1
|
Gediya P, Parikh PK, Vyas VK, Ghate MD. Histone deacetylase 2: A potential therapeutic target for cancer and neurodegenerative disorders. Eur J Med Chem 2021; 216:113332. [PMID: 33714914 DOI: 10.1016/j.ejmech.2021.113332] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
Histone deacetylases (HDACs) have been implicated in a number of diseases including cancer, cardiovascular disorders, diabetes mellitus, neurodegenerative disorders and inflammation. For the treatment of epigenetically altered diseases such as cancer, HDAC inhibitors have made a significant progress in terms of development of isoform selective inhibitiors. Isoform specific HDAC inhibitors have less adverse events and better safety profile. A HDAC isoform i.e., HDAC2 demonstrated significant role in the development of variety of diseases, mainly involved in the cancer and neurodegenerative disorders. Discovery and development of selective HDAC2 inhibitors have a great potential for the treatment of target diseases. In the present compilation, we have reviewed the role of HDAC2 in progression of cancer and neurodegenerative disorders, and information on the drug development opportunities for selective HDAC2 inhibition.
Collapse
Affiliation(s)
- Piyush Gediya
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Palak K Parikh
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India; Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Manjunath D Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
2
|
Li R, Ling D, Tang T, Huang Z, Wang M, Ding Y, Liu T, Wei H, Xu W, Mao F, Zhu J, Li X, Jiang L, Li J. Discovery of Novel Plasmodium falciparum HDAC1 Inhibitors with Dual-Stage Antimalarial Potency and Improved Safety Based on the Clinical Anticancer Drug Candidate Quisinostat. J Med Chem 2021; 64:2254-2271. [PMID: 33541085 DOI: 10.1021/acs.jmedchem.0c02104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Previously, we identified the clinical anticancer drug candidate quisinostat as a novel and potent antimalarial lead compound. To further enhance the antimalarial effect and improve safety, 31 novel spirocyclic hydroxamic acid derivatives were synthesized based on the structure of quisinostat, and their antimalarial activities and cytotoxicity were evaluated. Among them, compound 11 displayed broad potency in vitro against several multiresistant malarial parasites, especially two artemisinin-resistant clinical isolates. Moreover, 11 could eliminate both liver and erythrocytic parasites in vivo, kill all morphological erythrocytic parasites with specific potency against schizonts, and show acceptable metabolic stability and pharmacokinetic properties. Western blot analysis, PfHDAC gene knockdown, and enzymatic inhibition experiments collectively confirmed that PfHDAC1 was the target of 11. In summary, 11 is a structurally novel PfHDAC1 inhibitor with the potential to prevent and cure malaria, overcome multidrug resistance, and provide a prospective prototype for antimalarial drug research.
Collapse
Affiliation(s)
- Ruoxi Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Dazheng Ling
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Tongke Tang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Zhenghui Huang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Manjiong Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Ding
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Hanwen Wei
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jin Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaokang Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lubin Jiang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, P.R. China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.,College of Pharmacy and Chemistry, Dali University, 5 Xue Ren Road, Dali 671000, China.,Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China
| |
Collapse
|
3
|
Tapadar S, Fathi S, Wu B, Sun CQ, Raji I, Moore SG, Arnold RS, Gaul DA, Petros JA, Oyelere AK. Liver-Targeting Class I Selective Histone Deacetylase Inhibitors Potently Suppress Hepatocellular Tumor Growth as Standalone Agents. Cancers (Basel) 2020; 12:E3095. [PMID: 33114147 PMCID: PMC7690782 DOI: 10.3390/cancers12113095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 01/06/2023] Open
Abstract
Dysfunctions in epigenetic regulation play critical roles in tumor development and progression. Histone deacetylases (HDACs) and histone acetyl transferase (HAT) are functionally opposing epigenetic regulators, which control the expression status of tumor suppressor genes. Upregulation of HDAC activities, which results in silencing of tumor suppressor genes and uncontrolled proliferation, predominates in malignant tumors. Inhibition of the deacetylase activity of HDACs is a clinically validated cancer therapy strategy. However, current HDAC inhibitors (HDACi) have elicited limited therapeutic benefit against solid tumors. Here, we disclosed a class of HDACi that are selective for sub-class I HDACs and preferentially accumulate within the normal liver tissue and orthotopically implanted liver tumors. We observed that these compounds possess exquisite on-target effects evidenced by their induction of dose-dependent histone H4 hyperacetylation without perturbation of tubulin acetylation status and G0/G1 cell cycle arrest. Representative compounds 2 and 3a are relatively non-toxic to mice and robustly suppressed tumor growths in an orthotopic model of HCC as standalone agents. Collectively, our results suggest that these compounds may have therapeutic advantage against HCC relative to the current systemic HDACi. This prospect merits further comprehensive preclinical investigations.
Collapse
Affiliation(s)
- Subhasish Tapadar
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
- Sophia Bioscience, Inc. 311 Ferst Drive NW, Ste. L1325A, Atlanta, GA 30332, USA;
| | - Shaghayegh Fathi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
| | - Bocheng Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
| | - Carrie Q. Sun
- Department of Urology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA 30322, USA; (C.Q.S.); (R.S.A.)
| | - Idris Raji
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
| | - Samuel G. Moore
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
| | - Rebecca S. Arnold
- Department of Urology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA 30322, USA; (C.Q.S.); (R.S.A.)
| | - David A. Gaul
- Sophia Bioscience, Inc. 311 Ferst Drive NW, Ste. L1325A, Atlanta, GA 30332, USA;
| | - John A. Petros
- Department of Urology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA 30322, USA; (C.Q.S.); (R.S.A.)
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA 30332, USA; (S.T.); (S.F.); (B.W.); (I.R.); (S.G.M.)
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA
| |
Collapse
|
4
|
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: 36] [Impact Index Per Article: 7.2] [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
|
5
|
Janas A, Pecyna P, Gajecka M, Bartl F, Przybylski P. Synthesis and Antibacterial Activity of New
N
‐Alkylammonium and Carbonate‐Triazole Derivatives within Desosamine of 14‐ and 15‐Membered Lactone Macrolides. ChemMedChem 2020; 15:1529-1551. [DOI: 10.1002/cmdc.202000273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/21/2020] [Indexed: 02/03/2023]
Affiliation(s)
- Anna Janas
- Faculty of ChemistryAdam Mickiewicz University Uniwersytetu Poznańskiego 8 61-614 Poznań Poland
| | - Paulina Pecyna
- Chair and Department of Genetics and Pharmaceutical MicrobiologyPoznań University of Medical Sciences (PUMS) Święcickiego 4 60-781 Poznań Poland
| | - Marzena Gajecka
- Chair and Department of Genetics and Pharmaceutical MicrobiologyPoznań University of Medical Sciences (PUMS) Święcickiego 4 60-781 Poznań Poland
- Institute of Human GeneticsPolish Academy of Sciences Strzeszynska 32 60-479 Poznań Poland
| | - Franz Bartl
- Lebenswissenschaftliche Fakultät, Institut für Biologie Biophysikalische ChemieHumboldt-Universität zu Berlin Invalidenstrasse 42 10099 Berlin Germany
| | - Piotr Przybylski
- Faculty of ChemistryAdam Mickiewicz University Uniwersytetu Poznańskiego 8 61-614 Poznań Poland
| |
Collapse
|
6
|
Dzhemileva LU, D'yakonov VA, Islamov II, Yunusbaeva MM, Dzhemilev UM. New 1Z,5Z-diene macrodiolides: Catalytic synthesis, anticancer activity, induction of mitochondrial apoptosis, and effect on the cell cycle. Bioorg Chem 2020; 99:103832. [PMID: 32315897 DOI: 10.1016/j.bioorg.2020.103832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 02/03/2023]
Abstract
An original scheme was developed for the synthesis of previously undescribed unsaturated macrodiolides containing a 1Z,5Z-diene moiety in 44-80% yields and with high stereoselectivity (>95%) based on the intermolecular esterification of α,ω-diols with α,ω-alka-nZ,(n + 4)Z-dienedicarboxylic acids (1,12-dodeca-4Z,8Z-dienedicarboxylic acid, 1,14-tetradeca-5Z,9Z-dienedicarboxylic acid, 1,18-octadeca-7Z,11Z-dienedicarboxylic acid) catalyzed by hafnium triflate [Hf(OTf)4]. The unsaturated dicarboxylic acids were prepared via homo-cyclomagnesiation of tetrahydropyran ethers of O-containing 1,2-dienes with EtMgBr in the presence of Mg metal and the Cp2TiCl2 catalyst (10 mol.%) and the subsequent Jones oxidation of pyran ethers formed after the acid hydrolysis of magnesacyclopentanes. The thus prepared macrodiolides exhibit high cytotoxic activity in vitro against Jurkat, K562, U937, Hek293 and HeLa cancer cell lines. It was found that induction of the programmed cell death in Jurkat cells by macrodiolides corresponds to the mitochondrial apoptosis pathway. Also, it was shown that the prepared macrodiolides efficiently suppress phosphorylation of Akt and p38 kinases and CREB transcription factor in cancer cells.
