1
|
Chua HM, Moshawih S, Kifli N, Goh HP, Ming LC. Insights into the computer-aided drug design and discovery based on anthraquinone scaffold for cancer treatment: A systematic review. PLoS One 2024; 19:e0301396. [PMID: 38776291 PMCID: PMC11111074 DOI: 10.1371/journal.pone.0301396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND In the search for better anticancer drugs, computer-aided drug design (CADD) techniques play an indispensable role in facilitating the lengthy and costly drug discovery process especially when natural products are involved. Anthraquinone is one of the most widely-recognized natural products with anticancer properties. This review aimed to systematically assess and synthesize evidence on the utilization of CADD techniques centered on the anthraquinone scaffold for cancer treatment. METHODS The conduct and reporting of this review were done in accordance to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) 2020 guideline. The protocol was registered in the "International prospective register of systematic reviews" database (PROSPERO: CRD42023432904) and also published recently. The search strategy was designed based on the combination of concept 1 "CADD or virtual screening", concept 2 "anthraquinone" and concept 3 "cancer". The search was executed in PubMed, Scopus, Web of Science and MedRxiv on 30 June 2023. RESULTS Databases searching retrieved a total of 317 records. After deduplication and applying the eligibility criteria, the final review ended up with 32 articles in which 3 articles were found by citation searching. The CADD methods used in the studies were either structure-based alone (69%) or combined with ligand-based methods via parallel (9%) or sequential (22%) approaches. Molecular docking was performed in all studies, with Glide and AutoDock being the most popular commercial and public software used respectively. Protein data bank was used in most studies to retrieve the crystal structure of the targets of interest while the main ligand databases were PubChem and Zinc. The utilization of in-silico techniques has enabled a deeper dive into the structural, biological and pharmacological properties of anthraquinone derivatives, revealing their remarkable anticancer properties in an all-rounded fashion. CONCLUSION By harnessing the power of computational tools and leveraging the natural diversity of anthraquinone compounds, researchers can expedite the development of better drugs to address the unmet medical needs in cancer treatment by improving the treatment outcome for cancer patients.
Collapse
Affiliation(s)
- Hui Ming Chua
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Said Moshawih
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Nurolaini Kifli
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Hui Poh Goh
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
- School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
2
|
Abdulwahab HG, Mansour RES, Farghaly TA, El-Sehrawi HM. Discovery of novel benzimidazole derivatives as potent HDACs inhibitors against leukemia with (Thio)Hydantoin as zinc-binding moiety: Design, synthesis, enzyme inhibition, and cellular mechanistic study. Bioorg Chem 2024; 146:107284. [PMID: 38493640 DOI: 10.1016/j.bioorg.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/02/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Based on the well-established pharmacophoric features required for histone deacetylase (HDAC) inhibition, a novel series of easy-to-synthesize benzimidazole-linked (thio)hydantoin derivatives was designed and synthesized as HDAC6 inhibitors. All target compounds potently inhibited HDAC6 at nanomolar levels with compounds 2c, 2d, 4b and 4c (IC50s = 51.84-74.36 nM) being more potent than SAHA reference drug (IC50 = 91.73 nM). Additionally, the most potent derivatives were further assessed for their in vitro cytotoxic activity against two human leukemia cells. Hydantoin derivative 4c was equipotent/superior to SAHA against MOLT-4/CCRF-CEM leukemia cells, respectively and demonstrated safety profile better than that of SAHA against non-cancerous human cells. 4c was also screened against different HDAC isoforms. 4c was superior to SAHA against HDAC1. Cell-based assessment of 4c revealed a significant cell cycle arrest and apoptosis induction. Moreover, western blotting analysis showed increased levels of acetylated histone H3, histone H4 and α-tubulin in CCRF-CEM cells. Furthermore, docking study exposed the ability of title compounds to chelate Zn2+ located within HDAC6 active site. As well, in-silico evaluation of physicochemical properties showed that target compounds are promising candidates in terms of pharmacokinetic aspects.
Collapse
Affiliation(s)
- Hanan Gaber Abdulwahab
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| | - Reda El-Sayed Mansour
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Thoraya A Farghaly
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Hend M El-Sehrawi
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Drakontaeidi A, Pontiki E. A Review on Molecular Docking on HDAC Isoforms: Novel Tool for Designing Selective Inhibitors. Pharmaceuticals (Basel) 2023; 16:1639. [PMID: 38139766 PMCID: PMC10746130 DOI: 10.3390/ph16121639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 12/24/2023] Open
Abstract
Research into histone deacetylases (HDACs) has experienced a remarkable surge in recent years. These enzymes are key regulators of several fundamental biological processes, often associated with severe and potentially fatal diseases. Inhibition of their activity represents a promising therapeutic approach and a prospective strategy for the development of new therapeutic agents. A critical aspect of their inhibition is to achieve selectivity in terms of enzyme isoforms, which is essential to improve treatment efficacy while reducing undesirable pleiotropic effects. The development of computational chemistry tools, particularly molecular docking, is greatly enhancing the precision of designing molecules with inherent potential for specific activity. Therefore, it was considered necessary to review the molecular docking studies conducted on the major isozymes of the enzyme in order to identify the specific interactions associated with each selective HDAC inhibitor. In particular, the most critical isozymes of HDAC (1, 2, 3, 6, and 8) have been thoroughly investigated within the scope of this review.
Collapse
Affiliation(s)
| | - Eleni Pontiki
- Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
4
|
Yang Y, Liu Q, Wang X, Gou S. Design, Synthesis, and Biological Evaluation of Histone Deacetylase Inhibitors Derived from Erianin and Its Derivatives. ChemMedChem 2023; 18:e202300108. [PMID: 37058395 DOI: 10.1002/cmdc.202300108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/15/2023]
Abstract
Multi-target histone deacetylase (HDAC) inhibitors can be designed by introducing dominant structures of natural products to enhance activity and efficacy while avoiding the toxicity from other targets. In this study, we reported a series of novel HDAC inhibitors based on erianin and amino erianin upon pharmacophore fusion strategy. Two representative compounds, N-hydroxy-2-(2-methoxy-5- (3,4,5-trimethoxyphenethyl)phenoxy)acetamide and N-Hydroxy-8-((2-methoxy-5- (3,4,5-trimethoxyphenethyl)phenyl)amino)octanamide, possessed good inhibitory effect against five cancer cells tested (IC50 =0.30-1.29 μΜ, 0.29-1.70 μΜ) with strong HDAC inhibition, and low toxicity toward L02 cells, which were selected for subsequent biological studies in PANC-1 cells. They were also found to promote the intracellular generation of reactive oxygen species, cause DNA damage, block the cell cycle at G2/M phase, and activate the mitochondria-related apoptotic pathway to induce cell apoptosis, which are significant for the discovery of new HDAC inhibitors.
