1
|
El Omari N, Khalid A, Makeen HA, Alhazmi HA, Albratty M, Mohan S, Tan CS, Ming LC, Chook JB, Bouyahya A. Stochasticity of anticancer mechanisms underlying clinical effectiveness of vorinostat. Heliyon 2024; 10:e33052. [PMID: 39021957 PMCID: PMC11253278 DOI: 10.1016/j.heliyon.2024.e33052] [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: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
The Food and Drug Administration (FDA) has approved vorinostat, also called Zolinza®, for its effectiveness in fighting cancer. This drug is a suberoyl-anilide hydroxamic acid belonging to the class of histone deacetylase inhibitors (HDACis). Its HDAC inhibitory potential allows it to accumulate acetylated histones. This, in turn, can restore normal gene expression in cancer cells and activate multiple signaling pathways. Experiments have proven that vorinostat induces histone acetylation and cytotoxicity in many cancer cell lines, increases the level of p21 cell cycle proteins, and enhances pro-apoptotic factors while decreasing anti-apoptotic factors. Additionally, it regulates the immune response by up-regulating programmed death-ligand 1 (PD-L1) and interferon gamma receptor 1 (IFN-γR1) expression, and can impact proteasome and/or aggresome degradation, endoplasmic reticulum function, cell cycle arrest, apoptosis, tumor microenvironment remodeling, and angiogenesis inhibition. In this study, we sought to elucidate the precise molecular mechanism by which Vorinostat inhibits HDACs. A deeper understanding of these mechanisms could improve our understanding of cancer cell abnormalities and provide new therapeutic possibilities for cancer treatment.
Collapse
Affiliation(s)
- Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum, 11111, Sudan
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Postal Code 45142, Jazan, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ching Siang Tan
- School of Pharmacy, KPJ Healthcare University, Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Jack Bee Chook
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco
| |
Collapse
|
2
|
Chandrasekaran B, Tapadar S, Wu B, Saran U, Tyagi A, Johnston A, Gaul DA, Oyelere AK, Damodaran C. Antiandrogen-Equipped Histone Deacetylase Inhibitors Selectively Inhibit Androgen Receptor (AR) and AR-Splice Variant (AR-SV) in Castration-Resistant Prostate Cancer (CRPC). Cancers (Basel) 2023; 15:1769. [PMID: 36980655 PMCID: PMC10046692 DOI: 10.3390/cancers15061769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Epigenetic modification influences androgen receptor (AR) activation, often resulting in prostate cancer (PCa) development and progression. Silencing histone-modifying enzymes (histone deacetylases-HDACs) either genetically or pharmacologically suppresses PCa proliferation in preclinical models of PCa; however, results from clinical studies were not encouraging. Similarly, PCa patients eventually become resistant to androgen ablation therapy (ADT). Our goal is to develop dual-acting small molecules comprising antiandrogen and HDAC-inhibiting moieties that may overcome the resistance of ADT and effectively suppress the growth of castration-resistant prostate cancer (CRPC). METHODS Several rationally designed antiandrogen-equipped HDAC inhibitors (HDACi) were synthesized, and their efficacy on CRPC growth was examined both in vitro and in vivo. RESULTS While screening our newly developed small molecules, we observed that SBI-46 significantly inhibited the proliferation of AR+ CRPC cells but not AR- CRPC and normal immortalized prostate epithelial cells (RWPE1) or normal kidney cells (HEK-293 and VERO). Molecular analysis confirmed that SBI-46 downregulated the expressions of both AR+ and AR-splice variants (AR-SVs) in CRPC cells. Further studies revealed the downregulation of AR downstream (PSA) events in CRPC cells. The oral administration of SBI-46 abrogated the growth of C4-2B and 22Rv1 CRPC xenograft tumors that express AR or both AR and AR-SV in xenotransplanted nude mice models. Further, immunohistochemical analysis confirmed that SBI-46 inhibits AR signaling in xenografted tumor tissues. CONCLUSION These results demonstrate that SBI-46 is a potent agent that inhibits preclinical models of CRPC by downregulating the expressions of both AR and AR-SV. Furthermore, these results suggest that SBI-46 may be a potent compound for treating CRPC.