Collapse
Affiliation(s)
- Lilya U Dzhemileva
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation.
| | - Vladimir A D'yakonov
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation.
| | - Ilgiz I Islamov
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| | - Milyausha M Yunusbaeva
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| | - Usein M Dzhemilev
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| |
Collapse
|
7
|
Pyrimethamine conjugated histone deacetylase inhibitors: Design, synthesis and evidence for triple negative breast cancer selective cytotoxicity. Bioorg Med Chem 2020; 28:115345. [DOI: 10.1016/j.bmc.2020.115345] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/12/2020] [Accepted: 01/22/2020] [Indexed: 12/25/2022]
|
8
|
Janas A, Przybylski P. 14- and 15-membered lactone macrolides and their analogues and hybrids: structure, molecular mechanism of action and biological activity. Eur J Med Chem 2019; 182:111662. [DOI: 10.1016/j.ejmech.2019.111662] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 11/15/2022]
|
9
|
A nanodelivered Vorinostat derivative is a promising oral compound for the treatment of visceral leishmaniasis. Pharmacol Res 2019; 139:375-383. [DOI: 10.1016/j.phrs.2018.11.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
|
10
|
Histone deacetylase 8 (HDAC8) and its inhibitors with selectivity to other isoforms: An overview. Eur J Med Chem 2018; 164:214-240. [PMID: 30594678 DOI: 10.1016/j.ejmech.2018.12.039] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/04/2018] [Accepted: 12/16/2018] [Indexed: 01/08/2023]
Abstract
The histone deacetylases (HDACs) enzymes provided crucial role in transcriptional regulation of cells through deacetylation of nuclear histone proteins. Discoveries related to the HDAC8 enzyme activity signified the importance of HDAC8 isoform in cell proliferation, tumorigenesis, cancer, neuronal disorders, parasitic/viral infections and other epigenetic regulations. The pan-HDAC inhibitors can confront these conditions but have chances to affect epigenetic functions of other HDAC isoforms. Designing of selective HDAC8 inhibitors is a key feature to combat the pathophysiological and diseased conditions involving the HDAC8 activity. This review is concerned about the structural and positional aspects of HDAC8 in the HDAC family. It also covers the contributions of HDAC8 in the pathophysiological conditions, a preliminary discussion about the recent scenario of HDAC8 inhibitors. This review might help to deliver the structural, functional and computational information in order to identify and design potent and selective HDAC8 inhibitors for target specific treatment of diseases involving HDAC8 enzymatic activity.
Collapse
|
11
|
Luan Y, Li J, Bernatchez JA, Li R. Kinase and Histone Deacetylase Hybrid Inhibitors for Cancer Therapy. J Med Chem 2018; 62:3171-3183. [PMID: 30418766 DOI: 10.1021/acs.jmedchem.8b00189] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Histone deacetylases (HDACs), encompassing at least 18 members, are promising targets for anticancer drug discovery and development. To date, five histone deacetylase inhibitors (HDACis) have been approved for cancer treatment, and numerous others are undergoing clinical trials. It has been well validated that an agent that can simultaneously and effectively inhibit two or more targets may offer greater therapeutic benefits over single-acting agents in preventing resistance to treatment and in potentiating synergistic effects. A prime example of a bifunctional agent is the hybrid HDAC inhibitor. In this perspective, the authors review the majority of reported kinase/HDAC hybrid inhibitors.
Collapse
Affiliation(s)
- Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy , Qingdao University , Qingdao 266071 , Shandong Province , China
| | | | | | - Rongshi Li
- Department of Medicinal Chemistry, School of Pharmacy , Qingdao University , Qingdao 266071 , Shandong Province , China.,UNMC Center for Drug Discovery, Department of Pharmaceutical Sciences, College of Pharmacy, Fred and Pamela Buffett Cancer Center, and Center for Staphylococcal Research , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| |
Collapse
|
12
|
Targeted synthesis of macrodiolides containing bis-methylene-separated Z-double bonds and their antitumor activity in vitro. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Structure–activity relationships of hydroxamate-based histone deacetylase-8 inhibitors: reality behind anticancer drug discovery. Future Med Chem 2017; 9:2211-2237. [PMID: 29182018 DOI: 10.4155/fmc-2017-0130] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pan-histone deacetylase (HDAC) inhibitors comprise a fish-like structural orientation where hydrophobic aryl- and zinc-binding groups act as head and tail, respectively of a fish. The linker moiety correlates the body of the fish linking head and tail groups. Despite these pan-HDAC inhibitors, selective HDAC-8 inhibitors are still in demand as a safe remedy. HDAC-8 is involved in invasion and metastasis in cancer. This review deals with the rationale behind HDAC-8 inhibitory activity and selectivity along with detailed structure–activity relationships of diverse hydroxamate-based HDAC-8 inhibitors. HDAC-8 inhibitory potency may be increased by modifying the fish-like pharmacophoric features of such type of pan-HDAC inhibitors. This review may provide a preliminary basis to design and optimize new lead molecules with higher HDAC-8 inhibitory activity. This work may surely enlighten in providing useful information in the field of target-specific anticancer therapy.
Collapse
|
14
|
Sodium phenylbutyrate abrogates African swine fever virus replication by disrupting the virus-induced hypoacetylation status of histone H3K9/K14. Virus Res 2017; 242:24-29. [DOI: 10.1016/j.virusres.2017.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 02/08/2023]
|
15
|
Hailu GS, Robaa D, Forgione M, Sippl W, Rotili D, Mai A. Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives. J Med Chem 2017; 60:4780-4804. [DOI: 10.1021/acs.jmedchem.6b01595] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Gebremedhin S. Hailu
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Mariantonietta Forgione
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161 Rome, Italy
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Dante Rotili
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Antonello Mai
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Istituto
Pasteur, Fondazione Cenci-Bolognetti, “Sapienza” Università di Roma, 00185 Rome, Italy
| |
Collapse
|
16
|
Raji I, Ahluwalia K, Oyelere AK. Design, synthesis and evaluation of antiproliferative activity of melanoma-targeted histone deacetylase inhibitors. Bioorg Med Chem Lett 2017; 27:744-749. [PMID: 28131715 PMCID: PMC5314971 DOI: 10.1016/j.bmcl.2017.01.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/13/2017] [Indexed: 10/20/2022]
Abstract
The clinical validation of histone deacetylase inhibition as a cancer therapeutic modality has stimulated interest in the development of new generation of potent and tumor selective histone deacetylase inhibitors (HDACi). With the goal of selective delivery of the HDACi to melanoma cells, we incorporated the benzamide, a high affinity melanin-binding template, into the design of HDACi to generate a new series of compounds 10a-b and 11a-b which display high potency towards HDAC1 and HDAC6. However, these compounds have attenuated antiproliferative activities relative to the untargeted HDACi. An alternative strategy furnished compound 14, a prodrug bearing the benzamide template linked via a labile bond to a hydroxamate-based HDACi. This pro-drug compound showed promising antiproliferative activity and warrant further study.
