Generic placeholder image

Current Bioinformatics

Editor-in-Chief

ISSN (Print): 1574-8936
ISSN (Online): 2212-392X

Review Article

Bioinformatics Approach on Bioisosterism Softwares to be Used in Drug Discovery and Development

Author(s): Nelson José Freitas da Silveira*, Walter Filgueira de Azevedo, Rita Cardoso Guedes, Leandro Marcos Santos, Rodolfo Cabral Marcelino, Patrícia da Silva Antunes and Thiago Castilho Elias

Volume 17, Issue 1, 2022

Published on: 25 May, 2021

Page: [19 - 30] Pages: 12

DOI: 10.2174/1574893616666210525150747

Price: $65

Abstract

Background: In the rational drug development field, bioisosterism is a tool that improves lead compounds' performance, referring to molecular fragment substitution that has similar physical-chemical properties. Thus, it is possible to modulate drug properties such as absorption, toxicity, and half-life increase. This modulation is of pivotal importance in the discovery, development, identification, and interpretation of the mode of action of biologically active compounds.

Objective: Our purpose here is to review the development and application of bioisosterism in drug discovery. In this study history, applications, and use of bioisosteric molecules to create new drugs with high binding affinity in the protein-ligand complexes are described.

Methods: It is an approach for molecular modification of a prototype based on the replacement of molecular fragments with similar physicochemical properties, being related to the pharmacokinetic and pharmacodynamic phase, aiming at the optimization of the molecules.

Results: Discovery, development, identification, and interpretation of the mode of action of biologically active compounds are the most important factors for drug design. The strategy adopted for the improvement of leading compounds is bioisosterism.

Conclusion: Bioisosterism methodology is a great advance for obtaining new analogs to existing drugs, enabling the development of new drugs with reduced toxicity, in a comparative analysis with existing drugs. Bioisosterism has a wide spectrum to assist in several research areas.

Keywords: Protein-ligand interaction, bioisosterism, computational chemistry, medicinal chemistry, structural bioinformatics, pharmacokinetic.

