We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Novel quinazolinone-based 2,4-thiazolidinedione-3-acetic acid derivatives as potent aldose reductase inhibitors

    Kamel Metwally

    *Author for correspondence: Tel.: +20 101 499 2285;

    E-mail Address: kametwally@hotmail.com

    Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt

    ,
    Harris Pratsinis

    Laboratory of Cell Proliferation & Ageing, Institute of Biosciences & Applications, National Centre of Scientific Research ‘Demokritos’, Athens, Greece

    ,
    Dimitris Kletsas

    Laboratory of Cell Proliferation & Ageing, Institute of Biosciences & Applications, National Centre of Scientific Research ‘Demokritos’, Athens, Greece

    ,
    Luca Quattrini

    Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy

    ,
    Vito Coviello

    Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy

    ,
    Concettina La Motta

    **Author for correspondence: Tel.: +39 050 221 9593;

    E-mail Address: concettina.lamotta@unipi

    Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy

    ,
    Ahmed A El-Rashedy

    School of Health Sciences, University of KwaZulu-Natal, Westville, Durban 4001, South Africa

    &
    Mahmoud ES Soliman

    School of Health Sciences, University of KwaZulu-Natal, Westville, Durban 4001, South Africa

    Department of Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt

    Published Online:https://doi.org/10.4155/fmc-2017-0149

    Aim: Targeting aldose reductase enzyme with 2,4-thiazolidinedione-3-acetic acid derivatives having a bulky hydrophobic 3-arylquinazolinone residue. Materials & methods: All the target compounds were structurally characterized by different spectroscopic methods and microanalysis, their aldose reductase inhibitory activities were evaluated, and binding modes were studied by molecular modeling. Results: All the synthesized compounds proved to inhibit the target enzyme potently, exhibiting IC50 values in the nanomolar/low nanomolar range. Compound 5i (IC50 = 2.56 nM), the most active of the whole series, turned out to be almost 70-fold more active than the only marketed aldose reductase inhibitor epalrestat. Conclusion: This work represents a promising matrix for developing new potential therapeutic candidates for prevention of diabetic complications through targeting aldose reductase enzyme.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Diabetes Atlas. Gan D (Ed.). International Diabetes Federation, Brussels, Belgium (2003).
    • 2 Zimmet P, Alberti K, Shaw J. Global and societal implications of the diabetes epidemic. Nature 414, 782–787 (2001).
    • 3 Jain S, Saraf S. Type 2 diabetes mellitus – Its global prevalence and therapeutic strategies. Diabetes Metab. Syndr. Clin. Res. Rev. 4, 48–56 (2010).
    • 4 Bjork S. The cost of diabetes and diabetes care. Diabetes Res. Clin. Pract. 54, S13–S18 (2001). • Highlights the healthcare expenditure on managing diabetic complications.
    • 5 Lee AY, Chung SK, Chung SS. Demonstration that polyol accumulation is responsible for diabetic cataract by use of transgenic mice expressing the aldose reductase gene in the lens. Proc. Natl Acad. Sci. USA 92, 2780–2784 (1995).
    • 6 Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 414, 813–820 (2001).
    • 7 Srivastava S, Ramana K, Bhatnagar A. Role of aldose reductase and oxidative damage in diabetes and the consequent potential for therapeutic options. Endocr. Rev. 26, 380–392 (2005).
    • 8 Ramana K, Friedrich B, Srivastava S, Bhatnagar A, Srivastava S. Activation of nuclear factor-kappa B by hyperglycemia in vascular smooth muscle cells is regulated by aldose reductase. Diabetes 53, 2910–2920 (2004).
