Foti, RS, Dalvie, DK. Cytochrome P450 and non‐cytochrome P450 oxidative metabolism: contributions to the pharmacokinetics, safety, and efficacy of xenobiotics. Drug Metab and Dispos 2016, 44:1229–1245.
Onakpoya, IJ, Heneghan, CJ, Aronson, JK. Post‐marketing withdrawal of 462 medicinal products because of adverse drug reactions: a systematic review of the world literature. BMC Med 2017, 14:10.
Cerny, MA. Prevalence of non‐cytochrome P450 mediated metabolism in Food and Drug Administration‐approved oral and intravenous drugs: 2006–2015. Drug Metab Dispos 2016, 44:1246–1252.
Fan, PW, Zhang, D, Driscoll, JP, Halladay, JS, Khojasteh, C. Going beyond common drug metabolizing enzymes: case studies of biotransformation involving aldehyde oxidase, gamma‐glutamyl transpeptidase, cathepsin B, flavin‐containing monooxygenase, and ADP‐ribosyltransferase. Drug Metab Dispos 2016, 44:1253–1261.
Michael, JS, Paul, AS, John, OM, Peter, IM, Ross, AM. Recent advances in the in silico modelling of UDP glucuronosyltransferase substrates. Curr Drug Metab 2008, 9:60–69.
Hughes, TB, Miller, GP, Swamidass, SJ. Modeling epoxidation of drug‐like molecules with a deep machine learning network. ACS Cent Sci 2015, 1:168–180.
Ridder, L, Wagener, M. SyGMa: combining expert knowledge and empirical scoring in the prediction of metabolites. ChemMedChem 2008, 3:821–832.
Marchant, CA, Briggs, KA, Long, A. In silico tools for sharing data and knowledge on toxicity and metabolism: derek for windows, meteor, and vitic. Toxicol Mech Methods 2008, 18:177–187.
Meanwell, NA. Improving drug design: an update on recent applications of efficiency metrics, strategies for replacing problematic elements, and compounds in nontraditional drug space. Chem Res Toxicol 2016, 29:564–616.
Kirchmair, J, Goller, AH, Lang, D, Kunze, J, Testa, B, Wilson, ID, Glen, RC, Schneider, G. Predicting drug metabolism: experiment and/or computation? Nat Rev Drug Discov 2015, 14:387–404.
Kirchmair, J, Williamson, MJ, Tyzack, JD, Tan, L, Bond, PJ, Bender, A, Glen, RC. Computational prediction of metabolism: sites, products, SAR, P450 enzyme dynamics, and mechanisms. J Chem Inf Model 2012, 52:617–648.
Cruciani, G, Carosati, E, De Boeck, B, Ethirajulu, K, Mackie, C, Howe, T, Vianello, R. MetaSite: understanding metabolism in human cytochromes from the perspective of the chemist. J Med Chem 2005, 48:6970–6979.
Cruciani, G, Valeri, A, Goracci, L, Pellegrino, RM, Buonerba, F, Baroni, M. Flavin monooxygenase metabolism: why medicinal chemists should matter. J Med Chem 2014, 57:6183–6196.
Zaretzki, J, Matlock, M, Swamidass, SJ. XenoSite: accurately predicting CYP‐mediated sites of metabolism with neural networks. J Chem Inf Model 2013, 53:3373–3383.
Rydberg, P, Gloriam, DE, Zaretzki, J, Breneman, C, Olsen, L. SMARTCyp: a 2D method for prediction of cytochrome P450‐mediated drug metabolism. ACS Med Chem Lett 2010, 1:96–100.
Zaretzki, J, Bergeron, C, Rydberg, P, Huang, T‐W, Bennett, KP, Breneman, CM. RS‐Predictor: a new tool for predicting sites of cytochrome P450‐mediated metabolism applied to CYP 3A4. J Chem Inf Model 2011, 51:1667–1689.
Carlsson, L, Spjuth, O, Adams, S, Glen, RC, Boyer, S. Use of historic metabolic biotransformation data as a means of anticipating metabolic sites using MetaPrint2D and Bioclipse. BMC Bioinformatics 2010, 11:362.
ADMET Predictor. Simulations Plus Metabolite Software Module, Version 5.5, 2011.
JChem Metabolizer. ChemAxon, Kft. 2014. Available at: https://docs.chemaxon.com/display/docs/Metabolizer+Introduction. (Accessed June 25, 2016).
Rostkowski, M, Spjuth, O, Rydberg, P. WhichCyp: prediction of cytochromes P450 inhibition. Bioinformatics 2013, 29:2051–2052.
Shao, C‐Y, Su, B‐H, Tu, Y‐S, Lin, C, Lin, OA, Tseng, YJ. CypRules: a rule‐based P450 inhibition prediction server. Bioinformatics 2015, 31:1869–1871.
Vedani, A, Dobler, M, Hu, Z, Smiesko, M. OpenVirtualToxLab: a platform for generating and exchanging in silico toxicity data. Toxicol Lett 2015, 232:519–532.
Dixit, VA, Deshpande, S. Advances in computational prediction of regioselective and isoform‐specific drug metabolism catalyzed by CYP450s. ChemistrySelect 2016, 1:6571–6597.
Cashman, JR, Zhang, J. Human flavin‐containing monooxygenases. Annu Rev Pharmacol Toxicol 2006, 46:65–100.
Wagmann, L, Meyer, MR, Maurer, HH. What is the contribution of human FMO3 in the N‐oxygenation of selected therapeutic drugs and drugs of abuse? Toxicol Lett 2016, 258:55–70.
Testa, B, Pedretti, A, Vistoli, G. Reactions and enzymes in the metabolism of drugs and other xenobiotics. Drug Discov Today 2012, 17:549–560.
Strolin Benedetti, M, Whomsley, R, Baltes, EN. Involvement of enzymes other than CYPs in the oxidative metabolism of xenobiotics. Expert Opin Drug Metab Toxicol 2006, 2:895–921.
Alfieri, A, Malito, E, Orru, R, Fraaije, MW, Mattevi, A. Revealing the moonlighting role of NADP in the structure of a flavin‐containing monooxygenase. Proc Natl Acad Sci USA 2008, 105:6572–6577.
Chen, G‐P, Poulsen, LL, Ziegler, DM. Oxidation of aldehydes catalyzed by pig liver flavin‐containing monooxygenase. Drug Metab Dispos 1995, 23:1390–1393.