Collapse
Affiliation(s)
| | - Subhasish Tapadar
- Parker H. Petit Institute for Bioengineering & Biosciences, School of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Bocheng Wu
- Parker H. Petit Institute for Bioengineering & Biosciences, School of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Uttara Saran
- Rangel School of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | - Ashish Tyagi
- Rangel School of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | - Alexis Johnston
- Parker H. Petit Institute for Bioengineering & Biosciences, School of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - David A. Gaul
- Parker H. Petit Institute for Bioengineering & Biosciences, School of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Adegboyega K. Oyelere
- Parker H. Petit Institute for Bioengineering & Biosciences, School of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Chendil Damodaran
- Rangel School of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| |
Collapse
|
3
|
Pramanik SD, Kumar Halder A, Mukherjee U, Kumar D, Dey YN, R M. Potential of histone deacetylase inhibitors in the control and regulation of prostate, breast and ovarian cancer. Front Chem 2022; 10:948217. [PMID: 36034650 PMCID: PMC9411967 DOI: 10.3389/fchem.2022.948217] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play a role in chromatin remodeling and epigenetics. They belong to a specific category of enzymes that eliminate the acetyl part of the histones' -N-acetyl lysine, causing the histones to be wrapped compactly around DNA. Numerous biological processes rely on HDACs, including cell proliferation and differentiation, angiogenesis, metastasis, gene regulation, and transcription. Epigenetic changes, specifically increased expression and activity of HDACs, are commonly detected in cancer. As a result, HDACi could be used to develop anticancer drugs. Although preclinical outcomes with HDACs as monotherapy have been promising clinical trials have had mixed results and limited success. In both preclinical and clinical trials, however, combination therapy with different anticancer medicines has proved to have synergistic effects. Furthermore, these combinations improved efficacy, decreased tumor resistance to therapy, and decreased toxicity. In the present review, the detailed modes of action, classification of HDACs, and their correlation with different cancers like prostate, breast, and ovarian cancer were discussed. Further, the different cell signaling pathways and the structure-activity relationship and pharmaco-toxicological properties of the HDACi, and their synergistic effects with other anticancer drugs observed in recent preclinical and clinical studies used in combination therapy were discussed for prostate, breast, and ovarian cancer treatment.
Collapse
Affiliation(s)
- Siddhartha Das Pramanik
- Department of Pharmaceutical Engineering and Technology, IIT-BHU, Varanasi, Uttar Pradesh, India
| | - Amit Kumar Halder
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Ushmita Mukherjee
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Dharmendra Kumar
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Sasaram, Bihar, India
| | - Yadu Nandan Dey
- Dr. B.C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, West Bengal, India
| | - Mogana R
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI Education SDN.BHD., Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Deng T, Xiao Y, Dai Y, Xie L, Li X. Roles of Key Epigenetic Regulators in the Gene Transcription and Progression of Prostate Cancer. Front Mol Biosci 2021; 8:743376. [PMID: 34977151 PMCID: PMC8714908 DOI: 10.3389/fmolb.2021.743376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/25/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is a top-incidence malignancy, and the second most common cause of death amongst American men and the fifth leading cause of cancer death in men around the world. Androgen receptor (AR), the key transcription factor, is critical for the progression of PCa by regulating a series of target genes by androgen stimulation. A number of co-regulators of AR, including co-activators or co-repressors, have been implicated in AR-mediated gene transcription and PCa progression. Epigenetic regulators, by modifying chromatin integrity and accessibility for transcription regulation without altering DNA sequences, influence the transcriptional activity of AR and further regulate the gene expression of AR target genes in determining cell fate, PCa progression and therapeutic response. In this review, we summarized the structural interaction of AR and epigenetic regulators including histone or DNA methylation, histone acetylation or non-coding RNA, and functional synergy in PCa progression. Importantly, epigenetic regulators have been validated as diagnostic markers and therapeutic targets. A series of epigenetic target drugs have been developed, and have demonstrated the potential to treat PCa alone or in combination with antiandrogens.
