1
|
Raucci A, Castiello C, Mai A, Zwergel C, Valente S. Heterocycles-Containing HDAC Inhibitors Active in Cancer: An Overview of the Last Fifteen Years. ChemMedChem 2024; 19:e202400194. [PMID: 38726979 DOI: 10.1002/cmdc.202400194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/09/2024] [Indexed: 08/30/2024]
Abstract
Cancer is one of the primary causes of mortality worldwide. Despite nowadays are numerous therapeutic treatments to fight tumor progression, it is still challenging to completely overcome it. It is known that Histone Deacetylases (HDACs), epigenetic enzymes that remove acetyl groups from lysines on histone's tails, are overexpressed in various types of cancer, and their inhibition represents a valid therapeutic strategy. To date, some HDAC inhibitors have achieved FDA approval. Nevertheless, several other potential drug candidates have been developed. This review aims primarily to be comprehensive of the studies done so far regarding HDAC inhibitors bearing heterocyclic rings since their therapeutic potential is well known and has gained increasing interest in recent years. Hence, inserting heterocyclic moieties in the HDAC-inhibiting scaffold can be a valuable strategy to provide potent and/or selective compounds. Here, in addition to summarizing the properties of novel heterocyclic HDAC inhibiting compounds, we also provide ideas for developing new, more potent, and selective compounds for treating cancer.
Collapse
Affiliation(s)
- Alessia Raucci
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Carola Castiello
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| |
Collapse
|
2
|
Frühauf A, Behringer M, Meyer-Almes FJ. Significance of Five-Membered Heterocycles in Human Histone Deacetylase Inhibitors. Molecules 2023; 28:5686. [PMID: 37570656 PMCID: PMC10419652 DOI: 10.3390/molecules28155686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/15/2023] [Accepted: 07/15/2023] [Indexed: 08/13/2023] Open
Abstract
Five-membered heteroaromatic rings, in particular, have gained prominence in medicinal chemistry as they offer enhanced metabolic stability, solubility and bioavailability, crucial factors in developing effective drugs. The unique physicochemical properties and biological effects of five-membered heterocycles have positioned them as key structural motifs in numerous clinically effective drugs. Hence, the exploration of five-ring heterocycles remains an important research area in medicinal chemistry, with the aim of discovering new therapeutic agents for various diseases. This review addresses the incorporation of heteroatoms such as nitrogen, oxygen and sulfur into the aromatic ring of these heterocyclic compounds, enhancing their polarity and facilitating both aromatic stacking interactions and the formation of hydrogen bonds. Histone deacetylases are present in numerous multiprotein complexes within the epigenetic machinery and play a central role in various cellular processes. They have emerged as important targets for cancer, neurodegenerative diseases and other therapeutic indications. In histone deacetylase inhibitors (HDACi's), five-ring heterocycles perform various functions as a zinc-binding group, a linker or head group, contributing to binding activity and selective recognition. This review focuses on providing an up-to-date overview of the different five-membered heterocycles utilized in HDACi motifs, highlighting their biological properties. It summarizes relevant publications from the past decade, offering insights into the recent advancements in this field of research.
Collapse
Affiliation(s)
- Anton Frühauf
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| | - Martin Behringer
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| | - Franz-Josef Meyer-Almes
- Department of Chemical Engineering and Biotechnology, University of Applied Sciences, Haardtring 100, 64295 Darmstadt, Germany
| |
Collapse
|
3
|
Structural Variations in the Central Heterocyclic Scaffold of Tripartite 2,6-Difluorobenzamides: Influence on Their Antibacterial Activity against MDR Staphylococcus aureus. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196619. [PMID: 36235156 PMCID: PMC9573484 DOI: 10.3390/molecules27196619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
Five series of heterocyclic tripartite 2,6-difluorobenzamides, namely 1,2,3-triazoles, 1,2,4- and 1,3,4-oxadiazoles, analogs of reported model anti-staphylococcal compounds, were prepared. The purpose was to investigate the influence of the nature of the heterocyclic central scaffold on the biological activity against three strains of S. aureus, including two drug-resistant ones. Among the 15 compounds of the new collection, a 3-(4-tert-butylphenyl)-1,2,4-oxadiazole linked via a methylene group with a 2,6-difluorobenzamide moiety (II.c) exhibited a minimal inhibitory concentration between 0.5 and 1 µg/mL according to the strain. Subsequent studies on II.c demonstrated no human cytotoxicity, while targeting the bacterial divisome.
Collapse
|
4
|
HDAC Inhibitors for the Therapy of Triple Negative Breast Cancer. Pharmaceuticals (Basel) 2022; 15:ph15060667. [PMID: 35745586 PMCID: PMC9230362 DOI: 10.3390/ph15060667] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an urgent as well as huge medical challenge, which is associated with poor prognosis and responsiveness to chemotherapies. Since epigenetic changes are highly implicated in TNBC tumorigenesis and development, inhibitors of histone deacetylases (HDACIs) could represent a promising therapeutic strategy. Although clinical trials involving single HDACIs showed disappointing results against TNBC, recent studies emphasize the high potential impact of HDACIs in controlling TNBC. In addition, encouraging results stem from new compounds designed to obtain isoform selectivity and/or polypharmacological HDAC approach. The present review provides a discussion of the HDACIs pharmacophoric models and of the structural modifications, leading to compounds with a potent activity against TNBC progression.