Collapse
Affiliation(s)
- Yawen Yang
- Pharmaceutical Research Center and, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Qingqing Liu
- Pharmaceutical Research Center and, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
- School of Pharmacy, Jilin Medical University, Jilin City, 132013, Jilin Province, China
| | - Xinyi Wang
- Pharmaceutical Research Center and, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China
| |
Collapse
|
5
|
Khetmalis YM, Fathima A, Schweipert M, Debarnot C, Bandaru NVMR, Murugesan S, Jamma T, Meyer-Almes FJ, Sekhar KVGC. Design, Synthesis, and Biological Evaluation of Novel Quinazolin-4(3H)-One-Based Histone Deacetylase 6 (HDAC6) Inhibitors for Anticancer Activity. Int J Mol Sci 2023; 24:11044. [PMID: 37446224 DOI: 10.3390/ijms241311044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
A series of novel quinazoline-4-(3H)-one derivatives were designed and synthesized as histone deacetylase 6 (HDAC6) inhibitors based on novel quinazoline-4-(3H)-one as the cap group and benzhydroxamic acid as the linker and metal-binding group. A total of 19 novel quinazoline-4-(3H)-one analogues (5a-5s) were obtained. The structures of the target compounds were characterized using 1H-NMR, 13C-NMR, LC-MS, and elemental analyses. Characterized compounds were screened for inhibition against HDAC8 class I, HDAC4 class IIa, and HDAC6 class IIb. Among the compounds tested, 5b proved to be the most potent and selective inhibitor of HDAC6 with an IC50 value 150 nM. Some of these compounds showed potent antiproliferative activity in several tumor cell lines (HCT116, MCF7, and B16). Amongst all the compounds tested for their anticancer effect against cancer cell lines, 5c emerged to be most active against the MCF-7 line with an IC50 of 13.7 μM; it exhibited cell-cycle arrest in the G2 phase, as well as promoted apoptosis. Additionally, we noted a significant reduction in the colony-forming capability of cancer cells in the presence of 5c. At the intracellular level, selective inhibition of HDAC6 was enumerated by monitoring the acetylation of α-tubulin with a limited effect on acetyl-H3. Importantly, the obtained results suggested a potent effect of 5c at sub-micromolar concentrations as compared to the other molecules as HDAC6 inhibitors in vitro.
Collapse
Affiliation(s)
- Yogesh Mahadu Khetmalis
- Department of Chemistry, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Hyderabad 500078, Telangana, India
| | - Ashna Fathima
- Department of Biological Sciences, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Hyderabad 500078, Telangana, India
| | - Markus Schweipert
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Haardtring 100, 64295 Darmstadt, Germany
| | - Cécile Debarnot
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Haardtring 100, 64295 Darmstadt, Germany
| | | | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science Pilani, Pilani Campus, Pilani 333031, Rajasthan, India
| | - Trinath Jamma
- Department of Biological Sciences, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Hyderabad 500078, Telangana, India
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences Darmstadt, Haardtring 100, 64295 Darmstadt, Germany
| | | |
Collapse
|
6
|
Raghuveer D, Pai VV, Murali TS, Nayak R. Exploring Anthraquinones as Antibacterial and Antifungal agents. ChemistrySelect 2023. [DOI: 10.1002/slct.202204537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Dhanush Raghuveer
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - V. Varsha Pai
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - Thokur Sreepathy Murali
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - Roopa Nayak
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| |
Collapse
|
7
|
Khetmalis YM, Shree B, Kumar BVS, Schweipert M, Debarnot C, Ashna F, Sankaranarayanan M, Trinath J, Sharma V, Meyer-Almes FJ, Sekhar KVGC. Design, Synthesis, and Biological Evaluation of Tetrahydroisoquinoline Based Hydroxamate Derivatives as HDAC 6 Inhibitors For Cancer Therapy. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.134952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
8
|
Kaur S, Rajoria P, Chopra M. HDAC6: A unique HDAC family member as a cancer target. Cell Oncol (Dordr) 2022; 45:779-829. [PMID: 36036883 DOI: 10.1007/s13402-022-00704-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND HDAC6, a structurally and functionally distinct member of the HDAC family, is an integral part of multiple cellular functions such as cell proliferation, apoptosis, senescence, DNA damage and genomic stability, all of which when deregulated contribute to carcinogenesis. Among several HDAC family members known so far, HDAC6 holds a unique position. It differs from the other HDAC family members not only in terms of its subcellular localization, but also in terms of its substrate repertoire and hence cellular functions. Recent findings have considerably expanded the research related to the substrate pool, biological functions and regulation of HDAC6. Studies in HDAC6 knockout mice highlighted the importance of HDAC6 as a cell survival player in stressful situations, making it an important anticancer target. There is ample evidence stressing the importance of HDAC6 as an anti-cancer synergistic partner of many chemotherapeutic drugs. HDAC6 inhibitors have been found to enhance the effectiveness of conventional chemotherapeutic drugs such as DNA damaging agents, proteasome inhibitors and microtubule inhibitors, thereby highlighting the importance of combination therapies involving HDAC6 inhibitors and other anti-cancer agents. CONCLUSIONS Here, we present a review on HDAC6 with emphasis on its role as a critical regulator of specific physiological cellular pathways which when deregulated contribute to tumorigenesis, thereby highlighting the importance of HDAC6 inhibitors as important anticancer agents alone and in combination with other chemotherapeutic drugs. We also discuss the synergistic anticancer effect of combination therapies of HDAC6 inhibitors with conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Sumeet Kaur
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Prerna Rajoria
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
9
|
Chang Q, Yin D, Li H, Du X, Wang Z, Liu Y, Zhang J. HDAC6-specific inhibitor alleviates hashimoto's thyroiditis through inhibition of Th17 cell differentiation. Mol Immunol 2022; 149:39-47. [PMID: 35717700 DOI: 10.1016/j.molimm.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/26/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Hashimoto's thyroiditis (HT) is one of the commonest autoimmune disorders. This study was performed to investigate the potential effect of histone deacetylase 6-specific inhibitor (HDAC6i) on Th17 cell differentiation in animal model and the underlying mechanism. METHODS Experimental autoimmune thyroiditis (EAT) mouse model was established by subcutaneously immunization of porcine thyroglobulin (pTg) and adjuvant, and the HDACi Tubastatin A (TSA) or HDAC6i (ACY-1215) was intraperitoneally injected into mice in the following. The histological examination and immune analysis in EAT mice were carried out. Next, the CD4+ T cells were isolated from peripheral blood mononuclear cells (PBMCs) of EAT mice followed by Th17 cell differentiation assay. The associated factor levels, and the protein interaction between HDAC6 and PKM2 were examined. Subsequently, the effect of STAT3 activation on Th17 cell differentiation was explored. RESULTS ACY-1215 or TSA treatment reduced lymphocytic infiltration and alleviated thyroid tissue injury in EAT mice. Correspondingly, either ACY-1215 or TSA treatment reduced the levels of anti-thyroglobulin (Tg), anti-thyroid peroxidase (TPO), IL-17A, and IFN-γ in the serum, decreased Th17 cell differentiation, but enhanced TGF-β level and promoted Treg cell differentiation. In vitro, after induction of Th17 cell differentiation from CD4+ T cells, HDAC6 activity and Th17 cell differentiation were significantly decreased when treated with ACY-1215 or TSA. HDAC6 could interact with PKM2, and HDAC6 overexpression promoted the phosphorylation of STAT3 and PKM2 nuclear translocation. Furthermore, the STAT3 activator treatment reversed the effects of ACY-1215 or TSA treatment. CONCLUSION HDAC6i suppresses Th17 cell differentiation and attenuates HT via PKM2/STAT3 axis.