    • 9 Ramana K, Friedrich B, Tammali R, West M, Bhatnagar A, Srivastava S. Requirement of aldose reductase for the hyperglycemic activation of protein kinase C and formation of diacylglycerol in vascular smooth muscle cells. Diabetes 54, 818 (2005).
    • 10 Ramana K, Srivastava S. Mediation of aldose reductase in lipopolysaccharide-induced inflammatory signals in mouse peritoneal macrophages. Cytokine 36, 115–122 (2006).
    • 11 Tang W, Martin K, Hwa J. Aldose reductase, oxidative stress, and diabetic mellitus. Front Pharmacol. 3(87), 1–8 (2012).
    • 12 Costantino L, Rastelli G, Cignarella G, Vianello P, Barlocco D. New aldose reductase inhibitors as potential agents for the prevention of long-term diabetic complications. Exp. Opin. Ther. Patents 7, 843–858 (1997).
    • 13 Yabe-Nishimura C. Aldose reductase in glucose toxicity: a potential target for the prevention of diabetic complications. Pharmacol. Rev. 50, 21–33 (1998).
    • 14 Suzen S, Buyukbingol E. Recent studies of aldose reductase enzyme inhibition for diabetic complications. Curr. Med. Chem. 10, 1329–1352 (2003).
    • 15 Oates P. Polyol pathway and diabetic peripheral neuropathy. Int. Rev. Neurobiol. 50, 325–392 (2002).
    • 16 El-Kabbani O, Ruiz F, Darmanin C, Chung R. Aldose reductase structures: implications for mechanism and inhibition. Cell. Mol. Life Sci. 61, 750–762 (2004).
    • 17 Ishibashi Y, Matsui T, Matsumoto T, Kato H, Yamagishi S. Ranirestat has a stronger inhibitory activity on aldose reductase and suppresses inflammatory reactions in high glucose-exposed endothelial cells. Diab. Vasc. Dis. Res. 13, 312–315 (2016).
    • 18 Kao Y, Donahue K, Chan A, Knight J, Silink M. A novel polymorphism in the aldose reductase gene promoter region is strongly associated with diabetic retinopathy in adolescents with Type I diabetes. Diabetes 48, 1338–1340 (1999).
    • 19 Heesom A, Hibberd M, Millward A, Demaine A. Polymorphism in the 50-end of the aldose reductase gene is strongly associated with the development of diabetic nephropathy in Type I diabetes. Diabetes 46, 287–291 (1997).
    • 20 Moczulski D, Burak W, Doria A, Zychma M, Zukowska-Szczechowska E, Grzeszczak W. The role of aldose reductase gene in the susceptibility to diabetic nephropathy in Type II (non-insulin-dependent) diabetes mellitus. Diabetologia 42, 94–97 (1999).
    • 21 Oates P, Mylari B. Aldose reductase inhibitors: therapeutic implications for diabetic complications. Expert Opin. Inv. Drug 8, 1–15 (1999).
    • 22 Yamagishi M, Yamada Y, Ozaki K et al. Biological activities and quantitative structure–activity relationships of spiro[imidazolidine-4,4′(1′H)-quinazoline]-2,2′,5(3′H)-triones as aldose reductase inhibitors. J. Med. Chem. 35, 2085–2094 (1992).
    • 23 Kotani T, Nagaki Y, Ishii A et al. Highly selective aldose reductase inhibitors. 3. Structural diversity of 3-(arylmethyl)-2,4,5-trioxoimidazolidine-1-acetic acids. J. Med. Chem. 40, 684–694 (1997).
    • 24 Mylari B, Armento S, Beebe D et al. A novel series of non-carboxylic acid, non-hydantoin inhibitors of aldose reductase with potent oral activity in diabetic rat models: 6-(5-chloro-3-methylbenzofuran-2-sulfonyl)-2H-pyridazin-3-one and congeners. J. Med. Chem. 48, 6326–6339 (2005).
    • 25 Da Settimo F, Primofiore G, La Motta C et al. Naphtho[1, 2-d]isothiazoleacetic acid derivativesas a novelclass of selective aldose reductase inhibitors. J. Med. Chem. 48, 6897–6907 (2005).
    • 26 La Motta C, Sartini S, Salerno S et al. Acetic acid aldose reductase inhibitors bearing a five-membered heterocyclic core with potent topical activity in a visual impairment rat model. J. Med. Chem. 51, 3182–3193 (2008).
    • 27 Van Zandt M, Doan B, Sawicki D, Sredy J, Podjarny A. Discovery of [3-(4,5,7-trifluoro-benzothiazol-2-ylmethyl)-pyrrolo[2,3-b]pyridin-1-yl]acetic acids as highly potent and selective inhibitors of aldose reductase for treatment of chronic diabetic complications. Bioorg. Med. Chem. Lett. 19, 2006–2008 (2009).