Collapse
Affiliation(s)
- Idris Raji
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Kabir Ahluwalia
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
17
|
Zagni C, Floresta G, Monciino G, Rescifina A. The Search for Potent, Small-Molecule HDACIs in Cancer Treatment: A Decade After Vorinostat. Med Res Rev 2017; 37:1373-1428. [PMID: 28181261 DOI: 10.1002/med.21437] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022]
Abstract
Histone deacetylases (HDACs) play a crucial role in the remodeling of chromatin, and are involved in the epigenetic regulation of gene expression. In the last decade, inhibition of HDACs came out as a target for specific epigenetic changes associated with cancer and other diseases. Until now, more than 20 HDAC inhibitors (HDACIs) have entered clinical studies, and some of them (e.g., vorinostat, romidepsin) have been approved for the treatment of cutaneous T-cell lymphoma. This review provides an overview of current knowledge, progress, and molecular mechanisms of HDACIs, covering a period from 2011 until 2015.
Collapse
Affiliation(s)
- Chiara Zagni
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Giuseppe Floresta
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy.,Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Giulia Monciino
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonio Rescifina
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| |
Collapse
|
18
|
Raji I, Yadudu F, Janeira E, Fathi S, Szymczak L, Kornacki JR, Komatsu K, Li JD, Mrksich M, Oyelere AK. Bifunctional conjugates with potent inhibitory activity towards cyclooxygenase and histone deacetylase. Bioorg Med Chem 2017; 25:1202-1218. [PMID: 28057407 PMCID: PMC5291751 DOI: 10.1016/j.bmc.2016.12.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/17/2016] [Accepted: 12/20/2016] [Indexed: 12/16/2022]
Abstract
We herein disclose a series of compounds with potent inhibitory activities towards histone deacetylases (HDAC) and cyclooxygenases (COX). These compounds potently inhibited the growth of cancer cell lines consistent with their anti-COX and anti-HDAC activities. While compound 2b showed comparable level of COX-2 selectivity as celecoxib, compound 11b outperformed indomethacin in terms of selectivity towards COX-2 relative to COX-1. An important observation with our lead compounds (2b, 8, 11b, and 17b) is their enhanced cytotoxicity towards androgen dependent prostate cancer cell line (LNCaP) relative to androgen independent prostate cancer cell line (DU-145). Interestingly, compounds 2b and 17b arrested the cell cycle progression of LNCaP in the S-phase, while compound 8 showed a G0/G1 arrest, similar to SAHA. Relative to SAHA, these compounds displayed tumor-selective cytotoxicity as they have low anti-proliferative activity towards healthy cells (VERO); an attribute that makes them attractive candidates for drug development.
Collapse
Affiliation(s)
- Idris Raji
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Fatima Yadudu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Emily Janeira
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Shaghayegh Fathi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Lindsey Szymczak
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - James Richard Kornacki
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Kensei Komatsu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, USA
| | - Jian-Dong Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, USA
| | - Milan Mrksich
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
19
|
Maolanon AR, Kristensen HME, Leman LJ, Ghadiri MR, Olsen CA. Natural and Synthetic Macrocyclic Inhibitors of the Histone Deacetylase Enzymes. Chembiochem 2016; 18:5-49. [DOI: 10.1002/cbic.201600519] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Alex R. Maolanon
- Center for Biopharmaceuticals and; Department of Drug Design and Pharmacology; University of Copenhagen; Universitetsparken 2 2100 Copenhagen Denmark
| | - Helle M. E. Kristensen
- Center for Biopharmaceuticals and; Department of Drug Design and Pharmacology; University of Copenhagen; Universitetsparken 2 2100 Copenhagen Denmark
| | - Luke J. Leman
- Department of Chemistry; The Skaggs Institute for Chemical Biology; The Scripps Research Institute; 10550 North Torrey Pines Road La Jolla CA 92037 USA
| | - M. Reza Ghadiri
- Department of Chemistry; The Skaggs Institute for Chemical Biology; The Scripps Research Institute; 10550 North Torrey Pines Road La Jolla CA 92037 USA
| | - Christian A. Olsen
- Center for Biopharmaceuticals and; Department of Drug Design and Pharmacology; University of Copenhagen; Universitetsparken 2 2100 Copenhagen Denmark
| |
Collapse
|
20
|
Gaillard T, Madamet M, Tsombeng FF, Dormoi J, Pradines B. Antibiotics in malaria therapy: which antibiotics except tetracyclines and macrolides may be used against malaria? Malar J 2016; 15:556. [PMID: 27846898 PMCID: PMC5109779 DOI: 10.1186/s12936-016-1613-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/10/2016] [Indexed: 01/15/2023] Open
Abstract
Malaria, a parasite vector-borne disease, is one of the most significant health threats in tropical regions, despite the availability of individual chemoprophylaxis. Malaria chemoprophylaxis and chemotherapy remain a major area of research, and new drug molecules are constantly being developed before drug-resistant parasites strains emerge. The use of anti-malarial drugs is challenged by contra-indications, the level of resistance of Plasmodium falciparum in endemic areas, clinical tolerance and financial cost. New therapeutic approaches are currently needed to fight against this disease. Some antibiotics that have shown potential effects on malaria parasite have been recently studied in vitro or in vivo intensively. Two families, tetracyclines and macrolides and their derivatives have been particularly studied in recent years. However, other less well-known have been tested or are being used for malaria treatment. Some of these belong to older families, such as quinolones, co-trimoxazole or fusidic acid, while others are new drug molecules such as tigecycline. These emerging antibiotics could be used to prevent malaria in the future. In this review, the authors overview the use of antibiotics for malaria treatment.
Collapse
Affiliation(s)
- Tiphaine Gaillard
- Fédération des Laboratoires, Hôpital d'Instruction des Armées Saint Anne, Toulon, France.,Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France.,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Aix Marseille Université, UM 63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
| | - Marylin Madamet
- Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France.,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Aix Marseille Université, UM 63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France.,Centre National de Référence du Paludisme, Marseille, France
| | - Francis Foguim Tsombeng
- Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France.,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Aix Marseille Université, UM 63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
| | - Jérôme Dormoi
- Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France.,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Aix Marseille Université, UM 63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
| | - Bruno Pradines
- Unité de Parasitologie et d'Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France. .,Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Aix Marseille Université, UM 63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France. .,Centre National de Référence du Paludisme, Marseille, France.
| |
Collapse
|
21
|
Wu L, Bao K, Song R, Wang D, Zhang L, Wang W, Zhang W, Bin W. Development of Novel Erythromycin Derivatives with Inhibitory Activity against Proliferation of Tumor Cells. PLoS One 2016; 11:e0159503. [PMID: 27447724 PMCID: PMC4957748 DOI: 10.1371/journal.pone.0159503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/05/2016] [Indexed: 11/19/2022] Open
Abstract
In our continuing structure-activity relationship study of a new class of erythromycin A (EM-A) derivatives with antiproliferative activity, a new series of de(N-methyl) EM-A dimers jointed by a four-atom linker, -CH2CH = CHCH2-, were prepared and their antiproliferative activity against three human tumor cell lines was evaluated by MTT assay. The most active EM-A dimer, compound 1b, that carrying C6 methoxyl groups was further investigated and showed potent antiproliferative activity in six other human tumor cell lines. Flow cytometry analysis of 1b treated HeLa and MCF-7 cells indicated that the four-atom EM-A dimers induced the SubG1 phase cell cycle arrest and cell apoptosis, in time- and dose-dependent manners. Further experiments including morphologic observation, DNA agarose gel electrophoresis, mitochondrial potential alternation and western blot analysis revealed that the antiproliferative mechanism may involve the induction of apoptosis in activating the mitochondrial pathway, and regulation of apoptotic proteins.