Collapse
Affiliation(s)
- Tanggang Deng
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yugang Xiao
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Dai
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Xie
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiong Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
5
|
Barrett RRG, Nash C, Diennet M, Cotnoir-White D, Doyle C, Mader S, Thomson AA, Gleason JL. Dual-function antiandrogen/HDACi hybrids based on enzalutamide and entinostat. Bioorg Med Chem Lett 2021; 55:128441. [PMID: 34767912 DOI: 10.1016/j.bmcl.2021.128441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 11/02/2022]
Abstract
The combination of androgen receptor antagonists with histone deacetylase inhibitors (HDACi) has been shown to be more effective than antiandrogens alone in halting growth of prostate cancer cell lines. Here we have designed, synthesized and assessed a series of antiandrogen/HDACi hybrids by combining structural features of enzalutamide with either SAHA or entinostat. The hybrids are demonstrated to maintain bifunctionality using a fluorometric HDAC assay and a bioluminescence resonance energy transfer (BRET) antiandrogen assay. Antiproliferative assays showed that hybrids bearing o-aminoanilide-based HDACi motifs outperformed hydroxamic acid based HDACi's. The hybrids demonstrated selectivity for epithelial cell lines vs. stromal cell lines, suggesting a potentially useful therapeutic window.
Collapse
Affiliation(s)
- Ryan R G Barrett
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada
| | - Claire Nash
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Marine Diennet
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada
| | - David Cotnoir-White
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada
| | - Christopher Doyle
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada
| | - Sylvie Mader
- Institute for Research in Immunology and Cancer, Pavillon Marcelle Coutu, Université de Montréal, 2950 chemin de Polytechnique, Montreal, QC H3T1J4, Canada; Department of Biochemistry and Molecular Medicine, Pavillon Roger Gaudry, Université de Montréal, 2900 bd Edouard Montpetit, Montreal, QC H3T1J4, Canada
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - James L Gleason
- Department of Chemistry, McGill University, 801 Sherbrooke W., Montreal, QC H3A 0B8, Canada.
| |
Collapse
|
6
|
Liu P, Xiao J, Wang Y, Song X, Huang L, Ren Z, Kitazato K, Wang Y. Posttranslational modification and beyond: interplay between histone deacetylase 6 and heat-shock protein 90. Mol Med 2021; 27:110. [PMID: 34530730 PMCID: PMC8444394 DOI: 10.1186/s10020-021-00375-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
Posttranslational modification (PTM) and regulation of protein stability are crucial to various biological processes. Histone deacetylase 6 (HDAC6), a unique histone deacetylase with two functional catalytic domains (DD1 and DD2) and a ZnF-UBP domain (ubiquitin binding domain, BUZ), regulates a number of biological processes, including gene expression, cell motility, immune response, and the degradation of misfolded proteins. In addition to the deacetylation of histones, other nonhistone proteins have been identified as substrates for HDAC6. Hsp90, a molecular chaperone that is a critical modulator of cell signaling, is one of the lysine deacetylase substrates of HDAC6. Intriguingly, as one of the best-characterized regulators of Hsp90 acetylation, HDAC6 is the client protein of Hsp90. In addition to regulating Hsp90 at the post-translational modification level, HDAC6 also regulates Hsp90 at the gene transcription level. HDAC6 mainly regulates the Hsp90-HSF1 complex through the ZnF-UBP domain, thereby promoting the HSF1 entry into the nucleus and activating gene transcription. The mutual interaction between HDAC6 and Hsp90 plays an important role in the regulation of protein stability, cell migration, apoptosis and other functions. Plenty of of studies have indicated that blocking HDAC6/Hsp90 has a vital regulatory role in multifarious diseases, mainly in cancers. Therefore, developing inhibitors or drugs against HDAC6/Hsp90 becomes a promising development direction. Herein, we review the current knowledge on molecular regulatory mechanisms based on the interaction of HDAC6 and Hsp90 and inhibition of HDAC6 and/or Hsp90 in oncogenesis and progression, antiviral and immune-related diseases and other vital biological processes.
