1
|
Wagih N, Abdel-Rahman IM, El-Koussi NA, El-Din A Abuo-Rahma G. Anticancer benzimidazole derivatives as inhibitors of epigenetic targets: a review article. RSC Adv 2025; 15:966-1010. [PMID: 39807197 PMCID: PMC11726184 DOI: 10.1039/d4ra05014b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality worldwide. One of the primary causes of cancer development and progression is epigenetic dysregulation, which is a heritable modification that alters gene expression without changing the DNA sequence. Therefore, targeting these epigenetic changes has emerged as a promising therapeutic strategy. Benzimidazole derivatives have gained attention for their potent epigenetic modulatory effects as they interact with various epigenetic targets, including DNA methyltransferases, histone deacetylases and histone methyltransferases. This review provides a comprehensive overview of benzimidazole derivatives that inhibit different acetylation and methylation reader, writer and eraser epigenetic targets. Herein, we emphasize the therapeutic potential of these compounds in developing targeted, less toxic cancer therapies. Presently, some promising benzimidazole derivatives have entered clinical trials and shown great advancements in the fields of hematological and solid malignancy therapies. Accordingly, we highlight the recent advancements in benzimidazole research as epigenetic agents that could pave the way for designing new multi-target drugs to overcome resistance and improve clinical outcomes for cancer patients. This review can help researchers in designing new anticancer benzimidazole derivatives with better properties.
Collapse
Affiliation(s)
- Nardin Wagih
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| | - Islam M Abdel-Rahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| | - Nawal A El-Koussi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University 71526 Assiut Egypt
| | - Gamal El-Din A Abuo-Rahma
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University 61519 Minia Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Deraya University New Minia 61111 Egypt
| |
Collapse
|
2
|
Uba AI, Hryb M, Singh M, Bui-Linh C, Tran A, Atienza J, Misbah S, Mou X, Wu C. Discovery of novel inhibitors of histone deacetylase 6: Structure-based virtual screening, molecular dynamics simulation, enzyme inhibition and cell viability assays. Life Sci 2024; 338:122395. [PMID: 38181853 DOI: 10.1016/j.lfs.2023.122395] [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: 10/16/2023] [Revised: 12/23/2023] [Accepted: 12/26/2023] [Indexed: 01/07/2024]
Abstract
Histone deacetylase 6 (HDAC6) contributes to cancer metastasis in several cancers, including triple-negative breast cancer (TNBC)-the most lethal form that lacks effective therapy. Although several efforts have been invested to develop selective HDAC6 inhibitors, none have been approved by the FDA. Toward this goal, existing computational studies used smaller compound libraries and shorter MD simulations. Here, we conducted a structure-based virtual screening of ZINC "Druglike" library containing 17,900,742 compounds using a Glide virtual screening protocol comprising various filters with increasing accuracy. The top 20 hits were subjected to molecular dynamics simulation, MM-GBSA binding energy calculations, and further ADMET prediction. Furthermore, enzyme inhibition assay and cell viability assay were performed on six available compounds from the identified hits. C4 (ZINC000077541942) with a good profile of predicted drug properties was found to inhibit HDAC6 (IC50: 4.7 ± 11.6 μM) with comparative affinity to that of the known HDAC6 selective inhibitor Tubacin (TA) in our experiments. C4 also demonstrated cytotoxic effects against triple-negative breast cancer (TNBC) cell line MDA-MB-231 with EC50 of 40.6 ± 12.7 μM comparable to that of TA (2-20 μM). Therefore, this compound, with pharmacophore features comprising a non-hydroxamic acid zinc-binding group, heteroaromatic linker, and cap group, is proposed as a novel HDAC6 inhibitor.
