Integration of in silico and in vitro tools for scaffold optimization during drug discovery: predicting P-glycoprotein efflux.

In silico tools are regularly utilized for designing and prioritizing compounds to address challenges related to drug metabolism and pharmacokinetics (DMPK) during the process of drug discovery. P-Glycoprotein (P-gp) is a member of the ATP-binding cassette (ABC) transporters with broad substrate specificity that plays a significant role in absorption and distribution of drugs that are P-gp substrates. As a result, screening for P-gp transport has now become routine in the drug discovery process. Typically, bidirectional permeability assays are employed to assess in vitro P-gp efflux. In this article, we use P-gp as an example to illustrate a well-validated methodology to effectively integrate in silico and in vitro tools to identify and resolve key barriers during the early stages of drug discovery. A detailed account of development and application of in silico tools such as simple guidelines based on physicochemical properties and more complex quantitative structure-activity relationship (QSAR) models is provided. The tools were developed based on structurally diverse data for more than 2000 compounds generated using a robust P-gp substrate assay over the past several years. Analysis of physicochemical properties revealed a significantly lower proportion (<10%) of P-gp substrates among the compounds with topological polar surface area (TPSA) <60 Å(2) and the most basic cpKa <8. In contrast, this proportion of substrates was greater than 75% for compounds with TPSA >60 Å(2) and the most basic cpKa >8. Among the various QSAR models evaluated to predict P-gp efflux, the Bagging model provided optimum prediction performance for prospective validation based on chronological test sets. Four sequential versions of the model were built with increasing numbers of compounds to train the models as new data became available. Except for the first version with the smallest training set, the QSAR models exhibited robust prediction profiles with positive prediction values (PPV) and negative prediction values (NPV) exceeding 80%. The QSAR model demonstrated better concordance with the manual P-gp substrate assay than an automated P-gp substrate screen. The in silico and the in vitro tools have been effectively integrated during early stages of drug discovery to resolve P-gp-related challenges exemplified by several case studies. Key learning based on our experience with P-gp can be widely applicable across other DMPK-related challenges.

[1]  T. Tsuruo,et al.  Purification of the 170- to 180-kilodalton membrane glycoprotein associated with multidrug resistance. 170- to 180-kilodalton membrane glycoprotein is an ATPase. , 1988, The Journal of biological chemistry.

[2]  W. Loh,et al.  SPLIT SELECTION METHODS FOR CLASSIFICATION TREES , 1997 .

[3]  P. Selzer,et al.  Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. , 2000, Journal of medicinal chemistry.

[4]  Steven L. Dixon,et al.  Use of Robust Classification Techniques for the Prediction of Human Cytochrome P450 2D6 Inhibition. , 2003 .

[5]  Michal Vieth,et al.  Structure-guided expansion of kinase fragment libraries driven by support vector machine models. , 2010, Biochimica et biophysica acta.

[6]  Remigijus Didziapetris,et al.  Fragmental Methods in the Design of New Compounds. Applications of The Advanced Algorithm Builder , 2002 .

[7]  A. Tropsha,et al.  Beware of q2! , 2002, Journal of molecular graphics & modelling.

[8]  Yue Weng,et al.  Structure of P-Glycoprotein Reveals a Molecular Basis for Poly-Specific Drug Binding , 2009, Science.

[9]  A. Seelig A general pattern for substrate recognition by P-glycoprotein. , 1998, European journal of biochemistry.

[10]  Leo Breiman,et al.  Bagging Predictors , 1996, Machine Learning.

[11]  B. Kuhn,et al.  Intramolecular hydrogen bonding in medicinal chemistry. , 2010, Journal of medicinal chemistry.

[12]  P. Borst,et al.  What have we learnt thus far from mice with disrupted P-glycoprotein genes? , 1996, European journal of cancer.

[13]  Yiyu Cheng,et al.  Identifying P-Glycoprotein Substrates Using a Support Vector Machine Optimized by a Particle Swarm , 2007, J. Chem. Inf. Model..

[14]  Ronald T. Borchardt,et al.  Quantitative Approaches to Delineate Passive Transport Mechanisms in Cell Culture Monolayers , 1999 .

[15]  Bin Chen,et al.  Comparison of Random Forest and Pipeline Pilot Naïve Bayes in Prospective QSAR Predictions , 2012, J. Chem. Inf. Model..

[16]  Jeffrey W Cramer,et al.  Strategies for conducting ADME studies during lead generation in the drug discovery process. , 2010, IDrugs : the investigational drugs journal.

[17]  J. Greenwood Characterization of a rat retinal endothelial cell culture and the expression of P-glycoprotein in brain and retinal endothelium in vitro , 1992, Journal of Neuroimmunology.

[18]  Thomas J. Raub,et al.  In vitro models of the blood-brain barrier. , 1998, Alternatives to laboratory animals : ATLA.

[19]  J. Riordan,et al.  The mdr1 gene, responsible for multidrug-resistance, codes for P-glycoprotein. , 1986, Biochemical and biophysical research communications.

[20]  Gabriele Cruciani,et al.  A pharmacophore hypothesis for P-glycoprotein substrate recognition using GRIND-based 3D-QSAR. , 2005, Journal of medicinal chemistry.

[21]  Wei Zhou,et al.  High-Dimensional Descriptor Selection and Computational QSAR Modeling for Antitumor Activity of ARC-111 Analogues Based on Support Vector Regression (SVR) , 2012, International journal of molecular sciences.