Graphical Abstract
[1]
Srivastava P, Tiwari A. Critical role of computer simulations in drug discovery and development. Curr Top Med Chem 2017; 17(21): 2422-32.
[http://dx.doi.org/10.2174/1568026617666170403113541] [PMID: 28366137]
[2]
Freitas PG, Elias TC, Pinto IA, et al. Computational approach to the discovery of phytochemical molecules with therapeutic potential targets to the PKCZ protein. Lett Drug Des Discov 2017; 14: 1-12.
[3]
Yu W, Mackerell ADJ. Computer-Aided Drug Design Methods Methods in Molecular Biology. Humana Press Inc. 2017; Vol. 1520: pp. 85-106.
[4]
Moretti L, Sartori L. Software infrastructure for computer-aided drug discovery and development, a practical example with guidelines. Mol Inform 2016; 35(8-9): 382-90.
[http://dx.doi.org/10.1002/minf.201501037] [PMID: 27546042]
[5]
Wang Y, Du Y, Huang N. A survey of the role of nitrile groups in protein-ligand interactions. Future Med Chem 2018; 10(23): 2713-28.
[http://dx.doi.org/10.4155/fmc-2018-0252] [PMID: 30518255]
[6]
Nagarajan N, Yapp EKY, Le NQK, Yeh HY. In silico screening of sugar alcohol compounds to inhibit viral matrix protein VP40 of Ebola virus. Mol Biol Rep 2019; 46(3): 3315-24.
[http://dx.doi.org/10.1007/s11033-019-04792-w] [PMID: 30982214]
[7]
Nagarajan N, Yapp EKY, Le NQK, Kamaraj B, Al-Subaie AM, Yeh HY. Application of computational biology and artificial intelligence technologies in cancer precision drug discovery. BioMed Res Int 2019; 20198427042
[8]
Tsui V, Ortwine DF, Blaney JM. Enabling drug discovery project decisions with integrated computational chemistry and informatics. J Comput Aided Mol Des 2017; 31(3): 287-91.
[http://dx.doi.org/10.1007/s10822-016-9988-y] [PMID: 27796615]
[9]
Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res 2017; 45(D1): D353-61.
[http://dx.doi.org/10.1093/nar/gkw1092] [PMID: 27899662]
[10]
Croft RA, Mousseau JJ, Choi C, Bull JA. Lithium-catalyzed thiol alkylation with tertiary and secondary alcohols: synthesis of 3-sulfanyl-oxetanes as bioisosteres. Chemistry 2018; 24(4): 818-21.
[http://dx.doi.org/10.1002/chem.201705576] [PMID: 29181870]
[11]
Luty B, Rose PW. The need for scientific software engineering in the pharmaceutical industry. J Comput Aided Mol Des 2017; 31(3): 301-4.
[http://dx.doi.org/10.1007/s10822-016-9997-x] [PMID: 27995514]
[12]
Case DA, Cheatham TE III, Darden T, et al. The Amber biomolecular simulation programs. J Comput Chem 2005; 26(16): 1668-88.
[http://dx.doi.org/10.1002/jcc.20290] [PMID: 16200636]
[13]
Hess B, Kutzner C, van der Spoel D, Lindahl E. GROMACS 4:Algorithms for highly efficient, Load-balanced, and scalable molecular simulation. J Chem Theory Comput 2008; 4(3): 435-47.
[http://dx.doi.org/10.1021/ct700301q] [PMID: 26620784]
[14]
Labute P, Williams C, Feher M, Sourial E, Schmidt JM. Flexible alignment of small molecules. J Med Chem 2001; 44(10): 1483-90.
[http://dx.doi.org/10.1021/jm0002634] [PMID: 11334559]
[15]
Dixon SL, Smondyrev AM, Knoll EH, Rao SN, Shaw DE, Friesner RA. PHASE: a new engine for pharmacophore perception, 3D QSAR model development, and 3D database screening: 1. Methodology and preliminary results. J Comput Aided Mol Des 2006; 20(10-11): 647-71.
[http://dx.doi.org/10.1007/s10822-006-9087-6] [PMID: 17124629]
[16]
Steinbeck C, Han Y, Kuhn S, Horlacher O, Luttmann E, Willighagen E. The Chemistry Development Kit (CDK): an open-source Java library for Chemo- and Bioinformatics. J Chem Inf Comput Sci 2003; 43(2): 493-500.
[http://dx.doi.org/10.1021/ci025584y] [PMID: 12653513]
[17]
Burger A. Isosterism and bioisosterism in drug design. Prog Drug Res 1991; 37: 287-371.
[PMID: 1763185]
[18]
Meanwell NA. Fluorine and fluorinated motifs in the design and application of bioisosteres for drug design. J Med Chem 2018; 61(14): 5822-80.
[http://dx.doi.org/10.1021/acs.jmedchem.7b01788] [PMID: 29400967]
[19]
Mugnaini C, Pasquini S, Corelli F. The bioisosteric concept applied to cannabinoid ligands. Curr Med Chem 2012; 19(28): 4794-815.
[http://dx.doi.org/10.2174/092986712803341575] [PMID: 22830341]
[20]
Lin Y, Chen Z-Y, Yang F, et al. Application of molecular imaging technologies in antitumor drug development and therapy. Curr Pharm Des 2015; 21(16): 2136-46.