Collapse
Affiliation(s)
- Abdullahi Ibrahim Uba
- Complex Systems Division, Beijing Computational Science Research Center, Beijing 100193, China
| | - Mariya Hryb
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Mursalin Singh
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Candice Bui-Linh
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Annie Tran
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Jiancarlo Atienza
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Sarah Misbah
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Xiaoyang Mou
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA.
| | - Chun Wu
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA.
| |
Collapse
|
3
|
AbdElmoniem N, H. Abdallah M, M. Mukhtar R, Moutasim F, Rafie Ahmed A, Edris A, Ibraheem W, Makki AA, M. Elshamly E, Elhag R, Osman W, A. Mothana R, Alzain AA. Identification of Novel Natural Dual HDAC and Hsp90 Inhibitors for Metastatic TNBC Using e-Pharmacophore Modeling, Molecular Docking, and Molecular Dynamics Studies. Molecules 2023; 28:1771. [PMID: 36838758 PMCID: PMC9965823 DOI: 10.3390/molecules28041771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Breast cancer (BC) is one of the main types of cancer that endangers women's lives. The characteristics of triple-negative breast cancer (TNBC) include a high rate of recurrence and the capacity for metastasis; therefore, new therapies are urgently needed to combat TNBC. Dual targeting HDAC6 and Hsp90 has shown good synergistic effects in treating metastatic TNBC. The goal of this study was to find potential HDAC6 and Hsp90 dual inhibitors. Therefore, several in silico approaches have been used. An e-pharmacophore model generation based on the HDAC6-ligand complex and subsequently a pharmacophore-based virtual screening on 270,450 natural compounds from the ZINC were performed, which resulted in 12,663 compounds that corresponded to the obtained pharmacophoric hypothesis. These compounds were docked into HDAC6 and Hsp90. This resulted in the identification of three compounds with good docking scores and favorable free binding energy against the two targets. The top three compounds, namely ZINC000096116556, ZINC000020761262, and ZINC000217668954, were further subjected to ADME prediction and molecular dynamic simulations, which showed promising results in terms of pharmacokinetic properties and stability. As a result, these three compounds can be considered potential HDAC6 and Hsp90 dual inhibitors and are recommended for experimental evaluation.
Collapse
Affiliation(s)
- Nihal AbdElmoniem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Marwa H. Abdallah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Rua M. Mukhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Fatima Moutasim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Ahmed Rafie Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Alaa Edris
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Walaa Ibraheem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Alaa A. Makki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Eman M. Elshamly
- Department of Molecular Biotechnology, Hochschule Anhalt, 06846 Dessau-Roßlau, Germany
| | - Rashid Elhag
- Department of Biology, College of Science and Technology, Florida A & M University, Tallahassee, FL 32307, USA
| | - Wadah Osman
- Department of Pharmacognosy, Faculty of Pharmacy, University of Khartoum, Khartoum 11114, Sudan
| | - Ramzi A. Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulrahim A. Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| |
Collapse
|
4
|
Sixto-López Y, Gómez-Vidal JA, de Pedro N, Bello M, Rosales-Hernández MC, Correa-Basurto J. In silico design of HDAC6 inhibitors with neuroprotective effects. J Biomol Struct Dyn 2022; 40:14204-14222. [PMID: 34784487 DOI: 10.1080/07391102.2021.2001378] [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] [Indexed: 12/29/2022]
Abstract
HDAC6 has emerged as a molecular target to treat neurodegenerative disorders, due to its participation in protein aggregate degradation, oxidative stress process, mitochondrial transport, and axonal transport. Thus, in this work we have designed a set of 485 compounds with hydroxamic and bulky-hydrophobic moieties that may function as HDAC6 inhibitors with a neuroprotective effect. These compounds were filtered by their predicted ADMET properties and their affinity to HDAC6 demonstrated by molecular docking and molecular dynamics simulations. The combination of in silico with in vitro neuroprotective results allowed the identification of a lead compound (FH-27) which shows neuroprotective effect that could be due to HDAC6 inhibition. Further, FH-27 chemical moiety was used to design a second series of compounds improving the neuroprotective effect from 2- to 10-fold higher (YSL-99, YSL-109, YSL-112, YSL-116 and YSL-121; 1.25 ± 0.67, 1.82 ± 1.06, 7.52 ± 1.78, 5.59 and 5.62 ± 0.31 µM, respectively). In addition, the R enantiomer of FH-27 (YSL-106) was synthesized, showing a better neuroprotective effect (1.27 ± 0.60 µM). In conclusion, we accomplish the in silico design, synthesis, and biological evaluation of hydroxamic acid derivatives with neuroprotective effect as suggested by an in vitro model. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yudibeth Sixto-López