Collapse
Affiliation(s)
- Qungang Chang
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Discipline Laboratory of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Detao Yin
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Discipline Laboratory of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongqiang Li
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin Du
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zipeng Wang
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yihao Liu
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jieming Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
10
|
Kim JH, Ali KH, Oh YJ, Seo YH. Design, synthesis, and biological evaluation of histone deacetylase inhibitor with novel salicylamide zinc binding group. Medicine (Baltimore) 2022; 101:e29049. [PMID: 35512065 PMCID: PMC9276175 DOI: 10.1097/md.0000000000029049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/11/2021] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Histone deacetylases (HDACs) have emerged as important therapeutic targets for various diseases, such as cancer and neurological disorders. Although a majority of HDAC inhibitors use hydroxamic acids as zinc binding groups, hydroxamic acid zinc-binding groups suffer from poor bioavailability and nonspecific metal-binding properties, necessitating a new zinc-binding group. Salicylic acid and its derivatives, well-known for their therapeutic value, have also been reported to chelate zinc ions in a bidentate fashion. This drew our attention towards replacing hydroxamic acid with salicylamide as a zinc-binding group. METHODS In this study, for the first time, compound 5 possessing a novel salicylamide zinc-binding group was synthesized and evaluated biologically for its ability to inhibit various HDAC isoforms and induce acetylation upon α-tubulin and histone H3 among MDA-MB-231 cells. RESULTS Compound 5 exhibits selective inhibition against class I HDAC isoforms (HDAC1, 2, and 3) over class II and IV HDAC isoforms (HDAC4, 6, and 11). The exposure of MDA-MB-231 cells to compound 5 efficiently induced the acetylation of more histone H3 than α-tubulin, suggesting that compound 5 is a class I selective HDAC inhibitor. Moreover, the molecular docking study indicated that the salicylamide zinc-binding group of compound 5 coordinates the active zinc ion of class I HDAC2 in a bidentate fashion. CONCLUSION Overall, salicylamide represents a novel zinc-binding group for the development of class I selective HDAC inhibitors. GRAPHICAL ABSTRACT (http://links.lww.com/MD/G668).
Collapse
|
11
|
Peng X, Li L, Chen J, Ren Y, Liu J, Yu Z, Cao H, Chen J. Discovery of Novel Histone Deacetylase 6 (HDAC6) Inhibitors with Enhanced Antitumor Immunity of Anti-PD-L1 Immunotherapy in Melanoma. J Med Chem 2022; 65:2434-2457. [PMID: 35043615 DOI: 10.1021/acs.jmedchem.1c01863] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A series of 2-phenylthiazole analogues were designed and synthesized as potential histone deacetylase 6 (HDAC6) inhibitors based on compound 12c (an HDAC6/tubulin dual inhibitor discovered by us recently) and CAY10603 (a known HDAC6 inhibitor). Among them, compound XP5 was the most potent HDAC6 inhibitor with an IC50 of 31 nM and excellent HDAC6 selectivity (SI = 338 for HDAC6 over HDAC3). XP5 also displayed high antiproliferative activity against various cancer cell lines including the HDACi-resistant YCC3/7 gastric cancer cells (IC50 = 0.16-2.31 μM), better than CAY10603. Further, XP5 (50 mg/kg) exhibited significant antitumor efficacy in a melanoma tumor model with a tumor growth inhibition (TGI) of 63% without apparent toxicity. Moreover, XP5 efficiently enhanced the in vivo antitumor immune response when combined with a small-molecule PD-L1 inhibitor, as demonstrated by the increased tumor-infiltrating lymphocytes and reduced PD-L1 expression levels. Taken together, the above results suggest that XP5 is a promising HDAC6 inhibitor deserving further investigation.
Collapse
Affiliation(s)
- Xiaopeng Peng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Ling Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Jingxuan Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Yichang Ren
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Jin Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Ziwen Yu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Hao Cao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 516000, China
| |
Collapse
|
12
|
Qiu X, Zhu L, Wang H, Tan Y, Yang Z, Yang L, Wan L. From natural products to HDAC inhibitors: An overview of drug discovery and design strategy. Bioorg Med Chem 2021; 52:116510. [PMID: 34826681 DOI: 10.1016/j.bmc.2021.116510] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 02/08/2023]
Abstract
Histone deacetylases (HDACs) play a key role in the homeostasis of protein acetylation in histones and have recently emerged as a therapeutic target for numerous diseases. The inhibition of HDACs may block angiogenesis, arrest cell growth, and lead to differentiation and apoptosis in tumour cells. Thus, HDAC inhibitors (HDACi) have received increasing attention and many of which are developed from natural sources. In the past few decades, naturally occurring HDACi have been identified to have potent anticancer activities, some of which have demonstrated promising therapeutic effects on haematological malignancies. In this review, we summarized the discovery and modification of HDAC inhibitors from natural sources, novel drug design that uses natural products as parent nuclei, and dual target design strategies that combine HDAC with non-HDAC targets.
Collapse
Affiliation(s)
- Xiang Qiu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lv Zhu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Wan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
13
|
Ukey S, Choudhury C, Sharma P. Identification of unique subtype-specific interaction features in Class II zinc-dependent HDAC subtype binding pockets: A computational study. J Biosci 2021. [DOI: 10.1007/s12038-021-00197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
14
|
Zhang M, Wei W, Peng C, Ma X, He X, Zhang H, Zhou M. Discovery of novel pyrazolopyrimidine derivatives as potent mTOR/HDAC bi-functional inhibitors via pharmacophore-merging strategy. Bioorg Med Chem Lett 2021; 49:128286. [PMID: 34314844 DOI: 10.1016/j.bmcl.2021.128286] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 02/08/2023]
Abstract
The mTOR and HDAC dual suppression is meaningful for counteracting drug resistance resulted from kinase mutation and bypass mechanisms. Herein, we communicate our recent discovery of a novel structural series of mTOR/HDAC bi-functional inhibitors featuring the pyrazolopyrimidine core via pharmacophore-merging strategy. More than half of them exerted potent dual-target inhibitory activities. In particular, compound 50 exhibited IC50 values of 0.49 and 0.91 nM against mTOR and HDAC1, respectively, along with remarkably enhanced anti-proliferative activity (IC50 = 1.74 μM) against MV4-11 cell line than mTOR inhibitor MLN-0128 (IC50 = 5.84 μM) and HDAC inhibitor SAHA (IC50 = 8.44 μM). Its intracellular intervention of both mTOR signaling and HDAC was validated by the Western blot analysis. Moreover, as the first disclosed mTOR/HDAC dual inhibitor with selectivity for some specific HDAC subtypes, it has the potential to alleviate the adverse effects resulted from pan-HDAC inhibition. Attributed to its favorable in vitro performance, compound 50 is valuable for further functional investigation as a polypharmacological anti-cancer agent.