Collapse
Affiliation(s)
- Ping Liu
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Ji Xiao
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Yiliang Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Xiaowei Song
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Lianzhou Huang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhe Ren
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Department of Clinical Research Pharmacy, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, China.
| |
Collapse
|
7
|
Chan AM, Fletcher S. Shifting the paradigm in treating multi-factorial diseases: polypharmacological co-inhibitors of HDAC6. RSC Med Chem 2021; 12:178-196. [PMID: 34046608 PMCID: PMC8127619 DOI: 10.1039/d0md00286k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/28/2020] [Indexed: 01/20/2023] Open
Abstract
Multi-factorial diseases are illnesses that exploit multiple cellular processes, or stages within one process, and thus highly targeted therapies often succumb to the disease, losing efficacy as resistance sets in. Combination therapies have become a mainstay to battle these diseases, however these regimens are plagued with caveats. An emerging avenue to treat multi-factorial diseases is polypharmacology, wherein a single drug is rationally designed to bind multiple targets, and is widely touted to be superior to combination therapy by inherently addressing the latter's shortcomings, which include poor patient compliance, narrow therapeutic windows and spiraling healthcare costs. Through its roles in intracellular trafficking, cell motility, mitosis, protein folding and as a back-up to the proteasome pathway, HDAC6 has rapidly become an exciting new target for therapeutics, particularly in the discovery of new drugs to treat Alzheimer's disease and cancer. Herein, we describe recent efforts to marry together HDAC pharmacophores, with a particular emphasis on HDAC6 selectivity, with those of other targets towards the discovery of potent therapeutics to treat these evasive diseases. Such polypharmacological agents may supercede combination therapies through inherent synergism, permitting reduced dosing, wider therapeutic windows and improved compliance.
Collapse
Affiliation(s)
- Alexandria M Chan
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N Pine St Baltimore MD 21201 USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy 20 N Pine St Baltimore MD 21201 USA
- University of Maryland Greenebaum Cancer Center 22 S Greene St Baltimore MD 21201 USA
| |
Collapse
|
8
|
Zhou M, Zheng H, Li Y, Huang H, Min X, Dai S, Zhou W, Chen Z, Xu G, Chen Y. Discovery of a novel AR/HDAC6 dual inhibitor for prostate cancer treatment. Aging (Albany NY) 2021; 13:6982-6998. [PMID: 33621955 PMCID: PMC7993727 DOI: 10.18632/aging.202554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/23/2020] [Indexed: 11/25/2022]
Abstract
Androgen receptor (AR) and histone deacetylase 6 (HDAC6) are important targets for cancer therapy. Given that both AR antagonists and HDAC6 inhibitors modulate AR signaling, a novel AR/HDAC6 dual inhibitor is investigated for its anticancer effects in castration-resistant prostate cancer (CRPC). Zeta55 inhibits nuclear translocation of AR and suppresses androgen-induced PSA and TMPRSS2 expression. Meanwhile, Zeta55 selectively inhibits HDAC6 activity, leading to AR degradation. Zeta55 reduces the growth of AR-overexpressing VCaP prostate cancer cells both in vitro and in a CRPC xenograft model. These results provide preclinical proof of principle for Zeta55 as a promising therapeutic in prostate cancer treatment.
