1
|
Ibrahim HS, Guo M, Hilscher S, Erdmann F, Schmidt M, Schutkowski M, Sheng C, Sippl W. Probing class I histone deacetylases (HDAC) with proteolysis targeting chimera (PROTAC) for the development of highly potent and selective degraders. Bioorg Chem 2024; 153:107887. [PMID: 39423771 DOI: 10.1016/j.bioorg.2024.107887] [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: 07/18/2024] [Revised: 09/22/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Class I HDACs are considered promising targets for cancer due to their role in epigenetic modifications. The main challenges in developing a new, potent and non-toxic class I HDAC inhibitor are selectivity and appropriate pharmacokinetics. The PROTAC technique (Proteolysis Targeting Chimera) is a new method in drug development for the production of active substances that can degrade a protein of interest (POI) instead of inhibiting it. This technique will open the era to produce selective and potent drugs with a high margin of safety. Previously, we reported different inhibitors targeting class I HDACs functionalized with aminobenzamide or hydroxamate groups. In the current research work, we will employ PROTAC technique to develop class I HDAC degraders based on our previously reported inhibitors. We synthesized two series of aminobenzamide-based PROTACs and hydroxamate-based PROTACs and tested them in vitro against class I HDACs. To ensure their degradation, all of them were screened against HDAC2 as representative example of class I. The best candidates were evaluated at different concentrations at various HDAC subtypes. This resulted in the PROTAC (32a) (HI31.1) that degrades HDAC8 with a DC50 of 8.9 nM with a proper margin of selectivity against other isozymes. Moreover, PROTAC 32a is able to degrade HDAC6 with DC50 = 14.3 nM. Apoptotic study on leukemic cells (MV-4-11) displayed more than 50 % apoptosis took place at 100 nM. PROTAC 32a (HI31.1) showed a good margin of safety against normal cell line and proper chemical stability.
Collapse
Affiliation(s)
- Hany S Ibrahim
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11829, Egypt.
| | - Menglu Guo
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201203, China; The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China
| | - Sebatian Hilscher
- Department of Enzymology, Institute of Biochemistry, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Frank Erdmann
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Matthias Schmidt
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, China.
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
2
|
Design, Synthesis and Biological Characterization of Histone Deacetylase 8 (HDAC8) Proteolysis Targeting Chimeras (PROTACs) with Anti-Neuroblastoma Activity. Int J Mol Sci 2022; 23:ijms23147535. [PMID: 35886887 PMCID: PMC9322761 DOI: 10.3390/ijms23147535] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023] Open
Abstract
In addition to involvement in epigenetic gene regulation, histone deacetylases (HDACs) regulate multiple cellular processes through mediating the activity of non-histone protein substrates. The knockdown of HDAC8 isozyme is associated with the inhibition of cell proliferation and apoptosis enhancement in several cancer cell lines. As shown in several studies, HDAC8 can be considered a potential target in the treatment of cancer forms such as childhood neuroblastoma. The present work describes the development of proteolysis targeting chimeras (PROTACs) of HDAC8 based on substituted benzhydroxamic acids previously reported as potent and selective HDAC8 inhibitors. Within this study, we investigated the HDAC8-degrading profiles of the synthesized PROTACs and their effect on the proliferation of neuroblastoma cells. The combination of in vitro screening and cellular testing demonstrated selective HDAC8 PROTACs that show anti-neuroblastoma activity in cells.