Collapse
Affiliation(s)
- Mingming Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wei Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, China
| | - Chengjun Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Xiaodong Ma
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Department of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China.
| | - Xiao He
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Heng Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Mingkang Zhou
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
15
|
Valarmathi T, Premkumar R, Meera MR, Milton Franklin Benial A. Spectroscopic, quantum chemical and molecular docking studies on 1-amino-5-chloroanthraquinone: A targeted drug therapy for thyroid cancer. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 255:119659. [PMID: 33751957 DOI: 10.1016/j.saa.2021.119659] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/07/2021] [Accepted: 02/28/2021] [Indexed: 06/12/2023]
Abstract
The DFT studies of the 1-Amino-5-chloro-anthraquinone (ACAQ) molecule have been carried out with extensive and accurate investigations of detailed vibrational and spectroscopic investigations and validated by experimentally. The optimized molecular structure and harmonic resonance frequencies were computed based on DFT/B3LYP method with 6-311G++(d,p) basis set using the Gaussian 09 program. The experimental and calculated vibrational wavenumbers were assigned on the basis of PED calculations using VEDA 4.0 program. The 13C NMR isotropic chemical shifts of the molecule were calculated using Gauge-Invariant-Atomic Orbital (GIAO) method in DMSO solution and compared with the experimental data. The absorption spectrum of the molecule was computed in liquid phase (ethanol), which exhibits л to л* electronic transition and compared with observed UV-Vis spectrum. Frontier molecular orbitals analysis shows the molecular reactivity and kinetic stability of the molecule. The Mulliken atomic charge distribution and molecular electrostatic potential surface analysis of the molecule validate the reactive site of the molecule. The natural bond orbital analysis proves the bioactivity of the molecule. Molecular docking analysis indicate that ACAQ molecule inhibits the action of c-Met Kinase protein, which is associated with the thyroid cancer. Hence, the present study pave the way for the development of novel drugs in the treatment of thyroid cancer.
Collapse
Affiliation(s)
- T Valarmathi
- P.G. and Research Department of Physics, N.M.S.S.V.N. College, Madurai 625019, Tamil Nadu, India
| | - R Premkumar
- P.G. and Research Department of Physics, N.M.S.S.V.N. College, Madurai 625019, Tamil Nadu, India
| | - M R Meera
- Department of Physics, Sree Ayyappa College for Women, Chunkankadai, Kanyakumari 629003, Tamil Nadu, India
| | - A Milton Franklin Benial
- P.G. and Research Department of Physics, N.M.S.S.V.N. College, Madurai 625019, Tamil Nadu, India.
| |
Collapse
|
16
|
Sirazhetdinova NS, Savelyev VA, Baev DS, Golubeva TS, Klimenko LS, Tolstikova TG, Ganbaatar J, Shults EE. Synthesis, characterization and anticancer evaluation of nitrogen-substituted 1-(3-aminoprop-1-ynyl)-4-hydroxyanthraquinone derivatives. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02754-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
17
|
He X, Li Z, Zhuo XT, Hui Z, Xie T, Ye XY. Novel Selective Histone Deacetylase 6 (HDAC6) Inhibitors: A Patent Review (2016-2019). Recent Pat Anticancer Drug Discov 2021; 15:32-48. [PMID: 32065106 DOI: 10.2174/1574892815666200217125419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Many human diseases are associated with dysregulation of HDACs. HDAC6 exhibits deacetylase activity not only to histone protein but also to non-histone proteins such as α- tubulin, HSP90, cortactin, and peroxiredoxin. These unique functions of HDAC6 have gained significant attention in the medicinal chemistry community in recent years. Thus a great deal of effort has devoted to developing selective HDAC6 inhibitors for therapy with the hope to minimize the side effects caused by pan-HDAC inhibition. OBJECTIVE The review intends to analyze the structural feature of the scaffolds, to provide useful information for those who are interested in this field, as well as to spark the future design of the new inhibitors. METHODS The primary tool used for patent searching is SciFinder. All patents are retrieved from the following websites: the World Intellectual Property Organization (WIPO®), the United States Patent Trademark Office (USPTO®), Espacenet®, and Google Patents. The years of patents covered in this review are between 2016 and 2019. RESULTS Thirty-six patents from seventeen companies/academic institutes were classified into three categories based on the structure of ZBG: hydroxamic acid, 1,3,4-oxadiazole, and 1,2,4-oxadiazole. ZBG connects to the cap group through a linker. The cap group can tolerate different functional groups, including amide, urea, sulfonamide, sulfamide, etc. The cap group appears to modulate the selectivity of HDAC6 over other HDAC subtypes. CONCLUSION Selectively targeting HDAC6 over other subtypes represents two fold advantages: it maximizes the pharmacological effects and minimizes the side effects seen in pan-HDAC inhibitors. Many small molecule selective HDAC6 inhibitors have advanced to clinical studies in recent years. We anticipate the approval of selective HDAC6 inhibitors as therapeutic agents in the near future.
Collapse
Affiliation(s)
- Xingrui He
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Zhen Li
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiao-Tao Zhuo
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Zi Hui
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Tian Xie
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiang-Yang Ye
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang 311121, China.,Holistic Integrative Pharmacy Institutes (HIPI), School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
18
|
Sun S, Zhao W, Li Y, Chi Z, Fang X, Wang Q, Han Z, Luan Y. Design, synthesis and antitumor activity evaluation of novel HDAC inhibitors with tetrahydrobenzothiazole as the skeleton. Bioorg Chem 2021; 108:104652. [PMID: 33497873 DOI: 10.1016/j.bioorg.2021.104652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022]
Abstract
HDACs as important targets for cancer therapy have attracted extensive attentions. In this work, a series of sixteen hydroxamic acid based HDAC inhibitors were designed and synthesized with 4,5,6,7-tetrahydrobenzothiazole as the structural core. Majority of them exhibited potent inhibitory activities against HDACs and one leading compound 6h was dug out. 6h was proven to be a pan-HDAC inhibitor and displayed high cytotoxicity against seven human cancer cell lines with IC50 values in low micromolar range. 6h could arrest cell cycle in G2/M phase and induce apoptosis in A549 cells. Moreover, compound 6h exhibited remarkable anti-migration and anti-angiogenesis activities. At the same time, 6h was able to elevate the expression of acetylated α-tubulin and acetylated histone H3 in a dose-dependent manner. Docking simulation revealed that 6h fitted well into the active sites of HDAC2 and 6. Finally, compound 6h also exerted potent antitumor effects in an A549 zebrafish xenograft model. Our study demonstrated that compound 6h was a promising candidate for further preclinical studies.
