1,961
Views
13
CrossRef citations to date
0
Altmetric
Short Communication

Newer human inosine 5′-monophosphate dehydrogenase 2 (hIMPDH2) inhibitors as potential anticancer agents

ORCID Icon & ORCID Icon
Pages 972-977 | Received 20 Dec 2017, Accepted 02 May 2018, Published online: 24 May 2018

References

  • Jackson RC, Weber G, Morris HP. IMP dehydrogenase, an enzyme linked with proliferation and malignancy. Nature 1975;256:331–3.
  • Hedstrom L. IMP dehydrogenase: structure, mechanism, and inhibition. Chem Rev 2009;109:2903–28.
  • Jayaram HN, Dion RL, Glazer RI, et al. Initial studies on the mechanism of action of a new oncolytic thiazole nucleoside, 2-β-D-ribofuranosylthiazole-4-carboxamide (NSC 286193). Biochem Pharmacol 1982;31:2371–80.
  • Manzoli L, Billi AM, Gilmour RS, et al. Phosphoinositide signaling in nuclei of friend cells: tiazofurin down-regulates phospholipase C beta 1. Cancer Res 1995;55:2978–80.
  • Mandanas BRA, Leibowitz DS, Gharehbaghi K, et al. Role of p21 RAS in p210 bcr-abl transformation of murine myeloid cells. Blood 1993;82:1838–47.
  • Kharbanda SM, Sherman ML, Kufe DW. Effects of tiazofurin on guanine nucleotide binding regulatory proteins in HL-60 cells. Blood 1990;75:583–8.
  • Vitale M, Zamai L, Falcieri E, et al. IMP dehydrogenase inhibitor, tiazofurin, induces apoptosis in K562 human erythroleukemia cells. Commun Clin Cytom 1997;30:61–6.
  • Kim HR, Roe JS, Lee JE, et al. A p53-inducible microRNA-34a downregulates Ras signaling by targeting IMPDH. Biochem Biophys Res Commun 2012;418:682–8.
  • Messina E, Gazzaniga P, Micheli V, et al. Guanine nucleotide depletion triggers cell cycle arrest and apoptosis in human neuroblastoma cell lines. Int J Cancer 2004;108:812–7.
  • Konno Y, Natsumedasq Y, Nagai M, et al. Expression of human IMP dehydrogenase types I and I1 in Escherichia coli and distribution in human normal lymphocytes and leukemic cell lines. J Biol Chem 1991;266:506–9.
  • Chong CR, Qian DZ, Pan F, et al. Identification of type 1 inosine monophosphate dehydrogenase as an antiangiogenic drug target. J Med Chem 2006;49:2677–80.
  • Petrelli R, Vita P, Torquati I, et al. Novel inhibitors of inosine monophosphate dehydrogenase in patent literature of the last decade. Recent Pat Anticancer Drug Discov 2013;8:103–25.
  • Olah E, Kokeny S, Papp J, et al. Modulation of cancer pathways by inhibitors of guanylate metabolism. Adv Enzyme Regul 2006;46:176–90.
  • Bentley R. Mycophenolic acid: a one hundred year odyssey from antibiotic to immunosuppressant. Chem Rev 2000;100:3801–25.
  • Knight SR, Morris PJ. Does the evidence support the use of mycophenolate mofetil therapeutic drug monitoring in clinical practice? A systematic review. Transplantation 2008;85:1675–85.
  • Nair V, Shu Q. Inosine monophosphate dehydrogenase as a probe in antiviral drug discovery. Antivir Chem Chemother 2007;18:245–58.
  • Shah CP, Kharkar PS. Inosine 5'-monophosphate dehydrogenase inhibitors as antimicrobial agents: recent progress and future perspectives. Future Med Chem 2015;7:1415–29.
  • Shu Q, Nair V. Inosine monophosphate dehydrogenase (IMPDH) as a target in drug discovery. Med Res Rev 2008;28:219–32.