Collapse
|
3
|
Zeyen P, Zeyn Y, Herp D, Mahmoudi F, Yesiloglu TZ, Erdmann F, Schmidt M, Robaa D, Romier C, Ridinger J, Herbst-Gervasoni CJ, Christianson DW, Oehme I, Jung M, Krämer OH, Sippl W. Identification of histone deacetylase 10 (HDAC10) inhibitors that modulate autophagy in transformed cells. Eur J Med Chem 2022; 234:114272. [PMID: 35306288 PMCID: PMC9007901 DOI: 10.1016/j.ejmech.2022.114272] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/02/2023]
Abstract
Histone deacetylases (HDACs) are a family of 18 epigenetic modifiers that fall into 4 classes. Histone deacetylase inhibitors (HDACi) are valid tools to assess HDAC functions. HDAC6 and HDAC10 belong to the class IIb subgroup of the HDAC family. The targets and biological functions of HDAC10 are ill-defined. This lack of knowledge is due to a lack of specific and potent HDAC10 inhibitors with cellular activity. Here, we have synthesized and characterized piperidine-4-acrylhydroxamates as potent and highly selective inhibitors of HDAC10. This was achieved by targeting the acidic gatekeeper residue Glu274 of HDAC10 with a basic piperidine moiety that mimics the interaction of the polyamine substrate of HDAC10. We have confirmed the binding modes of selected inhibitors using X-ray crystallography. Promising candidates were selected based on their specificity by in vitro profiling using recombinant HDACs. The most promising HDAC10 inhibitors 10c and 13b were tested for specificity in acute myeloid leukemia (AML) cells with the FLT3-ITD oncogene. By immunoblot experiments we assessed the hyperacetylation of histones and tubulin-α, which are class I and HDAC6 substrates, respectively. As validated test for HDAC10 inhibition we used flow cytometry assessing autolysosome formation in neuroblastoma and AML cells. We demonstrate that 10c and 13b inhibit HDAC10 with high specificity over HDAC6 and with no significant impact on class I HDACs. The accumulation of autolysosomes is not a consequence of apoptosis and 10c and 13b are not toxic for normal human kidney cells. These data show that 10c and 13b are nanomolar inhibitors of HDAC10 with high specificity. Thus, our new HDAC10 inhibitors are tools to identify the downstream targets and functions of HDAC10 in cells.
Collapse
|
4
|
Ghazy E, Heimburg T, Lancelot J, Zeyen P, Schmidtkunz K, Truhn A, Darwish S, Simoben CV, Shaik TB, Erdmann F, Schmidt M, Robaa D, Romier C, Jung M, Pierce R, Sippl W. Synthesis, structure-activity relationships, cocrystallization and cellular characterization of novel smHDAC8 inhibitors for the treatment of schistosomiasis. Eur J Med Chem 2021; 225:113745. [PMID: 34392190 DOI: 10.1016/j.ejmech.2021.113745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 01/22/2023]
Abstract
Schistosomiasis is a major neglected parasitic disease that affects more than 265 million people worldwide and for which the control strategy consists of mass treatment with the only available drug, praziquantel. In this study, we chemically optimized our previously reported benzhydroxamate-based inhibitors of Schistosoma mansoni histone deacetylase 8 (smHDAC8). Crystallographic analysis provided insights into the inhibition mode of smHDAC8 activity by the highly potent inhibitor 5o. Structure-based optimization of the novel inhibitors was carried out using the available crystal structures as well as docking studies on smHDAC8. The compounds were evaluated in screens for inhibitory activity against schistosome and human HDACs (hHDAC). The in vitro and docking results were used for detailed structure activity relationships. The synthesized compounds were further investigated for their lethality against the schistosome larval stage using a fluorescence-based assay. The most promising inhibitor 5o showed significant dose-dependent killing of the schistosome larvae and markedly impaired egg laying of adult worm pairs maintained in culture.
Collapse
Affiliation(s)
- Ehab Ghazy
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Tino Heimburg
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Julien Lancelot
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017- CIIL - Centre d'Infection et d'Immunité de Lille, F-59000, Lille, France
| | - Patrik Zeyen
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Karin Schmidtkunz
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Anne Truhn
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Salma Darwish
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Conrad V Simoben
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Tajith B Shaik
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Biologie Structurale Intégrative, 67404, Illkirch Cedex, France
| | - Frank Erdmann
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Matthias Schmidt
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany
| | - Christophe Romier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Biologie Structurale Intégrative, 67404, Illkirch Cedex, France
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Raymond Pierce
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017- CIIL - Centre d'Infection et d'Immunité de Lille, F-59000, Lille, France
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120, Halle/Saale, Germany.