Collapse
Affiliation(s)
- Maojun Zhou
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hao Zheng
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine, Ministry of Educational of China, Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yubin Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Huichao Huang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiaoli Min
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shuyan Dai
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | | | - Zhuchu Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Guangyu Xu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine, Ministry of Educational of China, Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, National Center for Geriatrics Clinical Research, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
9
|
Hydroxamic acid hybrids as the potential anticancer agents: An Overview. Eur J Med Chem 2020; 205:112679. [PMID: 32791404 DOI: 10.1016/j.ejmech.2020.112679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
|
10
|
Bass AKA, El-Zoghbi MS, Nageeb ESM, Mohamed MFA, Badr M, Abuo-Rahma GEDA. Comprehensive review for anticancer hybridized multitargeting HDAC inhibitors. Eur J Med Chem 2020; 209:112904. [PMID: 33077264 DOI: 10.1016/j.ejmech.2020.112904] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/18/2020] [Accepted: 09/30/2020] [Indexed: 02/08/2023]
Abstract
Despite the encouraging clinical progress of chemotherapeutic agents in cancer treatment, innovation and development of new effective anticancer candidates still represents a challenging endeavor. With 15 million death every year in 2030 according to the estimates, cancer has increased rising of an alarm as a real crisis for public health and health systems worldwide. Therefore, scientist began to introduce innovative solutions to control the cancer global health problem. One of the promising strategies in this issue is the multitarget or smart hybrids having two or more pharmacophores targeting cancer. These rationalized hybrid molecules have gained great interests in cancer treatment as they are capable to simultaneously inhibit more than cancer pathway or target without drug-drug interactions and with less side effects. A prime important example of these hybrids, the HDAC hybrid inhibitors or referred as multitargeting HDAC inhibitors. The ability of HDAC inhibitors to synergistically improve the efficacy of other anti-cancer drugs and moreover, the ease of HDAC inhibitors cap group modification prompt many medicinal chemists to innovate and develop new generation of HDAC hybrid inhibitors. Notably, and during this short period, there are four HDAC inhibitor hybrids have entered different phases of clinical trials for treatment of different types of blood and solid tumors, namely; CUDC-101, CUDC-907, Tinostamustine, and Domatinostat. This review shed light on the most recent hybrids of HDACIs with one or more other cancer target pharmacophore. The designed multitarget hybrids include topoisomerase inhibitors, kinase inhibitors, nitric oxide releasers, antiandrogens, FLT3 and JAC-2 inhibitors, PDE5-inhibitors, NAMPT-inhibitors, Protease inhibitors, BRD4-inhibitors and other targets. This review may help researchers in development and discovery of new horizons in cancer treatment.
Collapse
Affiliation(s)
- Amr K A Bass
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - Mona S El-Zoghbi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - El-Shimaa M Nageeb
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt
| | - Mohamed Badr
- Department of Biochemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
| | - Gamal El-Din A Abuo-Rahma
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, New Minia, Minia, Egypt.
| |
Collapse
|
11
|
Vaidya GN, Rana P, Venkatesh A, Chatterjee DR, Contractor D, Satpute DP, Nagpure M, Jain A, Kumar D. Paradigm shift of "classical" HDAC inhibitors to "hybrid" HDAC inhibitors in therapeutic interventions. Eur J Med Chem 2020; 209:112844. [PMID: 33143937 DOI: 10.1016/j.ejmech.2020.112844] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
'Epigenetic' regulation of genes via post-translational modulation of proteins is the current mainstay approach for the disease therapies, particularly explored in the Histone Deacetylase (HDAC) class of enzymes. Mainly sight saw in cancer chemotherapeutics, HDAC inhibitors have also found a promising role in other diseases (neurodegenerative disorders, cardiovascular diseases, and viral infections) and successfully entered in various combination therapies (pre-clinical/clinical stages). The prevalent flexibility in the structural design of HDAC inhibitors makes them easily tuneable to merge with other pharmacophore modules for generating multi-targeted single hybrids as a novel tactic to overcome drawbacks of polypharmacy. Herein, we reviewed the putative role of prevalent HDAC hybrids inhibitors in the current and prospective stage as a translational approach to overcome the limitations of the existing conventional drug candidates (parent molecule) when used either alone (drug resistance, solubility issues, adverse side effects, selectivity profile) or in combination (pharmacokinetic interactions, patient compliance) for treating various diseases.
Collapse
Affiliation(s)
- Gargi Nikhil Vaidya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Pooja Rana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Ashwini Venkatesh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Darshan Contractor
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Dinesh Parshuram Satpute
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Mithilesh Nagpure
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India; Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi, India.
| | - Dinesh Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
12
|
Cerasuolo M, Maccarinelli F, Coltrini D, Mahmoud AM, Marolda V, Ghedini GC, Rezzola S, Giacomini A, Triggiani L, Kostrzewa M, Verde R, Paris D, Melck D, Presta M, Ligresti A, Ronca R. Modeling Acquired Resistance to the Second-Generation Androgen Receptor Antagonist Enzalutamide in the TRAMP Model of Prostate Cancer. Cancer Res 2020; 80:1564-1577. [PMID: 32029552 DOI: 10.1158/0008-5472.can-18-3637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 10/28/2019] [Accepted: 01/30/2020] [Indexed: 11/16/2022]
Abstract
Enzalutamide (MDV3100) is a potent second-generation androgen receptor antagonist approved for the treatment of castration-resistant prostate cancer (CRPC) in chemotherapy-naïve as well as in patients previously exposed to chemotherapy. However, resistance to enzalutamide and enzalutamide withdrawal syndrome have been reported. Thus, reliable and integrated preclinical models are required to elucidate the mechanisms of resistance and to assess therapeutic settings that may delay or prevent the onset of resistance. In this study, the prostate cancer multistage murine model TRAMP and TRAMP-derived cells have been used to extensively characterize in vitro and in vivo the response and resistance to enzalutamide. The therapeutic profile as well as the resistance onset were characterized and a multiscale stochastic mathematical model was proposed to link the in vitro and in vivo evolution of prostate cancer. The model showed that all therapeutic strategies that use enzalutamide result in the onset of resistance. The model also showed that combination therapies can delay the onset of resistance to enzalutamide, and in the best scenario, can eliminate the disease. These results set the basis for the exploitation of this "TRAMP-based platform" to test novel therapeutic approaches and build further mathematical models of combination therapies to treat prostate cancer and CRPC.Significance: Merging mathematical modeling with experimental data, this study presents the "TRAMP-based platform" as a novel experimental tool to study the in vitro and in vivo evolution of prostate cancer resistance to enzalutamide.
Collapse
Affiliation(s)
- Marianna Cerasuolo
- School of Mathematics and Physics, University of Portsmouth, Hampshire, United Kingdom
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Coltrini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ali Mokhtar Mahmoud
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Viviana Marolda
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Gaia Cristina Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Roberta Verde
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Dominique Melck
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy.
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
13
|
Abazid A, Martin B, Choinowski A, McNeill RV, Brandenburg LO, Ziegler P, Zimmermann U, Burchardt M, Erb H, Stope MB. The androgen receptor antagonist enzalutamide induces apoptosis, dysregulates the heat shock protein system, and diminishes the androgen receptor and estrogen receptor β1 expression in prostate cancer cells. J Cell Biochem 2019; 120:16711-16722. [PMID: 31297844 DOI: 10.1002/jcb.28929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 11/05/2022]
Abstract
Enzalutamide's accepted mode of action is by targeting the androgen receptor's (AR) activity. In clinical practice, enzalutamide demonstrates a good benefit-risk profile for the treatment of advanced prostate cancer (PC), even after poor response to standard antihormonal treatment. However, since both, well-established antiandrogens and enzalutamide, target AR functionality, we hypothesized that additional unknown mechanisms might be responsible for enzalutamide's superior anticancer activity. In the current study, PC cells were incubated with enzalutamide and enzalutamide-dependent modulation of apoptotic mechanisms were assessed via Western blot analysis, TDT-mediated dUTP-biotin nick end-labeling assay, and nuclear morphology assay. Alterations of heat shock protein (HSP), AR, and estrogen receptor (ER) expression were examined by Western blot analysis. Enzalutamide attenuated the proliferation of PC cells in a time- and dose-dependent manner. In the presence of enzalutamide, apoptosis occurred which was shown by increased BAX expression, decreased Bcl-2 expression, nuclear pyknosis, and genomic DNA fragmentation. Moreover, enzalutamide inhibited the expression of HSPs primarily involved in steroid receptor stabilization and suppressed AR and ERβ1 expression. This study demonstrates for the first time that enzalutamide treatment of PC cells triggers varying molecular mechanisms resulting in antiproliferative effects of the drug. In addition to the well-characterized antagonistic inhibition of AR functionality, we have shown that enzalutamide also affects the intracellular synthesis of steroid receptor-associated HSPs, thereby diminishing the expression of AR and ERβ1 proteins and inducing apoptotic pathways. According to an indirect attenuation of HSP-associated factors such as steroid receptors, endometrial carcinoma, uterine leiomyosarcoma, and mamma carcinoma cells also demonstrated inhibited cell growth in the presence of enzalutamide. Our data, therefore, suggest that enzalutamide's high efficacy is at least partially independent of AR and p53 protein expression, which are frequently lost in advanced PC.