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

[23]  Jörg Huwyler,et al.  Comparison of in vitro P-glycoprotein screening assays: recommendations for their use in drug discovery. , 2003, Journal of medicinal chemistry.

[24]  Robert P. Sheridan,et al.  Random Forest: A Classification and Regression Tool for Compound Classification and QSAR Modeling , 2003, J. Chem. Inf. Comput. Sci..

[25]  Lei Chen,et al.  ADME evaluation in drug discovery. 10. Predictions of P-glycoprotein inhibitors using recursive partitioning and naive Bayesian classification techniques. , 2011, Molecular pharmaceutics.

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

[27]  A. Seelig,et al.  P-glycoprotein senses its substrates and the lateral membrane packing density: consequences for the catalytic cycle. , 2008, Biochemistry.

[28]  J. Polli,et al.  Rational use of in vitro P-glycoprotein assays in drug discovery. , 2001, The Journal of pharmacology and experimental therapeutics.

[29]  Alexander Golbraikh,et al.  Predictive QSAR modeling based on diversity sampling of experimental datasets for the training and test set selection , 2002, J. Comput. Aided Mol. Des..

[30]  A. Dantzig,et al.  Reversal of P-glycoprotein-mediated multidrug resistance by a potent cyclopropyldibenzosuberane modulator, LY335979. , 1996, Cancer research.

[31]  M. Melamed,et al.  Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[32]  Carol L. Williams,et al.  Transepithelial transport of drugs by the multidrug transporter in cultured Madin-Darby canine kidney cell epithelia. , 1989, The Journal of biological chemistry.

[33]  D. Manallack,et al.  Theoretical hydrogen bonding parameters for drug design. , 2001, Journal of molecular graphics & modelling.

[34]  H. Kubinyi,et al.  Three-dimensional quantitative similarity-activity relationships (3D QSiAR) from SEAL similarity matrices. , 1998, Journal of medicinal chemistry.

[35]  R L Juliano,et al.  A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. , 1976, Biochimica et biophysica acta.

[36]  A. Seelig,et al.  Exploring the P-glycoprotein binding cavity with polyoxyethylene alkyl ethers. , 2010, Biophysical journal.

[37]  T. Tsuruo,et al.  Tissue distribution of P-glycoprotein encoded by a multidrug-resistant gene as revealed by a monoclonal antibody, MRK 16. , 1988, Cancer research.

[38]  M. Yamazaki,et al.  Clinical Relevance of P-Glycoprotein in Drug Therapy , 2003, Drug metabolism reviews.

[39]  Michael H. Abraham,et al.  Scales of solute hydrogen-bonding: their construction and application to physicochemical and biochemical processes , 2010 .

[40]  I. Pastan,et al.  ATP-dependent transport of vinblastine in vesicles from human multidrug-resistant cells. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[41]  Remigijus Didziapetris,et al.  Classification Analysis of P-Glycoprotein Substrate Specificity , 2003, Journal of drug targeting.

[42]  Ying Xue,et al.  Prediction of P‐Glycoprotein Substrates by a Support Vector Machine Approach. , 2004 .

[43]  Thomas J. Raub,et al.  P-glycoprotein recognition of substrates and circumvention through rational drug design. , 2006, Molecular pharmaceutics.

[44]  S. Hitchcock,et al.  Structural modifications that alter the P-glycoprotein efflux properties of compounds. , 2012, Journal of medicinal chemistry.

[45]  J. Hochman,et al.  Evaluation of drug interactions with P-glycoprotein in drug discovery: in vitro assessment of the potential for drug-drug interactions with P-glycoprotein. , 2002, Current drug metabolism.

[46]  Paola Gramatica,et al.  Principles of QSAR models validation: internal and external , 2007 .

[47]  Vijay K Gombar,et al.  Predicting P-glycoprotein substrates by a quantitative structure-activity relationship model. , 2004, Journal of pharmaceutical sciences.

[48]  Lei Chen,et al.  Computational models for predicting substrates or inhibitors of P-glycoprotein. , 2012, Drug discovery today.

[49]  M. Gleeson Generation of a set of simple, interpretable ADMET rules of thumb. , 2008, Journal of medicinal chemistry.

[50]  Geri A. Sawada,et al.  Early Preclinical Evaluation of Brain Exposure in Support of Hit Identification and Lead Optimization , 2006 .

[51]  D. Mudra,et al.  In silico, in vitro and in situ models to assess interplay between CYP3A and P-gp. , 2011, Current drug metabolism.

[52]  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..

[53]  M C Willingham,et al.  Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues. , 1987, Proceedings of the National Academy of Sciences of the United States of America.

[54]  Alexander Golbraikh,et al.  Combinatorial QSAR Modeling of P-Glycoprotein Substrates , 2006, J. Chem. Inf. Model..

[55]  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..

[56]  Thomas J. Raub,et al.  How hydrogen bonds impact P-glycoprotein transport and permeability. , 2012, Bioorganic & medicinal chemistry letters.

[57]  I. Wilhelm,et al.  In vitro models of the blood-brain barrier. , 2011, Acta neurobiologiae experimentalis.

[58]  I. Pastan,et al.  Human multidrug-resistant cell lines: increased mdr1 expression can precede gene amplification. , 1986, Science.