| |
Collapse
|
5
|
La MT, Jeong B, Kim H. Design and Synthesis of Novel
N
‐(2‐aminophenyl)benzamide Derivatives as Histone Deacetylase Inhibitors and Their Antitumor Activity Study. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Minh Thanh La
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center Jeonbuk National University Medical School and Hospital Jeonju 54907 Republic of Korea
| | - Byung‐Hoon Jeong
- Department of Bioactive Material Sciences Jeonbuk National University Jeonju Jeonbuk 54896 Republic of Korea
| | - Hee‐Kwon Kim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center Jeonbuk National University Medical School and Hospital Jeonju 54907 Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University‐Biomedical Research Institute of Jeonbuk National University Hospital Jeonju 54907 Republic of Korea
| |
Collapse
|
6
|
Design, synthesis, and biological evaluation of dual targeting inhibitors of histone deacetylase 6/8 and bromodomain BRPF1. Eur J Med Chem 2020; 200:112338. [PMID: 32497960 DOI: 10.1016/j.ejmech.2020.112338] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/09/2020] [Accepted: 04/13/2020] [Indexed: 01/19/2023]
Abstract
Histone modifying proteins, specifically histone deacetylases (HDACs) and bromodomains, have emerged as novel promising targets for anticancer therapy. In the current work, based on available crystal structures and docking studies, we designed dual inhibitors of both HDAC6/8 and the bromodomain and PHD finger containing protein 1 (BRPF1). Biochemical and biophysical tests showed that compounds 23a,b and 37 are nanomolar inhibitors of both target proteins. Detailed structure-activity relationships were deduced for the synthesized inhibitors which were supported by extensive docking and molecular dynamics studies. Cellular testing in acute myeloid leukemia (AML) cells showed only a weak effect, most probably because of the poor permeability of the inhibitors. We also aimed to analyse the target engagement and the cellular activity of the novel inhibitors by determining the protein acetylation levels in cells by western blotting (tubulin vs histone acetylation), and by assessing their effects on various cancer cell lines.
Collapse
|
7
|
Hirata Y, Sasaki T, Kanki H, Choong CJ, Nishiyama K, Kubo G, Hotei A, Taniguchi M, Mochizuki H, Uesato S. New 5-Aryl-Substituted 2-Aminobenzamide-Type HDAC Inhibitors with a Diketopiperazine Group and Their Ameliorating Effects on Ischemia-Induced Neuronal Cell Death. Sci Rep 2018; 8:1400. [PMID: 29362442 PMCID: PMC5780423 DOI: 10.1038/s41598-018-19664-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023] Open
Abstract
We previously synthesized new 5-thienyl-substituted 2-aminobenzamide-type HDAC1, 2 inhibitors with the (4-ethyl-2,3-dioxopiperazine-1-carboxamido) methyl group. K-560 (1a) protected against neuronal cell death in a Parkinson’s disease model by up-regulating the expression of XIAP. This finding prompted us to design new K-560-related compounds. We examined the structure activity relationship (SAR) for the neuronal protective effects of newly synthesized and known K-560 derivatives after cerebral ischemia. Among them, K-856 (8), containing the (4-methyl-2,5-dioxopiperazin-1-yl) methyl group, exhibited a promising neuronal survival activity. The SAR study strongly suggested that the attachment of a monocyclic 2,3- or 2,5-diketopiperazine group to the 2-amino-5-aryl (but not 2-nitro-5-aryl) scaffold is necessary for K-560-related compounds to exert a potent neuroprotective effect.
Collapse
Affiliation(s)
- Yoshiyuki Hirata
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan.,Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| | - Hideaki Kanki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Chi-Jing Choong
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Kumiko Nishiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Genki Kubo
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Ayana Hotei
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan
| | - Masahiko Taniguchi
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Shinichi Uesato
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35, Yamate-cho, Suita, Osaka, 564-8680, Japan. .,Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| |
Collapse
|
8
|
Heimburg T, Kolbinger FR, Zeyen P, Ghazy E, Herp D, Schmidtkunz K, Melesina J, Shaik TB, Erdmann F, Schmidt M, Romier C, Robaa D, Witt O, Oehme I, Jung M, Sippl W. Structure-Based Design and Biological Characterization of Selective Histone Deacetylase 8 (HDAC8) Inhibitors with Anti-Neuroblastoma Activity. J Med Chem 2017; 60:10188-10204. [DOI: 10.1021/acs.jmedchem.7b01447] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Tino Heimburg
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Fiona R. Kolbinger
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
| | - Patrik Zeyen
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Ehab Ghazy
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Daniel Herp
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Karin Schmidtkunz
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Jelena Melesina
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Tajith Baba Shaik
- Département
de Biologie Structurale Intégrative, Institut de Génétique
et Biologie Moléculaire et Cellulaire (IGBMC), Université
de Strasbourg (UDS), CNRS, INSERM, 67404 Illkirch Cedex, France
| | - Frank Erdmann
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Matthias Schmidt
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Christophe Romier
- Département
de Biologie Structurale Intégrative, Institut de Génétique
et Biologie Moléculaire et Cellulaire (IGBMC), Université
de Strasbourg (UDS), CNRS, INSERM, 67404 Illkirch Cedex, France