- Laboratorio de Modelado Molecular, Bioinformática y Diseño de fármacos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico.,Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Granada, Spain
| | - José Antonio Gómez-Vidal
- Departamento de Química Farmacéutica y Orgánica, Facultad de Farmacia, Universidad de Granada, Granada, Spain
| | - Nuria de Pedro
- Fundación MEDINA, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Martiniano Bello
- Laboratorio de Modelado Molecular, Bioinformática y Diseño de fármacos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - José Correa-Basurto
- Laboratorio de Modelado Molecular, Bioinformática y Diseño de fármacos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
5
|
Abdel-Atty MM, Farag NA, Serya RAT, Abouzid KAM, Mowafy S. Molecular design, synthesis and in vitro biological evaluation of thienopyrimidine-hydroxamic acids as chimeric kinase HDAC inhibitors: a challenging approach to combat cancer. J Enzyme Inhib Med Chem 2021; 36:1290-1312. [PMID: 34187263 PMCID: PMC8253220 DOI: 10.1080/14756366.2021.1933465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 11/21/2022] Open
Abstract
A series of thieno[2,3-d]pyrimidine-based hydroxamic acid hybrids was designed and synthesised as multitarget anti-cancer agents, through incorporating the pharmacophore of EGFR, VEGFR2 into the inhibitory functionality of HDAC6. Three compounds (12c, 15b and 20b) were promising hits, whereas (12c) exhibited potent VEGFR2 inhibition (IC50=185 nM), potent EGFR inhibition (IC50=1.14 µM), and mild HDAC6 inhibition (23% inhibition). Moreover, compound (15c) was the most potent dual inhibitor among all the synthesised compounds, as it exhibited potent EGFR and VEGFR2 inhibition (IC50=19 nM) and (IC50=5.58 µM), respectively. While compounds (20d) and (7c) displayed nanomolar selective kinase inhibition with EGFR IC50= 68 nM and VEGFR2 IC50= 191 nM, respectively. All of the synthesised compounds were screened in vitro for their cytotoxic effect on 60 human NCI tumour cell lines. Additionally, molecular docking studies and ADMET studies were carried out to gain further insight into their binding mode and predict the pharmacokinetic properties of all the synthesised inhibitors.
Collapse
Affiliation(s)
- Mona M. Abdel-Atty
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Nahla A. Farag
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Rabah A. T. Serya
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Khaled A. M. Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Organic and Medicinal Chemistry Department, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Samar Mowafy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
- Department of Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
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
|
7
|
Linciano P, Benedetti R, Pinzi L, Russo F, Chianese U, Sorbi C, Altucci L, Rastelli G, Brasili L, Franchini S. Investigation of the effect of different linker chemotypes on the inhibition of histone deacetylases (HDACs). Bioorg Chem 2020; 106:104462. [PMID: 33213894 DOI: 10.1016/j.bioorg.2020.104462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
Histone Deacetylases (HDACs) are among the most attractive and interesting targets in anticancer drug discovery. The clinical relevance of HDAC inhibitors (HDACIs) is testified by four FDA-approved drugs for cancer treatment. However, one of the main drawbacks of these drugs resides in the lack of selectivity against the different HDAC isoforms, resulting in severe side effects. Thus, the identification of selective HDACIs represents an exciting challenge for medicinal chemists. HDACIs are composed of a cap group, a linker region, and a metal-binding group interacting with the catalytic zinc ion. While the cap group has been extensively investigated, less information is available about the effect of the linker on isoform selectivity. To this aim, in this work, we explored novel linker chemotypes to direct isoform selectivity. A small library of 25 hydroxamic acids with hitherto unexplored linker chemotypes was prepared. In vitro tests demonstrated that, depending on the linker type, some candidates selectively inhibit HDAC1 over HDAC6 isoform or vice versa. Docking calculations were performed to rationalize the effect of the novel linker chemotypes on biologic activity. Moreover, four compounds were able to increase the levels of acetylation of histone H3 or tubulin. These compounds were also assayed in breast cancer MCF7 cells to test their antiproliferative effect. Three compounds showed a significant reduction of cancer proliferation, representing valuable starting points for further optimization.