Collapse
|
5
|
Atmaram UA, Roopan SM. Biological activity of oxadiazole and thiadiazole derivatives. Appl Microbiol Biotechnol 2022; 106:3489-3505. [PMID: 35562490 PMCID: PMC9106569 DOI: 10.1007/s00253-022-11969-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022]
Abstract
Abstract
The 5-membered oxadiazole and thiadiazole scaffolds are the most privileged and well-known heterocycles, being a common and essential feature of a variety of natural products and medicinal agents. These scaffolds take up the center position and are the core structural components of numerous drugs that belong to different categories. These include antimicrobial, anti-tubercular, anti-inflammatory, analgesic, antiepileptic, antiviral, and anticancer agents. In this review, we mostly talk about the isomers 1,2,4-oxadiazole and 1,3,4-thiadiazole because they have important pharmacological properties. This is partly because they are chemical and heat resistant, unlike other isomers, and they can be used as bio-isosteric replacements in drug design. We are reviewing the structural modifications of different oxadiazole and thiadiazole derivatives, more specifically, the anti-tubercular and anticancer pharmacological activities reported over the last 5 years, as we have undertaken this as a core area of research. This review article desires to do a thorough study and analysis of the recent progress made in the important biological isomers 1,2,4-oxadiazole and 1,3,4-thiadiazol. This will be a great place to start for future research. Key points • Five-membered heterocyclic compound chemistry and biological activity recent survey. • Synthesis and pharmacological evolution of 1,2,4-oxadiazole and 1,3,4-thiadiazole are discussed in detail. • The value and significance of heterocyclic compounds in the field of drug designing are highlighted. Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-11969-0.
Collapse
Affiliation(s)
- Upare Abhay Atmaram
- Chemistry of Heterocycles & Natural Product Research Laboratory, Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - Selvaraj Mohana Roopan
- Chemistry of Heterocycles & Natural Product Research Laboratory, Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
6
|
Ibrahim HS, Abdelsalam M, Zeyn Y, Zessin M, Mustafa AHM, Fischer MA, Zeyen P, Sun P, Bülbül EF, Vecchio A, Erdmann F, Schmidt M, Robaa D, Barinka C, Romier C, Schutkowski M, Krämer OH, Sippl W. Synthesis, Molecular Docking and Biological Characterization of Pyrazine Linked 2-Aminobenzamides as New Class I Selective Histone Deacetylase (HDAC) Inhibitors with Anti-Leukemic Activity. Int J Mol Sci 2021; 23:ijms23010369. [PMID: 35008795 PMCID: PMC8745332 DOI: 10.3390/ijms23010369] [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: 12/11/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Class I histone deacetylases (HDACs) are key regulators of cell proliferation and they are frequently dysregulated in cancer cells. We report here the synthesis of a novel series of class-I selective HDAC inhibitors (HDACi) containing a 2-aminobenzamide moiety as a zinc-binding group connected with a central (piperazin-1-yl)pyrazine or (piperazin-1-yl)pyrimidine moiety. Some of the compounds were additionally substituted with an aromatic capping group. Compounds were tested in vitro against human HDAC1, 2, 3, and 8 enzymes and compared to reference class I HDACi (Entinostat (MS-275), Mocetinostat, CI994 and RGFP-966). The most promising compounds were found to be highly selective against HDAC1, 2 and 3 over the remaining HDAC subtypes from other classes. Molecular docking studies and MD simulations were performed to rationalize the in vitro data and to deduce a complete structure activity relationship (SAR) analysis of this novel series of class-I HDACi. The most potent compounds, including 19f, which blocks HDAC1, HDAC2, and HDAC3, as well as the selective HDAC1/HDAC2 inhibitors 21a and 29b, were selected for further cellular testing against human acute myeloid leukemia (AML) and erythroleukemic cancer (HEL) cells, taking into consideration their low toxicity against human embryonic HEK293 cells. We found that 19f is superior to the clinically tested class-I HDACi Entinostat (MS-275). Thus, 19f is a new and specific HDACi with the potential to eliminate blood cancer cells of various origins.
Collapse
Affiliation(s)
- Hany S. Ibrahim
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11829, Egypt
| | - Mohamed Abdelsalam
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yanira Zeyn
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (A.-H.M.M.); (M.A.F.)
| | - Matthes Zessin
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
- Department of Enzymology, Institute of Biochemistry, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Al-Hassan M. Mustafa
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (A.-H.M.M.); (M.A.F.)
- Department of Zoology, Faculty of Science, Aswan University, Aswan 81528, Egypt
| | - Marten A. Fischer
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (A.-H.M.M.); (M.A.F.)
| | - Patrik Zeyen
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Ping Sun
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Emre F. Bülbül
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Anita Vecchio
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Frank Erdmann
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Matthias Schmidt
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Dina Robaa
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 25250 Vestec, Czech Republic;
| | - Christophe Romier
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS, INSERM, Université de Strasbourg, CEDEX, 67404 Illkirch, France;
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center, 55131 Mainz, Germany; (Y.Z.); (A.-H.M.M.); (M.A.F.)
- Correspondence: (O.H.K.); (W.S.)
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (H.S.I.); (M.A.); (M.Z.); (P.Z.); (P.S.); (E.F.B.); (A.V.); (F.E.); (M.S.); (D.R.)
- Correspondence: (O.H.K.); (W.S.)
| |
Collapse
|
7
|
Buonvicino D, Ranieri G, Chiarugi A. Treatment with Non-specific HDAC Inhibitors Administered after Disease Onset does not Delay Evolution in a Mouse Model of Progressive Multiple Sclerosis. Neuroscience 2021; 465:38-45. [PMID: 33862148 DOI: 10.1016/j.neuroscience.2021.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Drugs able to efficiently counteract progression of multiple sclerosis (MS) are still an unmet need. Several lines of evidence indicate that histone deacetylase inhibitors (HDACi) are clinically-available epigenetic drugs that might be repurposed for immunosuppression in MS therapy. Here, we studied the effects of HDACi on disease evolution in myelin oligodendrocyte glycoprotein (MOG)-immunized NOD mice, an experimental model of progressive experimental autoimmune encephalomyelitis (PEAE). To obtain data of potential clinical relevance, the HDACi panobinostat, givinostat and entinostat were administered orally adopting a daily treatment protocol after disease onset. We report that the 3 drugs efficiently reduced in vitro lymphocyte proliferation in a dose-dependent manner. Notably, however, none of the drugs delayed evolution of PEAE or reduced lethality in NOD mice. In striking contrast with this, however, the lymphocyte proliferation response to MOG as well as Th1 and Th17 spinal cord infiltrates were significantly lower in animals exposed to the HDACi compared to those receiving vehicle. When put into a clinical context, for the first time data cast doubt on the relevance of HDACi to treatment of progressive MS (PMS). Also, our findings further indicate that, akin to PMS, neuropathogensis of PEAE in NOD mice becomes independent from autoimmunity, thereby corroborating the relevance of this model to experimental PMS research.
Collapse
Affiliation(s)
- Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| | - Giuseppe Ranieri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
8
|
Catalytic thiomethylation of regioisomeric aminobenzamides using bis(N,N-dimethylamino)methane and α,ω-alkanedithiols. Russ Chem Bull 2021. [DOI: 10.1007/s11172-021-3147-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Khairullina RR, Tyumkina TV, Akhmetshina GA, Abdullin MF, Ibragimov AG. Catalytic thiomethylation of aminobenzamides using bis(N,N-dimethylamino)methane, hydrogen sulfide, and its sodium salts. Russ Chem Bull 2021. [DOI: 10.1007/s11172-021-3070-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
10
|
Kapoor G, Bhutani R, Pathak DP, Chauhan G, Kant R, Grover P, Nagarajan K, Siddiqui SA. Current Advancement in the Oxadiazole-Based Scaffolds as Anticancer Agents. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1886123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Garima Kapoor
- KIET School of Pharmacy, KIET Group of InstitutionsGhaziabad, Uttar Pradesh, India
| | - Rubina Bhutani
- School of Medical and Allied Sciences, GD Goenka University, Gurgaon, Haryana, India
| | - Dharam Pal Pathak
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), New Delhi, India
| | - Garima Chauhan
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), New Delhi, India
| | - Ravi Kant
- Lloyd Institute of Management and Technology, Greater Noida, India
| | - Parul Grover
- KIET School of Pharmacy, KIET Group of InstitutionsGhaziabad, Uttar Pradesh, India
| | - Kandasamy Nagarajan
- KIET School of Pharmacy, KIET Group of InstitutionsGhaziabad, Uttar Pradesh, India
| | | |
Collapse
|
11
|
Design, synthesis, antibacterial evaluation and molecular docking studies of novel pyrazole/1,2,4-oxadiazole conjugate ester derivatives. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
12
|
Benassi A, Doria F, Pirota V. Groundbreaking Anticancer Activity of Highly Diversified Oxadiazole Scaffolds. Int J Mol Sci 2020; 21:ijms21228692. [PMID: 33217987 PMCID: PMC7698752 DOI: 10.3390/ijms21228692] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/29/2022] Open
Abstract
Nowadays, an increasing number of heterocyclic-based drugs found application in medicinal chemistry and, in particular, as anticancer agents. In this context, oxadiazoles—five-membered aromatic rings—emerged for their interesting biological properties. Modification of oxadiazole scaffolds represents a valid strategy to increase their anticancer activity, especially on 1,2,4 and 1,3,4 regioisomers. In the last years, an increasing number of oxadiazole derivatives, with remarkable cytotoxicity for several tumor lines, were identified. Structural modifications, that ensure higher cytotoxicity towards malignant cells, represent a solid starting point in the development of novel oxadiazole-based drugs. To increase the specificity of this strategy, outstanding oxadiazole scaffolds have been designed to selectively interact with biological targets, including enzymes, globular proteins, and nucleic acids, showing more promising antitumor effects. In the present work, we aim to provide a comprehensive overview of the anticancer activity of these heterocycles, describing their effect on different targets and highlighting how their structural versatility has been exploited to modulate their biological properties.
Collapse
|
13
|
Hydroxamic acid hybrids as the potential anticancer agents: An Overview. Eur J Med Chem 2020; 205:112679. [PMID: 32791404 DOI: 10.1016/j.ejmech.2020.112679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
|
14
|
Mokenapelli S, Thalari G, Vadiyaala N, Yerrabelli JR, Irlapati VK, Gorityala N, Sagurthi SR, Chitneni PR. Synthesis, cytotoxicity, and molecular docking of substituted 3-(2-methylbenzofuran-3-yl)-5-(phenoxymethyl)-1,2,4-oxadiazoles. Arch Pharm (Weinheim) 2020; 353:e2000006. [PMID: 32309890 DOI: 10.1002/ardp.202000006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 11/09/2022]
Abstract
A series of new benzofuran/oxadiazole hybrids (8a-n) was synthesized from 2H-chromene-3-carbonitriles (3a-c) through the multistep synthetic methodology, and these hybrids are known to exhibit anticancer activities. All the compounds were evaluated for their in vitro cytotoxicity against the HCT116 and MIA PaCa2 cell lines. Compounds 6a (IC50 : 9.71 ± 1.9 μM), 6b (IC50 : 7.48 ± 0.6 μM), and 6c (IC50 : 3.27 ± 1.1 μM) displayed a significant cytotoxic activity, whereas compounds 8d and 8e exhibited good activity against both cell lines. The depletion of glycogen synthase kinase-3β (GSK3β) induces apoptosis through the inhibition of basal NF-κB activity in HCT116 colon cancer cells and MIA PaCa2 pancreatic cancer cells. Molecular docking of compounds 6a, 6b, 6c, 8d, and 8e with GSK3β demonstrated the best binding affinity, correlating with the biological activity assay. Furthermore, the structure-activity relationship of these novel compounds reveals promising features for their use in anticancer therapy.
Collapse
Affiliation(s)
- Sudhakar Mokenapelli
- Natural Products Laboratory, Department of Chemistry, Osmania University, Hyderabad, Telangana, India
| | - Gangadhar Thalari
- Natural Products Laboratory, Department of Chemistry, Osmania University, Hyderabad, Telangana, India
| | - Naveen Vadiyaala
- Natural Products Laboratory, Department of Chemistry, Osmania University, Hyderabad, Telangana, India
| | - Jayaprakash R Yerrabelli
- Natural Products Laboratory, Department of Chemistry, Osmania University, Hyderabad, Telangana, India
| | - Vamshi K Irlapati
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| | - Neelima Gorityala
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| | - Someswar R Sagurthi
- Department of Genetics and Biotechnology, Osmania University, Hyderabad, Telangana, India
| | - Prasad R Chitneni
- Natural Products Laboratory, Department of Chemistry, Osmania University, Hyderabad, Telangana, India
| |
Collapse
|
15
|
Ruan B, Zhang Y, Tadesse S, Preston S, Taki AC, Jabbar A, Hofmann A, Jiao Y, Garcia-Bustos J, Harjani J, Le TG, Varghese S, Teguh S, Xie Y, Odiba J, Hu M, Gasser RB, Baell J. Synthesis and structure-activity relationship study of pyrrolidine-oxadiazoles as anthelmintics against Haemonchus contortus. Eur J Med Chem 2020; 190:112100. [PMID: 32018095 DOI: 10.1016/j.ejmech.2020.112100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
Abstract
Parasitic roundworms (nematodes) are significant pathogens of humans and animals and cause substantive socioeconomic losses due to the diseases that they cause. The control of nematodes in livestock animals relies heavily on the use of anthelmintic drugs. However, their extensive use has led to a widespread problem of drug resistance in these worms. Thus, the discovery and development of novel chemical entities for the treatment of parasitic worms of humans and animals is needed. Herein, we describe our medicinal chemistry optimization efforts of a phenotypic hit against Haemonchus contortus based on a pyrrolidine-oxadiazole scaffold. This led to the identification of compounds with potent inhibitory activities (IC50 = 0.78-22.4 μM) on the motility and development of parasitic stages of H. contortus, and which were found to be highly selective in a mammalian cell counter-screen. These compounds could be used as suitable chemical tools for drug target identification or as lead compounds for further optimization.
Collapse
Affiliation(s)
- Banfeng Ruan
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia; Key Lab of Biofabrication of Anhui Higher Education, Institution Centre for Advanced Biofabrication, Hefei University, Hefei, 230601, PR China
| | - Yuezhou Zhang
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia; Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia; State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Solomon Tadesse
- Department of Pharmaceutical Chemistry and Pharmacognosy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Sarah Preston
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia; School of Health and Life Sciences, Federation University, Ballarat, Victoria, 3353, Australia
| | - Aya C Taki
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Abdul Jabbar
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andreas Hofmann
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yaqing Jiao
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jose Garcia-Bustos
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jitendra Harjani
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Thuy Giang Le
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Swapna Varghese
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Silvia Teguh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Yiyue Xie
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Jephthah Odiba
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria, 3052, Australia; School of Pharmaceutical Sciences, Nanjing Tech University, No. 30, South Puzhu Road, Nanjing, 211816, PR China; ARC Centre for Fragment-Based Design, Monash University, Parkville, VIC, 3052, Australia; Australian Translational Medicinal Chemistry Facility (ATMCF), Monash University, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
16
|
QSAR analysis of coumarin-based benzamides as histone deacetylase inhibitors using CoMFA, CoMSIA and HQSAR methods. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.126961] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
17
|
Banerjee S, Amin SA, Adhikari N, Jha T. Essential elements regulating HDAC8 inhibition: a classification based structural analysis and enzyme-inhibitor interaction study of hydroxamate based HDAC8 inhibitors. J Biomol Struct Dyn 2019; 38:5513-5525. [DOI: 10.1080/07391102.2019.1704881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sk. Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
18
|
Janaki Ramaiah M, Divyapriya K, Kartik Kumar S, Rajesh YBRD. Drug-induced modifications and modulations of microRNAs and long non-coding RNAs for future therapy against Glioblastoma Multiforme. Gene 2019; 723:144126. [PMID: 31589963 DOI: 10.1016/j.gene.2019.144126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
Non-coding RNAs are known to participate in cancer initiation, progression, and metastasis by regulating the status of chromatin epigenetics and gene expression. Although these non-coding RNAs do not possess defined protein-coding potential, they are involved in the expression and stability of messenger RNA (mRNA). The length of microRNAs (miRs) ranges between 20 and 22 nt, whereas, long non-coding RNAs (lncRNAs) length ranges between 200 nt to 1 Kb. In the case of circular RNAs (circRNAs), the size varies depending upon the length of the exon from where they were derived. Epigenetic regulations of miR and lncRNA genes will influence the gene expression by modulating histone acetylation and methylation patterns. Especially, lncRNAs will act as a scaffold for various epigenetic proteins, such as EZH2 and LSD1, and influence the chromatin epigenetic state at various genomic loci involved at silencing. Thus investigations on the expression of lncRNAs and designing drugs to modulate the expression of these genes will have a profound impact on future therapeutics against cancers such as Glioblastoma Multiforme (GBM) and also against various other diseases. With the recent advancements in genome-wide transcriptomic studies, scientists are focused on the non-coding RNAs and their regulations on various cellular processes involved in GBM and on other types of cancer as well as trying to understand possible epigenetic modulations that help in generating promising therapeutics for the future generations. In this review, the involvement of epigenetic proteins, enzymes that change chromatin architecture and epigenetic landscape and new roles of lncRNAs that are involved in GBM progression are elaborately discussed.
Collapse
Affiliation(s)
- M Janaki Ramaiah
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India.
| | - Karthikeyan Divyapriya
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - Sarwareddy Kartik Kumar
- Laboratory of Functional Genomics and Disease Biology, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - Y B R D Rajesh
- Organic Synthesis and Catalysis Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
19
|
Krishna S, Lakra AD, Shukla N, Khan S, Mishra DP, Ahmed S, Siddiqi MI. Identification of potential histone deacetylase1 (HDAC1) inhibitors using multistep virtual screening approach including SVM model, pharmacophore modeling, molecular docking and biological evaluation. J Biomol Struct Dyn 2019; 38:3280-3295. [DOI: 10.1080/07391102.2019.1654925] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Shagun Krishna
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amar Deep Lakra
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nidhi Shukla
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Saman Khan
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Durga Prasad Mishra
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shakil Ahmed
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
20
|
Khairullina RR, Tyumkina TV, Geniyatova AR, Abdullin MF, Ibragimov AG. Catalytic Cycloaminomethylation of Aminobenzamides with 1,3-Bis[dimethylamino(methoxy)methyl]thiourea. RUSS J GEN CHEM+ 2019. [DOI: 10.1134/s1070363219030022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Khairullina RR, Tyumkina TV, Geniyatova AR, Mullagil’dina FK, Ibragimov AG. Catalytic Aminomethylation of Aminobenzamides with Bis(N,N-dimethylamino)methane and Carbamides. RUSS J GEN CHEM+ 2019. [DOI: 10.1134/s1070363219020063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Li Y, Fang H, Hou Z, Sang L, Yang X. An in-line capillary electrophoresis assay for the high-throughput screening of histone deacetylase inhibitors. J Chromatogr A 2019; 1591:171-177. [DOI: 10.1016/j.chroma.2019.01.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/04/2023]
|
23
|
Histone deacetylase 8 (HDAC8) and its inhibitors with selectivity to other isoforms: An overview. Eur J Med Chem 2018; 164:214-240. [PMID: 30594678 DOI: 10.1016/j.ejmech.2018.12.039] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/04/2018] [Accepted: 12/16/2018] [Indexed: 01/08/2023]
Abstract
The histone deacetylases (HDACs) enzymes provided crucial role in transcriptional regulation of cells through deacetylation of nuclear histone proteins. Discoveries related to the HDAC8 enzyme activity signified the importance of HDAC8 isoform in cell proliferation, tumorigenesis, cancer, neuronal disorders, parasitic/viral infections and other epigenetic regulations. The pan-HDAC inhibitors can confront these conditions but have chances to affect epigenetic functions of other HDAC isoforms. Designing of selective HDAC8 inhibitors is a key feature to combat the pathophysiological and diseased conditions involving the HDAC8 activity. This review is concerned about the structural and positional aspects of HDAC8 in the HDAC family. It also covers the contributions of HDAC8 in the pathophysiological conditions, a preliminary discussion about the recent scenario of HDAC8 inhibitors. This review might help to deliver the structural, functional and computational information in order to identify and design potent and selective HDAC8 inhibitors for target specific treatment of diseases involving HDAC8 enzymatic activity.