| | - Dina Robaa
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Olaf Witt
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Department
of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Medical Center, 69120 Heidelberg, Germany
| | - Ina Oehme
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
| | - Manfred Jung
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| |
Collapse
|
9
|
Gao S, Zang J, Gao Q, Liang X, Ding Q, Li X, Xu W, Chou CJ, Zhang Y. Design, synthesis and anti-tumor activity study of novel histone deacetylase inhibitors containing isatin-based caps and o-phenylenediamine-based zinc binding groups. Bioorg Med Chem 2017; 25:2981-2994. [PMID: 28511906 PMCID: PMC5994915 DOI: 10.1016/j.bmc.2017.03.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 11/23/2022]
Abstract
As a hot topic of epigenetic studies, histone deacetylases (HDACs) are related to lots of diseases, especially cancer. Further researches indicated that different HDAC isoforms played various roles in a wide range of tumor types. Herein a novel series of HDAC inhibitors with isatin-based caps and o-phenylenediamine-based zinc binding groups have been designed and synthesized through scaffold hopping strategy. Among these compounds, the most potent compound 9n exhibited similar if not better HDAC inhibition and antiproliferative activities against multiple tumor cell lines compared with the positive control entinostat (MS-275). Additionally, compared with MS-275 (IC50 values for HDAC1, 2 and 3 were 0.163, 0.396 and 0.605µM, respectively), compound 9n with IC50 values of 0.032, 0.256 and 0.311µM for HDAC1, 2 and 3 respectively, showed a moderate HDAC1 selectivity.
Collapse
Affiliation(s)
- Shuai Gao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - Jie Zang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - Qianwen Gao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - Xuewu Liang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - Qinge Ding
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - Xiaoyang Li
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Wenfang Xu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China
| | - C James Chou
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yingjie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong University, 44 West Culture Road, 250012 Ji'nan, Shandong, PR China; Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, Peoples' Republic of China.
| |
Collapse
|
10
|
Traoré MM, Zwick V, Simões-Pires CA, Nurisso A, Issa M, Cuendet M, Maynadier M, Wein S, Vial H, Jamet H, Wong YS. Hydroxyl Ketone-Based Histone Deacetylase Inhibitors To Gain Insight into Class I HDAC Selectivity versus That of HDAC6. ACS OMEGA 2017; 2:1550-1562. [PMID: 30023639 PMCID: PMC6044785 DOI: 10.1021/acsomega.6b00481] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/29/2017] [Indexed: 06/07/2023]
Abstract
Little is known about the biological and structural features that govern the isoform selectivity for class I histone deacetylases (HDACs) over HDAC6. In addition to that for known inhibitors, like benzamides, psammaplin A, and cyclodepsipeptide-derived thiols, selectivity was also observed for naturally occurring cyclopeptide HDAC inhibitors with an aliphatic flexible linker and ketonelike zinc-binding group (ZBG). The present study reports that this isoform selectivity is mainly due to the linker and ZBG, as replacement of the cyclopeptide cap region by a simple aniline retained class I HDAC isoform selectivity toward HDAC6 in enzymatic assays. The best cyclopeptide-free analogues preserved efficacy against Plasmodium falciparum and cancer cell lines. Molecular modeling provided hypotheses to explain this selectivity and suggests different behaviors of the flexible linker on HDAC1 and HDAC6 pockets, which may influence, on the basis of the strength of the ZBG, its coordination with the zinc ion.
Collapse
Affiliation(s)
- Mohamed
D. M. Traoré
- Département
de Pharmacochimie Moléculaire, CNRS
UMR 5063, ICMG FR 2607, Univ. Grenoble Alpes, 470 rue de la chimie, 38041 Grenoble cedex 9, France
- Département
de Chimie Moléculaire, CNRS UMR 5250,
ICMG FR 2607, Univ. Grenoble Alpes, 301 rue de la chimie, 38041 Grenoble cedex 9, France
| | - Vincent Zwick
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Claudia A. Simões-Pires
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Alessandra Nurisso
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
- Laboratoire
Dynamique des Interactions Membranaires Normales et Pathologiques, UMR 5235, CNRS, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Mark Issa
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Muriel Cuendet
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Marjorie Maynadier
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Sharon Wein
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Henri Vial
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Helene Jamet
- Département
de Chimie Moléculaire, CNRS UMR 5250,
ICMG FR 2607, Univ. Grenoble Alpes, 301 rue de la chimie, 38041 Grenoble cedex 9, France
| | - Yung-Sing Wong
- Département
de Pharmacochimie Moléculaire, CNRS
UMR 5063, ICMG FR 2607, Univ. Grenoble Alpes, 470 rue de la chimie, 38041 Grenoble cedex 9, France
| |
Collapse
|
11
|
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
|
12
|
Tanwar B, Purohit P, Raju BN, Kumar D, Kommi DN, Chakraborti AK. An “all-water” strategy for regiocontrolled synthesis of 2-aryl quinoxalines. RSC Adv 2015. [DOI: 10.1039/c4ra16568c] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
CascadeN-aroylmethylation–reduction–condensation process as novel strategy of “all water chemistry” for first generalized regioselective synthesis of 2-aryl quinoxalines.
