2D SMARTCyp Reactivity-Based Site of Metabolism Prediction for Major Drug-Metabolizing Cytochrome P450 Enzymes

Cytochrome P450 (CYP) 3A4, 2D6, 2C9, 2C19, and 1A2 are the most important drug-metabolizing enzymes in the human liver. Knowledge of which parts of a drug molecule are subject to metabolic reactions catalyzed by these enzymes is crucial for rational drug design to mitigate ADME/toxicity issues. SMARTCyp, a recently developed 2D ligand structure-based method, is able to predict site-specific metabolic reactivity of CYP3A4 and CYP2D6 substrates with an accuracy that rivals the best and more computationally demanding 3D structure-based methods. In this article, the SMARTCyp approach was extended to predict the metabolic hotspots for CYP2C9, CYP2C19, and CYP1A2 substrates. This was accomplished by taking into account the impact of a key substrate-receptor recognition feature of each enzyme as a correction term to the SMARTCyp reactivity. The corrected reactivity was then used to rank order the likely sites of CYP-mediated metabolic reactions. For 60 CYP1A2 substrates, the observed major sites of CYP1A2 catalyzed metabolic reactions were among the top-ranked 1, 2, and 3 positions in 67%, 80%, and 83% of the cases, respectively. The results were similar to those obtained by MetaSite and the reactivity + docking approach. For 70 CYP2C9 substrates, the observed sites of CYP2C9 metabolism were among the top-ranked 1, 2, and 3 positions in 66%, 86%, and 87% of the cases, respectively. These results were better than the corresponding results of StarDrop version 5.0, which were 61%, 73%, and 77%, respectively. For 36 compounds metabolized by CYP2C19, the observed sites of metabolism were found to be among the top-ranked 1, 2, and 3 sites in 78%, 89%, and 94% of the cases, respectively. The computational procedure was implemented as an extension to the program SMARTCyp 2.0. With the extension, the program can now predict the site of metabolism for all five major drug-metabolizing enzymes with an accuracy similar to or better than that achieved by the best 3D structure-based methods. Both the Java source code and the binary executable of the program are freely available to interested users.

[1]  James R. Halpert,et al.  An open conformation of mammalian cytochrome P450 2B4 at 1.6-Å resolution , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[2]  Lars Olsen,et al.  Ligand-Based Site of Metabolism Prediction for Cytochrome P450 2D6. , 2012, ACS medicinal chemistry letters.

[3]  F. Guengerich,et al.  Common and uncommon cytochrome P450 reactions related to metabolism and chemical toxicity. , 2001, Chemical research in toxicology.

[4]  David E. Gloriam,et al.  SMARTCyp: A 2D Method for Prediction of Cytochrome P450-Mediated Drug Metabolism. , 2010, ACS medicinal chemistry letters.

[5]  G. Cruciani,et al.  MetaSite: understanding metabolism in human cytochromes from the perspective of the chemist. , 2005, Journal of medicinal chemistry.

[6]  Barry C. Jones,et al.  DRUG-DRUG INTERACTIONS FOR UDP-GLUCURONOSYLTRANSFERASE SUBSTRATES: A PHARMACOKINETIC EXPLANATION FOR TYPICALLY OBSERVED LOW EXPOSURE (AUCI/AUC) RATIOS , 2004, Drug Metabolism and Disposition.

[7]  H. Yamazaki,et al.  Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. , 1994, The Journal of pharmacology and experimental therapeutics.

[8]  F. Guengerich Cytochrome p450 and chemical toxicology. , 2008, Chemical research in toxicology.

[9]  David S. Wishart,et al.  DrugBank 3.0: a comprehensive resource for ‘Omics’ research on drugs , 2010, Nucleic Acids Res..

[10]  Olivier Taboureau,et al.  Virtual Screening and Prediction of Site of Metabolism for Cytochrome P450 1A2 Ligands , 2009, J. Chem. Inf. Model..

[11]  Barry C. Jones,et al.  Properties of cytochrome P450 isoenzymes and their substrates Part 1: active site characteristics , 1997 .

[12]  C David Stout,et al.  Adaptations for the Oxidation of Polycyclic Aromatic Hydrocarbons Exhibited by the Structure of Human P450 1A2*♦ , 2007, Journal of Biological Chemistry.

[13]  Arnout Ceulemans,et al.  Structure-based site of metabolism prediction for cytochrome P450 2D6. , 2011, Journal of medicinal chemistry.

[14]  A. Alex,et al.  A novel approach to predicting P450 mediated drug metabolism. CYP2D6 catalyzed N-dealkylation reactions and qualitative metabolite predictions using a combined protein and pharmacophore model for CYP2D6. , 1999, Journal of medicinal chemistry.

[15]  Sandor Vajda,et al.  Ensemble modeling of substrate binding to cytochromes P450: analysis of catalytic differences between CYP1A orthologs. , 2007, Biochemistry.

[16]  D E McRee,et al.  Mammalian microsomal cytochrome P450 monooxygenase: structural adaptations for membrane binding and functional diversity. , 2000, Molecular cell.

[17]  L. Pedersen,et al.  Identification of Residues 99, 220, and 221 of Human Cytochrome P450 2C19 as Key Determinants of Omeprazole Hydroxylase Activity (*) , 1996, The Journal of Biological Chemistry.

[18]  Elizabeth Yuriev,et al.  Challenges and advances in computational docking: 2009 in review , 2011, Journal of molecular recognition : JMR.

[19]  S. Asahi,et al.  Important amino acid residues that confer CYP2C19 selective activity to CYP2C9. , 2008, Journal of biochemistry.

[20]  William L. Jorgensen,et al.  Journal of Chemical Information and Modeling , 2005, J. Chem. Inf. Model..

[21]  Jin-Young Park,et al.  Construction and assessment of models of CYP2E1: predictions of metabolism from docking, molecular dynamics, and density functional theoretical calculations. , 2003, Journal of medicinal chemistry.

[22]  S. Ekins,et al.  Pharmacophore and three-dimensional quantitative structure activity relationship methods for modeling cytochrome p450 active sites. , 2001, Drug metabolism and disposition: the biological fate of chemicals.

[23]  T. Lynch,et al.  The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. , 2007, American family physician.

[24]  Jeffrey P. Jones,et al.  Charge and substituent effects on affinity and metabolism of benzbromarone-based CYP2C19 inhibitors. , 2004, Journal of medicinal chemistry.

[25]  Dennis A. Smith,et al.  Properties of cytochrome P450 isoenzymes and their substrates Part 2: properties of cytochrome P450 substrates , 1997 .

[26]  G. Wilkinson,et al.  Drug metabolism and variability among patients in drug response. , 2005, The New England journal of medicine.

[27]  Lars Carlsson,et al.  State-of-the-art Tools for Computational Site of Metabolism Predictions: Comparative Analysis, Mechanistical Insights, and Future Applications , 2007, Drug metabolism reviews.

[28]  Eric F. Johnson,et al.  The Structure of Human Cytochrome P450 2C9 Complexed with Flurbiprofen at 2.0-Å Resolution* , 2004, Journal of Biological Chemistry.

[29]  P. Danielson,et al.  The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans. , 2002, Current drug metabolism.

[30]  T. Richardson,et al.  Identification of amino acid substitutions that confer a high affinity for sulfaphenazole binding and a high catalytic efficiency for warfarin metabolism to P450 2C19. , 1998, Biochemistry.

[31]  Chris Oostenbrink,et al.  Catalytic site prediction and virtual screening of cytochrome P450 2D6 substrates by consideration of water and rescoring in automated docking. , 2006, Journal of medicinal chemistry.

[32]  Helmut Sigel,et al.  The ubiquitous roles of cytochrome P450 proteins , 2007 .

[33]  Guoying Tai,et al.  Re-engineering of CYP2C9 to Probe Acid-Base Substrate Selectivity , 2008, Drug Metabolism and Disposition.

[34]  Matthew J Sykes,et al.  Prediction of metabolism by cytochrome P450 2C9: alignment and docking studies of a validated database of substrates. , 2008, Journal of medicinal chemistry.

[35]  Diansong Zhou,et al.  COMPARISON OF METHODS FOR THE PREDICTION OF THE METABOLIC SITES FOR CYP3A4-MEDIATED METABOLIC REACTIONS , 2006, Drug Metabolism and Disposition.

[36]  F Peter Guengerich,et al.  Role of glutamic acid 216 in cytochrome P450 2D6 substrate binding and catalysis. , 2003, Biochemistry.

[37]  M H Tarbit,et al.  Molecular modelling of human CYP2C subfamily enzymes CYP2C9 and CYP2C19: rationalization of substrate specificity and site-directed mutagenesis experiments in the CYP2C subfamily. , 1998, Xenobiotica; the fate of foreign compounds in biological systems.

[38]  R. Sheridan,et al.  A model for predicting likely sites of CYP3A4-mediated metabolism on drug-like molecules. , 2003, Journal of medicinal chemistry.

[39]  C David Stout,et al.  Structure of Human Microsomal Cytochrome P450 2C8 , 2004, Journal of Biological Chemistry.

[40]  R L Slaughter,et al.  Recent Advances: the Cytochrome P450 Enzymes , 1995, The Annals of pharmacotherapy.

[41]  P. Goodford A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. , 1985, Journal of medicinal chemistry.

[42]  Helmut Sigel,et al.  The Ubiquitous Roles of Cytochrome P450 Proteins: Sigel/The Ubiquitous Roles of Cytochrome P450 Proteins , 2007 .

[43]  Chris Oostenbrink,et al.  Fast Prediction of Cytochrome P450 Mediated Drug Metabolism , 2009, ChemMedChem.

[44]  J. Goldstein,et al.  Identification of residues 99, 220, and 221 of human cytochrome P450 2C19 as key determinants of omeprazole activity. , 1996, The Journal of biological chemistry.

[45]  Hoa Le,et al.  Comparison of metabolic soft spot predictions of CYP3A4, CYP2C9 and CYP2D6 substrates using MetaSite and StarDrop. , 2011, Combinatorial chemistry & high throughput screening.