Comprehensive Characterization of Cytochrome P450 Isozyme Selectivity across Chemical Libraries

The cytochrome P450 (CYP) gene family catalyzes drug metabolism and bioactivation and is therefore relevant to drug development. We determined potency values for 17,143 compounds against five recombinant CYP isozymes (1A2, 2C9, 2C19, 2D6 and 3A4) using an in vitro bioluminescent assay. The compounds included libraries of US Food and Drug Administration (FDA)-approved drugs and screening libraries. We observed cross-library isozyme inhibition (30–78%) with important differences between libraries. Whereas only 7% of the typical screening library was inactive against all five isozymes, 33% of FDA-approved drugs were inactive, reflecting the optimized pharmacological properties of the latter. Our results suggest that low CYP 2C isozyme activity is a common property of drugs, whereas other isozymes, such as CYP 2D6, show little discrimination between drugs and unoptimized compounds found in screening libraries. We also identified chemical substructures that differentiated between the five isozymes. The pharmacological compendium described here should further the understanding of CYP isozymes.

[1]  P. Hollenberg Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes , 2002, Drug metabolism reviews.

[2]  Ruili Huang,et al.  Characterization of diversity in toxicity mechanism using in vitro cytotoxicity assays in quantitative high throughput screening. , 2008, Chemical research in toxicology.

[3]  Chris de Graaf,et al.  Cytochrome p450 in silico: an integrative modeling approach. , 2005, Journal of medicinal chemistry.

[4]  Erkki Oja,et al.  Computing with neural networks. , 1987, Science.

[5]  Teuvo Kohonen,et al.  Self-organizing neural projections , 2006, Neural Networks.

[6]  M H Tarbit,et al.  Structural determinants of cytochrome P450 substrate specificity, binding affinity and catalytic rate. , 1998, Chemico-biological interactions.

[7]  Paul J Hergenrother,et al.  Identification of promiscuous small molecule activators in high-throughput enzyme activation screens. , 2008, Journal of medicinal chemistry.

[8]  Noel Southall,et al.  A Cell-Based Assay for IκBα Stabilization Using A Two-Color Dual Luciferase-Based Sensor , 2007 .

[9]  M. Relling,et al.  Pharmacogenomics: translating functional genomics into rational therapeutics. , 1999, Science.

[10]  J B Houston,et al.  CYP3A4 drug interactions: correlation of 10 in vitro probe substrates. , 1999, British journal of clinical pharmacology.

[11]  D. Back,et al.  Differential selectivity of cytochrome P450 inhibitors against probe substrates in human and rat liver microsomes. , 1998, British journal of clinical pharmacology.

[12]  Li Di,et al.  Application of pharmaceutical profiling assays for optimization of drug-like properties. , 2005, Current opinion in drug discovery & development.

[13]  C. Lipinski Drug-like properties and the causes of poor solubility and poor permeability. , 2000, Journal of pharmacological and toxicological methods.

[14]  H. Yamazaki,et al.  Cytochrome P450-dependent drug oxidation activities in liver microsomes of various animal species including rats, guinea pigs, dogs, monkeys, and humans , 1997, Archives of Toxicology.

[15]  M. J. Coon,et al.  Cytochrome P-450 : multiplicity of isoforms, substrates, and catalytic and regulatory mechanisms , 1991 .

[16]  Thomas D. Y. Chung,et al.  A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays , 1999, Journal of biomolecular screening.

[17]  Teuvo Kohonen,et al.  The self-organizing map , 1990 .

[18]  Corwin Hansch,et al.  QSAR and ADME. , 2004, Bioorganic & medicinal chemistry.

[19]  J. Marois,et al.  Description of a 96-well plate assay to measure cytochrome P4503A inhibition in human liver microsomes using a selective fluorescent probe. , 1999, Analytical biochemistry.

[20]  Chris de Graaf,et al.  Binding mode prediction of cytochrome p450 and thymidine kinase protein-ligand complexes by consideration of water and rescoring in automated docking. , 2005, Journal of medicinal chemistry.

[21]  Ruili Huang,et al.  Compound Cytotoxicity Profiling Using Quantitative High-Throughput Screening , 2007, Environmental health perspectives.

