A probabilistic method to report predictions from a human liver microsomes stability QSAR model: a practical tool for drug discovery

Using data from the in vitro liver microsomes metabolic stability assay, we have developed QSAR models to predict in vitro human clearance. Models were trained using in house high-throughput assay data reported as the predicted human hepatic clearance by liver microsomes or pCLh. Machine learning regression methods were used to generate the models. Model output for a given molecule was reported as its probability of being metabolically stable, thus allowing for synthesis prioritization based on this prediction. Use of probability, instead of the regression value or categories, has been found to be an efficient way for both reporting and assessing predictions. Model performance is evaluated using prospective validation. These models have been integrated into a number of desktop tools, and the models are routinely used to prioritize the synthesis of compounds. We discuss two therapeutic projects at Genentech that exemplify the benefits of a probabilistic approach in applying the models. A three-year retrospective analysis of measured liver microsomes stability data on all registered compounds at Genentech reveals that the use of these models has resulted in an improved metabolic stability profile of synthesized compounds.

[1]  Johan Ulander,et al.  Impact of input parameters on the prediction of hepatic plasma clearance using the well-stirred model. , 2010, Current drug metabolism.

[2]  BinQing Wei,et al.  Discovery of a potent, selective, and orally available class I phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) kinase inhibitor (GDC-0980) for the treatment of cancer. , 2011, Journal of medicinal chemistry.

[3]  Li Li,et al.  In silico prediction of cytochrome P450-mediated drug metabolism. , 2011, Combinatorial chemistry & high throughput screening.

[4]  James G. Nourse,et al.  Reoptimization of MDL Keys for Use in Drug Discovery , 2002, J. Chem. Inf. Comput. Sci..

[5]  Lemont B. Kier,et al.  The E-State as the Basis for Molecular Structure Space Definition and Structure Similarity , 2000, J. Chem. Inf. Comput. Sci..

[6]  E. Hodgson,et al.  Organophosphorus Chemicals: Potent Inhibitors of the Human Metabolism of Steroid Hormones and Xenobiotics , 2006, Drug metabolism reviews.

[7]  M. Forina,et al.  Class-modeling techniques, classic and new, for old and new problems , 2008 .

[8]  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.

[9]  Douglas R. Henry,et al.  Optimization of MDL substructure search keys for the prediction of activity and toxicity , 2005 .

[10]  Li Di,et al.  Development of QSAR models for microsomal stability: identification of good and bad structural features for rat, human and mouse microsomal stability , 2010, J. Comput. Aided Mol. Des..

[11]  Robert P. Sheridan,et al.  Similarity to Molecules in the Training Set Is a Good Discriminator for Prediction Accuracy in QSAR , 2004, J. Chem. Inf. Model..

[12]  Yvonne C. Martin,et al.  Application of Belief Theory to Similarity Data Fusion for Use in Analog Searching and Lead Hopping , 2008, J. Chem. Inf. Model..

[13]  David B Duignan,et al.  High throughput ADME screening: practical considerations, impact on the portfolio and enabler of in silico ADME models. , 2008, Current drug metabolism.

[14]  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.

[15]  Scott P. Brown,et al.  A unified, probabilistic framework for structure- and ligand-based virtual screening. , 2011, Journal of medicinal chemistry.

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

[17]  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.

[18]  A. Ghose,et al.  Prediction of Hydrophobic (Lipophilic) Properties of Small Organic Molecules Using Fragmental Methods: An Analysis of ALOGP and CLOGP Methods , 1998 .

[19]  M. Jamei,et al.  Misuse of the Well-Stirred Model of Hepatic Drug Clearance , 2007, Drug Metabolism and Disposition.

[20]  J. Halladay,et al.  Metabolic stability screen for drug discovery using cassette analysis and column switching. , 2007, Drug metabolism letters.

[21]  P. Hajduk,et al.  Cheminformatic tools for medicinal chemists. , 2010, Journal of medicinal chemistry.

