1. Trang chủ
  2. » Giáo Dục - Đào Tạo

Statistical learning approaches for predicting pharmacological properties of pharmaceutical agents

234 358 0

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

THÔNG TIN TÀI LIỆU

Thông tin cơ bản

Định dạng
Số trang 234
Dung lượng 1,49 MB

Nội dung

STATISTICAL LEARNING APPROACHES FOR PREDICTING PHARMACOLOGICAL PROPERTIES OF PHARMACEUTICAL AGENTS LI HU (B.Sc, M.Sc, Jilin University) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF PHARMACY NATIONAL UNIVERSITY OF SINGAPORE 2007 ii Acknowledgements First and foremost, my heartfelt appreciation and thanks go to my supervisor and mentor, Associate Professor Chen Yu Zong. His innovative insights, excellent guidance, words of wisdom, constant support and patience throughout my study have been crucial to this research analysis. I would like to dedicate my thesis to my wife, Dr. Zhu Shizhen. The beautiful time and memories we have in Singapore are definitely great treasures in my life, I cherish it very much. And I am eternally grateful for everything you for me, I appreciate it very much. I want to praise my closest collaborator and friend Dr. Ung Choong Yong. I am proud that our collaboration could be another good story of inter-background collaboration. Many thanks to Dr. Xue Ying, Dr. Li Zerong and Dr. Yap Chun Wei, for their suggestions and contributions and my thanks to all the members in BIDD group, for their kind supports in one way or another. At the same time my thanks also to everyone else whose name may not be down here but knows who they are. I am lucky that I have the greatest families in the world. They always had total confidence in me that I could achieve what I set out to do. Thanks very much my dearest parents and all the family members. Finally, I am very grateful to the National University of Singapore for awarding me the Research Scholarship, the prestigious President Graduate Fellowship and the Best Graduate Researcher Award during my PhD candidature. iii Table of Contents Acknowledgements . ii Table of Contents . iii Summary viii List of Tables x List of Figures xiii List of Abbreviations xv List of Publications . xvii Chapter Introduction 1.1 Drug discovery and pharmacological properties of pharmaceutical agents . 1.2 Statistical learning methods in characterization of pharmacological properties of pharmaceutical agents . 1.3 Describing molecular properties using molecular descriptors 15 1.4 Feature selection methods . 15 1.5 Models studied in this work and the importance of these models 16 1.5.1 Pregnane X Receptor (PXR) activators 17 1.5.2 Blood brain barrier (BBB) agents .18 1.5.3 Estrogen receptor (ER) agonists .19 1.5.4 Genotoxicity agents 19 1.5.5 Tetrahymena pyriformis toxicity (TPT) agents .20 1.6 Objectives and outline of this work 21 Chapter Methods 26 2.1 Datasets . 26 2.1.1 Quality analysis .26 2.1.2 Statistical molecular design 27 iv 2.1.2.1 Introduction 27 2.1.2.2 Kennard and Stone algorithm 30 2.1.2.3 Removal-until-done algorithm .30 2.1.3 Diversity and representativity of datasets .31 2.2 Molecular descriptors 32 2.2.1 Types of molecular descriptors .32 2.2.2 Scaling .36 2.2.2.1 Auto-scaling .37 2.2.2.2 Range scaling (Normalization) 37 2.3 Feature selection method 38 2.3.1 Recursive feature elimination (RFE) 39 2.3.2 The procedure of RFE .41 2.4 Statistical learning methods 43 2.4.1 Methods .43 2.4.1.1 Logistic regression (LR) 43 2.4.1.2 Linear discriminate analysis (LDA) .43 2.4.1.3 C4.5 decision trees (DT) 45 2.4.1.4 k-nearest neighbor (k-NN) .47 2.4.1.5 Probabilistic neural network (PNN) 49 2.4.1.6 Support vector machine (SVM) 52 2.4.2 Parameters optimization 55 2.5 Model validation . 