Collapse
Affiliation(s)
- Alexander Abazid
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Benedikt Martin
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Anja Choinowski
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Rhiannon V McNeill
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany
| | | | - Patrick Ziegler
- Institute for Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany
| | - Uwe Zimmermann
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Holger Erb
- Department of Urology, University of Dresden, Dresden, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
14
|
Hu WY, Xu L, Chen B, Ou S, Muzzarelli KM, Hu DP, Li Y, Yang Z, Vander Griend DJ, Prins GS, Qin Z. Targeting prostate cancer cells with enzalutamide-HDAC inhibitor hybrid drug 2-75. Prostate 2019; 79:1166-1179. [PMID: 31135075 DOI: 10.1002/pros.23832] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The progression of castration-resistant prostate cancer (CRPC) still relies on the function of androgen receptor (AR), achieved by evolving mechanisms to reactivate AR signaling under hormonal therapy. Histone deacetylase inhibitors (HDACis) disrupt cytoplasmic AR chaperone heat shock protein 90 (Hsp90) via HDAC6 inhibition, leading to AR degradation and growth suppression of prostate cancer (PCa) cells. However, current HDACis are not effective in clinical trials treating CRPC. METHODS We designed hybrid molecules containing partial chemical scaffolds of AR antagonist enzalutamide (Enz) and HDACi suberoylanilide hydroxamic acid (SAHA) as new anti-PCa agents. We previously demonstrated that Enz-HDACi hybrid drug 2-75 targets both AR and Hsp90, which inhibits the growth of Enz-resistant C4-2 cells. In the current study, we further investigate the molecular and cellular actions of 2-75 and test its anti-PCa effects in vivo. RESULTS Compared with Enz, 2-75 had greater AR antagonistic effects by decreasing the stability, transcriptional activity, and nuclear translocation of intracellular AR. In addition to inhibition of full-length AR (FL AR), 2-75 downregulated the AR-V7 variant in multiple PCa cell lines. Mechanistic studies indicated that the AR affinity of 2-75 retains the drug in the cytoplasm of AR + PCa cells and further directs 2-75 to the AR-associated protein complex, which permits localized effects on AR-associated Hsp90. Further, unlike pan-HDACi SAHA, the cytoplasm-retaining property allows 2-75 to significantly inhibit cytoplasmic HDAC6 with limited impact on nuclear HDACs. These selective cytoplasmic actions of 2-75 overcome the unfavorable resistance and toxicity properties associated with classical AR antagonists, HDACis, and Hsp90 inhibitors. Finally, 2-75 showed greater antitumor activities than Enz in vivo on SQ xenografts derived from LNCaP cells. CONCLUSIONS Novel therapeutic strategy using newly designed 2-75 and related AR antagonist-HDACi hybrid drugs has great potential for effective treatment of CRPC.