[http://dx.doi.org/10.2174/1381612821666150109122915] [PMID: 25578891]
[21]
Barreiro EJ, Fraga CAM. Bioisosterism as a Strategy for Planning, Design, Molecular Modification and Optimization of Binders and Prototype Compounds. In: Medicinal chemistry: The molecular basis of drugs action Porto Alegre. 2015; p. 30.
[22]
Jampilek J. Heterocycles in medicinal chemistry. Molecules 2019; 24(21): 3839.
[http://dx.doi.org/10.3390/molecules24213839] [PMID: 31731387]
[23]
Hevey R. Bioisosteres of carbohydrate functional groups in glycomimetic design. Biomimetics (Basel) 2019; 4(3): 53.
[http://dx.doi.org/10.3390/biomimetics4030053] [PMID: 31357673]
[24]
Langmuir I. Isomorphism, isosterism and covalence. J Am Chem Soc 1919; 41: 1543-59.
[http://dx.doi.org/10.1021/ja02231a009]
[25]
Tseng CC, Baillie G, Donvito G, et al. The trifluoromethyl group as a bioisosteric replacement of the aliphatic nitro group in CB1 receptor positive allosteric modulators. J Med Chem 2019; 62(10): 5049-62.
[http://dx.doi.org/10.1021/acs.jmedchem.9b00252] [PMID: 31050898]
[26]
Barreiro EJ. Química medicinal: As bases moleculares da ação dos fármacos. 3rd ed. Porto Alegre: ARTMED 2015.
[27]
van Wijngaarden I, Kruse CG, van Hes R, van der Heyden JAM, Tulp MT. 2-Phenylpyrroles as conformationally restricted benzamide analogues. A new class of potential antipsychotics. 1. J Med Chem 1987; 30(11): 2099-104.
[http://dx.doi.org/10.1021/jm00394a028] [PMID: 2889830]
[28]
Loftsson T. Drug stability for pharmaceutical scientists. Oxford: Academic Press 2014.
[29]
Meng X-Y, Zhang H-X, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des 2011; 7(2): 146-57.
[http://dx.doi.org/10.2174/157340911795677602] [PMID: 21534921]
[30]
Duschinsky R, Pleven E, Heidelberger C. The synthesis of 5- fluoropyrimidines. J Am Chem Soc 1957; 79: 4559-60.
[http://dx.doi.org/10.1021/ja01573a087]
[31]
Kirk KL. Selective fluorination in drug design and development: an overview of biochemical rationales. Curr Top Med Chem 2006; 6(14): 1447-56.
[http://dx.doi.org/10.2174/156802606777951073] [PMID: 16918460]
[32]
Heidelberger C, Chaudhuri NK, Danneberg P, et al. Fluorinated pyrimidines, a new class of tumour-inhibitory compounds. Nature 1957; 179(4561): 663-6.
[http://dx.doi.org/10.1038/179663a0] [PMID: 13418758]
[33]
Binder D, Hromatka O, Geissler F, et al. Analogues and derivatives of tenoxicam. 1. Synthesis and antiinflammatory activity of analogues with different residues on the ring nitrogen and the amide nitrogen. J Med Chem 1987; 30(4): 678-82.
[http://dx.doi.org/10.1021/jm00387a017] [PMID: 3494124]
[34]
Ruetsch YA, Böni T, Borgeat A. From cocaine to ropivacaine: the history of local anesthetic drugs. Curr Top Med Chem 2001; 1(3): 175-82.
[http://dx.doi.org/10.2174/1568026013395335] [PMID: 11895133]
[35]
Mykhailiuk PK. Saturated bioisosteres of benzene: where to go next? Org Biomol Chem 2019; 17(11): 2839-49.
[http://dx.doi.org/10.1039/C8OB02812E] [PMID: 30672560]
[36]
Lin P, Marino D, Lo JL, et al. 2-(3,5-Dimethylphenyl)tryptamine derivatives that bind to the GnRH receptor. Bioorg Med Chem Lett 2001; 11(8): 1073-6.
[http://dx.doi.org/10.1016/S0960-894X(01)00134-2] [PMID: 11327593]
[37]
Kim KS, Kimball SD, Misra RN, et al. Discovery of aminothiazole inhibitors of cyclin-dependent kinase 2: synthesis, X-ray crystallographic analysis, and biological activities. J Med Chem 2002; 45(18): 3905-27.
[http://dx.doi.org/10.1021/jm0201520] [PMID: 12190313]
[38]
Swapna GVT, Jagannadh B, Gurjar MK, Kunwar AC. NMR investigation on the structure and conformation of 3′-azido-2′,3′-dideoxyribosylthymine (AZT), an inhibitor of the HIV (AIDS virus). Biochem Biophys Res Commun 1989; 164(3): 1086-92.
[http://dx.doi.org/10.1016/0006-291X(89)91780-4] [PMID: 2590188]
[39]
Vital DG, Damasceno FS, Rapado LN, et al. Application of bioisosterism in design of the semicarbazone derivatives as cruzain inhibitors: a theoretical and experimental study. J Biomol Struct Dyn 2017; 35(6): 1244-59.
[http://dx.doi.org/10.1080/07391102.2016.1176603] [PMID: 27064715]