Collapse
Affiliation(s)
- Simin Sun
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao, Shandong, China
| | - Wenwen Zhao
- Department of Pharmacology, School of Basic Medicine, Qingdao University Medical College, Qingdao, Shandong, China
| | - Yongliang Li
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao, Shandong, China
| | - Ziwei Chi
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao, Shandong, China
| | - Xixi Fang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao, Shandong, China
| | - Qiang Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China
| | - Zhiwu Han
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao, Shandong, China.
| |
Collapse
|
19
|
Song Y, Park SY, Wu Z, Liu KH, Seo YH. Hybrid inhibitors of DNA and HDACs remarkably enhance cytotoxicity in leukaemia cells. J Enzyme Inhib Med Chem 2021; 35:1069-1079. [PMID: 32314611 PMCID: PMC7191901 DOI: 10.1080/14756366.2020.1754812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chlorambucil is a nitrogen mustard-based DNA alkylating drug, which is widely used as a front-line treatment of chronic lymphocytic leukaemia (CLL). Despite its widespread application and success for the initial treatment of leukaemia, a majority of patients eventually develop acquired resistance to chlorambucil. In this regard, we have designed and synthesised a novel hybrid molecule, chloram-HDi that simultaneously impairs DNA and HDAC enzymes. Chloram-HDi efficiently inhibits the proliferation of HL-60 and U937 leukaemia cells with GI50 values of 1.24 µM and 1.75 µM, whereas chlorambucil exhibits GI50 values of 21.1 µM and 37.7 µM against HL-60 and U937 leukaemia cells, respectively. The mechanism behind its remarkably enhanced cytotoxicity is that chloram-HDi not only causes a significant DNA damage of leukaemia cells but also downregulates DNA repair protein, Rad52, resulting in the escalation of its DNA-damaging effect. Furthermore, chloram-HDi inhibits HDAC enzymes to induce the acetylation of α-tubulin and histone H3.
Collapse
Affiliation(s)
- Yoojin Song
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Sun You Park
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Zhexue Wu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Young Ho Seo
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
20
|
2-Methylquinazoline derivative 23BB as a highly selective histone deacetylase 6 inhibitor alleviated cisplatin-induced acute kidney injury. Biosci Rep 2020; 40:221748. [PMID: 31894849 PMCID: PMC6970081 DOI: 10.1042/bsr20191538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 12/13/2019] [Accepted: 12/31/2019] [Indexed: 02/05/2023] Open
Abstract
Histone deacetylases 6 (HDAC6) has been reported to be involved in the pathogenesis of cisplatin-induced acute kidney injury (AKI). Selective inhibition of HDAC6 might be a potential treatment for AKI. In our previous study, a highly selective HDAC6 inhibitor (HDAC6i) 23BB effectively protected against rhabdomyolysis-induced AKI with good safety. However, whether 23BB possessed favorable renoprotection against cisplatin-induced AKI and the involved mechanisms remained unknown. In the study, cisplatin-injected mice developed severe AKI symptom as indicated by acute kidney dysfunction and pathological changes, companied by the overexpression of HDAC6 in tubular epithelial cells. Pharmacological inhibition of HDAC6 by the treatment of 23BB significantly attenuated sCr, BUN and renal tubular damage. Mechanistically, 23BB enhanced the acetylation of histone H3 to reduce the HDAC6 activity. Cisplatin-induced AKI triggered multiple signal mediators of endoplasmic reticulum (ER) stress including PERK, ATF6 and IRE1 pathway, as well as CHOP, GRP78, p-JNK and caspase 12 proteins. Oral administration of our HDAC6i 23BB at a dose of 40 mg/kg/d for 3 days notably improved above-mentioned responses in the injured kidney tissues. HDAC6 inhibition also reduced the number of TUNEL-positive tubular cells and regulated apoptosis-related protein expression. Overall, these data highlighted that HDAC6 inhibitor 23BB modulated apoptosis via the inhibition of ER stress in the tubular epithelial cells of cisplatin-induced AKI.
Collapse
|
21
|
Pulya S, Amin SA, Adhikari N, Biswas S, Jha T, Ghosh B. HDAC6 as privileged target in drug discovery: A perspective. Pharmacol Res 2020; 163:105274. [PMID: 33171304 DOI: 10.1016/j.phrs.2020.105274] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/15/2020] [Accepted: 10/25/2020] [Indexed: 12/25/2022]
Abstract
HDAC6, a class IIB HDAC isoenzyme, stands unique in its structural and physiological functions. Besides histone modification, largely due to its cytoplasmic localization, HDAC6 also targets several non-histone proteins including Hsp90, α-tubulin, cortactin, HSF1, etc. Thus, it is one of the key regulators of different physiological and pathological disease conditions. HDAC6 is involved in different signaling pathways associated with several neurological disorders, various cancers at early and advanced stage, rare diseases and immunological conditions. Therefore, targeting HDAC6 has been found to be effective for various therapeutic purposes in recent years. Though several HDAC6 inhibitors (HDAC6is) have been developed till date, only two ACY-1215 (ricolinostat) and ACY-241 (citarinostat) are in the clinical trials. A lot of work is still needed to pinpoint strictly selective as well as potent HDAC6i. Considering the recent crystal structure of HDAC6, novel HDAC6is of significant therapeutic value can be designed. Notably, the canonical pharmacophore features of HDAC6is consist of a zinc binding group (ZBG), a linker function and a cap group. Significant modifications of cap function may lead to achieve better selectivity of the inhibitors. This review details the study about the structural biology of HDAC6, the physiological and pathological role of HDAC6 in several disease states and the detailed structure-activity relationships (SARs) of the known HDAC6is. This detailed review will provide key insights to design novel and highly effective HDAC6i in the future.