Fiorentini, F, Geier, M, Binda, C, Winkler, M, Faber, K, Hall, M, Mattevi, A. Biocatalytic characterization of human FMO5: unearthing Baeyer‐Villiger reactions in humans. ACS Chem Biol 2016, 11:1039–1048.
Celius, T, Pansoy, A, Matthews, J, Okey, AB, Henderson, MC, Krueger, SK, Williams, DE. Flavin‐containing monooxygenase‐3: induction by 3‐methylcholanthrene and complex regulation by xenobiotic chemicals in hepatoma cells and mouse liver. Toxicol Appl Pharmacol 2010, 247:60–69.
Boersma, MG, Cnubben, NH, van Berkel, WJ, Blom, M, Vervoort, J, Rietjens, IM. Role of cytochromes P‐450 and flavin‐containing monooxygenase in the biotransformation of 4‐fluoro‐N‐methylaniline. Drug Metab Dispos 1993, 21:218–230.
Lai, WG, Farah, N, Moniz, GA, Wong, YN. A Baeyer‐Villiger oxidation specifically catalyzed by human flavin‐containing monooxygenase 5. Drug Metab Dispos 2011, 39:61–70.
Pirovano, A, Brandmaier, S, Huijbregts, MAJ, Ragas, AMJ, Veltman, K, Hendriks, AJ. The utilisation of structural descriptors to predict metabolic constants of xenobiotics in mammals. Environ Toxicol Pharmacol 2015, 39:247–258.
Cronin, MTD, Schultz, TW. Pitfalls in QSAR. J Mol Struct (THEOCHEM) 2003, 622:39–51.
Guo, W‐XA, Poulsen, LL, Ziegler, DM. Use of thiocarbamides as selective substrate probes for isoforms of flavin‐containing monooxygenases. Biochem Pharmacol 1992, 44:2029–2037.
Lin, T‐H, Tsai, T‐L. Constructing a linear QSAR for some metabolizable drugs by human or pig flavin‐containing monooxygenases using some molecular features selected by a genetic algorithm trained SVM. J Theor Biol 2014, 356:85–97.
Baroni, M, Cruciani, G, Sciabola, S, Perruccio, F, Mason, JS. A common reference framework for analyzing/comparing proteins and ligands: fingerprints for ligands and Proteins (FLAP): theory and application. J Chem Inf Model 2007, 47:279–294.
C‐w, F, Lin, T‐H. Predicting the metabolic sites by flavin‐containing monooxygenase on drug molecules using SVM classification on computed quantum mechanics and circular fingerprints molecular descriptors. PLoS One 2017, 12:e0169910.
Chih‐Chung, C, Chih‐Jen, L. LIBSVM: a library for support vector machines. ACM Trans Intell Syst Technol 2011, 2:1–27.
Geha, RM, Rebrin, I, Chen, K, Shih, JC. Substrate and inhibitor specificities for human monoamine oxidase A and B are influenced by a single amino acid. J Biol Chem 2001, 276:9877–9882.
Fisar, Z. Drugs related to monoamine oxidase activity. Prog Neuropsychopharmacol Biol Psychiatry 2016, 69:112–124.
McMartin, KE, Sebastian, CS, Dies, D, Jacobsen, D. Kinetics and metabolism of fomepizole in healthy humans. Clin Toxicol 2012, 50:375–383.
Parajuli, B, Kimble‐Hill, AC, Khanna, M, Ivanova, Y, Meroueh, S, Hurley, TD. Discovery of novel regulators of aldehyde dehydrogenase isoenzymes. Chem Biol Interact 2011, 191:153–158.
Salva, M, Jansat, JM, Martinez‐Tobed, A, Palacios, JM. Identification of the human liver enzymes involved in the metabolism of the antimigraine agent almotriptan. Drug Metab Dispos 2003, 31:404–411.
Strolin Benedetti, M, Tipton, KF, Whomsley, R. Amine oxidases and monooxygenases in the in vivo metabolism of xenobiotic amines in humans: has the involvement of amine oxidases been neglected? Fundam Clin Pharmacol 2007, 21:467–480.
Edenberg, HJ. Regulation of the mammalian alcohol dehydrogenase genes. In: Moldave K, ed. Progress in Nucleic Acid Research and Molecular Biology, vol. 64. Cambridge, MA: Academic Press; 2000, 295–341.
Gibbons, BJ, Hurley, TD. Structure of three Class I human alcohol dehydrogenases complexed with isoenzyme specific formamide inhibitors. Biochemistry 2004, 43:12555–12562.
Walsh, JS, Reese, MJ, Thurmond, LM. The metabolic activation of abacavir by human liver cytosol and expressed human alcohol dehydrogenase isozymes. Chem Biol Interact 2002, 142:135–154.
Gunness, P, Aleksa, K, Bend, J, Koren, G. Acyclovir‐induced nephrotoxicity: the role of the acyclovir aldehyde metabolite. Transl Res 2011, 158:290–301.
Hansch, C, Klein, T, McClarin, J, Langridge, R, Cornell, NW. A quantitative structure‐activity relationship and molecular graphics analysis of hydrophobic effects in the interactions of inhibitors with alcohol dehydrogenase. J Med Chem 1986, 29:615–620.
Moreno, A, Farrés, J, Parés, X, Jörnvall, H, Persson, B. Molecular modelling of human gastric alcohol dehydrogenase (class IV) and substrate docking: differences towards the classical liver enzyme (class I). FEBS Lett 1996, 395:99–102.
Stone, CL, Hurley, TD, Peggs, CF, Kedishvili, NY, Davis, GJ, Thomasson, HR, Li, T‐K, Bosron, WF. Cimetidine inhibition of human gastric and liver alcohol dehydrogenase isoenzymes: identification of inhibitor complexes by kinetics and molecular modeling. Biochemistry 1995, 34:4008–4014.
Brown, J. Omeprazole, ranitidine and cimetidine have no effect on peak blood ethanol concentrations, first pass metabolism or area under the time–ethanol curve under ‘real‐life’ drinking conditions. Aliment Pharmacol Ther 1998, 12:141–145.
Lai, C‐L, Li, Y‐P, Liu, C‐M, Hsieh, H‐S, Yin, S‐J. Inhibition of human alcohol and aldehyde dehydrogenases by cimetidine and assessment of its effects on ethanol metabolism. Chem Biol Interact 2013, 202:275–282.