Collapse
Affiliation(s)
- Babita Tanwar
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| | - Priyank Purohit
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| | - Banothu Naga Raju
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| | - Dinesh Kumar
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| | - Damodara N. Kommi
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| | - Asit K. Chakraborti
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research (NIPER)
- Punjab
- India
| |
Collapse
|
13
|
Di Costanzo A, Del Gaudio N, Migliaccio A, Altucci L. Epigenetic drugs against cancer: an evolving landscape. Arch Toxicol 2014; 88:1651-68. [DOI: 10.1007/s00204-014-1315-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 07/17/2014] [Indexed: 02/08/2023]
|
14
|
Marek M, Kannan S, Hauser AT, Moraes Mourão M, Caby S, Cura V, Stolfa DA, Schmidtkunz K, Lancelot J, Andrade L, Renaud JP, Oliveira G, Sippl W, Jung M, Cavarelli J, Pierce RJ, Romier C. Structural basis for the inhibition of histone deacetylase 8 (HDAC8), a key epigenetic player in the blood fluke Schistosoma mansoni. PLoS Pathog 2013; 9:e1003645. [PMID: 24086136 PMCID: PMC3784479 DOI: 10.1371/journal.ppat.1003645] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/04/2013] [Indexed: 11/19/2022] Open
Abstract
The treatment of schistosomiasis, a disease caused by blood flukes parasites of the Schistosoma genus, depends on the intensive use of a single drug, praziquantel, which increases the likelihood of the development of drug-resistant parasite strains and renders the search for new drugs a strategic priority. Currently, inhibitors of human epigenetic enzymes are actively investigated as novel anti-cancer drugs and have the potential to be used as new anti-parasitic agents. Here, we report that Schistosoma mansoni histone deacetylase 8 (smHDAC8), the most expressed class I HDAC isotype in this organism, is a functional acetyl-L-lysine deacetylase that plays an important role in parasite infectivity. The crystal structure of smHDAC8 shows that this enzyme adopts a canonical α/β HDAC fold, with specific solvent exposed loops corresponding to insertions in the schistosome HDAC8 sequence. Importantly, structures of smHDAC8 in complex with generic HDAC inhibitors revealed specific structural changes in the smHDAC8 active site that cannot be accommodated by human HDACs. Using a structure-based approach, we identified several small-molecule inhibitors that build on these specificities. These molecules exhibit an inhibitory effect on smHDAC8 but show reduced affinity for human HDACs. Crucially, we show that a newly identified smHDAC8 inhibitor has the capacity to induce apoptosis and mortality in schistosomes. Taken together, our biological and structural findings define the framework for the rational design of small-molecule inhibitors specifically interfering with schistosome epigenetic mechanisms, and further support an anti-parasitic epigenome targeting strategy to treat neglected diseases caused by eukaryotic pathogens. Schistosomiasis, a neglected parasitic disease caused by flatworms of the genus Schistosoma, is responsible for hundreds of thousands of deaths yearly. Its treatment currently depends on a single drug, praziquantel, with reports of drug-resistant parasites. Human epigenetic enzymes, in particular histone deacetylases (HDACs), are predominantly attractive inhibitory targets for anti-cancer therapies. Validated scaffolds against these enzymes could also be used as leads in the search for novel specific drugs against schistosomiasis. In our study, we show that Schistosoma mansoni histone deacetylase 8 (smHDAC8) is a functional acetyl-L-lysine deacetylase that plays an important role in parasite infectivity and is therefore a relevant target for drug discovery. The determination of the atomic structures of smHDAC8 in complex with generic HDAC inhibitors revealed that the architecture of the smHDAC8 active site pocket differed significantly from its human counterparts and provided a framework for the development of inhibitors selectively interfering with schistosome epigenetic mechanisms. In agreement, this information enabled us to identify several small-molecule scaffolds that possess specific inhibitory effects on smHDAC8 and cause mortality in schistosomes. Our results provide the proof of concept that targeting epigenetic enzymes is a valid approach to treat neglected diseases caused by eukaryotic pathogens.