[22]  Wendy A. Warr,et al.  Guiding effective decisions: an interview with Matthew Segall, CEO of Optibrium , 2011, J. Comput. Aided Mol. Des..

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

[24]  Andreas Bender,et al.  Computational Prediction of Metabolism: Sites, Products, SAR, P450 Enzyme Dynamics, and Mechanisms , 2012, J. Chem. Inf. Model..

[25]  Ralph Kühne,et al.  External Validation and Prediction Employing the Predictive Squared Correlation Coefficient Test Set Activity Mean vs Training Set Activity Mean , 2008, J. Chem. Inf. Model..

[26]  Sean Ekins,et al.  Using Open Source Computational Tools for Predicting Human Metabolic Stability and Additional Absorption, Distribution, Metabolism, Excretion, and Toxicity Properties , 2010, Drug Metabolism and Disposition.

[27]  Jean Carletta,et al.  Assessing Agreement on Classification Tasks: The Kappa Statistic , 1996, CL.

[28]  Ryan H. Lilien,et al.  Applying Medicinal Chemistry Transformations and Multiparameter Optimization to Guide the Search for High-Quality Leads and Candidates , 2011, J. Chem. Inf. Model..

[29]  Yojiro Sakiyama,et al.  Predicting human liver microsomal stability with machine learning techniques. , 2008, Journal of molecular graphics & modelling.

[30]  Man-Ling Lee,et al.  DEGAS: Sharing and Tracking Target Compound Ideas with External Collaborators , 2012, J. Chem. Inf. Model..

[31]  John C. Davis,et al.  Significant Species Difference in Amide Hydrolysis of GDC-0834, a Novel Potent and Selective Bruton's Tyrosine Kinase Inhibitor , 2011, Drug Metabolism and Disposition.

[32]  David Rogers,et al.  Extended-Connectivity Fingerprints , 2010, J. Chem. Inf. Model..

[33]  Jane R Kenny,et al.  Discovery and optimization of C-2 methyl imidazopyrrolopyridines as potent and orally bioavailable JAK1 inhibitors with selectivity over JAK2. , 2012, Journal of medicinal chemistry.

[34]  Supa Hannongbua,et al.  In-silico ADME models: a general assessment of their utility in drug discovery applications. , 2011, Current topics in medicinal chemistry.

[35]  Jing Lu,et al.  Development of in silico models for human liver microsomal stability , 2007, J. Comput. Aided Mol. Des..

[36]  G R Wilkinson,et al.  Commentary: a physiological approach to hepatic drug clearance. , 1975, Clinical pharmacology and therapeutics.

[37]  Shane Weaver,et al.  The importance of the domain of applicability in QSAR modeling. , 2008, Journal of molecular graphics & modelling.

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

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

[40]  Gunnar Rätsch,et al.  Active Learning with Support Vector Machines in the Drug Discovery Process , 2003, J. Chem. Inf. Comput. Sci..

[41]  Jan M. Kriegl,et al.  Computational approaches to predict drug metabolism , 2009, Expert opinion on drug metabolism & toxicology.

[42]  Glenn Shafer,et al.  A Mathematical Theory of Evidence , 2020, A Mathematical Theory of Evidence.

[43]  Jane R Kenny,et al.  Identification of C-2 hydroxyethyl imidazopyrrolopyridines as potent JAK1 inhibitors with favorable physicochemical properties and high selectivity over JAK2. , 2013, Journal of medicinal chemistry.

[44]  Bohdan Waszkowycz,et al.  Identification of imidazo-pyrrolopyridines as novel and potent JAK1 inhibitors. , 2012, Journal of medicinal chemistry.

[45]  Ovidiu Ivanciuc,et al.  Applications of Support Vector Machines in Chemistry , 2007 .

[46]  Gary Box,et al.  The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer . , 2008, Journal of medicinal chemistry.