clin Endocrinol Metab 2022; 36:101629. [PMID: 35277356 DOI: 10.1016/j.beem.2022.101629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- David J Handelsman
- Professor of Reproductive Endocrinology and Andrology, ANZAC Research Institute, University of SydneyHead, Andrology Department, Concord RG Hospital, Australia.
| |
Collapse
|
12
|
Kuznik NC, Solozobova V, Lee II, Jung N, Yang L, Nienhaus K, Ntim EA, Rottenberg JT, Muhle-Goll C, Kumar AR, Peravali R, Gräßle S, Gourain V, Deville C, Cato L, Neeb A, Dilger M, Cramer von Clausbruch CA, Weiss C, Kieffer B, Nienhaus GU, Brown M, Bräse S, Cato ACB. A chemical probe for BAG1 targets androgen receptor-positive prostate cancer through oxidative stress signaling pathway. iScience 2022; 25:104175. [PMID: 35479411 PMCID: PMC9036123 DOI: 10.1016/j.isci.2022.104175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/01/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
BAG1 is a family of polypeptides with a conserved C-terminal BAG domain that functions as a nucleotide exchange factor for the molecular chaperone HSP70. BAG1 proteins also control several signaling processes including proteostasis, apoptosis, and transcription. The largest isoform, BAG1L, controls the activity of the androgen receptor (AR) and is upregulated in prostate cancer. Here, we show that BAG1L regulates AR dynamics in the nucleus and its ablation attenuates AR target gene expression especially those involved in oxidative stress and metabolism. We show that a small molecule, A4B17, that targets the BAG domain downregulates AR target genes similar to a complete BAG1L knockout and upregulates the expression of oxidative stress-induced genes involved in cell death. Furthermore, A4B17 outperformed the clinically approved antagonist enzalutamide in inhibiting cell proliferation and prostate tumor development in a mouse xenograft model. BAG1 inhibitors therefore offer unique opportunities for antagonizing AR action and prostate cancer growth. BAG1L interacts with a sequence overlapping a polyalanine tract in the AR NTD Knockdown of BAG1L increase AR dynamics in the nucleus BAG1L uses ROS pathway to regulate AR+ prostate cancer cell proliferation A small molecule BAG1 inhibitor inhibits prostate tumor growth in mouse xenografts
Collapse
Affiliation(s)
- Nane C Kuznik
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Valeria Solozobova
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Irene I Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nicole Jung
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Linxiao Yang
- Institute of Applied Physics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Karin Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Emmanuel A Ntim
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Jaice T Rottenberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Claudia Muhle-Goll
- Institute of Biological Interfaces 4, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Amrish Rajendra Kumar
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Ravindra Peravali
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Simone Gräßle
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Victor Gourain
- LabEx IGO "Immunotherapy, Graft, Oncology", Centre de Recherche en Transplantation et Immunologie - UMR1064, 44093 Nantes, France
| | - Célia Deville
- Department of Integrative Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM, U964, CNRS, UMR-7104, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - Laura Cato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Antje Neeb
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Marco Dilger
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Christina A Cramer von Clausbruch
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Carsten Weiss
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Bruno Kieffer
- Department of Integrative Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM, U964, CNRS, UMR-7104, Université de Strasbourg, 67404 Illkirch-Graffenstaden, France
| | - G Ulrich Nienhaus
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
- Institute of Applied Physics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Andrew C B Cato
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
13
|
Chai X, Sun H, Zhou W, Chen C, Shan L, Yang Y, He J, Pang J, Yang L, Wang X, Cui S, Fu Y, Xu X, Xu L, Yao X, Li D, Hou T. Discovery of N-(4-(Benzyloxy)-phenyl)-sulfonamide Derivatives as Novel Antagonists of the Human Androgen Receptor Targeting the Activation Function 2. J Med Chem 2022; 65:2507-2521. [PMID: 35077161 DOI: 10.1021/acs.jmedchem.1c01938] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Androgen receptor (AR) antagonists have been widely used for the treatment of prostate cancer (PCa). As a link between the AR and its transcriptional function, the activation function 2 (AF2) region has recently been revealed as a novel targeting site for developing AR antagonists. Here, we reported a series of N-(4-(benzyloxy)-phenyl)-sulfonamide derivatives as new-scaffold AR antagonists targeting the AR AF2. Therein, compound T1-12 showed excellent AR antagonistic activity (IC50 = 0.47 μM) and peptide displacement activity (IC50 = 18.05 μM). Furthermore, the in vivo LNCaP xenograft study confirmed that T1-12 offered effective inhibition on tumor growth when administered intratumorally. The study represents the first successful attempt to identify a small molecule targeting the AR AF2 with submicromolar AR antagonistic activity by structure-based virtual screening and provides important clues for the development of novel therapeutics for PCa treatment.
Collapse
Affiliation(s)
- Xin Chai
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Huiyong Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Wenfang Zhou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Changwei Chen
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Luhu 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, Zhejiang, China
| | - Yuhui Yang
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang, China
| | - Junzhao He
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang, China
| | - Jinping Pang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Liu Yang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xinyue Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yaqin Fu
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang, China
| | - Xiaohong 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, Zhejiang, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, Jiangsu, China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
| | - Dan Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
14
|
Overview of the development of selective androgen receptor modulators (SARMs) as pharmacological treatment for osteoporosis (1998–2021). Eur J Med Chem 2022; 230:114119. [DOI: 10.1016/j.ejmech.2022.114119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/20/2021] [Accepted: 01/09/2022] [Indexed: 02/08/2023]
|
15
|
AR Structural Variants and Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:195-211. [DOI: 10.1007/978-3-031-11836-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
16
|
Kuznik NC, Solozobova V, Jung N, Gräßle S, Lei Q, Lewandowski EM, Munuganti R, Zoubeidi A, Chen Y, Bräse S, Cato ACB. Development of a Benzothiazole Scaffold-Based Androgen Receptor N-Terminal Inhibitor for Treating Androgen-Responsive Prostate Cancer. ACS Chem Biol 2021; 16:2103-2108. [PMID: 34506104 DOI: 10.1021/acschembio.1c00390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
All current clinically approved androgen deprivation therapies for prostate cancer target the C-terminal ligand-binding domain of the androgen receptor (AR). However, the main transactivation function of the receptor is localized at the AR N-terminal domain (NTD). Targeting the AR NTD directly is a challenge because of its intrinsically disordered structure and the lack of pockets for drugs to bind. Here, we have taken an alternative approach using the cochaperone BAG1L, which interacts with the NTD, to develop a novel AR inhibitor. We describe the identification of 2-(4-fluorophenyl)-5-(trifluoromethyl)-1,3-benzothiazole (A4B17), a small molecule that inhibits BAG1L-AR NTD interaction, attenuates BAG1L-mediated AR NTD activity, downregulates AR target gene expression, and inhibits proliferation of AR-positive prostate cancer cells. This compound represents a prototype of AR antagonists that could be key in the development of future prostate cancer therapeutics.
Collapse
Affiliation(s)
- Nane C. Kuznik
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Valeria Solozobova
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Nicole Jung
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Simone Gräßle
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Qing Lei
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| | - Eric M. Lewandowski
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612-4799, United States
| | - Ravi Munuganti
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urology, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Yu Chen
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612-4799, United States
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, Functional Molecular Systems, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Andrew C. B. Cato
- Institute of Biological and Chemical Systems, Biological Information Processing, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
17
|
Li H, Liu Y, Wang Y, Zhao X, Qi X. Hormone therapy for ovarian cancer: Emphasis on mechanisms and applications (Review). Oncol Rep 2021; 46:223. [PMID: 34435651 PMCID: PMC8424487 DOI: 10.3892/or.2021.8174] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/04/2021] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer (OC) remains the leading cause of mortality due to gynecological malignancies. Epidemiological studies have demonstrated that steroid hormones released from the hypothalamic-pituitary-ovarian axis can play a role in stimulating or inhibiting OC progression, with gonadotropins, estrogens and androgens promoting OC progression, while gonadotropin-releasing hormone (GnRH) and progesterone may be protective factors in OC. Experimental studies have indicated that hormone receptors are expressed in OC cells and mediate the growth stimulatory or growth inhibitory effects of hormones on these cells. Hormone therapy agents have been evaluated in a number of clinical trials. The majority of these trials were conducted in patients with relapsed or refractory OC with average efficacy and limited side-effects. A better understanding of the mechanisms through which hormones affect cell growth may improve the efficacy of hormone therapy. In the present review article, the role of hormones (GnRH, gonadotropins, androgens, estrogens and progestins) and their receptors in OC tumorigenesis, and hormonal therapy in OC treatment is discussed and summarized.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children and Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Liu
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children and Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children and Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children and Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
18
|
Structure-property and structure-activity relationships of phenylferrocene derivatives as androgen receptor antagonists. Bioorg Med Chem Lett 2021; 46:128141. [PMID: 34048883 DOI: 10.1016/j.bmcl.2021.128141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/26/2022]
Abstract
Ferrocene is a representative organometallic compound having a sandwich structure with high stability and hydrophobicity. In this study, we determined the physicochemical properties of a series of nitro- and cyanophenylferrocenes, and evaluated their biological activity as androgen receptor (AR) antagonists. Ferrocene derivatives exhibited hydrophobicity parameter π values in the range between 2.54 and 3.23, depending on the substituents, indicating that the hydrophobicity of ferrocene is suitable for its application as a hydrophobic core structure of nuclear receptor ligands. The synthesized ferrocene derivatives showed AR-antagonistic activity, and among them, 3-nitrophenylferrocene 14 exhibited the most potent activity with an IC50 value of 0.28 μM. The developed compounds may be candidates for further structural development as AR antagonists. These findings also support the utility of organometallic species as structural options for drug discovery.
Collapse
|
19
|
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: 1.0] [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
|
20
|
An Overview of Next-Generation Androgen Receptor-Targeted Therapeutics in Development for the Treatment of Prostate Cancer. Int J Mol Sci 2021; 22:ijms22042124. [PMID: 33672769 PMCID: PMC7924596 DOI: 10.3390/ijms22042124] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Traditional endocrine therapy for prostate cancer (PCa) has been directed at suppression of the androgen receptor (AR) signaling axis since Huggins et al. discovered that diethylstilbestrol (DES; an estrogen) produced chemical castration and PCa tumor regression. Androgen deprivation therapy (ADT) still remains the first-line PCa therapy. Insufficiency of ADT over time leads to castration-resistant PCa (CRPC) in which the AR axis is still active, despite castrate levels of circulating androgens. Despite the approval and use of multiple generations of competitive AR antagonists (antiandrogens), antiandrogen resistance emerges rapidly in CRPC due to several mechanisms, mostly converging in the AR axis. Recent evidence from multiple groups have defined noncompetitive or noncanonical direct binding sites on AR that can be targeted to inhibit the AR axis. This review discusses new developments in the PCa treatment paradigm that includes the next-generation molecules to noncanonical sites, proteolysis targeting chimera (PROTAC), or noncanonical N-terminal domain (NTD)-binding of selective AR degraders (SARDs). A few lead compounds targeting each of these novel noncanonical sites or with SARD activity are discussed. Many of these ligands are still in preclinical development, and a few early clinical leads have emerged, but successful late-stage clinical data are still lacking. The breadth and diversity of targets provide hope that optimized noncanonical inhibitors and/or SARDs will be able to overcome antiandrogen-resistant CRPC.
Collapse
|
21
|
Sang H, Wang Y, Zhong Y, Gu S, Wang G, Sun J, Peng Y. Quantitative determination of proxalutamide in rat plasma and tissues using liquid chromatography/tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e9003. [PMID: 33169448 DOI: 10.1002/rcm.9003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/28/2020] [Accepted: 11/04/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Proxalutamide is a novel drug for the treatment of prostate cancer. However, to date, there are almost no reports on the pharmacokinetics of proxalutamide in vivo. This study developed a liquid chromatography/tandem mass spectrometry (LC/MS/MS) method to determine the concentrations of proxalutamide in biological samples for pharmacokinetic studies. METHODS Chromatographic separation was achieved on a Kromasil 100-5C8 column followed by gradient elution using a Shimadzu HPLC system. MS was performed in positive ion electrospray ionization mode using a SCIEX API 4000 triple quadrupole system. A simple and rapid one-step protein precipitation method was used for sample processing, and a low sample volume of 10 μL was used for processing and analysis. RESULTS The method was validated to show good selectivity, sensitivity, precision, and accuracy. Good linearity (r2 > 0.99) was observed for rat plasma (range: 2-5000 ng/mL) and rat tissue homogenates (range: 2-2000 ng/mL). The extraction recovery was above 98%, and no significant matrix effect was observed. This method was successfully applied to investigate the pharmacokinetics and tissue distribution of proxalutamide in rats. CONCLUSIONS A rapid and sensitive LC/MS/MS method was developed and validated to determine the quantity of proxalutamide in rat plasma and tissue homogenates and to further study the pharmacokinetic parameters of proxalutamide in a rat model. The results showed that proxalutamide had good oral bioavailability and wide tissue distribution in vivo.