Collapse
Affiliation(s)
- Lan Wu
- Department of Geratology, The First Affiliated Hospital of Chinese Medical University, Shenyang, China
- * E-mail: (LW); (WB)
| | - Kai Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Structure-Based Drug Design and Discovery Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Song
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Defa Wang
- Key Laboratory of Structure-Based Drug Design and Discovery Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Lei Zhang
- Department of Geratology, The First Affiliated Hospital of Chinese Medical University, Shenyang, China
| | - Weiyun Wang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Wen Bin
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- * E-mail: (LW); (WB)
| |
Collapse
|
22
|
Maolanon A, Madsen A, Olsen C. Innovative Strategies for Selective Inhibition of Histone Deacetylases. Cell Chem Biol 2016; 23:759-768. [DOI: 10.1016/j.chembiol.2016.06.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/24/2016] [Accepted: 06/22/2016] [Indexed: 01/22/2023]
|
23
|
Li J, Li X, Wang X, Hou J, Zang J, Gao S, Xu W, Zhang Y. PXD101 analogs with L-phenylglycine-containing branched cap as histone deacetylase inhibitors. Chem Biol Drug Des 2016; 88:574-84. [PMID: 27235003 DOI: 10.1111/cbdd.12787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) allow histones to wrap DNA more tightly and finally lead to the repression of some tumor suppressor genes. Histone deacetylase inhibitors (HDACIs) have been proved to have effects on tumorigenesis and tumor progression. In this study, we reported the design, synthesis, and in vitro activity evaluation of novel PXD101 analogs with L-phenylglycine-containing cap as HDACIs. Our results showed that HDACs inhibitory activities of compounds 10k, 10r, and 10s were not only superior to the first approved HDACI SAHA, but also comparable to their parent compound PXD101, a recently approved HDACI in 2014. However, all 6 selected PXD101 analogs exhibited moderate in vitro antiproliferative activities, less potent than PXD101 and SAHA. Representative compound 10s showed similar HDACs isoform selective profile to PXD101, which demonstrated that introduction of L-phenylglycine-containing branched cap group could not change the isoform selectivity of PXD101 dramatically.
Collapse
Affiliation(s)
- Jingyao Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Xiaoyang Li
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Xue Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Jinning Hou
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Jie Zang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Shuai Gao
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Wenfang Xu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China
| | - Yingjie Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Ji'nan, Shandong, China.
| |
Collapse
|
24
|
Ling Y, Xu C, Luo L, Cao J, Feng J, Xue Y, Zhu Q, Ju C, Li F, Zhang Y, Zhang Y, Ling X. Novel β-Carboline/Hydroxamic Acid Hybrids Targeting Both Histone Deacetylase and DNA Display High Anticancer Activity via Regulation of the p53 Signaling Pathway. J Med Chem 2015; 58:9214-27. [PMID: 26555243 DOI: 10.1021/acs.jmedchem.5b01052] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A novel series of hybrids from β-carboline and hydroxamic acid were designed and synthesized. Several compounds (5m, 11b-d, and 11h) not only exerted significant antiproliferation activity against four human colorectal cancer (CRC) cell lines but also showed histone deacetylase inhibitory effects in vitro. The most potent compound, 11c, exhibited anticancer potency sevenfold higher than that of SAHA. 11c triggered more significant cancer cell apoptosis than did SAHA by cleavage of both PARP and caspase 3 in a dose-dependent manner. Furthermore, 11c simultaneously increased the acetylation of histone H3 and α-tubulin, enhanced expression of DNA damage markers histone H2AX phosphorylation and p-p53 (Ser15), and activated p53 signaling pathway in HCT116 cells. Finally, 11c showed low acute toxicity in mice and inhibited the growth of implanted human CRC in mice more potently than did SAHA. Together, 11c possessed potent antitumor activity and may be a promising candidate for the potential treatment of human CRC.
Collapse
Affiliation(s)
- Yong Ling
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | - Chenjun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | | | | | | | | | | | | | - Fengzhi Li
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute , Buffalo, New York, USA
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing 210009, P.R. China
| | | | - Xiang Ling
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute , Buffalo, New York, USA
| |
Collapse
|
25
|
Tapadar S, Fathi S, Raji I, Omesiete W, Kornacki JR, Mwakwari SC, Miyata M, Mitsutake K, Li JD, Mrksich M, Oyelere AK. A structure-activity relationship of non-peptide macrocyclic histone deacetylase inhibitors and their anti-proliferative and anti-inflammatory activities. Bioorg Med Chem 2015; 23:7543-64. [PMID: 26585275 DOI: 10.1016/j.bmc.2015.10.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/21/2015] [Accepted: 10/31/2015] [Indexed: 10/22/2022]
Abstract
Inhibition of the enzymatic activity of histone deacetylase (HDAC) is a promising therapeutic strategy for cancer treatment and several distinct small molecule histone deacetylase inhibitors (HDACi) have been reported. We have previously identified a new class of non-peptide macrocyclic HDACi derived from 14- and 15-membered macrolide skeletons. In these HDACi, the macrocyclic ring is linked to the zinc chelating hydroxamate moiety through a para-substituted aryl-triazole cap group. To further delineate the depth of the SAR of this class of HDACi, we have synthesized series of analogous compounds and investigated the influence of various substitution patterns on their HDAC inhibitory, anti-proliferative and anti-inflammatory activities. We identified compounds 25b and 38f with robust anti-proliferative activities and compound 26f (IC50 47.2 nM) with superior anti-inflammatory (IC50 88 nM) activity relative to SAHA.
Collapse
Affiliation(s)
- Subhasish Tapadar
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Shaghayegh Fathi
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Idris Raji
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Wilson Omesiete
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - James R Kornacki
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA; Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Sandra C Mwakwari
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Masanori Miyata
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Kazunori Mitsutake
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jian-Dong Li
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Milan Mrksich
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA; Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
26
|
New macrocyclic analogs of the natural histone deacetylase inhibitor FK228; design, synthesis and preliminary biological evaluation. Bioorg Med Chem 2015; 23:6785-93. [PMID: 26481659 DOI: 10.1016/j.bmc.2015.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/08/2015] [Accepted: 10/05/2015] [Indexed: 11/23/2022]
Abstract
Among the natural histone deacetylase inhibitors (HDACi), the bicyclic depsipeptide macrolactone FK228 stands out for its unique chemical structure and mechanism of action. In order to expand the chemical diversity, exploiting the FK228 peculiar structure, we have synthesized a collection of 24 simplified novel analogs. A first series consists of bicyclic macrolactones, where the carboxy terminus of the natural compound was substituted by peptidomimetic aminomethylphenylacetic acid derivatives. These analogs, 7a-i, showed submicromolar cytotoxic activity, even though very low inhibitory activity against HDAC enzymes, suggesting that most probably they behave with a mechanism different from the natural compound. One of the most active members in the group, 7g, was evaluated in vivo and exhibited significant antitumor activity. This evidence supports that the activity is unrelated to HDAC inhibition and these compounds represent a novel series of promising active agents. Another analog series consists of monocyclic macrolactones, 9a-c and 10a-d which lack the disulfide bridge and bear the protected sulfur on the linear external chain; they showed similar cytotoxic activities compared to the natural compound, but proved to be very sensitive to the nature of the sulfur protection. In fact, when the sulfur was protected by an 1-octanoyl residue, like in 9b, the product displayed a one digit nanomolar activity. The results provide evidence that our approach may be followed to develop novel series of FK228 analogs.