Collapse
Affiliation(s)
- Martin Marek
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UDS), CNRS, INSERM, Illkirch, France
| | | | - Alexander-Thomas Hauser
- Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Marina Moraes Mourão
- Genomics and Computational Biology Group, Center for Excellence in Bioinformatics, National Institute of Science and Technology in Tropical Diseases, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Stéphanie Caby
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 – CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Vincent Cura
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UDS), CNRS, INSERM, Illkirch, France
| | - Diana A. Stolfa
- Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Karin Schmidtkunz
- Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Julien Lancelot
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 – CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
| | - Luiza Andrade
- Genomics and Computational Biology Group, Center for Excellence in Bioinformatics, National Institute of Science and Technology in Tropical Diseases, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Jean-Paul Renaud
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UDS), CNRS, INSERM, Illkirch, France
| | - Guilherme Oliveira
- Genomics and Computational Biology Group, Center for Excellence in Bioinformatics, National Institute of Science and Technology in Tropical Diseases, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Wolfgang Sippl
- Institut für Pharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
- Freiburg Institute of Advanced Studies (FRIAS), Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Manfred Jung
- Institut für Pharmazeutische Wissenschaften, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Freiburg Institute of Advanced Studies (FRIAS), Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Jean Cavarelli
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UDS), CNRS, INSERM, Illkirch, France
| | - Raymond J. Pierce
- Center for Infection and Immunity of Lille (CIIL), INSERM U1019 – CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille, France
- * E-mail: (RJP); (CR)
| | - Christophe Romier
- Département de Biologie Structurale Intégrative, Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UDS), CNRS, INSERM, Illkirch, France
- * E-mail: (RJP); (CR)
| |
Collapse
|
15
|
Microbial natural products: molecular blueprints for antitumor drugs. J Ind Microbiol Biotechnol 2013; 40:1181-210. [PMID: 23999966 DOI: 10.1007/s10295-013-1331-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 08/07/2013] [Indexed: 12/18/2022]
Abstract
Microbes from two of the three domains of life, the Prokarya, and Eukarya, continue to serve as rich sources of structurally complex chemical scaffolds that have proven to be essential for the development of anticancer therapeutics. This review describes only a handful of exemplary natural products and their derivatives as well as those that have served as elegant blueprints for the development of novel synthetic structures that are either currently in use or in clinical or preclinical trials together with some of their earlier analogs in some cases whose failure to proceed aided in the derivation of later compounds. In every case, a microbe has been either identified as the producer of secondary metabolites or speculated to be involved in the production via symbiotic associations. Finally, rapidly evolving next-generation sequencing technologies have led to the increasing availability of microbial genomes. Relevant examples of genome mining and genetic manipulation are discussed, demonstrating that we have only barely scratched the surface with regards to harnessing the potential of microbes as sources of new pharmaceutical leads/agents or biological probes.
Collapse
|
16
|
Marek L, Hamacher A, Hansen FK, Kuna K, Gohlke H, Kassack MU, Kurz T. Histone deacetylase (HDAC) inhibitors with a novel connecting unit linker region reveal a selectivity profile for HDAC4 and HDAC5 with improved activity against chemoresistant cancer cells. J Med Chem 2013; 56:427-36. [PMID: 23252603 DOI: 10.1021/jm301254q] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The synthesis and biological evaluation of new potent hydroxamate-based HDAC inhibitors with a novel alkoxyamide connecting unit linker region are described. Biological evaluation includes MTT and cellular HDAC assays on sensitive and chemoresistant cancer cell lines as well as HDAC profiling of selected compounds. Compound 19i (LMK235) (N-((6-(hydroxyamino)-6-oxohexyl)oxy)-3,5-dimethylbenzamide) showed similar effects compared to vorinostat on inhibition of cellular HDACs in a pan-HDAC assay but enhanced cytotoxic effects against the human cancer cell lines A2780, Cal27, Kyse510, and MDA-MB231. Subsequent HDAC profiling yielded a novel HDAC isoform selectivity profile of 19i in comparison to vorinostat or trichostatin A (TSA). 19i shows nanomolar inhibition of HDAC4 and HDAC5, whereas vorinostat and TSA inhibit HDAC4 and HDAC5 in the higher micromolar range.
Collapse
Affiliation(s)
- Linda Marek
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich Heine Universität , Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
17
|
M. Jung. ChemMedChem 2012. [DOI: 10.1002/cmdc.201200280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|