55 2.5.1 Performance evaluation of a pharmacological property prediction model .55 2.5.2 Performance evaluation methods 57 2.5.3 Overfitting .59 v Chapter Prediction of pharmacokinetics properties of pharmaceutical agents 62 3.1 Pregnane X receptor activators model 62 3.1.1 Introduction .62 3.1.2 Methods .66 3.1.2.1 Collection of PXR activators and non-activators 66 3.1.2.2 Construction of training and testing sets .68 3.1.2.3 Molecular descriptors 69 3.1.2.4 Computational parameters and performance evaluation 69 3.1.3 Results and discussion 70 3.1.3.1Promiscuity nature of PXR activator structures and the selected molecular descriptors for classifying PXR activators 70 3.1.3.2 Performance of SLMs for predicting PXR activators 77 3.1.3.3 Relevance of molecular descriptors to the activity of PXR activators .81 3.1.4 Conclusion 87 3.2 Blood brain barrier agents model 89 3.2.1 Introduction .89 3.2.2 Methods .91 3.2.2.1 Selection of BBB+ and BBB- agents 91 3.2.2.2 Construction of training and testing sets .92 3.2.2.3 Molecular descriptors 92 3.2.3 Results and discussion 93 3.2.3.1 Molecular descriptors selected for BBB penetration prediction .93 3.2.3.2 Prediction accuracy for BBB+ and BBB- agents 98 3.2.4 Conclusion 104 Chapter Prediction of pharmacodynamics properties of pharmaceutical agents . 106 vi 4.1. Introduction 106 4.2 Methods . 110 4.2.1 Data collection of ER agonists and ER non-agonists .110 4.2.2 Structural diversity 112 4.2.3 Construction of training and testing sets .112 4.2.4 Molecular descriptors 113 4.3 Results and discussion 113 4.3.1 Overall prediction accuracies and merit of the statistical learning methods 113 4.3.2 Molecular descriptors associated with ER agonism .119 4.3.3 Misclassified ER agonists and non-agonists from independent test sets 128 4.4 Conclusion 130 Chapter Prediction of toxicological properties of pharmaceutical agents . 132 5.1 Genotoxicity model . 132 5.1.1 Introduction .132 5.1.2 Methods .136 5.1.2.1 Selection of GT+ and GT- agents 136 5.1.2.2 Construction of training and testing sets .136 5.1.2.3 Molecular descriptors 137 5.1.2.4 Parameter for feature selection .137 5.1.3 Results and discussion 138 5.1.3.1 Overall prediction accuracies 138 5.1.3.2 Relevance of selected features to genotoxicty study 140 5.1.3.3 Performance evaluation .144 5.1.3.4 Misclassified GT+ and GT- agents from independent test sets .146 5.1.4 Conclusion 152 vii 5.2 Tetrahymena pyriformis toxicity Model . 154 5.2.1 Introduction .154 5.2.2 Methods .157 5.2.2.1 Selection of TPT and non-TPT- agents 157 5.2.2.2 Construction of training and testing sets .159 5.2.2.3 Molecular descriptors 159 5.2.3 Results and discussion 159 5.2.3.1 Overall prediction accuracies 159 5.2.3.2 Relevance of selected molecular descriptors to Tetrahymena pyriformis toxicity prediction 165 5.2.3.3 Performance evaluation .173 5.2.4 Conclusion 174 Chapter Concluding remarks . 176 6.1 Major findings . 176 6.1.1 Merits of SLMs in the studies of pharmacological properties 176 6.1.2 Merits of RFE in the studies of pharmacological properties 177 6.1.3 The pharmacokinetic models: PXR activators and BBB agents .177 6.1.4 The pharmacodynamic model: ER agonists .179 6.1.5 The toxicity models: Genotoxicity agents and TPT agents 180 6.2 Contributions . 181 6.3 Limitations 184 6.4 Suggestions for future studies . 189 Bibliography . 191 viii Summary Drug development is aimed at therapeutic agents that possess desirable pharmacological properties, which include pharmacokinetic, pharmacodynamic and toxicological profiles. Historically, inappropriate pharmacological properties have been one of the primary reasons for the failure of drug candidates in the later stages of drug development. Thus tools for predicting pharmacological properties in early drug discovery stages are desirable for fast elimination of agents with undesirable properties so that development efforts can be focused on the most promising candidates. As part of the efforts for developing such tools, computational approaches have been explored for predicting various pharmacological properties of pharmaceutical agents. In particular, statistical learning methods (SLMs) have shown promise for these tasks by statistically analyzing the correlation between chemical structures and a specific property to derive statistical models or rules for predicting whether an agent possesses a specific property or not. Previously, pharmacological property prediction models were frequently built upon limited number of structurally related compound sets and by using linear regression methods. Hence they may not be suitable for the prediction of pharmacological properties of structurally diverse compounds and for pharmacological properties that are regulated by multiple mechanisms. Moreover, some pharmacological properties, which are pharmacologically and clinically important, are insufficiently studied by different computational approaches. Thus it is of interest and necessary to examine the potential of using enlarged and more diverse groups of compounds and non-linear SLMs in improving the quality of pharmacological property prediction models and in applying the SLMs on those important but insufficiently studied pharmacological properties. This ix work aims at studying the applicability of SLMs, such as support vector machine (SVM), probabilistic neural network (PNN), k nearest neighbor (k-NN), C4.5 decision tree (C4.5 DT), linear discriminate analysis (LDA) and logistic regression (LR) to classify compounds of diverse structures into different pharmacological property categories. Specifically, the pharmacokinetic models explored in this work are activators for pregnane X receptor (PXR) and blood brain barrier (BBB) agents. The pharmacodynamic model studied in this work is agonists of estrogen receptor (ER) and the toxicity models studied are genotoxicity (GT) and Tetrahymena pyriformis toxicity (TPT) agents. A set of 199 molecular descriptors are used to describe the molecular pysicochemical properties of those pharmaceutical agents studied in this work. A feature selection method, recursive feature elimination (RFE), is incorporated to improve the prediction performance. The results show that SLMs could improve the quality of these pharmacological property prediction models by using enlarged and more diverse groups of compounds. RFE is able to identify a group of relevant molecular descriptors that reflect the pharmacological property of studied models and are consistent to quantitive structure activity relationship (QSAR), pharmacophore and X-ray crystallographic studies. In addition, selection of appropriate molecular descriptors can lead to substantially more balanced prediction accuracies and enhance the overall accuracies. Moreover, SLMs are found to be useful for developing prediction models and characterizing relevant physicochemical features for PXR activators and ER agonists, which are very important pharmacological properties of drug candidates but insufficiently explored in previous studies. x List of Tables Table 1.1 Performance of regression-based statistical learning methods for predicting compounds of specific pharmacokinetic, pharmacodynamic or toxicological property . Table 1.2 Performance of classification-based statistical learning methods for predicting compounds of specific pharmacokinetic, pharmacodynamic or toxicological property . 11 Table 2.1 Methods for selecting training and validation sets . 29 Table 2.2 Molecular descriptors used in this work 35 Table 2.3 Common descriptor selection methods used in pharmacological properties classification studies . 39 Table 2.4 Commonly used kernel functions . 53 Table 3.1 Diversity index (DI) for the compounds in several chemical groups, and the number of molecular descriptors selected by RFE for predicting each group of compounds by using a SLM classification system. 71 Table 3.2 RFE selected 83 molecular descriptors for SLMs classification of PXR activators . 73 Table 3.3 Performance of three statistical learning methods (k-NN, PNN and SVM) for predicting PXR and hPXR activators and non-activators determined by a 10fold cross validation study. . 77 Table 3.4 Performance of the PXR and hPXR activator prediction systems for predicting the 15 recently published hPXR activators . 80 Table 3.5 The Euclidean distance of the 15 PXR activators in the independent set to the 28 ambiguous PXR activators and the 98 human PXR activators 81 Table 3.6 Prediction accuracies of BBB penetrating (BBB+) and non-penetrating agents (BBB-) from different earlier studies reported in the literatures. . 90 Table 3.7 Thirty-seven molecular descriptors selected from the RFE feature selection method for classification of blood-brain barrier penetrating and nonpenetrating agents . 94 Table 3.8 Important descriptor classes selected for the prediction of blood-brain barrier penetrating and non-penetrating agents 97 Table 3.9 Differences in the values of descriptors important for distinguishing between blood-brain barrier penetrating (BBB+) agents and non-penetrating (BBB-) agents 98 BIBLIOGRAPHY 201 Kliewer SA, Goodwin B, Willson TM (2002). The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocrine Reviews 23(5):687-702. Klopman G, Stefan LR, Saiakhov RD (2002). ADME evaluation. 2. A computer model for the prediction of intestinal absorption in humans. European Journal of Pharmaceutical Sciences 17(4-5):253-263. Kohavi R, John GH (1997a). Wrappers for feature subset selection. Artificial Intelligence 97(1-2):273-324. Kohavi R, John GH (1997b). Wrappers for Feature Subset Selection. Artificial Intelligence in Medicine 97:273-324. Kohonen T. (1995). Self-Organizing Maps. Berlin, Heidelberg: Springer. Kojima H, Katsura E, Takeuchi S, Niiyama K, Kobayashi K (2004). Screening for estrogen and androgen receptor activities in 200 pesticides by in vitro reporter gene assays using Chinese hamster ovary cells. Environmental Health Perspectives 112(5):524-531. Kononenko I. (1994) Estimating attributes: analysis and extensions of RELIEF. Kozak A, Kozak R (2003). Does cross validation provide additional information in the evaluation of regression models? Canadian Journal of Forest Research 33(6):976-987. Kramer PJ (1998). Genetic toxicology. Journal of Pharmacy and Pharmacology 50(4):395-405. Krasowski MD, Yasuda K, Hagey LR, Schuetz EG (2005). Evolutionary selection across the nuclear hormone receptor superfamily with a focus on the NR1I subfamily (vitamin D, pregnane X, and constitutive androstane receptors). Nuclear receptor 3:2. Kubat M, R.C. H, Matwin S (1998). Machine learning for the detection of oil spills in statellite radar images. Machine Learning 30(2-3):195-215. Kubinyi H (2003). Drug research: myths, hype and reality. Nature reviews. Drug discovery 2(8):665-668. Kupfer D (1975). Effects of pesticides and related compounds on steroid metabolism and function. Critical Reviews in Toxicology 4(1):83-124. Lacy CF, Armstrong LL, Goldman MP, Lance LL. (2004). Drug Information Handbook. Hudson, Cleveland: Lexi-Comp. Lam RLH, Welch WJ, Young SS (2002). Uniform coverage designs for molecule selection. Technometrics 44(2):99-109. BIBLIOGRAPHY 202 Leach AR, Gillet VJ. (2003). Selecting diverse sets of compounds. An introduction to chemoinformatics. Boston: Kluwer Academic Publisher. p 123-145. Leardia R, González AL (1998). Genetic algorithms applied to feature selection in PLS regression: How and when to use them. Chemometrics and Intelligent Laboratory Systems 41(2):195-207. Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT, Kliewer SA (1998). The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. Journal of Clinical Investigation 102,(5):1016-1023. Lemaire G, Mnif W, Pascussi JM, Pillon A, Rabenoelina F, Fenet H, Gomez E, Casellas C, Nicolas JC, Cavailles V and others (2006). Identification of New Human PXR Ligands among Pesticides Using a Stable Reporter Cell System. Toxicological Sciences:[Epub ahead of print]. Lengauer T, Lemmen C, Rarey M, Zimmermann M (2004). Novel technologies for virtual screening. Drug Discovery Today 9(1):27-34. Lepp Z, Kinoshita T, Chuman H (2006). Screening for new antidepressant leads of multiple activities by support vector machines. Journal of Chemical Information and Modeling. 46(1):158-167. Li AP (2002). Early ADME/Tox studies and in silico screen. Drug Discovery Today 7(1):25-27. Li H, Ung CY, Yap CW, Xue Y, Li ZR, Cao ZW, Chen YZ (2005a). Prediction of Genotoxicity of Chemical Compounds by Statistical Learning Methods. Chemical Research in Toxicology 18(6):1071-1080. Li H, Ung CY, Yap CW, Xue Y, Li ZR, Chen YZ (2006a). Prediction of estrogen receptor agonists and characterization of associated molecular descriptors by statistical learning methods. Journal of Molecular Graphics & Modelling 25(3):313-323. Li H, Yap CW, Ung CY, Xue Y, Cao ZW, Chen YZ (2005b). Effect of Selection of Molecular Descriptors on the Prediction of Blood-Brain Barrier Penetrating and Nonpenetrating Agents by Statistical Learning Methods. Journal of Chemical Information and Modeling. 45(5):1376-1384. Li H, Yap CW, Xue Y, Li ZR, Ung CY, Han LY, Chen YZ (2006b). Statistical learning approach for predicting specific pharmacodynamic, pharmacokinetic or toxicological properties of pharmaceutical agents. Drug Development Research 66(4):245-259. Li ZR, Han LY, Xue Y, Yap CW, Li H, Jiang L, Chen YZ (2006c). MODEL-Molecular Descriptor Lab: A web-based server for computing structural and physicochemical features of compounds. Biotechnology and Bioengineering:DOI: BIBLIOGRAPHY 203 10.1002/bit.21214. Linnan He, Peter C. Jurs, Laura L. Custer, Stephen K. Durham, Greg M. Pearl (2003). Predicting the Genotoxicity of Polycyclic Aromatic Compounds from Molecular Structure with Different Classifiers. Chemical Research in Toxicology 16(12):1567-1580. Lissin LW, Cooke JP (2000). Phytoestrogens and cardiovascular health. Journal of the American College of Cardiology 35(6):1403-1410. Liu HX, Zhang RS, Yao XJ, Liu MC, Hu ZD, Fan BT (2004). QSAR and classification models of a novel series of COX-2 selective inhibitors: 1,5-diarylimidazoles based on support vector machines. Journal of Computer-Aided Molecular Design 18(6):389-399. Liu R, So SS (2001a). Development of quantitative structure-property relationship models for early ADME evaluation in drug discovery. 1. Aqueous solubility. Journal of Chemical Information and Computer Sciences 41(6):1633-1639. Liu R, Sun H, So SS (2001b). Development of quantitative structure-property relationship models for early ADME evaluation in drug discovery. 2. Blood-brain barrier penetration. Journal of Chemical Information and Computer Sciences 41(6):1623-1632. Liu X, Tu M, Kelly RS, Chen C, Smith BJ (2004). Development of a computational approach to predict blood-brain barrier permeability. Drug Metab Dispos. 32(1):132-139. Liu Y (2004). A comparative study on feature selection methods for drug discovery. Journal of Chemical Information and Computer Sciences 44(5):1823-1828. Livingstone DJ. (1995). Data analysis for chemists: Applications to QSAR and chemical product design. Oxford: Oxford University Press. Livingstone DJ, Manallack DT (2003). Neural networks in 3D QSAR. QSAR & Combinatorial Science 22(5):510-518. Livingstone DJ, Rahr E (1989). Corchop - An interative routine for the dimension reduction of large QSAR data sets. Quantitative Structure-Activity Relationships 8:103-108. Lobell M, Molnár L, Keserü GM (2003a). Recent advances in the prediction of bloodbrain partitioning from molecular structure. Journal of Pharmaceutical Sciences 92(2):360-370. Lobell M, Sivarajah V (2003b). In silico prediction of aqueous solubility, human plasma protein binding and volume of distribution of compounds from calculated pKa and AlogP98 values. Molecular Diversity 7(1):69-87. BIBLIOGRAPHY 204 Lombardo F, Blake JF, Curatolo WJ (1996). Computation of brain-blood partitioning of organic solutes via free energy calculations. Journal of Medicinal Chemistry 39(24):4750-4755. Lucasius CB, Kateman G (1993). Understanding and using genetic algorithms. Part 1. Concepts, properties and context. Chemometrics and Intelligent Laboratory Systems 19(1):1-33. Luco JM (1999). Prediction of the brain-blood distribution of a large set of drugs from structurally derived descriptors using partial least-squares (PLS) modeling. Journal of Chemical Information and Computer Sciences 39(2):396-404. MacGregor JI, Jordan VC (1998). Basic guide to the mechanisms of antiestrogen action. Pharmacological Reviews 50(2):151-196. Manallack D, Pitt W, Gancia E, Montana J, Livingstone D, Ford M, Whitley D (2002). Selecting screening candidates for kinase and G protein-coupled receptor targets using neural networks. Journal of Chemical Information and Computer Sciences 42(5):1256-1262. Manallack DT, Livingstone DJ (1999). Neural networks in drug discovery: have they lived up to their promise? European Journal of Medicinal Chemistry 34(3):195208. Manly BFJ. (1997). Randomization bootstrap and Monte Carlo methods in biology. London: Chapman and Hall. Marchant CA (1996). Prediction of rodent carcinogenicity using the DEREK system for 30 chemicals currently being tested by the National Toxicology Program. The DEREK Collaborative Group. Environmental Health Perspectives 104(Suppl 5):1065-1073. Matthews BW (1975). Comparison of the predicted and observed secondary structure of T4 phage lysozyme. Biochimica et Biophysica Acta 405(2):442-451. Mattioni BE, Kauffman GW, Jurs PC, Custer LL, Durham SK, Pearl GM. (2003). Predicting the Genotoxicity of Secondary and Aromatic Amines Using Data Subsetting To Generate a Model Ensemble. Journal of Chemical Information and Computer Sciences 43:949-963. Mattioni BE, Kauffman GW, Jurs PC, Custer LL, Durham SK, Pearl GM (2003). Predicting the Genotoxicity of Secondary and Aromatic Amines Using Data Subsetting To Generate a Model Ensemble. Journal of Chemical Information and Computer Sciences 43:949-963. McDuffie HH (2005). Host factors and genetic susceptibility: a paradigm of the conundrum of pesticide exposure and cancer associations. Reviews on Environmental Health 20(2):77-101. BIBLIOGRAPHY 205 Melagraki G, Afantitis A, Makridima K, Sarimveis H, Igglessi-Markopoulou O (2005). Prediction of toxicity using a novel RBF neural network training methodology. Journal of Molecular Modeling 8:1-9. Menard PR, Mason JS, Morize I, Bauerschmidt S (1998). Chemical space metrics in diversity analysis, library design, and compound selection. Journal of Chemical Information and Computer Sciences 38(6):1204-1213. Meyer D, Leischa F, Hornik K (2003). The support vector machine under test. Neurocomputing 55(1-2):169-186. MICROMEDEX. (2003). Greenwood Village, Colorado: Thomson MICROMEDEX. Mignon P, Loverix S, Steyaert J, Geerlings P (2005). Influence of the pi-pi interaction on the hydrogen bonding capacity of stacked DNA/RNA bases. Nucleic Acids Research 33(6):1779-1789. Miller JL, Bradley EK, Teig SL (2003). Luddite: an information-theoretic library design tool. Journal of Chemical Information and Computer Sciences 43(1):47-54. Mitchell TJ (1974). An algorithm for the construction of "D-optimal" experimental designs. Technometrics 16:203-210. Molnar L, Keseru GM (2002a). A neural network based virtual screening of cytochrome P450 3A4 inhibitors. Bioorganic and Medicinal Chemistry Letters 12(3):419-421. Molnar L, Keseru GM (2002b). A neural network based virtual screening of cytochrome P450 3A4 inhibitors. Bioorganic & Medicinal Chemistry Letters 12(3):419-421. Moore LB, Maglich JM, McKee DD, Wisely B, Willson TM, Kliewer SA, Lambert MH, Moore JT (2002). Pregnane X receptor (PXR), constitutive androstane receptor (CAR), and benzoate X receptor (BXR) define three pharmacologically distinct classes of nuclear receptors. Molecular Endocrinology 16(5):977-986. Morales AH, Perez MA, Combes RD, Gonzalez MP (2006). Quantitative structure activity relationship for the computational prediction of nitrocompounds carcinogenicity. Toxicology 220(1):51-62. Mosier PD, Jurs PC (2002). QSAR/QSPR studies using probabilistic neural networks and generalized regression neural networks. Journal of Chemical Information and Computer Sciences 42(6):1460-1470. Mosier PD, Jurs PC, Custer LL, Durham SK, Pearl GM (2003). Predicting the Genotoxicity of Thiophene Derivatives from Molecular Structure. Chemical Research in Toxicology 16:721-732. Mount J, Ruppert J, Welch W, Jain AN (1999). IcePick: a flexible surface-based system for molecular diversity. Journal of Medicinal Chemistry 42(1):60-66. BIBLIOGRAPHY 206 Mueller SO, Katzenellenbogen JA, Korach KS (2004). Endogenous estrogen receptor beta is transcriptionally active in primary ovarian cells from estrogen receptor knockout mice. Steroids 69(10):681-686. Nakazato A, Sakagami K, Yasuhara A, Ohta H, Yoshikawa R, Itoh M, Nakamura M, Chaki S (2004). Synthesis, in vitro pharmacology, structure-activity relationships, and pharmacokinetics of 3-alkoxy-2-amino-6-fluorobicyclo[3.1.0]hexane-2,6dicarboxylic acid derivatives as potent and selective group II metabotropic glutamate receptor antagonists. Journal of Medicinal Chemistry 47(18):45704587. Nandagere AK, Jones B. (2003). Prediction of bioavailability. In: van de Waterbeemd H, Lennernas H, Artursson P, editors. Drug bioavailability: Estimation of solubility, permeability, absorption and bioavailability. Weinheim: Wiley-VCH. p 444-460. Narendra PM, Fukunaga K (1977). A branch and bound algorithm for feature subset selection. IEEE Transactions on Computers 26:917-922. Needham LL, Barr DB, Caudill SP, Pirkle JL, Turner WE, Osterloh J, Jones RL, Sampson EJ (2005). Concentrations of environmental chemicals associated with neurodevelopmental effects in U.S. population. Neurotoxicology 26(4):531-545. Netzeva TI, Aptula AO, Chaudary SH, Duffy JC, Schultz TW, Schüürmann G, Cronin MTD (2003a). Structure-Activity Relationships for the Toxicity of Substituted Poly-hydroxylated Benzenes to Tetrahymena pyriformis: Influence of Free Radical Formation. QSAR & Combinatorial Science 22(6):575-582. Netzeva TI, Schultz TW (2005). QSARs for the aquatic toxicity of aromatic aldehydes from Tetrahymena data. Chemosphere 61(11):1632-1643. Netzeva TI, Schultz TW, Aptula AO, Cronin MT (2003b). Partial least squares modelling of the acute toxicity of aliphatic compounds to Tetrahymena pyriformis. SAR and QSAR in Environmental Research 14(4):265-283. Niculescu SP, Kaiser KL, Schultz TW (2000). Modeling the toxicity of chemicals to Tetrahymena pyriformis using molecular fragment descriptors and probabilistic neural networks. Archives of Environmental Contamination and Toxicology 39(3):289-298. Nishihara T, Nishikawa J, Kanayama T, Dakeyama F, Saito K, Imagawa M, Takatori S, Kitagawa Y, Hori S, Utsumi H (2000). Estrogenic Activities of 517 Chemicals by Yeast Two-Hybrid Assay. Journal of Health Science 46(4):282-298. Norinder U, Haeberlein M (2002). Computational approaches to the prediction of the blood-brain distribution. Advanced Drug Delivery Reviews 54(3):291-313. Norinder U, Österberg T, Artursson P (1999). Theoretical calculation and prediction of intestinal absorption of drugs in humans using MolSurf parametrization and PLS BIBLIOGRAPHY 207 statistics. European Journal of Pharmaceutical Sciences 8(1):49-56. Norinder U, Sjöberg P, Österberg T (1998). Theoretical calculation and prediction of brain-blood partitioning of organic solutes using MolSurf parametrization and PLS statistics. Journal of Pharmaceutical Sciences 87(8):952-959. Oh WK (2002). The evolving role of estrogen therapy in prostate cancer. Clinical Prostate Cancer 1(2):81-89. Ohlstein EH, Ruffolo RRJ, Elliott JD (2000). Drug discovery in the next millennium. Annual Review of Pharmacology and Toxicology 40:177-191. Olsson IM, Gottfries J, Wold S (2004). D-optimal onion designs in statistical molecular design. Chemometrics and Intelligent Laboratory Systems 73(1):37-46. Ooms F, Weber P, Carrupt PA, Testa B (2002). A simple model to predict blood-brain barrier permeation from 3D molecular fields. Biochimica et Biophysica Acta 1587(2-3):118-125. Oprea TI, Gottfries J (1999). Toward minimalistic modeling of oral drug absorption. Journal of Molecular Graphics and Modelling 17(5-6):261-274. Oprea TI, Matter H (2004). Integrating virtual screening in lead discovery. Current Opinion in Chemical Biology 8(4):349-358. Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS (1997). Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science 277(5331):1508-1510. Pan D, Iyer M, Liu J, Li Y, Hopfinger AJ (2004). Constructing optimum blood brain barrier QSAR models using a combination of 4D-molecular similarity measures and cluster analysis. Journal of Chemical Information and Computer Sciences 44(6):2083-2098. Park BK, Kitteringham NR, Powell H, Pirmohamed M (2000). Advances in molecular toxicology-towards understanding idiosyncratic drug toxicity. Toxicology 153(13):39-60. Parzen E (1962). On estimation of a probability density function and mode. Annals of Mathematical Statistics 33:1065-1076. Pearlman RS. CONCORD User's Manual. St. Louis, MO: Tripos. Perez JJ (2005). Managing molecular diversity. Chemical Society Reviews 34(2):143-152. Philip D. Mosier, Peter C. Jurs, Laura L. Custer, Stephen K. Durham, Greg M. Pearl (2003). Predicting the Genotoxicity of Thiophene Derivatives from Molecular Structure. Chemical Research in Toxicology 16:721-732. BIBLIOGRAPHY 208 Pike AC, Brzozowski AM, Hubbard RE, Bonn T, Thorsell AG, Engstrom O, Ljunggren J, Gustafsson JA, Carlquist M (1999). Structure of the ligand-binding domain of oestrogen receptor beta in the presence of a partial agonist and a full antagonist. The EMBO Journal 18:4608-4618. Pike AC, Brzozowski AM, Walton J, Hubbard RE, Thorsell AG, Li YL, Gustafsson JA, Carlquist M (2001). Structural insights into the mode of action of a pure antiestrogen. Structure 9(2):145-153. Platts JA, Abraham MH, Zhao YH, Hersey A, Ijaz L, Butina D (2001). Correlation and prediction of a large blood-brain distribution data set - an LFER study. European Journal of Medicinal Chemistry 36(9):719-730. Potter T, Matter H (1998). Random or rational design? Evaluation of diverse compound subsets from chemical structure databases. Journal of Medicinal Chemistry 41(4):478-488. Prentis RA, Lis Y, Walker SR (1988). Pharmaceutical innovation by the seven UK-owned pharmaceutical companies (1964-1985). British Journal of Pharmacology 25(3):387-396. PubChem. 