Collapse
|
27
|
Sugawara A, Maita N, Gouda H, Yamamoto T, Hirose T, Kimura S, Saito Y, Nakano H, Kasai T, Nakano H, Shiomi K, Hirono S, Watanabe T, Taniguchi H, O̅mura S, Sunazuka T. Creation of Customized Bioactivity within a 14-Membered Macrolide Scaffold: Design, Synthesis, and Biological Evaluation Using a Family-18 Chitinase. J Med Chem 2015; 58:4984-97. [DOI: 10.1021/acs.jmedchem.5b00175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Akihiro Sugawara
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Nobuo Maita
- Institute
for Enzyme Research, University of Tokushima, 3-18-15 Kuramotocho, Tokushima City, Tokushima, 770-8503, Japan
| | - Hiroaki Gouda
- School
of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsuyoshi Yamamoto
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoyasu Hirose
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Saori Kimura
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Yoshifumi Saito
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Hayato Nakano
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takako Kasai
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Hirofumi Nakano
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kazuro Shiomi
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Shuichi Hirono
- School
of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Takeshi Watanabe
- Department
of Applied Biological Chemistry, Faculty of Agriculture, Niigata University, 8050 Ikarashi-2, Niigata 950-2181, Japan
| | - Hisaaki Taniguchi
- Institute
for Enzyme Research, University of Tokushima, 3-18-15 Kuramotocho, Tokushima City, Tokushima, 770-8503, Japan
| | - Satoshi O̅mura
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Toshiaki Sunazuka
- The
Kitasato Institute, Kitasato Institute for Life Sciences and Graduate
School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
28
|
Washington AZ, Tapadar S, George A, Oyelere AK. Exploiting translational stalling peptides in an effort to extend azithromycin interaction within the prokaryotic ribosome nascent peptide exit tunnel. Bioorg Med Chem 2015; 23:5198-209. [PMID: 26037612 DOI: 10.1016/j.bmc.2015.04.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/18/2015] [Accepted: 04/29/2015] [Indexed: 10/23/2022]
Abstract
The ribosome is the primary protein synthesis machine in the cell and is a target for treatment of a variety of diseases including bacterial infection and cancer. The ribosomal peptide exit tunnel, the route of egress for the nascent peptide, is an inviting site for drug design. Toward a rational engagement of the nascent peptide components for the design of small molecule inhibitors of ribosome function, we designed and disclosed herein a set of N-10 indole functionalized azithromycin analogs. The indole moiety of these compounds is designed to mimic the translation stalling interaction of SecM W155 side-chain with the prokaryotic (Escherichia coli) ribosome A751 residue. Many of these N-10 functionalized compounds have enhanced translation inhibition activities against E. coli ribosome relative to azithromycin while a subset inhibited the growth of representative susceptible bacteria strains to about the same extent as azithromycin. Moreover, the inclusion of bovine serum in the bacterial growth media enhanced the anti-bacterial potency of the N-10 functionalized azithromycin analogs by as high as 10-fold.
Collapse
Affiliation(s)
- Arren Z Washington
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Subhasish Tapadar
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Alex George
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
29
|
Ling Y, Wang X, Wang C, Xu C, Zhang W, Zhang Y, Zhang Y. Hybrids from Farnesylthiosalicylic Acid and Hydroxamic Acid as Dual Ras-Related Signaling and Histone Deacetylase (HDAC) Inhibitors: Design, Synthesis and Biological Evaluation. ChemMedChem 2015; 10:971-6. [PMID: 25882299 DOI: 10.1002/cmdc.201500019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/18/2015] [Indexed: 01/05/2023]
Abstract
A novel series of hybrids was designed and synthesized by combining key elements from farnesylthiosalicylic acid (FTS) and hydroxamic acid. Several 3,7,11-trimethyldodeca-2,6,10-trien-1-yl) thio)benzamide derivatives, particularly those with branched and linear aliphatic linkers between the hydroxamic zinc binding group (ZBG) and the benzamide core, not only displayed significant antitumor activities against six human cancer cells but also exhibited histone deacetylase (HDAC) inhibitory effects in vitro. Among them, N-(4-(hydroxyamino)-4-oxobutyl)-2-(((2E,6E)-3,7,11-trimethyldodeca-2,6, 10-trien-1-yl)thio)benzamide (8 d) was the most potent, with IC50 values of 4.9-7.6 μM; these activities are eight- to sixteen-fold more potent than FTS and comparable to that of suberoylanilide hydroxamic acid (SAHA). Derivative 8 d induced cell cycle arrest in the G0/G1 phase, inhibited the acetylation of histone H3 and α-tubulin, and blocked Ras-related signaling pathways in a dose-dependent manner. The improved tumor growth inhibition and cell-cycle arrest in vitro might result from the dual inhibition. These findings suggest dual inhibitors of Ras-related signaling pathway and HDAC hold promise as therapeutic agents for the treatment of cancer.
Collapse
Affiliation(s)
- Yong Ling
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China). .,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009 (P. R. China).
| | - Xuemin Wang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Chenniu Wang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Chenjun Xu
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Wei Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China)
| | - Yihua Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China).,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009 (P. R. China)
| | - Yanan Zhang
- School of Pharmacy, Nantong University, Nantong 226001 (P. R. China).
| |
Collapse
|
30
|
An efficient synthesis of SK-658 and its analogs as potent histone deacetylase inhibitors. Bioorg Chem 2015; 59:145-50. [DOI: 10.1016/j.bioorg.2015.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/03/2015] [Accepted: 02/08/2015] [Indexed: 11/18/2022]
|
31
|
Washington AZ, Benicewicz DB, Canzoneri JC, Fagan CE, Mwakwari SC, Maehigashi T, Dunham CM, Oyelere AK. Macrolide-peptide conjugates as probes of the path of travel of the nascent peptides through the ribosome. ACS Chem Biol 2014; 9:2621-31. [PMID: 25198768 PMCID: PMC4245169 DOI: 10.1021/cb5003224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Despite
decades of research on the bacterial ribosome, the ribosomal
exit tunnel is still poorly understood. Although it has been suggested
that the exit tunnel is simply a convenient route of egress for the
nascent chain, specific protein sequences serve to slow the rate of
translation, suggesting some degree of interaction between the nascent
peptide chain and the exit tunnel. To understand how the ribosome
interacts with nascent peptide sequences, we synthesized and characterized
a novel class of probe molecules. These peptide–macrolide (or
“peptolide”) conjugates were designed to present unique
peptide sequences to the exit tunnel. Biochemical and X-ray structural
analyses of the interactions between these probes and the ribosome
reveal interesting insights about the exit tunnel. Using translation
inhibition and RNA structure probing assays, we find the exit tunnel
has a relaxed preference for the directionality (N → C or C
→ N orientation) of the nascent peptides. Moreover, the X-ray
crystal structure of one peptolide derived from a positively charged,
reverse Nuclear Localization Sequence peptide, bound to the 70S bacterial
ribosome, reveals that the macrolide ring of the peptolide binds in
the same position as other macrolides. However, the peptide tail folds
over the macrolide ring, oriented toward the peptidyl transferase
center and interacting in a novel manner with 23S rRNA residue C2442
and His69 of ribosomal protein L4. These data suggest that these peptolides
are viable probes for interrogating nascent peptide–exit tunnel
interaction.