Collapse
Affiliation(s)
- Pasquale Linciano
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Rosaria Benedetti
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Luca Pinzi
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Fabiana Russo
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Ugo Chianese
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Claudia Sorbi
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy.
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Giulio Rastelli
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Livio Brasili
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Silvia Franchini
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy.
| |
Collapse
|
8
|
Dawood M, Hegazy MEF, Elbadawi M, Fleischer E, Klinger A, Bringmann G, Kuntner C, Shan L, Efferth T. Vitamin K 3 chloro derivative (VKT-2) inhibits HDAC6, activates autophagy and apoptosis, and inhibits aggresome formation in hepatocellular carcinoma cells. Biochem Pharmacol 2020; 180:114176. [PMID: 32721508 DOI: 10.1016/j.bcp.2020.114176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/25/2022]
Abstract
Epigenetics plays a vital role in regulating gene expression and determining the specific phenotypes of eukaryotic cells. Histone deacetylases (HDACs) are important epigenetic regulatory proteins effecting multiple biological functions. Particularly, HDAC6 has become a promising anti-cancer drug target because of its regulation of cell mobility, protein trafficking, degradation of misfolded proteins, cell growth, apoptosis, and metastasis. In this study, we identified one out of six vitamin K3 derivatives, VKT-2, as HDAC6 inhibitor using molecular docking and cell viability assays in HDAC6-overexpressing HuH-7 cancer cells. Microscale thermophoresis and HDAC6 enzymatic assays revealed that VKT-2 bound to HDAC6 and inhibited its function. We further identified its cytotoxic activity. VKT-2 hyperacetylated HDAC6 substrates and disturbed tubulin integrity leading to significant inhibition of tumor migration in both HuH-7 spheroids and U2OS-GFP-α-tubulin cells. Moreover, VKT-2 induced autophagic and apoptotic cell death in HuH-7, while aggresome formation was restrained after VKT-2 treatment. A HuH-7 cell-xenograft model in zebrafish larvae provided evidence that VKT-2 inhibited the tumor growth in vivo. To best of our knowledge, it is the first time to demonstrate that vitamin k3 derivatives (VKT-2) inhibits HDAC6 in solid tumor cells. These unique findings suggested that VKT-2 is a promising anti-cancer agent targeting HDAC6.
Collapse
Affiliation(s)
- Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany; Department of Molecular Biology, Faculty of Medical Laboratory Science, Al-Neelain University, Khartoum, Sudan
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany; Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | | | | | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Claudia Kuntner
- AIT Austrian Institute of Technology GmbH, Preclinical Molecular Imaging, Seibersdorf, Austria
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
9
|
Dawood M, Fleischer E, Klinger A, Bringmann G, Shan L, Efferth T. Inhibition of cell migration and induction of apoptosis by a novel class II histone deacetylase inhibitor, MCC2344. Pharmacol Res 2020; 160:105076. [PMID: 32659428 DOI: 10.1016/j.phrs.2020.105076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022]
Abstract