Collapse
Affiliation(s)
- Sravani Pulya
- Epigenetic Research Laboratory, Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India
| | - Sk Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur University, Kolkata 700032, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur University, Kolkata 700032, India
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur University, Kolkata 700032, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India.
| |
Collapse
|
22
|
Recent advances in small molecular modulators targeting histone deacetylase 6. FUTURE DRUG DISCOVERY 2020. [DOI: 10.4155/fdd-2020-0023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a unique isozyme in the HDAC family with various distinguished characters. HDAC6 is predominantly localized in the cytoplasm and has several specific nonhistone substrates, such as α-tubulin, cortactin, Hsp90, tau and peroxiredoxins. Accumulating evidence reveals that targeting HDAC6 may serve as a promising therapeutic strategy for the treatment of cancers, neurological disorders and immune diseases, making the development of HDAC6 inhibitors particularly attractive. Recently, multitarget drug design and proteolysis targeting chimera technology have also been applied in the discovery of novel small molecular modulators targeting HDAC6. In this review, we briefly describe the structural features and biological functions of HDAC6 and discuss the recent advances in HDAC6 modulators, including selective inhibitors, chimeric inhibitors and proteolysis targeting chimeras for multiple therapeutic purposes.
Collapse
|
23
|
Dual inhibitors of histone deacetylases and other cancer-related targets: A pharmacological perspective. Biochem Pharmacol 2020; 182:114224. [PMID: 32956642 DOI: 10.1016/j.bcp.2020.114224] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Epigenetic enzymes histone deacetylases (HDACs) are clinically validated anticancer drug targets which have been studied intensively in the past few decades. Although several drugs have been approved in this field, they are still limited to a subset of hematological malignancies (in particular T-cell lymphomas), with therapeutic potential not fully realized and the drug-resistance occurred after a certain period of use. To maximize the therapeutic potential of these classes of anticancer drugs, and to extend their application to solid tumors, numerous combination therapies containing an HDACi and an anticancer agent from other mechanisms are currently ongoing in clinical trials. Recently, dual targeting strategy comprising the HDACs component has emerged as an alternative approach for combination therapies. In this perspective, we intend to gather all HDACs-containing dual inhibitors related to cancer therapy published in literature since 2015, classify them into five categories based on targets' biological functions, and discuss the rationale why dual acting agents should work better than combinatorial therapies using two separate drugs. The article discusses the pharmacological aspects of these dual inhibitors, including in vitro biological activities, pharmacokinetic studies, in vivo efficacy studies, as well as available clinical trials. The review of the current status and advances should provide better analysis for future opportunities and challenges of this field.
Collapse
|
24
|
Zou Y, Cao Z, Wang J, Chen X, Chen YQ, Li Y, Liu J, Zhao Y, Wang A, He B. A Series of Novel HDAC Inhibitors with Anthraquinone as a Cap Group. Chem Pharm Bull (Tokyo) 2020; 68:613-617. [PMID: 32611998 DOI: 10.1248/cpb.c20-00206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although anthraquinone derivatives possess significant antitumor activity, most of them also displayed those side effects like cardiotoxicity, mainly owing to their inhibition of topoisomerase II of DNA repair mechanisms. Our raised design strategy by switching therapeutic target from topoisomerase II to histone deacetylase (HDAC) has been applied to the design of anthraquinone derivatives in current study. Consequently, a series of novel HDAC inhibitors with a tricylic diketone of anthraquinone as a cap group have been synthesized. After screening and evaluation, compounds 4b, 4d, 7b and 7d have displayed the comparable inhibition in enzymatic activity and cell proliferation than that of Vorinostat (SAHA). Notably, compound 4b showed certain selectivity of antiproliferative effects on cancer cell lines over non-cancer cell lines.
Collapse
Affiliation(s)
- Yefang Zou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Zhuoxian Cao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Jie Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Xiaoxue Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Yan-Qin Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Yan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Basic Medicine, Guizhou Medical University
| | - Jingzi Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Yonglong Zhao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Aimin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| | - Bin He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University.,School of Pharmacy, Guizhou Medical University
| |
Collapse
|
25
|
Tikhomirov AS, Litvinova VA, Andreeva DV, Tsvetkov VB, Dezhenkova LG, Volodina YL, Kaluzhny DN, Treshalin ID, Schols D, Ramonova AA, Moisenovich MM, Shtil AA, Shchekotikhin AE. Amides of pyrrole- and thiophene-fused anthraquinone derivatives: A role of the heterocyclic core in antitumor properties. Eur J Med Chem 2020; 199:112294. [PMID: 32428792 DOI: 10.1016/j.ejmech.2020.112294] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/21/2022]
Abstract
Heteroarene-fused anthraquinone derivatives represent a class of perspective anticancer drug candidates capable of targeting multiple vital processes including drug resistance. Taking advantage of previously demonstrated potential of amide derivatives of heteroarene-fused anthraquinones, we herein dissected the role of the heterocyclic core in antitumor properties. A new series of naphtho[2,3-f]indole-3- and anthra[2,3-b]thiophene-3-carboxamides was synthesized via coupling the respective acids with cyclic diamines. New compounds demonstrated a submicromolar antiproliferative potency close to doxorubicin (Dox) against five tumor cell lines of various tissue origin. In contrast to Dox, the new compounds were similarly cytotoxic for HCT116 colon carcinoma cells (wild type p53) and their isogenic p53 knockout counterparts. Modification of the heterocyclic core changed the targeting properties: the best-in-series naphtho[2,3-f]indole-3-carboxamide 8 formed more affine complexes with DNA duplex than furan and thiophene analogs, a property that can be translated into a stronger inhibition of topoisomerase 1 mediated DNA unwinding. At tolerable doses the water soluble derivative 8 significantly inhibited tumor growth (up to 79%) and increased the lifespan (153%) of mice bearing P388 lymphoma transplants. Together with better solubility for parenteral administration and well tolerance by animals of the indole derivative 8 indicates prospects for further search of new antitumor drug candidates among the heteroarene-fused anthraquinones.
