Metabolic Stability for Drug Discovery and Development

Metabolic stability refers to the susceptibility of compounds to biotransformation in the context of selecting and/or designing drugs with favourable pharmacokinetic properties. Metabolic stability results are usually reported as measures of intrinsic clearance, from which secondary pharmacokinetic parameters such as bioavailability and half-life can be calculated when other data on volume of distribution and fraction absorbed are available. Since these parameters are very important in defining the pharmacological and toxicological profile of drugs as well as patient compliance, the pharmaceutical industry has a particular interest in optimising for metabolic stability during the drug discovery and development process. In the early phases of drug discovery, new chemical entities cannot be administered to humans; hence, predictions of these properties have to be made from in vivo animal, in vitro cellular/subcellular and computational systems. The utility of these systems to define the metabolic stability of compounds that is predictive of the human situation will be reviewed here. The timing of performing the studies in the discovery process and the impact of recent advances in research on drug absorption, distribution, metabolism and excretion (ADME) will be evaluated with respect to the scope and depth of metabolic stability issues.Quantitative prediction of in vivo clearance from in vitro metabolism data has, for many compounds, been shown to be poor in retrospective studies. One explanation for this may be that there are components used in the equations for scaling that are missing or uncertain and should be an area of more research. For example, as a result of increased biochemical understanding of drug metabolism, old assumptions (e.g. that the liver is the principal site of first-pass metabolism) need revision and new knowledge (e.g. the relationship between transporters and drug metabolising enzymes) needs to be incorporated into in vitro-in vivo correlation models. With ADME parameters increasingly being determined on automated platforms, instead of using results from high throughput screening (HTS) campaigns as simple go/no-go filters, the time saved and the many compounds analysed using the robots should be invested in careful processing of the data. A logical step would be to investigate the potential to construct computational models to understand the factors governing metabolic stability. A rational approach to the use of HTS assays should aim to screen for many properties (e.g. physicochemical parameters, absorption, metabolism, protein binding, pharmacokinetics in animals and pharmacology) in an integrated manner rather than screen against one property on many compounds, since it is likely that the final drug will represent a global average of these properties.

[1]  R. Wattiaux,et al.  Tissue fractionation studies. 6. Intracellular distribution patterns of enzymes in rat-liver tissue. , 1955, The Biochemical journal.

[2]  Grant R. Wilkinson,et al.  A physiological approach to hepatic drug clearance , 1975 .

[3]  M. Rowland,et al.  Hepatic elimination--dispersion model. , 1985, Journal of pharmaceutical sciences.

[4]  C. Hansch,et al.  Quantitative structure-activity relationships of cytochrome P-450. , 1993, Drug metabolism reviews.

[5]  J B Houston,et al.  Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. , 1994, Biochemical pharmacology.

[6]  Jeffrey P. Jones,et al.  Designing safer chemicals: predicting the rates of metabolism of halogenated alkanes. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[7]  C. Waller,et al.  Modeling the cytochrome P450-mediated metabolism of chlorinated volatile organic compounds. , 1996, Drug metabolism and disposition: the biological fate of chemicals.

[8]  K. Korzekwa,et al.  Predicting the rates and regioselectivity of reactions mediated by the P450 superfamily. , 1996, Methods in enzymology.

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

[10]  J B Houston,et al.  Prediction of hepatic clearance from microsomes, hepatocytes, and liver slices. , 1997, Drug metabolism reviews.

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

[12]  D J Rance,et al.  The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. , 1997, The Journal of pharmacology and experimental therapeutics.

[13]  T Ishizaki,et al.  Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data. , 1997, Pharmacology & therapeutics.

[14]  J. Strong,et al.  Effect of albumin on the estimation, in vitro, of phenytoin Vmax and Km values: implications for clinical correlation. , 1997, The Journal of pharmacology and experimental therapeutics.

[15]  J B Houston,et al.  Scaling factors to relate drug metabolic clearance in hepatic microsomes, isolated hepatocytes, and the intact liver: studies with induced livers involving diazepam. , 1997, Drug metabolism and disposition: the biological fate of chemicals.

[16]  P. Ortiz de Montellano,et al.  Structure-function of cytochromes P450 and flavin-containing monooxygenases: implications for drug metabolism. , 1998, Drug metabolism and disposition: the biological fate of chemicals.

[17]  Y. Sugiyama,et al.  Quantitative prediction of in vivo drug clearance and drug interactions from in vitro data on metabolism, together with binding and transport. , 1998, Annual review of pharmacology and toxicology.

[18]  F. Oesch,et al.  Drug metabolizing capacity of cryopreserved human, rat, and mouse liver parenchymal cells in suspension. , 1999, Drug Metabolism And Disposition.

[19]  J. Venhorst,et al.  Design, synthesis, and characterization of 7-methoxy-4-(aminomethyl)coumarin as a novel and selective cytochrome P450 2D6 substrate suitable for high-throughput screening. , 1999, Chemical research in toxicology.

[20]  N. Bodor,et al.  Quantitative structure-metabolism relationships: steric and nonsteric effects in the enzymatic hydrolysis of noncongener carboxylic esters. , 1999, Journal of medicinal chemistry.

[21]  G. L. Kedderis,et al.  Present status of the application of cryopreserved hepatocytes in the evaluation of xenobiotics: consensus of an international expert panel. , 1999, Chemico-biological interactions.

[22]  D. Lewis,et al.  Cytochrome P450 Substrate Specificities, Substrate Structural Templates and Enzyme Active Site Geometries , 1999, Drug metabolism and drug interactions.

[23]  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.

[24]  R. Obach,et al.  Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[25]  J. Houston,et al.  Microsomal prediction of in vivo clearance of CYP2C9 substrates in humans. , 1999, British journal of clinical pharmacology.

[26]  D. Lewis,et al.  On the recognition of mammalian microsomal cytochrome P450 substrates and their characteristics: towards the prediction of human p450 substrate specificity and metabolism. , 2000, Biochemical pharmacology.

[27]  J. Miners,et al.  Nonspecific binding of drugs to human liver microsomes. , 2000, British journal of clinical pharmacology.

[28]  D. Greenblatt,et al.  Microsomal binding of amitriptyline: effect on estimation of enzyme kinetic parameters in vitro. , 2000, The Journal of pharmacology and experimental therapeutics.

[29]  David W. Opitz,et al.  Use of Statistical and Neural Net Approaches in Predicting Toxicity of Chemicals , 2000, J. Chem. Inf. Comput. Sci..

[30]  I. A. D. de Graaf,et al.  Increased post-thaw viability and phase I and II biotransformation activity in cryopreserved rat liver slices after improvement of a fast-freezing method. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[31]  John G. Topliss,et al.  QSAR Model for Drug Human Oral Bioavailability1 , 2000 .

[32]  S. Ekins,et al.  Three-dimensional quantitative structure activity relationship computational approaches for prediction of human in vitro intrinsic clearance. , 2000, The Journal of pharmacology and experimental therapeutics.

[33]  R. E. White,et al.  High-throughput screening in drug metabolism and pharmacokinetic support of drug discovery. , 2000, Annual review of pharmacology and toxicology.

[34]  A. Sohlenius-Sternbeck,et al.  High conservation of both phase I and II drug-metabolizing activities in cryopreserved rat liver slices , 2000, Xenobiotica; the fate of foreign compounds in biological systems.

[35]  D. Lewis Modelling Human Cytochromes P450 for Evaluating Drug Metabolism: An Update , 2000, Drug metabolism and drug interactions.

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

[37]  J. Barrett MOLECULAR AND ENVIRONMENTAL CAUSES OF CANCER , 2000, Drug metabolism reviews.

[38]  D. Cross,et al.  A COMMENTARY ON THE USE OF HEPATOCYTES IN DRUG METABOLISM STUDIES DURING DRUG DISCOVERY AND DEVELOPMENT* , 2000, Drug metabolism reviews.

[39]  Y. Ishii,et al.  A convenient in vitro screening method for predicting in vivo drug metabolic clearance using isolated hepatocytes suspended in serum. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[40]  P. Jeffrey,et al.  Utility of metabolic stability screening: comparison of in vitro and in vivo clearance , 2001, Xenobiotica; the fate of foreign compounds in biological systems.

[41]  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.

[42]  S. Surapaneni,et al.  Role of drug metabolism in drug discovery and development , 2001, Medicinal research reviews.

[43]  C. Masimirembwa,et al.  In vitro high throughput screening of compounds for favorable metabolic properties in drug discovery. , 2001, Combinatorial chemistry & high throughput screening.

[44]  W. L. Nelson,et al.  Importance of amine pKa and distribution coefficient in the metabolism of fluorinated propranolol analogs: metabolism by CYP1A2. , 2001, Drug metabolism and disposition: the biological fate of chemicals.

[45]  C. Masimirembwa,et al.  Competitive CYP2C9 inhibitors: enzyme inhibition studies, protein homology modeling, and three-dimensional quantitative structure-activity relationship analysis. , 2001, Molecular pharmacology.

[46]  D. Greenblatt,et al.  Application of the relative activity factor approach in scaling from heterologously expressed cytochromes p450 to human liver microsomes: studies on amitriptyline as a model substrate. , 2001, The Journal of pharmacology and experimental therapeutics.

[47]  T N Thompson,et al.  Optimization of metabolic stability as a goal of modern drug design , 2001, Medicinal research reviews.

[48]  Gerd Folkers,et al.  Pharmacokinetic optimization in drug research , 2001 .

[49]  Sean Ekins,et al.  Pharmacophore modeling of cytochromes P450. , 2002, Advanced drug delivery reviews.

[50]  Anthony Long,et al.  Computer systems for the prediction of xenobiotic metabolism. , 2002, Advanced drug delivery reviews.

[51]  N. Takenaga,et al.  Structure-activity relationship in O-glucuronidation of indolocarbazole analogs. , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[52]  Sean Ekins,et al.  A pharmacophore for human pregnane X receptor ligands. , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[53]  Gabriele Cruciani,et al.  Predicting drug metabolism: a site of metabolism prediction tool applied to the cytochrome P450 2C9. , 2003, Journal of medicinal chemistry.

[54]  M. Morris,et al.  Membrane Transport in Hepatic Clearance of Drugs I: Extended Hepatic Clearance Models Incorporating Concentration-Dependent Transport and Elimination Processes , 1997, Pharmaceutical Research.

[55]  R. J. Riley,et al.  Development of a Generalized, Quantitative Physicochemical Model of CYP3A4 Inhibition for Use in Early Drug Discovery , 2001, Pharmaceutical Research.

[56]  Michael S. Roberts,et al.  A dispersion model of hepatic elimination: 1. Formulation of the model and bolus considerations , 1986, Journal of Pharmacokinetics and Biopharmaceutics.

[57]  Malcolm Rowland,et al.  Hepatic clearance of drugs. I. Theoretical considerations of a “well-stirred” model and a “parallel tube” model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance , 1977, Journal of Pharmacokinetics and Biopharmaceutics.

[58]  Malcolm Rowland,et al.  Clearance concepts in pharmacokinetics , 1973, Journal of Pharmacokinetics and Biopharmaceutics.