Park, DH, Plapp, BV. Interconversion of E and S isoenzymes of horse liver alcohol dehydrogenase: several residues contribute indirectly to catalysis. J Biol Chem 1992, 267:5527–5533.
Adolph, H‐W, Zwart, P, Meijers, R, Hubatsch, I, Kiefer, M, Lamzin, V, Cedergren‐Zeppezauer, E. Structural basis for substrate specificity differences of horse liver alcohol dehydrogenase isozymes. Biochemistry 2000, 39:12885–12897.
Murali, C, Creaser, EH. Protein engineering of alcohol dehydrogenase. 1. Effects of two amino acid changes in the active site of yeast ADH‐1. Protein Eng 1986, 1:55–57.
Esterbauer, H, Schaur, R Jr, Zollner, H. Chemistry and biochemistry of 4‐hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 1991, 11:81–128.
Rizzo, WB, Carney, G. Sjögren‐Larsson syndrome: diversity of mutations and polymorphisms in the fatty aldehyde dehydrogenase gene (ALDH3A2). Hum Mutat 2005, 26:1–10.
Farrant, RD, Walker, V, Mills, GA, Mellor, JM, Langley, GJ. Pyridoxal phosphate de‐activation by pyrroline‐5‐carboxylic acid: increased risk of vitamin b6 deficiency and seizures in hyperprolinemia type II. J Biol Chem 2001, 276:15107–15116.
Boyles, AL, DeRoo, LA, Lie, RT, Taylor, JA, Jugessur, A, Murray, JC, Wilcox, AJ. Maternal alcohol consumption, alcohol metabolism genes, and the risk of oral clefts: a population‐based case–control Study in Norway, 1996–2001. Am J Epidemiol 2010, 172:924–931.
Haynes, RL, Szweda, L, Pickin, K, Welker, ME, Townsend, AJ. Structure‐activity relationships for growth inhibition and induction of apoptosis by 4‐hydroxy‐2‐nonenal in raw 264.7 cells. Mol Pharmacol 2000, 58:788–794.
Condello, S, Morgan, CA, Nagdas, S, Cao, L, Turek, J, Hurley, TD, Matei, D. [β]‐Catenin‐regulated ALDH1A1 is a target in ovarian cancer spheroids. Oncogene 2015, 34:2297–2308.
Feldman, RI, Weiner, H. Horse liver aldehyde dehydrogenase. II. Kinetics and mechanistic implications of the dehydrogenase and esterase activity. J Biol Chem 1972, 247:267–272.
Kimble‐Hill, AC, Parajuli, B, Chen, C‐H, Mochly‐Rosen, D, Hurley, TD. Development of selective inhibitors for aldehyde dehydrogenases based on substituted indole‐2,3‐diones. J Med Chem 2014, 57:714–722.
Morgan, CA, Hurley, TD. Characterization of two distinct structural classes of selective aldehyde dehydrogenase 1A1 inhibitors. J Med Chem 2015, 58:1964–1975.
Chen, Z, Zhang, J, Stamler, JS. Identification of the enzymatic mechanism of nitroglycerin bioactivation. Proc Natl Acad Sci USA 2002, 99:8306–8311.
Goldstein, DS, Sullivan, P, Holmes, C, Miller, GW, Alter, S, Strong, R, Mash, DC, Kopin, IJ, Sharabi, Y. Determinants of buildup of the toxic dopamine metabolite DOPAL in Parkinson`s disease. J Neurochem 2013, 126:591–603.
Fitzmaurice, AG, Rhodes, SL, Cockburn, M, Ritz, B, Bronstein, JM. Aldehyde dehydrogenase variation enhances effect of pesticides associated with Parkinson disease. Neurology 2014, 82:419–426.
Kalgutkar, AS, Dalvie, DK, Castagnoli, N, Taylor, TJ. Interactions of nitrogen‐containing xenobiotics with monoamine oxidase (MAO) isozymes A and B: SAR studies on MAO substrates and inhibitors. Chem Res Toxicol 2001, 14:1139–1162.
Brunner, HG, Nelen, MR, van Zandvoort, P, Abeling, NG, van Gennip, AH, Wolters, EC, Kuiper, MA, Ropers, HH, van Oost, BA. X‐linked borderline mental retardation with prominent behavioral disturbance: phenotype, genetic localization, and evidence for disturbed monoamine metabolism. Am J Hum Genet 1993, 52:1032–1039.
De Colibus, L, Li, M, Binda, C, Lustig, A, Edmondson, DE, Mattevi, A. Three‐dimensional structure of human monoamine oxidase A (MAO A): relation to the structures of rat MAO A and human MAO B. Proc Natl Acad Sci USA 2005, 102:12684–12689.
Cesura, AM, Gottowik, J, Lahm, H‐W, Lang, G, Imhof, R, Malherbe, P, Röthlisberger, U, Prada, MD. Investigation on the structure of the active site of monoamine oxidase‐B by affinity labeling with the selective inhibitor lazabemide and by site‐directed mutagenesis. Eur J Biochem 1996, 236:996–1002.
Geha, RM, Chen, K, Wouters, J, Ooms, FDR, Shih, JC. Analysis of conserved active site residues in monoamine oxidase A and B and their three‐dimensional molecular modeling. J Biol Chem 2002, 277:17209–17216.
Silverman, RB, Zelechonok, Y. Evidence for a hydrogen atom transfer mechanism or a fast proton/electron transfer mechanism for monoamine oxidase. J Org Chem 1992, 57:6373–6374.
Abad, E, Zenn, RK, Kӓstner, J. Reaction mechanism of monoamine oxidase from QM/MM calculations. J Phys Chem B 2013, 117:14238–14246.
Mavri, J, Matute, RA, Chu, ZT, Vianello, R. Path integral simulation of the H/D kinetic isotope effect in monoamine oxidase b catalyzed decomposition of dopamine. J Phys Chem B 2016, 120:3488–3492.
Son, S‐Y, Ma, J, Kondou, Y, Yoshimura, M, Yamashita, E, Tsukihara, T. Structure of human monoamine oxidase A at 2.2 angstrom resolution: the control of opening the entry for substrates/inhibitors. Proc Natl Acad Sci USA 2008, 105:5739–5744.
Olesen, OV, Linnet, K. Studies on the stereoselective metabolism of citalopram by human liver microsomes and cDNA‐expressed cytochrome P450 enzymes. Pharmacology 1999, 59:298–309.
Kosel, M, Gnerre, C, Voirol, P, Amey, M, Rochat, B, Bouras, C, Testa, B, Baumann, P. In vitro biotransformation of the selective serotonin reuptake inhibitor citalopram, its enantiomers and demethylated metabolites by monoamine oxidase in rat and human brain preparations. Mol Psychiatry 2002, 7:181–188.
Youdim, MBH, Edmondson, D, Tipton, KF. The therapeutic potential of monoamine oxidase inhibitors. Nat Rev Neurosci 2006, 7:295–309.
Baker, GB, Urichuk, LJ, McKenna, KF, Kennedy, SH. Metabolism of monoamine oxidase inhibitors. Cell Mol Neurobiol 1999, 19:411–426.
Vilar, S, Ferino, G, Quezada, E, Santana, L, Friedman, C. Predicting monoamine oxidase inhibitory activity through ligand‐based models. Curr Top Med Chem 2012, 12:2258–2274.
Nunez, MB, Maguna, FP, Okulik, NB, Castro, EA. QSAR modeling of the MAO inhibitory activity of xanthones derivatives. Bioorg Med Chem Lett 2004, 14:5611–5617.
La Regina, G, Silvestri, R, Artico, M, Lavecchia, A, Novellino, E, Befani, O, Turini, P, Agostinelli, E. New pyrrole inhibitors of monoamine oxidase: synthesis, biological evaluation, and structural determinants of MAO‐A and MAO‐B selectivity. J Med Chem 2007, 50:922–931.
Ogunrombi, MO, Malan, SF, Terrea Blanche, G, Castagnoli, N Jr, Bergh, JJ, Petzer, JP. Structure activity relationships in the inhibition of monoamine oxidase B by 1‐methyl‐3‐phenylpyrroles. Bioorg Med Chem 2008, 16:2463–2472.
Altomare, C, Cellamare, S, Summo, L, Catto, M, Carotti, A, Thull, U, Carrupt, P‐A, Testa, B, Stoeckli‐Evans, H. Inhibition of monoamine oxidase‐B by condensed pyridazines and pyrimidines: effects of lipophilicity and structure–activity relationships. J Med Chem 1998, 41:3812–3820.
Vlok, N, Malan, SF, Castagnoli, N Jr, Bergh, JJ, Petzer, JP. Inhibition of monoamine oxidase B by analogues of the adenosine A2A receptor antagonist (E)‐8‐(3‐chlorostyryl)caffeine (CSC). Bioorg Med Chem 2006, 14:3512–3521.
Norinder, U, Florvall, L, Ross, SB. A PLS quantitative structure‐activity relationship study of some monoamine oxidase inhibitors of the phenyl alkylamine type. Eur J Med Chem 1994, 29:191–195.
Hasegawa, K, Kimura, T, Miyashita, Y, Funatsu, K. Nonlinear partial least squares modeling of phenyl alkylamines with the monoamine oxidase inhibitory activities. J Chem Inf Comput Sci 1996, 36:1025–1029.
Deeb, O, Clare, BW. Comparison of AM1 and B3LYP‐DFT for Inhibition of MAO‐A by phenylisopropylamines: a QSAR study. Chem Biol Drug Des 2008, 71:352–362.
Scorza, MC, Carrau, C, Silveira, R, Zapata‐Torres, G, Cassels, BK, Reyes‐Parada, M. Monoamine oxidase inhibitory properties of some methoxylated and alkylthio amphetamine derivatives: structure‐activity relationships. Biochem Pharmacol 1997, 54:1361–1369.
Hall, LH, Mohney, BK, Kier, LB. Comparison of electrotopological state indexes with molecular orbital parameters: inhibition of MAO by hydrazides. Quant Struct‐Act Relat 1993, 12:44–48.
Gnerre, C, Thull, U, Gaillard, P, Carrupt, PA, Testa, B, Fernandes, E, Silva, F, Pinto, M, Pinto, MMM, Wolfender, JL. Natural and synthetic xanthones as monoamine oxidase inhibitors: biological assay and 3D QSAR. Helv Chim Acta 2001, 84:552–570.
Medvedev, AE, Ivanov, AS, Kamyshanskaya, NS, Kirkel, AZ, Moskvitina, TA, Gorkin, VZ, Li, NY, Marshakov, VY. Interaction of indole derivatives with monoamine oxidase A and B: studies on the structure‐inhibitory activity relationship. Biochem Mol Biol Int 1995, 36:113–122.
Bautista‐Aguilera, OM, Samadi, A, Chioua, M, Nikolic, K, Filipic, S, Agbaba, D, Soriano, E, de Andres, L, Rodreguez‐Franco, MI, Alcaro, S, et al. N‐Methyl‐N‐((1‐methyl‐5‐(3‐(1‐(2‐methylbenzyl)piperidin‐4‐yl)propoxy)‐1H‐indol‐2‐yl)methyl)prop‐2‐yn‐1‐amine, a new cholinesterase and monoamine oxidase dual inhibitor. J Med Chem 2014, 57:10455–10463.
Medvedev, AE, Veselovsky, AV, Shvedov, VI, Tikhonova, OV, Moskvitina, TA, Fedotova, OA, Axenova, LN, Kamyshanskaya, NS, Kirkel, AZ, Ivanov, AS. Inhibition of monoamine oxidase by pirlindole analogues: 3D‐QSAR and CoMFA analysis. J Chem Inf Comput Sci 1998, 38:1137–1144.
Moron, JA, Campillo, M, Perez, V, Unzeta, M, Pardo, L. Molecular determinants of MAO selectivity in a series of indolylmethylamine derivatives: biological activities, 3D‐QSAR/CoMFA analysis, and computational simulation of ligand recognition. J Med Chem 2000, 43:1684–1691.
Gallardo‐Godoy, A, Fierro, A, McLean, TH, Castillo, M, Cassels, BK, Reyes‐Parada, M, Nichols, DE. Sulfur‐substituted alpha‐alkyl phenethylamines as selective and reversible MAO‐A inhibitors: biological activities, CoMFA analysis, and active site modeling. J Med Chem 2005, 48:2407–2419.
Kumar, V, Chadha, N, Tiwari, AK, Sehgal, N, Mishra, AK. Prospective atom‐based 3D‐QSAR model prediction, pharmacophore generation, and molecular docking study of carbamate derivatives as dual inhibitors of AChE and MAO‐B for Alzheimer`s disease. Med Chem Res 2014, 23:1114–1122.