Collapse
Affiliation(s)
- Alexander S Tikhomirov
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia; Mendeleyev University of Chemical Technology, 9 Miusskaya Square, Moscow, 125047, Russian Federation
| | - Valeria A Litvinova
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia
| | - Daria V Andreeva
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia
| | - Vladimir B Tsvetkov
- Computational Oncology Group, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya, 119991, Moscow, Russia; Research and Clinical Center for Physical Chemical Medicine, 1A M. Pirogovskaya Street, Moscow, 119435, Russia
| | - Lyubov G Dezhenkova
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia
| | - Yulia L Volodina
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia; Blokhin National Medical Center of Oncology, 24 Kashirskoye Shosse, Moscow, 115478, Russia
| | - Dmitry N Kaluzhny
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, Moscow, 119991, Russia
| | - Ivan D Treshalin
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia
| | - Dominique Schols
- Rega Institute for Medical Research, K.U. Leuven, 3000, Leuven, Belgium
| | - Alla A Ramonova
- Department of Biology, Moscow State University, 1 Leninskie Gory, Moscow, 119234, Russia
| | - Mikhail M Moisenovich
- Department of Biology, Moscow State University, 1 Leninskie Gory, Moscow, 119234, Russia
| | - Alexander A Shtil
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, Moscow, 119021, Russia; Blokhin National Medical Center of Oncology, 24 Kashirskoye Shosse, Moscow, 115478, Russia
| | | |
Collapse
|
26
|
Chen D, Soh CK, Goh WH, Wang Z, Wang H. Synthesis and biological evaluation of 6-phenylpurine linked hydroxamates as novel histone deacetylase inhibitors. Bioorg Chem 2020; 98:103724. [PMID: 32171983 DOI: 10.1016/j.bioorg.2020.103724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/23/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022]
Abstract
A series of 6-phenylpurine based hydroxamates have been designed, synthesized and evaluated. Compound 3b and its analogs are potent histone deacetylase (HDAC) but weak PI3K/mTOR inhibitors. These compounds demonstrated broad anti-cancer activities against 38 cancer cell lines with leukemia, lymphoma, and the majority of liver cancer cell lines exhibiting the most sensitivity towards these compounds. Compound 3b demonstrated modulation of HDAC targets in vitro in a dose-dependent manner. It has good in vitro ADME profile that translated into a greatly improved pharmacokinetic profile. 3b also demonstrated modulation of HDACs in tumors in a PC-3 xenograft model. It was further evaluated in combination therapies in vitro. It exhibited additive or synergistic growth inhibition effect in HepG2 cells when combined with a number of approved drugs such as sorafenib, sunitinib, and erlotinib. Hence, 3b has the potential to be combined with the above to treat advanced liver cancer. As such, current data warrant further evaluation, optimization, and subsequent in vivo validation of the potential combination therapies.
Collapse
Affiliation(s)
- Dizhong Chen
- Drug Development Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Chang Kai Soh
- Drug Development Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Wei Huang Goh
- Drug Development Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Zilong Wang
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30318, United States
| | - Haishan Wang
- Drug Development Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore; Probit Pharmaceuticals Pte. Ltd., 10 Anson Road #27-15, Singapore 079903, Republic of Singapore.
| |
Collapse
|
27
|
Huang M, Xie X, Gong P, Wei Y, Du H, Xu Y, Xu Q, Jing Y, Zhao L. A 18β-glycyrrhetinic acid conjugate with Vorinostat degrades HDAC3 and HDAC6 with improved antitumor effects. Eur J Med Chem 2019; 188:111991. [PMID: 31883490 DOI: 10.1016/j.ejmech.2019.111991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Semisynthetic 18β-glycyrrhetinic acid (GA) analogues bearing 1-en-2-cyano-3-oxo substitution on ring A have enhanced antitumor effects with reduced levels of HDAC3 and HDAC6 proteins. Aiming to inhibit both HDAC protein and activity, we developed a hybrid molecule by tethering active GA analogue methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate (CDODA-Me) and Vorinostat (SAHA). We tested the proper hybrid approaches of GA with hydroxamic acid and turned out that GA conjugated with SAHA by a piperazine linker was the best. The conjugate (15) of CDODA-Me and SAHA linked through a piperazine group was a potent cytotoxic agent against cancer cells with apoptosis induction. Compound 15 was more effective than the simple combination of CDODA-Me and SAHA to induce apoptosis. Mechanistic studies revealed that 15 was less effective than SAHA to inhibit HDAC activity, but was more effective than CDODA-Me to decrease the levels of HDAC3 and HDAC6 proteins with upregulated levels of acetylated H3 and acetylated α-tubulin. Compound 15 represents a new HDAC3 and HDAC6 inhibitor by reducing protein levels.
Collapse
Affiliation(s)
- Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaorui Xie
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ping Gong
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yunfei Wei
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Heliang Du
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuanbo Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qihao Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongkui Jing
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
28
|
Sharma C, Oh YJ, Park B, Lee S, Jeong CH, Lee S, Seo JH, Seo YH. Development of Thiazolidinedione-Based HDAC6 Inhibitors to Overcome Methamphetamine Addiction. Int J Mol Sci 2019; 20:ijms20246213. [PMID: 31835389 PMCID: PMC6940941 DOI: 10.3390/ijms20246213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Thiazolidinedione is a five-membered heterocycle that is widely used in drug discovery endeavors. In this study, we report the design, synthesis, and biological evaluation of a series of thiazolidinedione-based HDAC6 inhibitors. In particular, compound 6b exerts an excellent inhibitory activity against HDAC6 with an IC50 value of 21 nM, displaying a good HDAC6 selectivity over HDAC1. Compound 6b dose-dependently induces the acetylation level of α-tubulin via inhibition of HDAC6 in human neuroblastoma SH-SY5Y cell line. Moreover, compound 6b efficiently reverses methamphetamine-induced morphology changes of SH-SY5Y cells via regulating acetylation landscape of α-tubulin. Collectively, compound 6b represents a novel HDAC6-isoform selective inhibitor and demonstrates promising therapeutic potential for the treatment of methamphetamine addiction.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Yong Jin Oh
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Sangkil Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu 42601, Korea;
| | - Young Ho Seo
- College of Pharmacy, Keimyung University, Daegu 42601, Korea; (C.S.); (Y.J.O.); (B.P.); (S.L.); (C.-H.J.); (S.L.)
- Correspondence: ; Tel.: +82-053-580-6639
| |
Collapse
|
29
|
Vergani B, Sandrone G, Marchini M, Ripamonti C, Cellupica E, Galbiati E, Caprini G, Pavich G, Porro G, Rocchio I, Lattanzio M, Pezzuto M, Skorupska M, Cordella P, Pagani P, Pozzi P, Pomarico R, Modena D, Leoni F, Perego R, Fossati G, Steinkühler C, Stevenazzi A. Novel Benzohydroxamate-Based Potent and Selective Histone Deacetylase 6 (HDAC6) Inhibitors Bearing a Pentaheterocyclic Scaffold: Design, Synthesis, and Biological Evaluation. J Med Chem 2019; 62:10711-10739. [DOI: 10.1021/acs.jmedchem.9b01194] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Barbara Vergani
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Giovanni Sandrone
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Mattia Marchini
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Chiara Ripamonti
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Edoardo Cellupica
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Elisabetta Galbiati
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Gianluca Caprini
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Gianfranco Pavich
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Giulia Porro
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Ilaria Rocchio
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Maria Lattanzio
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Marcello Pezzuto
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Malgorzata Skorupska
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Paola Cordella
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Paolo Pagani
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Pietro Pozzi
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Roberta Pomarico
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Daniela Modena
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Flavio Leoni
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Raffaella Perego
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Gianluca Fossati
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Christian Steinkühler
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| | - Andrea Stevenazzi
- Preclinical R&D, Italfarmaco Group, Via dei Lavoratori 54, I-20092 Cinisello Balsamo, Milan, Italy
| |
Collapse
|
30
|
Choi MA, Park SY, Chae HY, Song Y, Sharma C, Seo YH. Design, synthesis and biological evaluation of a series of CNS penetrant HDAC inhibitors structurally derived from amyloid-β probes. Sci Rep 2019; 9:13187. [PMID: 31515509 PMCID: PMC6742641 DOI: 10.1038/s41598-019-49784-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/19/2019] [Indexed: 12/17/2022] Open
Abstract
To develop novel CNS penetrant HDAC inhibitors, a new series of HDAC inhibitors having benzoheterocycle were designed, synthesized, and biologically evaluated. Among the synthesized compounds, benzothiazole derivative 9b exhibited a remarkable anti-proliferative activity (GI50 = 2.01 μM) against SH-SY5Y cancer cell line in a dose and time-dependent manner, better than the reference drug SAHA (GI50 = 2.90 μM). Moreover, compound 9b effectively promoted the accumulation of acetylated Histone H3 and α-tubulin through inhibition of HDAC1 and HDAC6 enzymes, respectively. HDAC enzyme assay also confirmed that compound 9b efficiently inhibited HDAC1 and HDAC6 isoforms with IC50 values of 84.9 nM and 95.9 nM. Furthermore, compound 9b inhibited colony formation capacity of SH-SY5Y cells, which is considered a hallmark of cell carcinogenesis and metastatic potential. The theoretical prediction, in vitro PAMPA-BBB assay, and in vivo brain pharmacokinetic studies confirmed that compound 9b had much higher BBB permeability than SAHA. In silico docking study demonstrated that compound 9b fitted in the substrate binding pocket of HDAC1 and HDAC6. Taken together, compound 9b provided a novel scaffold for developing CNS penetrant HDAC inhibitors and therapeutic potential for CNS-related diseases.