[22]  Slobodan Petar Rendic Summary of information on human CYP enzymes: human P450 metabolism data , 2002, Drug metabolism reviews.

[23]  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.

[24]  Christopher P Austin,et al.  A high-throughput screen for aggregation-based inhibition in a large compound library. , 2007, Journal of medicinal chemistry.

[25]  David Raunig,et al.  In vitro drug interactions of cytochrome p450: an evaluation of fluorogenic to conventional substrates. , 2003, Drug metabolism and disposition: the biological fate of chemicals.

[26]  David J. Liu,et al.  Luminogenic cytochrome P450 assays , 2006, Expert opinion on drug metabolism & toxicology.

[27]  L. Wienkers,et al.  Predicting in vivo drug interactions from in vitro drug discovery data , 2005, Nature Reviews Drug Discovery.

[28]  Yuko Ito,et al.  Human cytochromes P450 in the metabolism of drugs: new molecular models of enzyme–substrate interactions , 2008 .

[29]  Christopher P Austin,et al.  Identification of pregnane X receptor ligands using time-resolved fluorescence resonance energy transfer and quantitative high-throughput screening. , 2009, Assay and drug development technologies.

[30]  Corwin Hansch,et al.  QSAR of Cytochrome P450 , 2004, Drug metabolism reviews.

[31]  Principal Component Analysis of CYP2C9 and CYP3A4 Probe Substrate/Inhibitor Panels , 2008, Drug Metabolism and Disposition.

[32]  M. Schulz,et al.  Therapeutic and toxic blood concentrations of more than 800 drugs and other xenobiotics. , 2003, Die Pharmazie.

[33]  Christopher P Austin,et al.  Three classes of glucocerebrosidase inhibitors identified by quantitative high-throughput screening are chaperone leads for Gaucher disease , 2007, Proceedings of the National Academy of Sciences.

[34]  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.

[35]  Sam Michael,et al.  Compound Management for Quantitative High-Throughput Screening , 2008, JALA.

[36]  Jan M. Kriegl,et al.  Chapter 5 Linear Quantitative Structure–Activity Relationships for the Interaction of Small Molecules with Human Cytochrome P450 Isoenzymes , 2007 .

[37]  D. Rock,et al.  CYP2C9 Inhibition: Impact of Probe Selection and Pharmacogenetics on in Vitro Inhibition Profiles , 2006, Drug Metabolism and Disposition.

[38]  D. Greenblatt,et al.  Phenacetin O-deethylation by human liver microsomes in vitro: inhibition by chemical probes, SSRI antidepressants, nefazodone and venlafaxine , 1996, Psychopharmacology.

[39]  P. R. Montellano Cytochrome P-450 , 1986, Springer US.

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

[41]  Brian K Shoichet,et al.  Interpreting steep dose-response curves in early inhibitor discovery. , 2006, Journal of medicinal chemistry.

[42]  Christopher P Austin,et al.  A basis for reduced chemical library inhibition of firefly luciferase obtained from directed evolution. , 2009, Journal of medicinal chemistry.

[43]  Adam Yasgar,et al.  Quantitative high-throughput screening: a titration-based approach that efficiently identifies biological activities in large chemical libraries. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[44]  Kim E. Garbison,et al.  The Minimum Significant Ratio: A Statistical Parameter to Characterize the Reproducibility of Potency Estimates from Concentration-Response Assays and Estimation by Replicate-Experiment Studies , 2006, Journal of biomolecular screening.

[45]  R. Foti,et al.  CYP2C19 Inhibition: The Impact of Substrate Probe Selection on in Vitro Inhibition Profiles , 2008, Drug Metabolism and Disposition.

[46]  Ortiz de Montellano,et al.  Cytochrome P-450: Structure, Mechanism, and Biochemistry , 1986 .

[47]  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.

[48]  David Weininger,et al.  SMILES, a chemical language and information system. 1. Introduction to methodology and encoding rules , 1988, J. Chem. Inf. Comput. Sci..

[49]  G. Zlokarnik,et al.  High throughput P450 inhibition screens in early drug discovery. , 2005, Drug discovery today.

[50]  Eric M. Gifford,et al.  Development of CYP 3 A 4 Inhibition Models : Comparisons of Machine-Learning Techniques and , 2005 .