[40]
Bentler P, Bergander K, Daniliuc CG, et al. Inverting small molecule-protein recognition by the fluorine Gauche effect: Selectivity regulated by multiple H→F bioisosterism. Angew Chem Int Ed Engl 2019; 58(32): 10990-4.
[http://dx.doi.org/10.1002/anie.201905452] [PMID: 31157945]
[41]
Ramu D, Jain R, Kumar RR, et al. Design and synthesis of imidazolidinone derivatives as potent anti-leishmanial agents by bioisosterism. Arch Pharm (Weinheim) 2019; 352(4)e1800290
[http://dx.doi.org/10.1002/ardp.201800290] [PMID: 30801775]
[42]
Kang D, Feng D, Sun Y, et al. Structure-based bioisosterism Yields HIV-1 NNRTIs with improved drug-resistance profiles and favorable pharmacokinetic properties. J Med Chem 2020; 63(9): 4837-48.
[http://dx.doi.org/10.1021/acs.jmedchem.0c00117] [PMID: 32293182]
[43]
Khanim F, Davies N, Veliça P, et al. Selective AKR1C3 inhibitors do not recapitulate the anti-leukaemic activities of the pan-AKR1C inhibitor medroxyprogesterone acetate. Br J Cancer 2014; 110(6): 1506-16.
[http://dx.doi.org/10.1038/bjc.2014.83] [PMID: 24569460]
[44]
Liu R, Li X, Lam KS. Combinatorial chemistry in drug discovery. Curr Opin Chem Biol 2017; 38: 117-26.
[45]
Pandeya SN, Thakkar D. Combinatorial chemistry: A novel method in drug discovery and its application. Indian J Chem Nova Deli 2005; 44B: 335-48.
[46]
Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 2002; 1(7): 493-502.
[http://dx.doi.org/10.1038/nrd839] [PMID: 12120256]
[47]
Barreiro EJ, Fraga CAM. Química Medicinal: As bases moleculares da ação dos fármacos 2. Porto Alegre: Artmed 2008.
[48]
Kim KS, Kimball SD, Misra RN, et al. Discovery of aminothiazole inhibitors of cyclin-dependent kinase 2: synthesis, X-ray crystallographic analysis, and biological activities. J Med Chem 2002; 45(18): 3905-27.
[http://dx.doi.org/10.1021/jm0201520] [PMID: 12190313]
[49]
Wirth M, Zoete V, Michielin O, Sauer WHB. SwissBioisostere: a database of molecular replacements for ligand design. Nucleic Acids Res 2013; 41(Database issue): D1137-43.
[http://dx.doi.org/10.1093/nar/gks1059] [PMID: 23161688]
[50]
Lešnik S, Škrlj B, Eržen N, et al. BoBER: web interface to the base of bioisosterically exchangeable replacements. J Cheminform 2017; 9(1): 62.
[http://dx.doi.org/10.1186/s13321-017-0251-x] [PMID: 29234984]
[51]
Elias TC, de Oliveira HCB, da Silveira NJF. MB-Isoster: A software for bioisosterism simulation. J Comput Chem 2018; 39(29): 2481-7.
[http://dx.doi.org/10.1002/jcc.25581] [PMID: 30318630]
[52]
Shan J, Ji C. MolOpt: a web server for drug design using bioisosteric transformation. Curr Comput Aided Drug Des 2020; 16(4): 460-6.
[53]
Gaulton A, Bellis LJ, Bento AP, et al. ChEMBL: A large-scale bioactivity database for drug discovery. Nucleic Acids Res 2011; 40.
[PMID: 21948594]
[54]
Morris GM, Huey R, Lindstrom W, et al. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J Comput Chem 2009; 30(16): 2785-91.
[http://dx.doi.org/10.1002/jcc.21256] [PMID: 19399780]
[55]
Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31(2): 455-61.
[PMID: 19499576]
[56]
Coelho CM, Santos T, Freitas PG, et al. Design, synthesis, biological evaluation and molecular modeling studies of novel eugenol esters as leishmanicidal agents. J Braz Chem Soc 2018; 29: 715-28.
[57]
Freitas PG, Castilho TE, Almeida L, et al. An in silico study of Benzophenone derivatives as potential non-competitive inhibitors of trypanosoma cruzi and leishmania amazonensis cysteine proteinases. J Braz Chem Soc 2018; 29(3): 515-27.
[http://dx.doi.org/10.21577/0103-5053.20170164]
[58]
Henrique T, José Freitas da Silveira N, Henrique Cunha Volpato A, et al. HNdb: an integrated database of gene and protein information on head and neck squamous cell carcinoma. Database (Oxford) 2016; 2016: 1-11.
[http://dx.doi.org/10.1093/database/baw026] [PMID: 27013077]
[59]
Alves Pinto I, Freitas Da Silveira NJ. In silico identification of potential inhibitors of the wnt signaling pathway in human breast cancer. J Comput Biol 2020; 27(7): 999-1010.
[http://dx.doi.org/10.1089/cmb.2019.0311] [PMID: 31647315]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy