26
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      Drug Design, Development and Therapy (submit here)

      This international, peer-reviewed Open Access journal by Dove Medical Press focuses on the design and development of drugs, as well as the clinical outcomes, patient safety, and programs targeted at the effective and safe use of medicines. Sign up for email alerts here.

      88,007 Monthly downloads/views I 4.319 Impact Factor I 6.6 CiteScore I 1.12 Source Normalized Impact per Paper (SNIP) I 0.784 Scimago Journal & Country Rank (SJR)

       

      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Design, Synthesis and Biological Evaluation of New HDAC1 and HDAC2 Inhibitors Endowed with Ligustrazine as a Novel Cap Moiety

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Histone deacetylases (HDACs) represent one of the most validated cancer targets. The inhibition of HDACs has been proven to be a successful strategy for the development of novel anticancer candidates.

          Methods

          This work describes design and synthesis of a new set of HDAC inhibitors ( 7a-c and 8a, b) utilizing ligustrazine as a novel cap moiety, and achieving the pharmacophoric features required to induce the desired inhibition.

          Results

          The newly synthesized derivatives were evaluated for their potential inhibitory activity toward two class I histone deacetylases, namely HDAC1 and HDAC2. The tested ligustrazine-based compounds were more potent toward HDAC2 (IC 50 range: 53.7–205.4 nM) than HDAC1 (IC 50 range: 114.3–2434.7 nM). Furthermore, the antiproliferative activities against two HDAC-expressing cancer cell lines; HT-29 and SH-SY5Y were examined by the MTT assay. Moreover, a molecular docking study of the designed HDAC inhibitors ( 7a-c and 8a,b) was carried out to investigate their binding pattern within their prospective targets; HDAC1 (PDB-ID: 4BKX) and HDAC2 (PDB-ID: 6G3O).

          Discussion

          Compound 7a was found to be the most potent analog in this study toward HDAC1 and HDAC2 with IC 50 values equal 114.3 and 53.7 nM, respectively. Moreover, it was the most effective counterpart (IC 50 = 1.60 µM), with 4.7-fold enhanced efficiency than reference drug Gefitinib (IC 50 = 7.63 µM) against SH-SY5Y cells. Whereas, compound 8a (IC 50 = 1.96 µM) was the most active member toward HT-29 cells, being 2.5-times more potent than Gefitinib (IC 50 = 4.99 µM). Collectively, these results suggest that 7a merits further optimization and development as an effective new HDACI lead compound.

          Most cited references22

          • Record: found
          • Abstract: found
          • Article: not found

          Histone-deacetylase inhibitors: novel drugs for the treatment of cancer.

          The opposing actions of histone acetyltransferases (HATs) and histone deacetylases (HDACs) allow gene expression to be exquisitely regulated through chromatin remodelling. Aberrant transcription due to altered expression or mutation of genes that encode HATs, HDACs or their binding partners, is a key event in the onset and progression of cancer. HDAC inhibitors can reactivate gene expression and inhibit the growth and survival of tumour cells. The remarkable tumour specificity of these compounds, and their potency in vitro and in vivo, underscore the potential of HDAC inhibitors as exciting new agents for the treatment of cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Medicinal chemistry and the molecular operating environment (MOE): application of QSAR and molecular docking to drug discovery.

            The search for new compounds with a given biological activity requires enormous effort in terms of manpower and cost. This effort arises from the large number of compounds that need to be synthesized and subsequently biologically evaluated. For this reason the pharmaceutical industry has shown great interest in theoretical methods that enable the rational design of pharmaceutical agents. In the last years bioinformatics has experienced a great evolution due to the development of specialized software and to the increasing computer power. The codification of the structural information of molecules through molecular descriptors and the subsequent data analysis allow establishing QSAR models (Quantitative Structure-Activity Relationship) that can be applied to the design and the virtual screening of new drugs. The development of sophisticated Docking methodologies also allows a more accurate predict of the biological activity of molecules. Moreover, through this type of computational techniques and theoretical approaches, it is possible to develop explanatory hypothesis on the mechanism of action of drugs. This work provides a brief description of a series of studies implemented in the software MOE (Molecular Operating Environment) with particular attention to the medicinal chemistry aspects.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              NMR Chemical Shifts of Common Laboratory Solvents as Trace Impurities.

                Bookmark

                Author and article information

                Journal
                Drug Des Devel Ther
                Drug Des Devel Ther
                DDDT
                dddt
                Drug Design, Development and Therapy
                Dove
                1177-8881
                04 February 2020
                2020
                : 14
                : 497-508
                Affiliations
                [1 ]Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University , Sakaka, Aljouf 2014, Saudi Arabia
                [2 ]Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST) , Daejeon, Korea
                [3 ]Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University , Sohag 82524, Egypt
                [4 ]Department of Pharmacology, College of Pharmacy, Jouf University , Sakaka, Aljouf 2014, Saudi Arabia
                [5 ]Department of Biochemistry, Faculty of Pharmacy, Nahda University , Beni-Suef, Egypt
                [6 ]Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy , Palakkad, Kerala 678557, India
                [7 ]Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University , Assiut 71526, Egypt
                [8 ]Department of Pharmacology, College of Pharmacy, Jouf University , Sakakah 72341, Saudi Arabia
                [9 ]Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University , Kafrelsheikh, Egypt
                [10 ]Department of Pharmaceutical Organic Chemistry, Beni-Suef University , Beni-Suef 62514, Egypt
                [11 ]Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University , Kafrelsheikh, Egypt
                Author notes
                Correspondence: Mohammad M Al-Sanea Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University , Sakaka, Aljouf2014, Saudi ArabiaTel +966 594076460 Email mmalsanea@ju.edu.sa
                Author information
                http://orcid.org/0000-0002-6834-6548
                http://orcid.org/0000-0003-0393-1890
                http://orcid.org/0000-0001-9035-5638
                Article
                237957
                10.2147/DDDT.S237957
                7008064
                bc18562e-02b5-405b-96e2-a2a81667ca5e
                © 2020 Al-Sanea et al.

                This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License ( http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms ( https://www.dovepress.com/terms.php).

                History
                : 09 November 2019
                : 06 January 2020
                Page count
                Figures: 7, Tables: 4, References: 31, Pages: 12
                Categories
                Original Research

                Pharmacology & Pharmaceutical medicine
                hdacs inhibitors,ligustrazine,anticancer agents,in silico study,synthesis

                Comments

                Comment on this article