Collapse
Affiliation(s)
- Arren Z. Washington
- School
of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Derek B. Benicewicz
- School
of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Joshua C. Canzoneri
- School
of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Crystal E. Fagan
- Department
of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Sandra C. Mwakwari
- School
of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | - Tatsuya Maehigashi
- Department
of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Christine M. Dunham
- Department
of Biochemistry, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Adegboyega K. Oyelere
- School
of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| |
Collapse
|
32
|
Yang F, Zhang T, Wu H, Yang Y, Liu N, Chen A, Li Q, Li J, Qin L, Jiang B, Wang X, Pang X, Yi Z, Liu M, Chen Y. Design and Optimization of Novel Hydroxamate-Based Histone Deacetylase Inhibitors of Bis-Substituted Aromatic Amides Bearing Potent Activities against Tumor Growth and Metastasis. J Med Chem 2014; 57:9357-69. [DOI: 10.1021/jm5012148] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Feifei Yang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tao Zhang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Haigang Wu
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yang Yang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ning Liu
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ang Chen
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qiang Li
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jingjie Li
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Liwen Qin
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Beier Jiang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xin Wang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiufeng Pang
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of
Regulatory Biology, The Institute of Biomedical Sciences and School
of Life Sciences, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
33
|
Chan CT, Qi J, Smith W, Paranol R, Mazitschek R, West N, Reeves R, Chiosis G, Schreiber SL, Bradner JE, Paulmurugan R, Gambhir SS. Syntheses and discovery of a novel class of cinnamic hydroxamates as histone deacetylase inhibitors by multimodality molecular imaging in living subjects. Cancer Res 2014; 74:7475-86. [PMID: 25320008 DOI: 10.1158/0008-5472.can-14-0197] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Histone deacetylases (HDAC) that regulate gene expression are being explored as cancer therapeutic targets. In this study, we focused on HDAC6 based on its ability to inhibit cancerous Hsp90 chaperone activities by disrupting Hsp90/p23 interactions. To identify novel HDAC6 inhibitors, we used a dual-luciferase reporter system in cell culture and living mice by bioluminescence imaging (BLI). On the basis of existing knowledge, a library of hydrazone compounds was generated for screening by coupling cinnamic hydroxamates with aldehydes and ketones. Potency and selectivity were determined by in vitro HDAC profiling assays, with further evaluation to inhibit Hsp90(α/β)/p23 interactions by BLI. In this manner, we identified compound 1A12 as a dose-dependent inhibitor of Hsp90(α/β)/p23 interactions, UKE-1 myeloid cell proliferation, p21(waf1) upregulation, and acetylated histone H3 levels. 1A12 was efficacious in tumor xenografts expressing Hsp90(α)/p23 reporters relative to carrier control-treated mice as determined by BLI. Small animal (18)F-FDG PET/CT imaging on the same cohort showed that 1A12 also inhibited glucose metabolism relative to control subjects. Ex vivo analyses of tumor lysates showed that 1A12 administration upregulated acetylated-H3 by approximately 3.5-fold. Taken together, our results describe the discovery and initial preclinical validation of a novel selective HDAC inhibitor.
Collapse
Affiliation(s)
- C T Chan
- Department of Radiology, Stanford University School of Medicine, Stanford, California. Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California. Bio-X Program, Stanford University School of Medicine, Stanford, California
| | - J Qi
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - W Smith
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - R Paranol
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - R Mazitschek
- Harvard Medical School, Boston, Massachusetts. Massachusetts General Hospital, Boston, Massachusetts. Broad Institute, Cambridge, Massachusetts
| | - N West
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - R Reeves
- Department of Radiology, Stanford University School of Medicine, Stanford, California. Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California. Bio-X Program, Stanford University School of Medicine, Stanford, California
| | - G Chiosis
- Department of Medicine and Program in Molecular Pharmacology and Medical Chemistry, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - J E Bradner
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Broad Institute, Cambridge, Massachusetts
| | - R Paulmurugan
- Department of Radiology, Stanford University School of Medicine, Stanford, California. Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California. Bio-X Program, Stanford University School of Medicine, Stanford, California
| | - S S Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, California. Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California. Bio-X Program, Stanford University School of Medicine, Stanford, California. Department of Bioengineering, Stanford University School of Medicine, Stanford, California. Division of Nuclear Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
34
|
Sodji Q, Patil V, Jain S, Kornacki JR, Mrksich M, Tekwani BL, Oyelere AK. The antileishmanial activity of isoforms 6- and 8-selective histone deacetylase inhibitors. Bioorg Med Chem Lett 2014; 24:4826-30. [PMID: 25240614 PMCID: PMC4225773 DOI: 10.1016/j.bmcl.2014.08.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 08/27/2014] [Indexed: 11/23/2022]
Abstract
Histone deacetylase inhibitors (HDACi) pleiotropy is largely due to their nonselective inhibition of various cellular HDAC isoforms. Connecting inhibition of a specific isoform to biological responses and/or phenotypes is essential toward deconvoluting HDACi pleiotropy. The contribution of classes I and II HDACs to the antileishmanial activity of HDACi was investigated using the amastigote and promastigote forms of Leishmania donovani. We observed that the antileishmanial activities of HDACi are largely due to the inhibition of HDAC6-like activity. This observation could facilitate the development of HDACi as antileishmanial agents.
Collapse
Affiliation(s)
- Quaovi Sodji
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Vishal Patil
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Surendra Jain
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - James R Kornacki
- Department of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Evanston, IL 60208-3113, USA
| | - Milan Mrksich
- Department of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Evanston, IL 60208-3113, USA
| | - Babu L Tekwani
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA.
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
35
|
Villalba M, Lopez-Royuela N, Krzywinska E, Rathore MG, Hipskind RA, Haouas H, Allende-Vega N. Chemical metabolic inhibitors for the treatment of blood-borne cancers. Anticancer Agents Med Chem 2014; 14:223-32. [PMID: 24237221 DOI: 10.2174/18715206113136660374] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/20/2013] [Accepted: 10/07/2013] [Indexed: 12/16/2022]
Abstract
Tumor cells, including leukemic cells, remodel their bioenergetic system in favor of aerobic glycolysis. This process is called "the Warburg effect" and offers an attractive pharmacological target to preferentially eliminate malignant cells. In addition, recent results show that metabolic changes can be linked to tumor immune evasion. Mouse models demonstrate the importance of this metabolic remodeling in leukemogenesis. Some leukemias, although treatable, remain incurable and resistance to chemotherapy produces an elevated percentage of relapse in most leukemia cases. Several groups have targeted the specific metabolism of leukemia cells in preclinical and clinical studies to improve the prognosis of these patients, i.e. using L-asparaginase to treat pediatric acute lymphocytic leukemia (ALL). Additional metabolic drugs that are currently being used to treat other diseases or tumors could also be exploited for leukemia, based on preclinical studies. Finally, we discuss the potential use of several metabolic drugs in combination therapies, including immunomodulatory drugs (IMiDs) or immune cell-based therapies, to increase their efficacy and reduce side effects in the treatment of hematological cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nerea Allende-Vega
- INSERM U1040, Institut de Recherche en Biothérapie, 80, avenue Augustin Fliche. 34295 Montpellier Cedex 5, France.
| |
Collapse
|
36
|
Chen Z, Yan Q, Yi H, Liu Z, Lei A, Zhang Y. Efficient Synthesis of 1,2,3‐Triazoles by Copper‐Mediated CN and NN Bond Formation Starting From
N
‐Tosylhydrazones and Amines. Chemistry 2014; 20:13692-7. [DOI: 10.1002/chem.201403515] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Zhengkai Chen
- ZJU‐NHU United R&D Center, Department of Chemistry, Zhejiang University, Hangzhou 310027 (P. R. China)
| | - Qiangqiang Yan
- ZJU‐NHU United R&D Center, Department of Chemistry, Zhejiang University, Hangzhou 310027 (P. R. China)
| | - Hong Yi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072 (P. R. China)
| | - Zhanxiang Liu
- ZJU‐NHU United R&D Center, Department of Chemistry, Zhejiang University, Hangzhou 310027 (P. R. China)
| | - Aiwen Lei
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072 (P. R. China)
| | - Yuhong Zhang
- ZJU‐NHU United R&D Center, Department of Chemistry, Zhejiang University, Hangzhou 310027 (P. R. China)
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000 (P. R. China)
| |
Collapse
|
37
|
Current trends in the development of histone deacetylase inhibitors: a review of recent patent applications. Pharm Pat Anal 2014; 1:75-90. [PMID: 24236715 DOI: 10.4155/ppa.11.3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Histone deacetylases (HDACs) have become an important target for the treatment of cancer and other diseases. Currently, more than ten HDAC inhibitors have entered clinical studies and two of them have already reached the market. The hydroxamic acid derivative SAHA (also known as vorinostat or Zolinza®) and the cyclic depsipeptide FK228 (romidepsin or Istodax®) have gained approval from the US FDA for the treatment of cutaneous T-cell lymphoma. Nevertheless, there has been a continuous effort aimed at discovering a new generation of clinical candidates with improved pharmaceutical properties. This review provides a summary of the most recent patents published from mid-2009 to mid-2011.
Collapse
|
38
|
Zhang QW, Feng J, Li JQ. Cap-Modified Hydroxamate Analogues as Histone Deacetylases Inhibitors and Antitumor Agents. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.1.129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Sodji QH, Patil V, Kornacki JR, Mrksich M, Oyelere AK. Synthesis and structure-activity relationship of 3-hydroxypyridine-2-thione-based histone deacetylase inhibitors. J Med Chem 2013; 56:9969-81. [PMID: 24304348 PMCID: PMC4029159 DOI: 10.1021/jm401225q] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously identified 3-hydroxypyridine-2-thione (3HPT) as a novel zinc binding group for histone deacetylase (HDAC) inhibition. Early structure-activity relationship (SAR) studies led to various small molecules possessing selective inhibitory activity against HDAC6 or HDAC8 but devoid of HDAC1 inhibition. To delineate further the depth of the SAR of 3HPT-derived HDAC inhibitors (HDACi), we have extended the SAR studies to include the linker region and the surface recognition group to optimize the HDAC inhibition. The current efforts resulted in the identification of two lead compounds, 10d and 14e, with potent HDAC6 and HDAC8 activities that are inactive against HDAC1. These new HDACi possess anticancer activities against various cancer cell lines including Jurkat J.γ1 for which SAHA and the previously disclosed 3HPT-derived HDACi were inactive.
Collapse
Affiliation(s)
- Quaovi H. Sodji
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Vishal Patil
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - James R. Kornacki
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113
| | - Milan Mrksich
- Departments of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| |
Collapse
|
40
|
Gryder BE, Rood MK, Johnson KA, Patil V, Raftery ED, Yao LPD, Rice M, Azizi B, Doyle DF, Oyelere AK. Histone deacetylase inhibitors equipped with estrogen receptor modulation activity. J Med Chem 2013; 56:5782-96. [PMID: 23786452 DOI: 10.1021/jm400467w] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We describe a set of novel histone deacetylase inhibitors (HDACi) equipped with either an antagonist or an agonist of the estrogen receptor (ER) to confer selective activity against breast cancers. These bifunctional compounds potently inhibit HDAC at nanomolar concentrations and either agonize or antagonize ERα and ERβ. The ER antagonist activities of tamoxifen-HDACi conjugates (Tam-HDACi) are nearly identical to those of tamoxifen. Conversely, ethynyl-estradiol-HDACi conjugates (EED-HDACi) have attenuated ER agonist activities relative to the parent ethynyl-estradiol. In silico docking analysis provides structural basis for the trends of ER agonism/antagonism and ER subtype selectivity. Excitingly, lead Tam-HDACi conjugates show anticancer activity that is selectively more potent against MCF-7 (ERα positive breast cancer) compared to MDA-MB-231 (triple negative breast cancer), DU145 (prostate cancer), or Vero (noncancerous cell line). This dual-targeting approach illustrates the utility of designing small molecules with an emphasis on cell-type selectivity, not merely improved potency, working toward a higher therapeutic index at the earliest stages of drug development.
Collapse
Affiliation(s)
- Berkley E Gryder
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology , Atlanta, Georgia, 30332-0400, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Guerrant W, Patil V, Canzoneri JC, Yao LP, Hood R, Oyelere AK. Dual-acting histone deacetylase-topoisomerase I inhibitors. Bioorg Med Chem Lett 2013; 23:3283-7. [PMID: 23622981 PMCID: PMC3657756 DOI: 10.1016/j.bmcl.2013.03.108] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/25/2013] [Accepted: 03/27/2013] [Indexed: 12/20/2022]
Abstract
Current chemotherapy regimens are comprised mostly of single-target drugs which are often plagued by toxic side effects and resistance development. A pharmacological strategy for circumventing these drawbacks could involve designing multivalent ligands that can modulate multiple targets while avoiding the toxicity of a single-targeted agent. Two attractive targets, histone deacetylase (HDAC) and topoisomerase I (Topo I), are cellular modulators that can broadly arrest cancer proliferation through a range of downstream effects. Both are clinically validated targets with multiple inhibitors in therapeutic use. We describe herein the design and synthesis of dual-acting histone deacetylase-topoisomerase I inhibitors. We also show that these dual-acting agents retain activity against HDAC and Topo I, and potently arrest cancer proliferation.
Collapse
Affiliation(s)
- William Guerrant
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Vishal Patil
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Joshua C. Canzoneri
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Li-Pan Yao
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Rebecca Hood
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332-0400 USA
| |
Collapse
|
42
|
Patil V, Sodji QH, Kornacki JR, Mrksich M, Oyelere AK. 3-Hydroxypyridin-2-thione as novel zinc binding group for selective histone deacetylase inhibition. J Med Chem 2013; 56:3492-506. [PMID: 23547652 DOI: 10.1021/jm301769u] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Small molecules bearing hydroxamic acid as the zinc binding group (ZBG) have been the most effective histone deacetylase inhibitors (HDACi) to date. However, concerns about the pharmacokinetic liabilities of the hydroxamic acid moiety have stimulated research efforts aimed at finding alternative nonhydroxamate ZBGs. We have identified 3-hydroxypyridin-2-thione (3-HPT) as a novel ZBG that is compatible with HDAC inhibition. 3-HPT inhibits HDAC 6 and HDAC 8 with an IC50 of 681 and 3675 nM, respectively. Remarkably, 3-HPT gives no inhibition of HDAC 1. Subsequent optimization led to several novel 3HPT-based HDACi that are selective for HDAC 6 and HDAC 8. Furthermore, a subset of these inhibitors induces apoptosis in various cancer cell lines.
Collapse
Affiliation(s)
- Vishal Patil
- School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332-0400, United States
| | | | | | | | | |
Collapse
|
43
|
Ruan ZX, Huangfu DS, Xu XJ, Sun PH, Chen WM. 3D-QSAR and molecular docking for the discovery of ketolide derivatives. Expert Opin Drug Discov 2013; 8:427-44. [DOI: 10.1517/17460441.2013.774369] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Zhi-Xiong Ruan
- Jinan University, College of Pharmacy, Department of Medicinal Chemistry,
Guangzhou 510632, P. R. China ;
| | - De-Sheng Huangfu
- Jinan University, College of Pharmacy, Department of Medicinal Chemistry,
Guangzhou 510632, P. R. China ;
| | - Xing-Jun Xu
- Jinan University, College of Pharmacy, Department of Medicinal Chemistry,
Guangzhou 510632, P. R. China ;
| | - Ping-Hua Sun
- Jinan University, College of Pharmacy, Department of Medicinal Chemistry,
Guangzhou 510632, P. R. China ;
| | - Wei-Min Chen
- Jinan University, College of Pharmacy, Department of Medicinal Chemistry,
Guangzhou 510632, P. R. China ;
| |
Collapse
|
44
|
A novel two-step QSAR modeling work flow to predict selectivity and activity of HDAC inhibitors. Bioorg Med Chem Lett 2013; 23:929-33. [PMID: 23321563 DOI: 10.1016/j.bmcl.2012.12.067] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 11/22/2022]
Abstract
A two-step modeling approach was employed to study the selectivity and activity of histone deacetylase inhibitors. First, according to the activity difference against HDAC1 and HDAC6, a binary classification model was established to classify two kinds of inhibitors. Then two continuous models were built for each subclass to predict the activity value of HDAC1 and HDAC6 inhibitors. The three models were all built with the GA-kNN method combined with dragon descriptors. They were external validated by using external prediction set and Y-randomization test. The highly predictive models were generated for all three data sets. For the classification model, the classification accuracies of the models were as high as 100% for the external test set. For HDAC1 and HDAC6 inhibitor consecutive models, external R(2) values are 0.947 and 0.911, respectively. The results proved the reliability of these models. All models were used to screen 1000 compounds included in PubMed dataset. Virtual screening resulted in 8 and 13 structurally unique consensus hits that were considered novel putative HDAC1 and HDAC6 inhibitors, respectively.
Collapse
|
45
|
Patil V, Canzoneri JC, Samatov TR, Lührmann R, Oyelere AK. Molecular architecture of zinc chelating small molecules that inhibit spliceosome assembly at an early stage. RNA (NEW YORK, N.Y.) 2012; 18:1605-11. [PMID: 22832025 PMCID: PMC3425776 DOI: 10.1261/rna.034819.112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The removal of intervening sequences (introns) from a primary RNA transcript is catalyzed by the spliceosome, a large ribonucleoprotein complex. At the start of each splicing cycle, the spliceosome assembles anew in a sequentially ordered manner on the pre-mRNA intron to be removed. We describe here the identification of a series of naphthalen-2-yl hydroxamate compounds that inhibit pre-mRNA splicing in vitro with mid- to high-micromolar values of IC(50). These hydroxamates stall spliceosome assembly at the A complex stage. A structure-activity analysis of lead compounds revealed three pharmacophores that are essential for splicing inhibition. Specifically, a hydroxamate as a zinc-binding group and a 6-methoxynaphthalene cap group are both critical, and a linker chain comprising eight to nine methylene groups is also important, for the specific binding to the docking site of a target protein molecule and precise positioning of the zinc binding group. As we found no correlation between the inhibition patterns of known histone deacetylases on the one hand and pre-mRNA splicing on the other, we conclude that these compounds may function through the inhibition of the activities of other, at present, unknown spliceosome-associated zinc metalloprotein(s).
Collapse
Affiliation(s)
- Vishal Patil
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, USA
| | - Josh C. Canzoneri
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, USA
| | - Timur R. Samatov
- Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | - Reinhard Lührmann
- Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
- Corresponding authorsE-mail E-mail
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, USA
- Corresponding authorsE-mail E-mail
| |
Collapse
|
46
|
Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Med Chem 2012; 4:505-24. [PMID: 22416777 DOI: 10.4155/fmc.12.3] [Citation(s) in RCA: 296] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Histone deacetylase inhibitors (HDACis) have now emerged as a powerful new class of small-molecule therapeutics acting through the regulation of the acetylation states of histone proteins (a form of epigenetic modulation) and other non-histone protein targets. Over 490 clinical trials have been initiated in the last 10 years, culminating in the approval of two structurally distinct HDACis - SAHA (vorinostat, Zolinza™) and FK228 (romidepsin, Istodax™). However, the current HDACis have serious limitations, including ineffectively low concentrations in solid tumors and cardiac toxicity, which is hindering their progress in the clinic. Herein, we review the primary paradigms being pursued to overcome these hindrances, including HDAC isoform selectivity, localized administration, and targeting cap groups to achieve selective tissue and cell type distribution.
Collapse
|
47
|
Guerrant W, Patil V, Canzoneri JC, Oyelere AK. Dual targeting of histone deacetylase and topoisomerase II with novel bifunctional inhibitors. J Med Chem 2012; 55:1465-77. [PMID: 22260166 DOI: 10.1021/jm200799p] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Strategies to ameliorate the flaws of current chemotherapeutic agents, while maintaining potent anticancer activity, are of particular interest. Agents which can modulate multiple targets may have superior utility and fewer side effects than current single-target drugs. To explore the prospect in cancer therapy of a bivalent agent that combines two complementary chemo-active groups within a single molecular architecture, we have synthesized dual-acting histone deacetylase and topoisomerase II inhibitors. These dual-acting agents are derived from suberoylanilide hydroxamic acid (SAHA) and anthracycline daunorubicin, prototypical histone deacetylase (HDAC) and topoisomerase II (Topo II) inhibitors, respectively. We report herein that these agents present the signatures of inhibition of HDAC and Topo II in both cell-free and whole-cell assays. Moreover, these agents potently inhibit the proliferation of representative cancer cell lines.
Collapse
Affiliation(s)
- William Guerrant
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0400, United States
| | | | | | | |
Collapse
|
48
|
Abstract
Parasitic diseases cause significant global morbidity and mortality, particularly in underdeveloped regions of the world. Malaria alone causes ~800000 deaths each year, with children and pregnant women being at highest risk. There is no licensed vaccine available for any human parasitic disease and drug resistance is compromising the efficacy of many available anti-parasitic drugs. This is driving drug discovery research on new agents with novel modes of action. Histone deacetylase (HDAC) inhibitors are being investigated as drugs for a range of diseases, including cancers and infectious diseases such as HIV/AIDS, and several parasitic diseases. This review focuses on the current state of knowledge of HDAC inhibitors targeted to the major human parasitic diseases malaria, schistosomiasis, trypanosomiasis, toxoplasmosis and leishmaniasis. Insights are provided into the unique challenges that will need to be considered if HDAC inhibitors are to be progressed towards clinical development as potential new anti-parasitic drugs.
Collapse
Affiliation(s)
- Katherine T Andrews
- Eskitis Institute for Cell and Molecular Therapies, Griffith University, Nathan, Queensland, Australia.
| | | | | |
Collapse
|
49
|
Lombardi PM, Cole KE, Dowling DP, Christianson DW. Structure, mechanism, and inhibition of histone deacetylases and related metalloenzymes. Curr Opin Struct Biol 2011; 21:735-43. [PMID: 21872466 DOI: 10.1016/j.sbi.2011.08.004] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 08/01/2011] [Indexed: 10/17/2022]
Abstract
Metal-dependent histone deacetylases (HDACs) catalyze the hydrolysis of acetyl-L-lysine side chains in histone and nonhistone proteins to yield l-lysine and acetate. This chemistry plays a critical role in the regulation of numerous biological processes. Aberrant HDAC activity is implicated in various diseases, and HDACs are validated targets for drug design. Two HDAC inhibitors are currently approved for cancer chemotherapy, and other inhibitors are in clinical trials. To date, X-ray crystal structures are available for four human HDACs (2, 4, 7, and 8) and three HDAC-related deacetylases from bacteria (histone deacetylase-like protein (HDLP); histone deacetylase-like amidohydrolase (HDAH); acetylpolyamine amidohydrolase (APAH)). Structural comparisons among these enzymes reveal a conserved constellation of active site residues, suggesting a common mechanism for the metal-dependent hydrolysis of acetylated substrates. Structural analyses of HDACs and HDAC-related deacetylases guide the design of tight-binding inhibitors, and future prospects for developing isozyme-specific inhibitors are quite promising.
Collapse
Affiliation(s)
- Patrick M Lombardi
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | | | | | | |
Collapse
|
50
|
Shao C, Wang X, Zhang Q, Luo S, Zhao J, Hu Y. Acid-base jointly promoted copper(I)-catalyzed azide-alkyne cycloaddition. J Org Chem 2011; 76:6832-6. [PMID: 21793533 DOI: 10.1021/jo200869a] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this novel acid-base jointly promoted CuAAC, the combination of CuI/DIPEA/HOAc was developed as a highly efficient catalytic system. The functions of DIPEA and HOAc have been assigned, and HOAc was recognized to accelerate the conversions of the C-Cu bond-containing intermediates and buffer the basicity of DIPEA. As a result, all drawbacks occurring in the popular catalytic system CuI/NR(3) were overcome easily.
Collapse
Affiliation(s)
- Changwei Shao
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | | | | | | | | | | |
Collapse
|