Collapse
|
24
|
Yang F, Shan P, Zhao N, Ge D, Zhu K, Jiang CS, Li P, Zhang H. Development of hydroxamate-based histone deacetylase inhibitors containing 1,2,4-oxadiazole moiety core with antitumor activities. Bioorg Med Chem Lett 2018; 29:15-21. [PMID: 30455152 DOI: 10.1016/j.bmcl.2018.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/17/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
Histone deacetylases (HDACs) has proved to be promising target for the development of antitumor drugs. In this study, we reported the design and synthesis of a class of novel hydroxamate-based bis-substituted aromatic amide HDAC inhibitors with 1,2,4-oxadiazole core. Most newly synthesized compounds displayed excellent HDAC1 inhibitory effects and significant anti-proliferative activities. Among them, compounds 11a and 11c increased acetylation of histone H3 and H4 in dose-dependent manner. Furthermore, 11a and 11c remarkably induced apoptosis in HepG2 cancer cells. Finally, the high potency of compound 11a was rationalized by molecular docking studies.
Collapse
Affiliation(s)
- Feifei Yang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Peipei Shan
- Institute for Translation Medicine, Qingdao University, Qingdao, Shandong Province 266071, China
| | - Na Zhao
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Kongkai Zhu
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China
| | - Peifeng Li
- Institute for Translation Medicine, Qingdao University, Qingdao, Shandong Province 266071, China.
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, Shandong Province 250022, China.
| |
Collapse
|
25
|
M JR, S V. BMI1 and PTEN are key determinants of breast cancer therapy: A plausible therapeutic target in breast cancer. Gene 2018; 678:302-311. [PMID: 30096458 DOI: 10.1016/j.gene.2018.08.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/11/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
BMI-1 (B-lymphoma Mo-MLV insertion region 1) is a key protein partner in polycomb repressive complex 1 (PRC1) that helps in maintaining the integrity of the complex. It is also a key player in ubiquitination of histone H2A which affects gene expression pattern involved in various cellular processes such as cell proliferation, growth, DNA repair, apoptosis and senescence. In many cancers, Overexpression of BMI1correlates with advanced stages of disease, aggressive clinicopathological behavior, poor prognosis resistance to radiation and chemotherapy. BMI1 is emerging as a key player in EMT, chemo-resistance and cancer stemness. Overexpression is observed in various cancer types such as breast, primary hepatocellular carcinoma (HCC), gastric, ovarian, head and neck, pancreatic and lung cancer. Studies have shown that experimental reduction of BMI protein level in tumor cells results in inhibition of cell proliferation, induction of apoptosis and/or senescence, and increases susceptibility to cytotoxic agents and radiation therapy. Thus, inhibition of BMI1 expression particularly in breast cancer stem cells can be used as a potential strategy for the complete elimination of tumor and to prevent disease relapse. On other hand PTEN is known to be an important tumor suppressor next to p53. In many cancers particularly in breast cancer, p53 and PTEN undergo mutations. Studies have indicated the functional and mechanistic link between the BMI-1oncoprotein and tumor suppressor PTEN in the development and progression of cancer. The current review focuses on recent findings of how oncogenicity and chemo-resistance are caused by BMI1. It also highlights the transcriptional regulation between BMI1 and PTEN that dictates the therapeutic outcome in cancers where the functional p53 is absent. Herein, we have clearly demonstrated the regulation of transcription at genomic loci of BMI1 and PTEN in cancerous tissue or cells and the possible epigenetic regulation by histone deacetylase inhibitors (HDACi) at BMI1 and PTEN loci that may provide some clue for the possible therapy against TNBC in near future.
Collapse
Affiliation(s)
- Janaki Ramaiah M
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, India.
| | - Vaishnave S
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, India
| |
Collapse
|
26
|
Recent advance in oxazole-based medicinal chemistry. Eur J Med Chem 2018; 144:444-492. [DOI: 10.1016/j.ejmech.2017.12.044] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023]
|
27
|
Structure–activity relationships of hydroxamate-based histone deacetylase-8 inhibitors: reality behind anticancer drug discovery. Future Med Chem 2017; 9:2211-2237. [PMID: 29182018 DOI: 10.4155/fmc-2017-0130] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pan-histone deacetylase (HDAC) inhibitors comprise a fish-like structural orientation where hydrophobic aryl- and zinc-binding groups act as head and tail, respectively of a fish. The linker moiety correlates the body of the fish linking head and tail groups. Despite these pan-HDAC inhibitors, selective HDAC-8 inhibitors are still in demand as a safe remedy. HDAC-8 is involved in invasion and metastasis in cancer. This review deals with the rationale behind HDAC-8 inhibitory activity and selectivity along with detailed structure–activity relationships of diverse hydroxamate-based HDAC-8 inhibitors. HDAC-8 inhibitory potency may be increased by modifying the fish-like pharmacophoric features of such type of pan-HDAC inhibitors. This review may provide a preliminary basis to design and optimize new lead molecules with higher HDAC-8 inhibitory activity. This work may surely enlighten in providing useful information in the field of target-specific anticancer therapy.