Collapse
Affiliation(s)
- Hua Sang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, The Affiliated Hospital of Nantong University, Nantong, China
- School of Pharmacy, Nantong University, Nantong, China
| | - Yu Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yunxi Zhong
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Shiyin Gu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianguo Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ying Peng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
22
|
Masamrekh RA, Filippova TA, Sherbakov KA, Veselovsky AV, Shumyantseva VV, Kuzikov AV. Interactions of galeterone and its 3-keto-Δ4 metabolite (D4G) with one of the key enzymes of corticosteroid biosynthesis - steroid 21-monooxygenase (CYP21A2). Fundam Clin Pharmacol 2020; 35:423-431. [PMID: 33012006 DOI: 10.1111/fcp.12607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/11/2020] [Accepted: 09/28/2020] [Indexed: 11/30/2022]
Abstract
We have investigated interactions of galeterone and its pharmacologically active metabolite - 3-keto-Δ4-galeterone (D4G) - with one of the key enzymes of corticosteroid biosynthesis - steroid 21-monooxygenase (CYP21A2). It was shown by absorption spectroscopy that both compounds induce type I spectral changes of CYP21A2. Spectral dissociation constants (KS ) of complexes of CYP21A2 with galeterone or D4G were calculated as 3.1 ± 0.7 μm and 4.6 ± 0.4 μm, respectively. It was predicted by molecular docking that both ligands similarly bind to the active site of CYP21A2. We have revealed using reconstituted monooxygenase system that galeterone is a competitive inhibitor of CYP21A2 with the inhibition constant (Ki ) value of 12 ± 3 μm, while D4G at the concentrations of 10 and 25 μm does not inhibit the enzyme. Summarizing, based on the in vitro analyses we detected inhibition of CYP21A2 by galeterone and lack of the influence of D4G on this enzyme.
Collapse
Affiliation(s)
- Rami A Masamrekh
- Pirogov Russian National Research Medical University, Ostrovitianov Street, 1, Moscow, 117997, Russia.,Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| | - Tatiana A Filippova
- Pirogov Russian National Research Medical University, Ostrovitianov Street, 1, Moscow, 117997, Russia.,Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| | - Kirill A Sherbakov
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| | - Alexander V Veselovsky
- Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| | - Victoria V Shumyantseva
- Pirogov Russian National Research Medical University, Ostrovitianov Street, 1, Moscow, 117997, Russia.,Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| | - Alexey V Kuzikov
- Pirogov Russian National Research Medical University, Ostrovitianov Street, 1, Moscow, 117997, Russia.,Institute of Biomedical Chemistry, Pogodinskaya Street, 10, Build 8, Moscow, 119121, Russia
| |
Collapse
|
23
|
Dual inhibitors of histone deacetylases and other cancer-related targets: A pharmacological perspective. Biochem Pharmacol 2020; 182:114224. [PMID: 32956642 DOI: 10.1016/j.bcp.2020.114224] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Epigenetic enzymes histone deacetylases (HDACs) are clinically validated anticancer drug targets which have been studied intensively in the past few decades. Although several drugs have been approved in this field, they are still limited to a subset of hematological malignancies (in particular T-cell lymphomas), with therapeutic potential not fully realized and the drug-resistance occurred after a certain period of use. To maximize the therapeutic potential of these classes of anticancer drugs, and to extend their application to solid tumors, numerous combination therapies containing an HDACi and an anticancer agent from other mechanisms are currently ongoing in clinical trials. Recently, dual targeting strategy comprising the HDACs component has emerged as an alternative approach for combination therapies. In this perspective, we intend to gather all HDACs-containing dual inhibitors related to cancer therapy published in literature since 2015, classify them into five categories based on targets' biological functions, and discuss the rationale why dual acting agents should work better than combinatorial therapies using two separate drugs. The article discusses the pharmacological aspects of these dual inhibitors, including in vitro biological activities, pharmacokinetic studies, in vivo efficacy studies, as well as available clinical trials. The review of the current status and advances should provide better analysis for future opportunities and challenges of this field.