Catto, M, Nicolotti, O, Leonetti, F, Carotti, A, Favia, AD, Soto‐Otero, R, Mendez‐Alvarez, E, Carotti, A. Structural insights into monoamine oxidase inhibitory potency and selectivity of 7‐substituted coumarins from ligand‐ and target‐based approaches. J Med Chem 2006, 49:4912–4925.
Chimenti, F, Secci, D, Bolasco, A, Chimenti, P, Granese, A, Carradori, S, Maccioni, E, Cardia, MC, Yáñez, M, Orallo, F, et al. Synthesis, semipreparative HPLC separation, biological evaluation, and 3D‐QSAR of hydrazothiazole derivatives as human monoamine oxidase B inhibitors. Bioorg Med Chem 2010, 18:5063–5070.
Franco, C, Adriana, B, Fedele, M, Daniela, S, Paola, C, Arianna, G, Olivia, B, Paola, T, Roberto, C, Francesco La, T, et al. Synthesis, biological evaluation and 3D‐QSAR of 1,3,5‐trisubstituted‐4,5‐dihydro‐(1H)‐pyrazole derivatives as potent and highly selective monoamine oxidase A inhibitors. Curr Med Chem 2006, 13:1411–1428.
Gritsch, S, Guccione, S, Hoffmann, RM, Cambria, A, Raciti, G, Langer, T. A 3D QSAR study of monoamino oxidase‐B inhibitors using the chemical function based pharmacophore generation approach. J Enzyme Inhib 2001, 16:199–215.
Sairam, KVVM, Khar, RK, Mukherjee, R, Jain, SK. Three dimensional pharmacophore modelling of monoamine oxidase‐A (MAO‐A) inhibitors. Int J Mol Sci 2007, 8:894–919.
Suryawanshi, MR, Kulkarni, VM, Mahadik, KR, Bhosale, SH. Pharmacophore modeling and atom‐based 3D‐QSAR studies of tricyclic selective monoamine oxidase A inhibitors. Der Pharma Chemica 2010, 2:171–182.
Shelke, SM, Bhosale, SH, Dash, RC, Suryawanshi, MR, Mahadik, KR. Exploration of new scaffolds as potential MAO‐A inhibitors using pharmacophore and 3D‐QSAR based in silico screening. Bioorg Med Chem Lett 2011, 21:2419–2424.
John, OM, Paul, AS, Michael, JS, Ross, AM, Peter, IM. Predicting human drug glucuronidation parameters: application of in vitro and in silico modeling approaches. Annu Rev Pharmacol Toxicol 2004, 44:1–25.
Zamek‐Gliszczynski, MJ, Day, JS, Hillgren, KM, Phillips, DL. Efflux transport is an important determinant of ethinyl estradiol glucuronide and ethinyl estradiol sulfate pharmacokinetics. Drug Metab Dispos 2011, 39:1794–1800.
De Gregori, S, De Gregori, M, Ranzani, GN, Allegri, M, Minella, C, Regazzi, M. Morphine metabolism, transport and brain disposition. Metab Brain Dis 2012, 27:1–5.
Sturgill, MG, Lambert, GH. Xenobiotic‐induced hepatotoxicity: mechanisms of liver injury and methods of monitoring hepatic function. Clin Chem 1997, 43:1512–1526.
Wu, B, Kulkarni, K, Basu, S, Zhang, S, Hu, M. First‐pass metabolism via UDP‐glucuronosyltransferase: a barrier to oral bioavailability of phenolics. J Pharm Sci 2011, 100:3655–3681.
Miley, MJ, Zielinska, AK, Keenan, JE, Bratton, SM, Radominska‐Pandya, A, Redinbo, MR. Crystal structure of the cofactor‐binding domain of the human phase ii drug‐metabolism enzyme UDP‐glucuronosyltransferase 2B7. J Mol Biol 2007, 369:498–511.
Argikar, UA. Unusual glucuronides. Drug Metab Dispos 2012, 40:1239–1251.
Richter, WJ, Alt, KO, Dieterle, W, Faigle, JW, Kriemler, H‐P, Mory, H, Winkler, T. C‐glucuronides, a novel type of drug metabolites. Helv Chim Acta 1975, 58:2512–2517.
Robert, HT, Christian, PS. Human UDP‐glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol 2000, 40:581–616.
Ouzzine, M, Gulberti, S, Ramalanjaona, N, Magdalou, J, Fournel‐Gigleux, S. The UDP‐glucuronosyltransferases of the blood–brain barrier: their role in drug metabolism and detoxication. Front Cell Neurosci 2014, 8:349.
Ouzzine, M, Magdalou, J, Burchell, B, Fournel‐Gigleux, S. An internal signal sequence mediates the targeting and retention of the human UDP‐glucuronosyltransferase 1A6 to the endoplasmic reticulum. J Biol Chem 1999, 274:31401–31409.
Williams, JA, Hyland, R, Jones, BC, Smith, DA, Hurst, S, Goosen, TC, Peterkin, V, Koup, JR, Ball, SE. Drug–drug interactions for UDP‐glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUC‐I/AUC) ratios. Drug Metab Dispos 2004, 32:1201–1208.
Boase, S, Miners, JO. In vitro–in vivo correlations for drugs eliminated by glucuronidation: investigations with the model substrate zidovudine. Br J Clin Pharmacol 2002, 54:493–503.
Knights, KM, Spencer, SM, Fallon, JK, Chau, N, Smith, PC, Miners, JOCMPR. Scaling factors for the in vitro–in vivo extrapolation (IV–IVE) of renal drug and xenobiotic glucuronidation clearance. Br J Clin Pharmacol 2016, 81:1153–1164.
Boersma, MG, van der Woude, H, Bogaards, J, Boeren, S, Vervoort, J, Cnubben, NHP, van Iersel, MLPS, van Bladeren, PJ, Rietjens, IMCM. Regioselectivity of Phase II metabolism of luteolin and quercetin by UDP‐glucuronosyltransferases. Chem Res Toxicol 2002, 15:662–670.
Wong, YC, Zhang, L, Lin, G, Zuo, Z. Structure‐activity relationships of the glucuronidation of flavonoids by human glucuronosyltransferases. Expert Opin Drug Metab Toxicol 2009, 5:1399–1419.
Peer, CJ, Sissung, TM, Kim, A, Jain, L, Woo, S, Gardner, ER, Kirkland, CT, Troutman, SM, English, BC, Richardson, ED, et al. Sorafenib is an inhibitor of UGT1A1 but is metabolized by UGT1A9: implications of genetic variants on pharmacokinetics and hyperbilirubinemia. Clin Cancer Res 2012, 18:2099–2107.
Zhang, D, Chando, TJ, Everett, DW, Patten, CJ, Dehal, SS, Humphreys, WG. In vitro inhibition of UDP glucuronosyltransferases by atazanavir and other HIV protease inhibitors and the relationship of this property to in vivo bilirubin glucuronidation. Drug Metab Dispos 2005, 33:1729–1739.
Kuehl, GE, Lampe, JW, Potter, JD, Bigler, J. Glucuronidation of nonsteroidal anti‐inflammatory drugs: identifying the enzymes responsible in human liver microsomes. Drug Metab Dispos 2005, 33:1027–1035.
Uchaipichat, V, Mackenzie, PI, Guo, X‐H, Gardner‐Stephen, D, Galetin, A, Houston, JB, Miners, JO. Human UDP‐glucuronosyltransferases: isoform selectivity and kinetics of 4‐methylumbelliferone and 1‐naphthol glucuronidation, effects of organic solvents, and inhibition by diclofenac and probenecid. Drug Metab Dispos 2004, 32:413–423.
Ghemtio, L, Soikkeli, A, Yliperttula, M, Hirvonen, J, Finel, M, Xhaard, H. SVM classification and CoMSIA modeling of UGT1A6 interacting molecules. J Chem Inf Model 2014, 54:1011–1026.
Zhang, N, Liu, Y, Jeong, H. Drug‐drug interaction potentials of tyrosine kinase inhibitors via inhibition of UDP‐glucuronosyltransferases. Sci Rep 2015, 5:17778.
Lewinsky, RH, Smith, PA, Mackenzie, PI. Glucuronidation of bioflavonoids by human UGT1A10: structure function relationships. Xenobiotica 2005, 35:117–129.
Ramirez, J, Mirkov, S, House LK, Ratain, MJ. Glucuronidation of OTS167 in humans is catalyzed by UDP‐glucuronosyltransferases UGT1A1, UGT1A3, UGT1A8, and UGT1A10. Drug Metab Dispos 2015, 43:928–935.
Takeda, S, Kitajima, Y, Ishii, Y, Nishimura, Y, Mackenzie, PI, Oguri, K, Yamada, H. Inhibition of UDP‐glucuronosyltransferase 2b7‐catalyzed morphine glucuronidation by ketoconazole: dual mechanisms involving a novel noncompetitive mode. Drug Metab Dispos 2006, 34:1277–1282.
Green, MD, Tephly, TR. Glucuronidation of amines and hydroxylated xenobiotics and endobiotics catalyzed by expressed human UGT1.4 protein. Drug Metab Dispos 1996, 24:356–363.
Mullen, W, Edwards, CA, Crozier, A. Absorption, excretion and metabolite profiling of methyl‐, glucuronyl‐, glucosyl‐ and sulpho‐conjugates of quercetin in human plasma and urine after ingestion of onions. Br J Nutr 2006, 96:107–116.
Ramirez, J, Liu, W, Mirkov, S, Desai, AA, Chen, P, Das, S, Innocenti, F, Ratain, MJ. Lack of association between common polymorphisms in UGT1A9 and gene expression and activity. Drug Metab Dispos 2007, 35:2149–2153.
Oliver Chen, CY, Blumberg, J. Are there age‐related changes in flavonoid bioavailability? In: Meskin MS, Bidlack WR, Randolph RK, eds. Phytochemicals. Boca Raton, FL: CRC Press; 2008, 19–37.
Lan, K, He, J‐l, Tian, Y, Tan, F, X‐h, J, Wang, L, L‐m, Y. Intra‐herb pharmacokinetics interaction between quercetin and isorhamentin. Acta Pharmacol Sin 2008, 29:1376–1382.
Dong, D, Wu, B. In silico modeling of UDP‐glucuronosyltransferase 1A10 substrates using the Volsurf approach. J Pharm Sci 2012, 101:3531–3539.
Ako, R, Dong, D, Wu, B. 3D‐QSAR studies on UDP‐glucuronosyltransferase 2B7 substrates using the pharmacophore and VolSurf approaches. Xenobiotica 2012, 42:891–900.
Czernik, PJ, Little, JM, Barone, GW, Raufman, J‐P, Radominska‐Pandya, A. Glucuronidation of estrogens and retinoic acid and expression of UDP‐glucuronosyltransferase 2B7 in human intestinal mucosa. Drug Metab Dispos 2000, 28:1210–1216.
Buchheit, D, Dragan, C‐A, Schmitt, EI, Bureik, M. Production of ibuprofen acyl glucosides by human UGT2B7. Drug Metab Dispos 2011, 39:2174–2181.
Wu, Z, Zhang, X, Ma, Z, Wu, B. Establishment of pharmacophore and VolSurf models to predict the substrates of UDP‐glucuronosyltransferase1A3. Xenobiotica 2015, 45:653–662.
Sorich, MJ, Miners, JO, McKinnon, RA, Smith, PA. Multiple pharmacophores for the investigation of human UDP‐glucuronosyltransferase isoform substrate selectivity. Mol Pharmacol 2004, 65:301–308.
Sorich, MJ, Miners, JO, McKinnon, RA, Winkler, DA, Burden, FR, Smith, PA. Comparison of linear and nonlinear classification algorithms for the prediction of drug and chemical metabolism by human UDP‐glucuronosyltransferase Isoforms. J Chem Inf Comput Sci 2003, 43:2019–2024.
Sorich, MJ, McKinnon, RA, Miners, JO, Winkler, DA, Smith, PA. Rapid prediction of chemical metabolism by human UDP‐glucuronosyltransferase isoforms using quantum chemical descriptors derived with the electronegativity equalization method. J Med Chem 2004, 47:5311–5317.
Peng, J, Lu, J, Shen, Q, Zheng, M, Luo, X, Zhu, W, Jiang, H, Chen, K. In silico site of metabolism prediction for human UGT‐catalyzed reactions. Bioinformatics 2013, 30:398–405.
O`Boyle, NM, Banck, M, James, CA, Morley, C, Vandermeersch, T, Hutchison, GR. Open Babel: an open chemical toolbox. J Cheminf 2011, 3:33.
Rudik, A, Dmitriev, A, Lagunin, A, Filimonov, D, Poroikov, V. SOMP: web‐server for in silico prediction of sites of metabolism for drug‐like compounds. Bioinformatics 2015, 31:2046–2048.
Rudik, AV, Dmitriev, AV, Lagunin, AA, Filimonov, DA, Poroikov, VV. Metabolism site prediction based on xenobiotic structural formulas and PASS prediction algorithm. J Chem Inf Model 2014, 54:498–507.
Dang, NL, Hughes, TB, Krishnamurthy, V, Swamidass, SJ. A simple model predicts UGT‐mediated metabolism. Bioinformatics 2016, 32:3183–3189.
Thompson, MJ, Sievers, SA, Karanicolas, J, Ivanova, MI, Baker, D, Eisenberg, D. The 3D profile method for identifying fibril‐forming segments of proteins. Proc Natl Acad Sci USA 2006, 103:4074–4078.
Gamage, N, Barnett, A, Hempel, N, Duggleby, RG, Windmill, KF, Martin, JL, McManus, ME. Human sulfotransferases and their role in chemical metabolism. Toxicol Sci 2006, 90:5–22.
Allali‐Hassani, A, Pan, PW, Dombrovski, L, Najmanovich, R, Tempel, W, Dong, A, Loppnau, P, Martin, F, Thonton, J, Edwards, AM. Structural and chemical profiling of the human cytosolic sulfotransferases. PLoS Biol 2007, 5:e97.
Cook, I, Wang, T, Leyh, TS. Sulfotransferase 1A1 substrate selectivity: a molecular clamp mechanism. Biochemistry 2015, 54:6114–6122.
Moroy, G, Martiny, VY, Vayer, P, Villoutreix, BO, Miteva, MA. Toward in silico structure‐based ADMET prediction in drug discovery. Drug Discov Today 2012, 17:44–55.
Meechan, AJ, Henderson, C, Bates, CD, Grant, MH, Tettey, JNA. Metabolism of troglitazone in hepatocytes isolated from experimentally induced diabetic rats. J Pharm Pharmacol 2006, 58:1359–1365.
Kauffman, FC. Sulfonation in Pharmacology and Toxicology. Drug Metab Rev 2004, 36:823–843.
Dixit, VA, Bharatam, PV. Toxic metabolite formation from troglitazone (TGZ): new insights from a DFT study. Chem Res Toxicol 2011, 24:1113–1122.
Rakers, C, Schumacher, F, Meinl, W, Glatt, H, Kleuser, B, Wolber, G. In silico prediction of human sulfotransferase 1E1 activity guided by pharmacophores from molecular dynamics simulations. J Biol Chem 2016, 291:58–71.
Iyer, LV, Ramamoorthy, A, Rutkowska, E, Furimsky, AM, Tang, L, Catz, P, Green, CE, Jozwiak, K, Wainer, IW. The stereoselective sulfate conjugation of 4`‐methoxyfenoterol stereoisomers by sulfotransferase enzymes. Chirality 2012, 24:796–803.
Ning, J, Cui, Y, Wang, C, Dong, P, Ge, G, Tian, X, Hou, J, Huo, X, Zhang, B, Ma, T, et al. Characterization of regio‐ and stereo‐selective sulfation of bufadienolides: exploring the mechanism and providing insight into the structure‐sulfation relationship by experimentation and molecular docking analysis. RSC Adv 2016, 6:5774–5783.
Sharma, V, Duffel, MW. A comparative molecular field analysis‐based approach to prediction of sulfotransferase catalytic specificity. Methods Enzymol 2005, 400:249–263.
Dajani, R, Cleasby, A, Neu, M, Wonacott, AJ, Jhoti, H, Hood, AM, Modi, S, Hersey, A, Taskinen, J, Cooke, RM, et al. X‐ray crystal structure of human dopamine sulfotransferase, SULT1A3: molecular modeling and quantitative structure‐activity relationship analysis demonstrate a molecular basis for sulfotransferase substrate specificity. J Biol Chem 1999, 274:37862–37868.
Townsend, DM, Manevich, Y, He, L, Hutchens, S, Pazoles, CJ, Tew, KD. Novel role for glutathione S‐transferase: regulator of protein S‐glutathionylation following oxidative and nitrosative stress. J Biol Chem 2009, 284:436–445.
Nebert, DW, Vasiliou, V. Analysis of the glutathione S‐transferase (GST) gene family. Hum Genomics 2004, 1:460.
Le Trong, I, Stenkamp, RE, Ibarra, C, Atkins, WM, Adman, ET. 1.3‐Å resolution structure of human glutathione S‐transferase with S‐hexyl glutathione bound reveals possible extended ligand binding site. Proteins 2002, 48:618–627.
Wu, B, Dong, D. Human cytosolic glutathione transferases: structure, function, and drug discovery. Trends Pharmacol Sci 2012, 33:656–668.
Morgenstern, R, Svensson, R, Bernat, BA, Armstrong, RN. Kinetic analysis of the slow ionization of glutathione by microsomal glutathione transferase MGST1. Biochemistry 2001, 40:3378–3384.
Valadez, JG, Liu, L, Loktionova, NA, Pegg, AE, Guengerich, FP. Activation of bis‐electrophiles to mutagenic conjugates by human O6‐alkylguanine‐DNA alkyltransferase. Chem Res Toxicol 2004, 17:972–982.
Ruzza, P, Calderan, A. Glutathione transferase (GST)‐activated prodrugs. Pharmaceutics 2013, 5:220–231.
Townsend, DM, Tew, KD. The role of glutathione‐S‐transferase in anti‐cancer drug resistance. Oncogene 2003, 22:7369–7375.
Jolivette, LJ, Anders, MW. Structure activity relationship for the biotransformation of haloalkenes by rat liver microsomal glutathione transferase. Chem Res Toxicol 2002, 15:1036–1041.
Soffers, AEMF, Ploemen, JHTM, Moonen, MJH, Wobbes, T, van Ommen, B, Vervoort, J, van Bladeren, PJ, Rietjens, IMCM. Regioselectivity and quantitative structure activity relationships for the conjugation of a series of fluoronitrobenzenes by purified glutathione S‐transferase enzymes from rat and man. Chem Res Toxicol 1996, 9:638–646.