Collapse
|
28
|
Abdizadeh T, Kalani MR, Abnous K, Tayarani-Najaran Z, Khashyarmanesh BZ, Abdizadeh R, Ghodsi R, Hadizadeh F. Design, synthesis and biological evaluation of novel coumarin-based benzamides as potent histone deacetylase inhibitors and anticancer agents. Eur J Med Chem 2017; 132:42-62. [PMID: 28340413 DOI: 10.1016/j.ejmech.2017.03.024] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/06/2017] [Accepted: 03/14/2017] [Indexed: 01/26/2023]
Abstract
Histone deacetylases (HDACs) are attractive therapeutic targets for the treatment of cancer and other diseases. It has four classes (I-IV), among them especially class I isozyme are involved in promoting tumor cells proliferation, angiogenesis, differentiation, invasion and metastasis and also viable targets for cancer therapeutics. A novel series of coumarin-based benzamides was designed and synthesized as HDAC inhibitors. The cytotoxic activity of the synthesized compounds (8a-u) was evaluated against six human cancer cell lines including HCT116, A2780, MCF7, PC3, HL60 and A549 and a single normal cell line (Huvec). We evaluated their inhibitory activities against pan HDAC and HDAC1 isoform. Four compounds (8f, 8q, 8r and 8u) showed significant cytotoxicity with IC50 in the range of 0.53-57.59 μM on cancer cells and potent pan-HDAC inhibitory activity (consists of HDAC isoenzymes) (IC50 = 0.80-14.81 μM) and HDAC1 inhibitory activity (IC50 = 0.47-0.87 μM and also, had no effect on Huvec (human normal cell line) viability (IC50 > 100 μM). Among them, 8u displayed a higher potency for HDAC1 inhibition with IC50 value of 0.47 ± 0.02 μM near equal to the reference drug Entinostat (IC50 = 0.41 ± 0.06 μM). Molecular docking studies and Molecular dynamics simulation of compound 8a displayed possible mode of interaction between this compound and HDAC1enzyme.
Collapse
Affiliation(s)
- Tooba Abdizadeh
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Kalani
- School of Cell and Molecular Biology, University of Illinois at Urbana-Champaign, Urbana, United States; Department of Molecular Medicine, Golestan University of Medical Sciences, Golestan, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Zahra Khashyarmanesh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rahman Abdizadeh
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Farzin Hadizadeh
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Moniot S, Forgione M, Lucidi A, Hailu GS, Nebbioso A, Carafa V, Baratta F, Altucci L, Giacché N, Passeri D, Pellicciari R, Mai A, Steegborn C, Rotili D. Development of 1,2,4-Oxadiazoles as Potent and Selective Inhibitors of the Human Deacetylase Sirtuin 2: Structure–Activity Relationship, X-ray Crystal Structure, and Anticancer Activity. J Med Chem 2017; 60:2344-2360. [DOI: 10.1021/acs.jmedchem.6b01609] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sébastien Moniot
- Department
of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Mariantonietta Forgione
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Alessia Lucidi
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Gebremedhin S. Hailu
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Angela Nebbioso
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Vincenzo Carafa
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Francesca Baratta
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- Department
of Biochemistry, Biophysics and General Pathology, Second University of Naples, Vico L. de Crecchio 7, 80138 Naples, Italy
| | - Nicola Giacché
- TES Pharma S.r.l., Via P. Togliatti 20, 06073 Corciano, Perugia, Italy
| | - Daniela Passeri
- TES Pharma S.r.l., Via P. Togliatti 20, 06073 Corciano, Perugia, Italy
| | | | - Antonello Mai
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| | - Clemens Steegborn
- Department
of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany
| | - Dante Rotili
- Department
of Drug Chemistry and Technologies, Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Sapienza University of Rome, P. le A. Moro
5, 00185 Rome, Italy
| |
Collapse
|
30
|
Tolmachev A, Bogolubsky AV, Pipko SE, Grishchenko AV, Ushakov DV, Zhemera AV, Viniychuk OO, Konovets AI, Zaporozhets OA, Mykhailiuk PK, Moroz YS. Expanding Synthesizable Space of Disubstituted 1,2,4-Oxadiazoles. ACS COMBINATORIAL SCIENCE 2016; 18:616-624. [PMID: 27548754 DOI: 10.1021/acscombsci.6b00103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
One-pot synthesis of 3,5-disubstituted 1,2,4-oxadiazoles from carboxylic acids and nitriles was optimized to parallel chemistry. The method was validated on a 141 member library; the desired products were recovered with a high success rate and in moderate yields. Practical application of the approach was demonstrated in the synthesis of bioactive compound pifexole and agonists of free fatty acid receptor 1. A library of 4 948 100 synthesizable drug-like 3,5-disubstituted 1,2,4-oxadiazoles was enumerated based on the method and available validated reagents.