Collapse
Affiliation(s)
- Wen-Yang Hu
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Bailing Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Siyu Ou
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Kendall M Muzzarelli
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan
| | - Dan-Ping Hu
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois
| | - Ye Li
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan
| | | | - Gail S Prins
- Department of Urology, University of Illinois at Chicago, Chicago, Illinois
| | - Zhihui Qin
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| |
Collapse
|
15
|
Hesham HM, Lasheen DS, Abouzid KA. Chimeric HDAC inhibitors: Comprehensive review on the HDAC-based strategies developed to combat cancer. Med Res Rev 2018; 38:2058-2109. [DOI: 10.1002/med.21505] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/29/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Heba M. Hesham
- Faculty of Pharmacy, Pharmaceutical Chemistry Department; Ain Shams University; Abbassia Cairo Egypt
| | - Deena S. Lasheen
- Faculty of Pharmacy, Pharmaceutical Chemistry Department; Ain Shams University; Abbassia Cairo Egypt
| | - Khaled A.M. Abouzid
- Faculty of Pharmacy, Pharmaceutical Chemistry Department; Ain Shams University; Abbassia Cairo Egypt
| |
Collapse
|
16
|
Jadhavar PS, Ramachandran SA, Riquelme E, Gupta A, Quinn KP, Shivakumar D, Ray S, Zende D, Nayak AK, Miglani SK, Sathe BD, Raja M, Farias O, Alfaro I, Belmar S, Guerrero J, Bernales S, Chakravarty S, Hung DT, Lindquist JN, Rai R. Targeting prostate cancer with compounds possessing dual activity as androgen receptor antagonists and HDAC6 inhibitors. Bioorg Med Chem Lett 2016; 26:5222-5228. [PMID: 27717544 DOI: 10.1016/j.bmcl.2016.09.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 01/19/2023]
Abstract
While enzalutamide and abiraterone are approved for treatment of metastatic castration-resistant prostate cancer (mCRPC), approximately 20-40% of patients have no response to these agents. It has been stipulated that the lack of response and the development of secondary resistance to these drugs may be due to the presence of AR splice variants. HDAC6 has a role in regulating the androgen receptor (AR) by modulating heat shock protein 90 (Hsp90) acetylation, which controls the nuclear localization and activation of the AR in androgen-dependent and independent scenarios. With dual-acting AR-HDAC6 inhibitors it should be possible to target patients who don't respond to enzalutamide. Herein, we describe the design, synthesis and biological evaluation of dual-acting compounds which target AR and are also specific towards HDAC6. Our efforts led to compound 10 which was found to have potent dual activity (HDAC6 IC50=0.0356μM and AR binding IC50=<0.03μM). Compound 10 was further evaluated for antagonist and other cell-based activities, in vitro stability and pharmacokinetics.
Collapse
Affiliation(s)
- Pradeep S Jadhavar
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Sreekanth A Ramachandran
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Eduardo Riquelme
- Fundación Ciencia y Vida, Avenida Zañartu 1482, Ñuñoa, Santiago 7780272, Chile
| | - Ashu Gupta
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Kevin P Quinn
- Medivation, 525 Market Street, 36th Floor, San Francisco, CA 94105, USA
| | | | | | - Dnyaneshwar Zende
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Anjan K Nayak
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Sandeep K Miglani
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Balaji D Sathe
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Mohd Raja
- Integral BioSciences Pvt. Ltd, C-64, Hosiery Complex Phase II Extension, Noida, Uttar Pradesh 201306, India
| | - Olivia Farias
- Fundación Ciencia y Vida, Avenida Zañartu 1482, Ñuñoa, Santiago 7780272, Chile
| | - Ivan Alfaro
- Fundación Ciencia y Vida, Avenida Zañartu 1482, Ñuñoa, Santiago 7780272, Chile
| | - Sebastián Belmar
- Fundación Ciencia y Vida, Avenida Zañartu 1482, Ñuñoa, Santiago 7780272, Chile
| | - Javier Guerrero
- Fundación Ciencia y Vida, Avenida Zañartu 1482, Ñuñoa, Santiago 7780272, Chile
| | | | | | - David T Hung
- Medivation, 525 Market Street, 36th Floor, San Francisco, CA 94105, USA
| | | | - Roopa Rai
- Medivation, 525 Market Street, 36th Floor, San Francisco, CA 94105, USA.
| |
Collapse
|