  • Pankiewicz KW, Petrelli R, Singh R, Felczak K. Nicotinamide adenine dinucleotide based therapeutics, update. Curr Med Chem 2015;22:3991–4028.
  • Cuny GD, Suebsuwong C, Ray SS. Inosine-5’-monophosphate dehydrogenase (IMPDH) inhibitors: a patent and scientific literature review (2002–2016). Expert Opin Ther Pat 2017;27:677–90.
  • Cholewinski G, Iwaszkiewicz-Grzes D, Trzonkowski P, Dzierzbicka K. Synthesis and biological activity of ester derivatives of mycophenolic acid and acridines/acridones as potential immunosuppressive agents. J Enzyme Inhib Med Chem 2016;31:974–82.
  • Chen L, Petrelli R, Olesiak M, et al. Bis (sulfonamide) isosters of mycophenolic adenine dinucleotide analogues: inhibition of inosine monophosphate dehydrogenase. Bioorganic Med Chem 2008;16:7462–9.
  • Dunkern T, Chavan S, Bankar D, et al. Design, synthesis and biological evaluation of novel inosine 5′-monophosphate dehydrogenase (IMPDH) inhibitors. J Enzyme Inhib Med Chem 2014;29:408–19.
  • Dunkern T, Prabhu A, Kharkar PS, et al. Virtual and experimental high-throughput screening (HTS) in search of novel inosine 5′-monophosphate dehydrogenase II (IMPDH II) inhibitors. J Comput Aided Mol Des 2012;26:1277–92.
  • Cholewiński G, Iwaszkiewicz-Grześ D, Prejs M, et al. Synthesis of the inosine 5′-monophosphate dehydrogenase (IMPDH) inhibitors. J Enzyme Inhib Med Chem 2015;30:550–63.
  • Chen L, Wilson DJ, Labello NP, et al. Mycophenolic acid analogs with a modified metabolic profile. Bioorganic Med Chem 2008;16:9340–5.
  • Jones DF, Mills SD. Preparation and antitumor properties of analogs and derivatives of mycophenolic acid. J Med Chem 1971;14:305–11.
  • Suzui S, Takaku S, Mori T. Antitumor activity of derivatives of mycophenolic acid. J Antibiot 1976;29:275–85.
  • Dzierzbicka K, Kołodziejczyk AM. Synthesis and antitumor activity of conjugates of muramyldipeptide or normuramyldipeptide with hydroxyacridine/acridone derivatives. J Med Chem 2003;46:183–9.
  • Malachowska-Ugarte M, Cholewinski G, Dzierzbicka K, Trzonkowski P. Synthesis and biological activity of novel mycophenolic acid conjugates containing nitro-acridine/acridone derivatives. Eur J Med Chem 2012;54:197–201.
  • Iwaszkiewicz-Grzes D, Cholewinski G, Kot-Wasik A, et al. Synthesis and biological activity of mycophenolic acid-amino acid derivatives. Eur J Med Chem 2013;69:863–71.
  • Domhan S, Muschal S, Schwager C, et al. Molecular mechanisms of the antiangiogenic and antitumor effects of mycophenolic acid. Mol Cancer Ther 2008;7:1656–68.
  • Muraoka T, Ide M, Irie M, et al. Development of a method for converting a TAK1 type I inhibitor into a type II or c-helix-out inhibitor by structure-based drug design (SBDD). Chem Pharm Bull 2016;64:1622–9.
  • Gudipati R, Reddy Anreddy RN, Manda S. Synthesis, anticancer and antioxidant activities of some novel N-(benzo [d]oxazol-2-yl)-2-(7-or 5-substituted-2-oxoindolin-3-ylidene) hydrazinecarboxamide derivatives. J Enzyme Inhib Med Chem 2011;26:813–8.
  • Kharkar P. Drugs acting on central nervous system (CNS) targets as leads for non-CNS targets. F1000Res 2014;3:40.