Epigenetic modifiers provide a new target for the development of anti-cancer drugs. The eraser histone deacetylase 6 (HDAC6) is a class IIb histone deacetylase that targets various non-histone proteins such as transcription factors, nuclear receptors, cytoskeletal proteins, DNA repair proteins, and molecular chaperones. Therefore, it became an attractive target for cancer treatment. In this study, virtual screening was applied to the MicroCombiChem database with 1162 drug-like compounds to identify new HDAC6 inhibitors. Five compounds were tested in silico and in vitro as HDAC6 inhibitors. Both analyses revealed 1-cyclohexene-1-carboxamide, 2-hydroxy-4,4-dimethyl-N-1-naphthalenyl-6-oxo- (MCC2344) as the best HDAC6 inhibitor among the five ligands. The binding affinity of MCC2344 to HDAC6 was further confirmed by microscale thermophoresis. Additionally, the anti-cancer activity of MCC2344 was tested in several tumor cell lines. Leukemia cells were the most sensitive cells towards MCC2344, particularly the P-glycoprotein-overexpressing multidrug-resistant cell line CEM/ADR5000 exhibited remarkable collateral sensitivity towards MCC2344. Transcriptome analysis using microarray hybridization was performed for investigating downstream mechanisms of action of MCC2344 in leukemia cells. MCC2344 affected microtubule dynamics and suppressed cell migration in the wound healing assay as well as in a spheroid model by hyper-acetylation of tubulin and HSP-90. MCC2344 induced cell death in CEM/ADR5000 cells by activation of PARP, caspase-3, and p21 in addition to the downregulation of p62. MCC2344 significantly inhibited tumor growth in vivo in zebrafish larvae without mortality until 20 pM. We propose MCC2344 as a novel HDAC6 inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany; Department of Molecular Biology, Faculty of Medical Laboratory Sciences, Al-Neelain University, Khartoum, Sudan
| | | | | | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, D-97074, Würzburg, Germany
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
10
|
Dawood M, Elbadawi M, Böckers M, Bringmann G, Efferth T. Molecular docking-based virtual drug screening revealing an oxofluorenyl benzamide and a bromonaphthalene sulfonamido hydroxybenzoic acid as HDAC6 inhibitors with cytotoxicity against leukemia cells. Biomed Pharmacother 2020; 129:110454. [PMID: 32768947 DOI: 10.1016/j.biopha.2020.110454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
HDAC6 is a crucial epigenetic modifier that plays a vital role in tumor progression and carcinogenesis due to its multiple biological functions. It is a unique member of class-II HDAC enzymes. It possesses two catalytic domains, which function independently of the overall enzyme activity. Up to date, there are only a few selective HDAC6 inhibitors with anti-cancer activity. In this study, 175,204 ligands obtained from the ZINC15 and OTAVAchemical databases were used for virtual drug screening against HDAC6. Molecular docking studies were performed for 100 selected compounds. Furthermore, the top 10 compounds obtained from docking were tested for their efficacy to inhibit the function of HDAC6. Five compounds (N-(9-oxo-9H-fluoren-3-yl)benzamide, 2-hydroxy-5-[(5-oxo-6-phenyl-4,5-dihydro-1,2,4-triazin-3-yl)amino]benzoic acid, 5-(4-bromonaphthalene-1-sulfonamido)-2-hydroxybenzoic acid, 2-(naphthalen-2-yl)-N-(1H-1,2,3,4-tetrazol-5-yl)cyclopropane-1-carboxamide, and 4-oxa-5,6 diazapentacyclo[10.7.1.0³,⁷.0⁸,²⁰.0¹⁴,¹⁹]icosa-1,3(7),5,8(20),9,11,14,16,18-nonaen-13-one) inhibited enzymatic activity by more than 50 % compared to DMSO as the control. Two candidates, (N-(9-oxo-9H-fluoren-3-yl)benzamide and 5-(4-bromonaphthalene-1-sulfonamido)-2-hydroxybenzoic acid), were identified with considerable cytotoxicity towards drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells. Microscale thermophoresis revealed the binding of N-(9-oxo-9H-fluoren-3-yl)benzamide and 5-(4-bromonaphthalene-1-sulfonamido)-2-hydroxybenzoic acid to purified HDAC6 protein. Both compounds induced apoptosis in a dose-dependent manner as analyzed by flow cytometry. In conclusion, we demonstrate for the first time that these two compounds bind to HDAC6, inhibit its function, and exert cytotoxic activity by apoptosis induction.
Collapse
Affiliation(s)
- Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Madeleine Böckers
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
11
|
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
|