Collapse
Affiliation(s)
- Myeong A Choi
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Sun You Park
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Hye Yun Chae
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Yoojin Song
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | | | - Young Ho Seo
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea.
| |
Collapse
|
31
|
Liang T, Hou X, Zhou Y, Yang X, Fang H. Design, Synthesis, and Biological Evaluation of 2,4-Imidazolinedione Derivatives as HDAC6 Isoform-Selective Inhibitors. ACS Med Chem Lett 2019; 10:1122-1127. [PMID: 31413795 DOI: 10.1021/acsmedchemlett.9b00084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/05/2019] [Indexed: 01/23/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) has emerged as a promising drug target for various human diseases, including diverse neurodegenerative diseases and cancer. Herein, we reported a series of 2,4-imidazolinedione derivatives as novel HDAC6 isoform-selective inhibitors based on structure-based drug design. Most target compounds exhibit good profiles in a preliminary screening concerning HDAC6 inhibitory activities. Moreover, the most active compound 10c increases the acetylation level of α-tubulin with little effect on the acetylation of histone H3. Further biological evaluation suggested that potent compound 10c, which possesses good antiproliferative activity, could induce apoptosis in HL-60 cell by activating caspase 3.
Collapse
Affiliation(s)
- Tao Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Xuben Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Yi Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Xinying Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Hao Fang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| |
Collapse
|
32
|
Grünstein E, Sellmer A, Mahboobi S. Enantioselective synthesis and biological investigation of tetrahydro‐β‐carboline‐based HDAC6 inhibitors with improved solubility. Arch Pharm (Weinheim) 2019; 352:e1900026. [DOI: 10.1002/ardp.201900026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/12/2019] [Accepted: 03/17/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Elisabeth Grünstein
- Institute of Pharmacy, Faculty of Chemistry and PharmacyUniversity of RegensburgRegensburg Germany
| | - Andreas Sellmer
- Institute of Pharmacy, Faculty of Chemistry and PharmacyUniversity of RegensburgRegensburg Germany
| | - Siavosh Mahboobi
- Institute of Pharmacy, Faculty of Chemistry and PharmacyUniversity of RegensburgRegensburg Germany
| |
Collapse
|
33
|
Zhao C, Gao J, Zhang L, Su L, Luan Y. Novel HDAC6 selective inhibitors with 4-aminopiperidine-1- carboxamide as the core structure enhanced growth inhibitory activity of bortezomib in MCF-7 cells. Biosci Trends 2019; 13:91-97. [PMID: 30867374 DOI: 10.5582/bst.2019.01049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In epigenetics, histone deacetylases (HDACs) are well validated targets for the development of anticancer drugs. In this work, we reported the design and synthesis of a series of twenty two novel (E)-N-hydroxycinnamamide-based HDAC inhibitors with 4-aminopiperidine1-carboxamide as the core structure. Most newly synthesized compounds displayed high inhibition rates toward HDAC at the concentration of 1 μM. Among them, the inhibition rates of compounds LYP-2, LYP-3, LYP-6, and LYP-15 were more than 75%. Furthermore, compounds LYP-2, LYP-3, and LYP-6 potently inhibited the activity of HDAC6 with selectivity over HDAC1. We chose LYP-2 and LYP-6 to test its antiproliferative effect on breast cancer cells MCF-7. Either LYP-2 or LYP-6 alone moderately suppressed the cell growth, but could synergistically enhance the inhibitory effect of bortezomib. These results suggested that combined HDAC6 inhibitor and bortezomib regimen might be an option for breast cancer treatment.
Collapse
Affiliation(s)
- Chenru Zhao
- Department of Pharmacology, School of Pharmacy, Qingdao University.,Department of Medicinal Chemistry, School of Pharmacy, Qingdao University
| | - Jianjun Gao
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Li Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University
| | - Li Su
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University
| | - Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University
| |
Collapse
|
34
|
Yang F, Zhao N, Ge D, Chen Y. Next-generation of selective histone deacetylase inhibitors. RSC Adv 2019; 9:19571-19583. [PMID: 35519364 PMCID: PMC9065321 DOI: 10.1039/c9ra02985k] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
Histone deacetylases (HDACs) are clinically validated epigenetic drug targets for cancer treatment. HDACs inhibitors (HDACis) have been successfully applied against a series of cancers. First-generation inhibitors are mainly pan-HDACis that target multiple isoforms which might lead to serious side effects. At present, the next-generation HDACis are mainly focused on being class- or isoform-selective which can provide improved risk–benefit profiles compared to non-selective inhibitors. Because of the rapid development in next-generation HDACis, it is necessary to have an updated and state-of-the-art overview. Here, we summarize the strategies and achievements of the selective HDACis. Histone deacetylases (HDACs) are clinically validated epigenetic drug targets for cancer treatment.![]()
Collapse
Affiliation(s)
- Feifei Yang
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
- Shanghai Key Laboratory of Regulatory Biology
| | - Na Zhao
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
| | - Di Ge
- School of Biological Science and Technology
- University of Jinan
- Jinan
- China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology
- The Institute of Biomedical Sciences and School of Life Sciences
- East China Normal University
- Shanghai
- China
| |
Collapse
|