Van der Aar, EM, Bouwman, T, Commandeur, JNM, Vermeulen, NPE. Structure‐activity relationships for chemical and glutathione S‐transferase‐catalysed glutathione conjugation reactions of a series of 2‐substituted 1‐chloro‐4‐nitrobenzenes. Biochem J 1996, 320:531–540.
Appiah‐Opong, R, Commandeur, JNM, Istyastono, E, Bogaards, JJ, Vermeulen, NPE. Inhibition of human glutathione S‐transferases by curcumin and analogues. Xenobiotica 2009, 39:302–311.
Dong, GQ, Calhoun, S, Fan, H, Kalyanaraman, C, Branch, MC, Mashiyama, ST, London, N, Jacobson, MP, Babbitt, PC, Shoichet, BK, et al. Prediction of substrates for glutathione transferases by covalent docking. J Chem Inf Model 2014, 54:1687–1699.
Lannutti, F, Marrone, A, Re, N. Binding of GSH conjugates to pi‐GST: a cross‐docking approach. J Mol Graph Model 2012, 32:9–18.
Jancova, P, Anzenbacher, P, Anzenbacherova, E. Phase II drug metabolizing enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2010, 154:103–116.
Weinshilboum, R. Pharmacogenetics of methylation: relationship to drug metabolism. Clin Biochem 1988, 21:201–210.
Kiss, LE, Soares‐da‐Silva, P. Medicinal chemistry of catechol O‐methyltransferase (COMT) inhibitors and their therapeutic utility. J Med Chem 2014, 57:8692–8717.
Tsao, D, Liu, S, Dokholyan, NV. Regioselectivity of catechol O‐methyltransferase confers enhancement of catalytic activity. Chem Phys Lett 2011, 506:135–138.
Mollard, P, Pfister, G. Catechol derivatives for treatment of oxidative stress diseases, 2016. Available at: https://www.google.com/patents/US9464016
Cao, Y, Chen, Z‐J, Jiang, H‐D, Chen, J‐Z. Computational studies of the regioselectivities of COMT‐catalyzed meta‐/para‐O‐methylations of luteolin and quercetin. J Phys Chem B 2014, 118:470–481.
Oselin, K, Anier, K. Inhibition of human thiopurine S‐methyltransferase by various nonsteroidal anti‐inflammatory drugs in vitro: a mechanism for possible drug interactions. Drug Metab Dispos 2007, 35:1452–1454.
Srimartpirom, S, Tassaneeyakul, W, Kukongviriyapan, V, Tassaneeyakul, W. Thiopurine S‐methyltransferase genetic polymorphism in the Thai population. Br J Clin Pharmacol 2004, 58:66–70.
Weinshilboum, RM, Otterness, DM, Szumlanski, CL. Methylation pharmacogenetics: catechol O‐methyltransferase, thiopurine methyltransferase, and histamine N‐methyltransferase. Annu Rev Pharmacol Toxicol 1999, 39:19–52.
Männistö, PT, Kaakkola, S. Catechol‐O‐methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol Rev 1999, 51:593–628.
Marsala, SZ, Gioulis, M, Ceravolo, R, Tinazzi, M. A systematic review of catechol‐O‐methyltransferase inhibitors: efficacy and safety in clinical practice. Clin Neuropharmacol 2012, 35:185–190.
Palma, PN, Bonifácio, MJ, Loureiro, AI, Wright, LC, Learmonth, DA, Soares‐da‐Silva, P. Molecular modeling and metabolic studies of the interaction of catechol‐O‐methyltransferase and a new nitrocatechol inhibitor. Drug Metab Dispos 2003, 31:250–258.
Palma, PN, Rodrigues, ML, Archer, M, Bonifácio, MJ, Loureiro, AI, Learmonth, DA, Carrondo, MA, Soares‐da‐Silva, P. Comparative Study of ortho and meta‐nitrated inhibitors of catechol‐O‐methyltransferase: interactions with the active site and regioselectivity of O‐methylation. Mol Pharmacol 2006, 70:143–153.
Chen, D, Wang, CY, Lambert, JD, Ai, N, Welsh, WJ, Yang, CS. Inhibition of human liver catechol‐O‐methyltransferase by tea catechins and their metabolites: structure‐activity relationship and molecular‐modeling studies. Biochem Pharmacol 2005, 69:1523–1531.
Tervo, AJ, Nyronen, TH, Ronkko, T, Poso, A. A structure‐activity relationship study of catechol‐O‐methyltransferase inhibitors combining molecular docking and 3D QSAR methods. J Comput Aided Mol Des 2003, 17:797–810.
Ai, C, Wang, Y, Li, Y, Li, Y, Yang, L. A 3D QSAR study of catechol‐O‐methyltransferase inhibitors using CoMFA and CoMSIA. Mol Inform 2008, 27:1183–1192.
He, S, Li, M, Ye, X, Wang, H, Yu, W, He, W, Wang, Y, Qiao, Y. Site of metabolism prediction for oxidation reactions mediated by oxidoreductases based on chemical bond. Bioinformatics 2016, 33:363–372.
García, I, Fall, Y, Gómez, G, González‐Díaz, H. First computational chemistry multi‐target model for anti‐Alzheimer, anti‐parasitic, anti‐fungi, and anti‐bacterial activity of GSK‐3 inhibitors in vitro, in vivo, and in different cellular lines. Mol Divers 2011, 15:561–567.
Speck‐Planche, A, Cordeiro, MNDS. Simultaneous virtual prediction of anti‐Escherichia coli activities and ADMET profiles: a chemoinformatic complementary approach for high‐throughput screening. ACS Comb Sci 2014, 16:78–84.
Speck‐Planche, A, Nds, CM. Computer‐aided discovery in antimicrobial research: in silico model for virtual screening of potent and safe anti‐pseudomonas agents. Comb Chem High Throughput Screen 2015, 18:305–314.
Speck‐Planche, A, Cordeiro, MNDS. Chemoinformatics for medicinal chemistry: in silico model to enable the discovery of potent and safer anti‐cocci agents. Future Med Chem 2014, 6:2013–2028.
Bohnert, T, Patel, A, Templeton, I, Chen, Y, Lu, C, Lai, G, Leung, L, Tse, S, Einolf, HJ, Wang, Y‐H, et al. Evaluation of a new molecular entity as a victim of metabolic drug–drug interactions: an industry perspective. Drug Metab Dispos 2016, 44:1399–1423.