    • 28 Ohmura C, Watada H, Azuma K et al. Aldose reductase inhibitor, epalrestat, reduces lipid hydroperoxides in Type 2 diabetes. Endocr. J. 56, 149–156 (2009).
    • 29 Del Corso A, Cappiello M, Mura U. From a dull enzyme to something else: facts and perspectives regarding aldose reductase. Curr. Med. Chem. 15, 1452–1461 (2008).
    • 30 Stefek M, Soltesova P, Majekova M, Rechlin C, Heine A, Klebe G. Identification of novel aldose reductase inhibitors based on carboxymethylated mercaptotriazinoindole scaffold. J. Med. Chem. 58, 2649–2657 (2015).
    • 31 Maccari R, Ottanà R. Targeting aldose reductase for the treatment of diabetes complications and inflammatory diseases: new insights and future directions. J. Med. Chem. 58, 2047–2067 (2015). •• Reviews the implication of aldose reductase inhibitors in both diabetic complications and inflammatory disorders.
    • 32 Chatzopoulou M, Pegklidou K, Papastavrou N, Demopoulos V. Development of aldose reductase inhibitors for the treatment of inflammatory disorders. Expert Opin. Drug Discov. 8, 1365–1380 (2013).
    • 33 Grewal A, Bhardwaj S, Pandita D, Lather V, Sekhon B. Updates on aldose reductase inhibitors for management of diabetic complications and non-diabetic diseases. Mini Rev. Med. Chem. 16, 120–162 (2016).
    • 34 Banditelli S, Boldrini E, Vilardo P et al. A new approach against sugar cataract through aldose reductase Inhibitors. Exp. Eye Res. 69, 533–538 (1999).
    • 35 Hotta N, Toyota T, Matsuoka K et al. Clinical efficacy of fidarestat, a novel aldose reductase inhibitor, for diabetic peripheral neuropathy. Diabetes Care 24, 1776–1782 (2001).
    • 36 Asano T, Saito Y, Kawakami M, Yamada N. Fidarestat (SNK-860) a potent aldose reductase inhibitor, normalizes the elevated sorbitol accumulation in erythrocytes of diabetic patients. J. Diabet. Complicat. 16, 133–138 (2002).
    • 37 Okayama N, Omi H, Okouchi M et al. Mechanisms of inhibitory activity of the aldose reductase inhibitor, Epalrestat, on high glucose-mediated endothelial injury: neutrophil-endothelial cell adhesion and surface expression of endothelial adhesion molecules. J. Diabet. Complicat. 16, 321–326 (2002).
    • 38 Nakahara M, Miyata K, Otani S et al. A randomised placebo controlled clinical trial of the aldose reducatse inhibitor CT-112 as management of corneal epithelial disorders in diabetic patients. Br. J. Ophthalm. 89, 266–268 (2005).
    • 39 Hotta N, Akanuma Y, Kawamori R et al. Long-term clinical effects of epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy. Diabetes Care 29, 1538–1544 (2006). • Highlights the mild and tolerable side effects of epalrestat.
    • 40 Oyama T, Miyasita Y, Watanabe H, Shirai K. The role of polyol pathway in high glucose-induced endothelial cell damages. Diabetes Res. Clin. Pract. 73, 227–234 (2006).
    • 41 Lorenzi M. The polyol pathway as a mechanism for diabetic retinopathy: attractive, elusive, and resilient. Exp. Diabetes Res. 2007, 61038 (2007).
    • 42 Matsumoto T, Ono Y, Kurono M, Kuromiya A, Nakamura K, Bril V. Ranirestat (AS-3201) a potent aldose reductase inhibitor, reduces sorbinil levels and improves motor nerve conduction velocity in streptozotocin-diabetes rats. J. Pharmacol. Sci. 107, 231–237 (2008).
    • 43 Schemmel K, Padiyara R, D'Souza J. Aldose reductase inhibitors in the treatment of diabetic peripheral neuropathy: a review. J. Diabet. Complicat. 24, 354–360 (2010).
    • 44 Bozdağ-Dündar O, Evranos B, Daş-Evcimen N, Sarikaya M, Ertan R. Synthesis and aldose reductase inhibitory activity of some new chromonyl-2,4-thiazolidinediones. Eur. J. Med. Chem. 43, 2412–2417 (2008).
    • 45 Daş-Evcimen N, Bozdağ-Dündar O, Sarikaya M, Ertan R. In vitro aldose reductase inhibitory activity of some flavonyl-2,4-thiazolidinediones. J. Enzyme Inhib. Med. Chem. 3, 297–301 (2008).
    • 46 Bozdag-Dundar O, Evcimen M, Ceylan-Unlusoy M, Rahmiye E, Mutlu S. Some new thiazolyl thiazolidinedione derivatives as aldose reductase inhibitors. Med. Chem. Res. 16, 39–47 (2007).
    • 47 Sambasivarao S, Soni L, Gupta A, Hanumantharao P, Kaskhedikar S. Quantitative structure–activity analysis of 5-arylidene-2,4-thiazolidinediones as aldose reductase inhibitors. Bioorg. Med. Chem. Lett. 16, 512–520 (2006).
    • 48 Maccari R, Ottanà R, Curinga C et al. Structure–activity relationships and molecular modelling of 5-arylidene-2,4-thiazolidinediones active as aldose reductase inhibitors. Bioorg. Med. Chem. 13, 2809–2823 (2005).
    • 49 Rakowitz D, Maccari R, Ottanà R, Vigorita M. In vitro aldose reductase inhibitory activity of 5-benzyl-2,4-thiazolidinediones. Bioorg. Med. Chem. 14, 567–574 (2006).
    • 50 Maccari R, Ottanà R, Ciurleo R et al. Synthesis, induced-fit docking investigations, and in vitro aldose reductase inhibitory activity of non-carboxylic acid containing 2,4-thiazolidinedione derivatives. Bioorg. Med. Chem. 16, 5840–5852 (2008).
    • 51 Maccari R, Ciurleo R, Giglio M et al. Identification of new non-carboxylic acid containing inhibitors of aldose reductase. Bioorg. Med. Chem. 18, 4049–4055 (2010).
    • 52 Maccari R, Ottanà R, Ciurleo R et al. Evaluation of in vitro aldose redutase inhibitory activity of 5-arylidene-2,4-thiazolidinediones. Bioorg. Med. Chem. Lett. 17, 3886–3893 (2007).
    • 53 Zentgraf M, Steuber H, Koch C et al. How reliable are current docking approaches for structure-based drug design? Lessons from aldose reductase. Angew. Chem. Int. Ed. Engl. 46, 3575–3578 (2007).
    • 54 Steuber H, Zentgraf M, La Motta C, Sartini S, Heine A, Klebe G. Evidence for a novel binding site conformer of aldose reductase in ligand-bound state. J. Mol. Biol. 369, 186–197 (2007).
    • 55 Metwally K, Pratsinis H, Kletsas D. Novel 2, 4-thiazolidinediones: synthesis, in vitro cytotoxic activity, and mechanistic investigation. Eur. J. Med. Chem. 133, 340–350 (2017).
    • 56 Del-Corso A, Balestri F, Di Bugno E et al. A new approach to control the enigmatic activity of aldose reductase. PLoS ONE 8, e74076 (2013).
    • 57 Ramunno A, Cosconati S, Sartini S et al. Progresses in the pursuit of aldose reductase inhibitors: the structure-based lead optimization step. Eur. J. Med. Chem. 5, 216–26 (2012).
    • 58 Sartini S, Cosconati S, Marinelli L et al. Benzofuroxane derivatives as multi-effective agents for the treatment of cardiovascular diabetic complications. Synthesis, functional evaluation, and molecular modeling studies. J. Med. Chem. 55, 10523–10531 (2012).
    • 59 Trott O, Olson A. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455–461 (2010).
    • 60 Morris G, Huey R, Lindstrom W et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J. Comput. Chem. 30, 2785–2791 (2009).
    • 61 Howard E, Sanishvili R, Cachau R et al. Ultrahigh resolution drug design I: details of interactions in human aldose reductase-inhibitor complex at 0.66 A. Proteins 55, 792–804 (2004).
    • 62 Van Zandt M, Sibley E, McCann E et al. Design and synthesis of highly potent and selective (2-arylcarbamoyl-phenoxy)-acetic acid inhibitors of aldose reductase for treatment of chronic diabetic complications. Bioorg. Med. Chem. 12, 5661–5675 (2004).
    • 63 Wang L, Gu Q, Zheng X et al. Discovery of new selective human aldose reductase inhibitors through virtual screening multiple binding pocket conformations. J. Chem. Inf. Model 53, 2409–2422 (2013).