Collapse
|
24
|
Koga H, Negishi M, Kinoshita M, Fujii S, Mori S, Ishigami-Yuasa M, Kawachi E, Kagechika H, Tanatani A. Development of Androgen-Antagonistic Coumarinamides with a Unique Aromatic Folded Pharmacophore. Int J Mol Sci 2020; 21:ijms21155584. [PMID: 32759847 PMCID: PMC7432827 DOI: 10.3390/ijms21155584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 01/06/2023] Open
Abstract
First-generation nonsteroidal androgen receptor (AR) antagonists, such as flutamide (2a) and bicalutamide (3), are effective for most prostate cancer patients, but resistance often appears after several years due to the mutation of AR. Second-generation AR antagonists are effective against some of these castration-resistant prostate cancers, but their structural variety is still limited. In this study, we designed and synthesized 4-methyl-7-(N-alkyl-arylcarboxamido)coumarins as AR antagonist candidates and evaluated their growth-inhibitory activity toward androgen-dependent SC-3 cells. Coumarinamides with a secondary amide bond did not show inhibitory activity, but their N-methylated derivatives exhibited AR-antagonistic activity. Especially, 19b and 31b were more potent than the lead compound 7b, which was comparable to hydroxyflutamide (2b). Conformational analysis showed that the inactive coumarinamides with a secondary amide bond have an extended structure with a trans-amide bond, while the active N-methylated coumarinamides have a folded structure with a cis-amide bond, in which the two aromatic rings are placed face-to-face. Docking study suggested that this folded structure is important for binding to AR. Selected coumarinamide derivatives showed AR-antagonistic activity toward LNCaP cells with T877A AR, and they had weak progesterone receptor (PR)-antagonistic activity. The folded coumarinamide structure appears to be a unique pharmacophore, different from those of conventional AR antagonists.
Collapse
Affiliation(s)
- Hitomi Koga
- Department of Chemistry, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan; (H.K.); (M.N.); (M.K.)
| | - Mai Negishi
- Department of Chemistry, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan; (H.K.); (M.N.); (M.K.)
| | - Marie Kinoshita
- Department of Chemistry, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan; (H.K.); (M.N.); (M.K.)
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; (S.F.); (S.M.); (M.I.-Y.); (E.K.)
| | - Shuichi Mori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; (S.F.); (S.M.); (M.I.-Y.); (E.K.)
| | - Mari Ishigami-Yuasa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; (S.F.); (S.M.); (M.I.-Y.); (E.K.)
| | - Emiko Kawachi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; (S.F.); (S.M.); (M.I.-Y.); (E.K.)
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan; (S.F.); (S.M.); (M.I.-Y.); (E.K.)
- Correspondence: (H.K.); (A.T.); Tel.: +81-3-5280-8032 (H.K.); +81-3-5978-2716 (A.T.)
| | - Aya Tanatani
- Department of Chemistry, Faculty of Science, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan; (H.K.); (M.N.); (M.K.)
- Institute of Human Life Innovation, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan
- Correspondence: (H.K.); (A.T.); Tel.: +81-3-5280-8032 (H.K.); +81-3-5978-2716 (A.T.)
| |
Collapse
|
25
|
Boukovala M, Spetsieris N, Efstathiou E. An evaluation of apalutamide for the treatment of prostate cancer. Expert Opin Pharmacother 2020; 21:1537-1546. [DOI: 10.1080/14656566.2020.1770726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Myrto Boukovala
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Nicholas Spetsieris
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
26
|
Thevis M, Walpurgis K, Thomas A. Analytical Approaches in Human Sports Drug Testing: Recent Advances, Challenges, and Solutions. Anal Chem 2019; 92:506-523. [DOI: 10.1021/acs.analchem.9b04639] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mario Thevis
- Center for Preventive Doping Research, Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne 50933, Germany
- European Monitoring Center for Emerging Doping Agents (EuMoCEDA), Cologne 50933, Germany
| | - Katja Walpurgis
- Center for Preventive Doping Research, Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne 50933, Germany
| | - Andreas Thomas
- Center for Preventive Doping Research, Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne 50933, Germany
| |
Collapse
|