Collapse
Affiliation(s)
- Andrey Tolmachev
- Enamine Ltd., 78 Chervonotkatska
Street, Kyiv, 02094, Ukraine
- ChemBioCenter, Kyiv National Taras Shevchenko University, 61 Chervonotkatska Street, Kyiv, 02094, Ukraine
| | | | - Sergey E. Pipko
- ChemBioCenter, Kyiv National Taras Shevchenko University, 61 Chervonotkatska Street, Kyiv, 02094, Ukraine
- UkrOrgSyntez Ltd. (UORSY), 29 Schorsa
Street, Kyiv, 01133, Ukraine
| | | | | | | | | | - Anzhelika I. Konovets
- Enamine Ltd., 78 Chervonotkatska
Street, Kyiv, 02094, Ukraine
- The
Institute of High Technologies, Kyiv National Taras Shevchenko University, 4 Glushkov Street, Building 5, Kyiv, 03187, Ukraine
| | - Olga A. Zaporozhets
- Department
of Chemistry, Kyiv National Taras Shevchenko University, 64 Volodymyrska
Street, Kyiv, 01601, Ukraine
| | - Pavel K. Mykhailiuk
- Enamine Ltd., 78 Chervonotkatska
Street, Kyiv, 02094, Ukraine
- Department
of Chemistry, Kyiv National Taras Shevchenko University, 64 Volodymyrska
Street, Kyiv, 01601, Ukraine
| | - Yurii S. Moroz
- ChemBioCenter, Kyiv National Taras Shevchenko University, 61 Chervonotkatska Street, Kyiv, 02094, Ukraine
| |
Collapse
|
31
|
Xing S, Bing Q, Song L, Li G, Liu J, Shi Z, Feng S, Xu R. The Uncommon Channel-Based Ln-MOFs for Highly Selective Fe3+
Detection and Superior Rhodamine B Adsorption. Chemistry 2016; 22:16230-16235. [DOI: 10.1002/chem.201603102] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Shanghua Xing
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| | - Qiming Bing
- Institute of Theoretical Chemistry; Jilin University; Changchun 130023 P.R. China
| | - Lifei Song
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| | - Guanghua Li
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| | - Jingyao Liu
- Institute of Theoretical Chemistry; Jilin University; Changchun 130023 P.R. China
| | - Zhan Shi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| | - Ruren Xu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry; College of Chemistry; Jilin University; Changchun 130012 P.R. China
| |
Collapse
|
32
|
Synthesis and Cytotoxic Activity of (4-Substituted-benzylidene)-(3-Phenyl-1,2,4-Oxadiazol-5-YL)Methylamines. Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1429-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Paul S, Roy A, Deka SJ, Panda S, Trivedi V, Manna D. Nitrobenzofurazan derivatives of N'-hydroxyamidines as potent inhibitors of indoleamine-2,3-dioxygenase 1. Eur J Med Chem 2016; 121:364-375. [PMID: 27267006 DOI: 10.1016/j.ejmech.2016.05.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 11/16/2022]
Abstract
Tryptophan metabolism through the kynurenine pathway is considered as a crucial mechanism in immune tolerance. Indoleamine 2,3-dioxygenase 1 (IDO1) plays a key role in tryptophan catabolism in the immune system and it is also considered as an important therapeutic target for the treatment of cancer and other diseases that are linked with kynurenine pathway. In this study, a series of nitrobenzofurazan derivatives of N'-hydroxybenzimidamides (1) and N'-hydroxy-2-phenylacetimidamides (2) were synthesized and their inhibitory activities against human IDO1 enzyme were tested using in-vitro and cellular enzyme activity assay. The optimization leads to the identification of potent compounds, 1d, 2i and 2k (IC50 = 39-80 nM), which are either competitive or uncompetitive inhibitors of IDO1 enzyme. These compounds also showed IDO1 inhibition potencies in the nanomolar range (IC50 = 50-71 nM) in MDA-MB-231 cells with no/negligible amount of cytotoxicity. The stronger selectivity of the potent compounds for IDO1 enzyme over tryptophan 2,3-dioxygenase (TDO) enzyme (312-1593-fold) also makes them very attractive for further immunotherapeutic applications.
Collapse
Affiliation(s)
- Saurav Paul
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Ashalata Roy
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Suman Jyoti Deka
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Subhankar Panda
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Vishal Trivedi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Debasis Manna
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
34
|
Orally available stilbene derivatives as potent HDAC inhibitors with antiproliferative activities and antitumor effects in human tumor xenografts. Eur J Med Chem 2016; 108:274-286. [DOI: 10.1016/j.ejmech.2015.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/05/2015] [Accepted: 11/07/2015] [Indexed: 11/22/2022]
|
35
|
Design, synthesis and biological evaluation of N-phenylquinazolin-4-amine hybrids as dual inhibitors of VEGFR-2 and HDAC. Eur J Med Chem 2015; 109:1-12. [PMID: 26741358 DOI: 10.1016/j.ejmech.2015.12.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 01/01/2023]
Abstract
A single agent that simultaneously inhibits multiple targets may offer greater therapeutic benefits in cancer than single-acting agents through interference with multiple pathways and potential synergistic action. In this work, a series of hybrids bearing N-phenylquinazolin-4-amine and hydroxamic acid moieties were designed and identified as dual VEGFR-2/HDAC inhibitors. Compound 6fd exhibited the most potent inhibitory activity against HDAC with IC50 of 2.2 nM and strong inhibitory effect against VEGFR-2 with IC50 of 74 nM. It also showed the most potent inhibitory activity against a human breast cancer cell line MCF-7 with IC50 of 0.85 μM. Docking simulation supported the initial pharmacophoric hypothesis and suggested a common mode of interaction at the active binding sites of VEGFR-2 and HDLP ((Histone Deacetylase-Like Protein), which demonstrates that compound 6fd is a potential agent for cancer therapy deserving further researching.
Collapse
|