    • 64 Cosconati S, Forli S, Perryman A, Harris R, Goodsell D, Olson A. Virtual screening with AutoDock: theory and practice. Expert Opin. Drug Discov. 5, 597–607 (2010).
    • 65 Sanner M. The python interpreter as a framework for integrating scientific computing software-components. Scripps Res. Inst. 26, 1–12 (2008).
    • 66 Case D, Cheatham T, Darden T et al. The amber biomolecular simulation programs. J. Comput. Chem. 26, 1668–1688 (2005).
    • 67 Lindorff-Larsen K, Piana S, Palmo K et al. Improved side-chain torsion potentials for the Amber ff99SB protein force field. Proteins 78, 1950–1958 (2010).
    • 68 Hanwell M, Curtis D, Lonie D, Vandermeersch T, Zurek E, Hutchison G. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J. Chem. inform. 4, 17 (2012).
    • 69 Wang J, Wolf R, Caldwell J, Kollman P, Case D. Development and testing of a general amber force field. J. Comput. Chem. 25, 1157–1174 (2004).
    • 70 Jorgensen W, Chandrasekhar J, Madura J, Impey R, Klein M. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).
    • 71 Essmann U, Perera L, Berkowitz M, Darden T, Lee H, Pedersen L. A smooth particlemesh Ewald method. J. Chem. Phys. 103, 8577–8593 (1995).
    • 72 Grest G, Kremer K. Molecular dynamics simulation for polymers in the presence of a heat bath. Phys. Rev. A Gen. Phys. 33, 3628–3631 (1986).
    • 73 Berendsen H, Postma J, van Gunsteren W, DiNola A, Haak J. Molecular dynamics with coupling to an external bath. J. Chem. Phys. 81, 3684–3690 (1984).
    • 74 Ryckaert J, Ciccotti G, Berendsen H. Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes. J. Comput. Phys. 23, 327–341 (1977).
    • 75 Pettersen E, Goddard T, Huang C et al. UCSF Chimera–a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).
    • 76 Wriggers W, Schulten K. Protein domain movements: detection of rigid domains and visualization of hinges in comparisons of atomic coordinates. Proteins 29, 1–14 (1997).
    • 77 Laskowski R, Swindells M. LigPlot+: multiple ligand-protein interaction diagrams for drug discovery. J. Chem. Inf. Model 51, 2778–2786 (2011).
    • 78 Kollman A, Massova I, Reyes C et al. Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models. Acc. Chem. Res. 33, 889–897 (2000).
    • 79 Tsui V, Case D. Theory and applications of the generalized born solvation model in macromolecular simulations. Biopolymers 56, 275–291 (2000).
    • 80 Onufriev A, Bashford D, Case D. Modification of the generalized born model suitable for macromolecules. J. Phys. Chem. B 104, 3712–3720 (2000).
    • 81 Domanig F. Synthese und einige Reaktionen yon 2-Azidomethyl-3-aryl-4-chinazolinonen. Monatshefte fur Chemie 112, 1195–1202 (1981).
    • 82 Fetter J, Czuppon T, Hornyak G, Feller A. The synthesis of some 3-amino-2-halomethyl-, 2- halomethyl-3-(subst.amino)- and 2-halomethyl-3-hetarylquinazolin-4(3)-ones as potential plant protecting agents. Tetrahedron 47, 9393–9410 (1991).
    • 83 Tani J, Yamada Y, Oine T, Ochiai T, Ishida R, Inoue I. Studies on biologically active halogenated compounds. 1. Synthesis and central nervous system depressant activity of 2- (fluoromethyl)-3-aryl-4(3H)-quinazolinone derivatives. J. Med. Chem. 22, 95–99 (1979).
    • 84 Giri R, Thaker H, Giordano T et al. Design, synthesis and evaluation of novel 2- thiophen-5-yl-3H-quinazolin-4-one analogues as inhibitors of transcription factors NfkappaB and AP-1 mediated transcriptional activation: their possible utilization as antiinflammatory and anti-cancer agents. Bioorg. Med. Chem. 18, 2796–2808 (2010).
    • 85 Hymavati V, Sobhia M. Implication of crystal water molecules in inhibitor binding at ALR2 active site. Comput. Math. Methods Med. 2012, 541594 (2012).
    • 86 Adcock S, McCammon J. Molecular dynamics: survey of methods for simulating the activity of proteins. Chem. Rev. 106, 1589–1615 (2006).