2004. http://pubchem.ncbi.nlm.nih.gov/. National Institutes of Health (NIH). Pudil P, Novoviova J, Kittler J (1994). Floating search methods in feature selection. Pattern Recognition Letters 15(11):1119-1125. Quillardet P, Hofnung M (1993). The SOS chromotest: a review. Mutation Research 297(3):235-279. Quinlan JR. (1993). C4.5: programs for machine learning. San Mateo, Calif: Morgan Kaufmann. Rajer-Kanduc K, Zupan J, Majcen N Separation of data on the training and test set for modeling: a case study for modelling of five colour properties of a white pigment. Chemometrics and Intelligent Laboratory Systems 65(2):221-229. Rang HP, Dale MM, Ritter JM, Moore PK. (2003). Pharmacology. Edinburgh: Churchill Livingstone. Ren S (2003a). Ecotoxicity prediction using mechanism- and non-mechanism-based QSARs: a preliminary study. Chemosphere 53(9):1053-1065. Ren S (2003b). Modeling the toxicity of aromatic compounds to tetrahymena pyriformis: the response surface methodology with nonlinear methods. Journal of Chemical Information and Computer Sciences 43(5):1679-1687. Ren S, Schultz TW (2002). Identifying the mechanism of aquatic toxicity of selected compounds by hydrophobicity and electrophilicity descriptors. Toxicology Letters BIBLIOGRAPHY 209 129(1-2):151-160. Reunanen J (2003). Overfitting in making comparisons between variable selection methods. Journal of Machine Learning Research 3:1371-1382. Robotka H, Nemeth H, Somlai C, Vecsei L TJ (2005). Systemically administered glucosamine-kynurenic acid, but not pure kynurenic acid, is effective in decreasing the evoked activity in area CA1 of the rat hippocampus. European Journal of Pharmacology 513(1-2):75-80. Rose K, Hall LH, Kier LB (2002). Modeling blood-brain barrier partitioning using the electrotopological state. Journal of Chemical Information and Computer Sciences 42(3):651-666. Roulston JE (2002). Screening with tumor markers. Molecular Pharmacology 20:153162. Roy DR, Parthasarathi R, Maiti B, Subramanian V, Chattaraj PK (2005). Electrophilicity as a possible descriptor for toxicity prediction. Bioorganic & Medicinal Chemistry 13(10):3405-3412. Saeys Y, Degroeve S, Aeyels D, Rouzé P, Peer YVD (2004). Feature selection for splice site prediction: A new method using EDA-based feature ranking. BMC bioinformatics 5(1):64. Safe S (1990). Polychlorinated biphenyls (PCBs), dibenzo-p-dioxins (PCDDs), dibenzofurans (PCDFs), and related compounds: environmental and mechanistic considerations which support the development of toxic equivalency factors (TEFs). Critical Reviews in Toxicology 21(1):51-88. Safe SH, Pallaroni L, Yoon K, Gaido K, Ross S, Saville B, McDonnellc D (2001). Toxicology of environmental estrogens. Reproduction, Fertility, and Development 13(4):307-315. Sauvant MP, Pepin D, Bohatier J, Groliere CA (1995). Microplate technique for screening and assessing cytotoxicity of xenobiotics with Tetrahymena pyriformis. Ecotoxicology and Environmental Safety 32(2):159-165. Sauvant MP, Pepin D, Piccinni E (1999). Tetrahymena pyriformis: a tool for toxicological studies. A review. Chemosphere 38(7):1631-1669. Schultz TW (1997). Tetrahymena pyriformis population growth impairment endpoint: a surrogate for fish lethality. Toxicology Methods 7:289-309. Schultz TW (1999). Structure-toxicity relationships for benzenes evaluated with Tetrahymena pyriformis. Chemical Research in Toxicology 12(12):1262-1267. Schultz TW, Netzeva TI, Cronin MT (2003). Selection of data sets for QSARs: analyses BIBLIOGRAPHY 210 of Tetrahymena toxicity from aromatic compounds. SAR and QSAR in Environmental Research 14(1):59-81. Schultz TW, Netzeva TI, Roberts DW, Cronin MT (2005). Structure-toxicity relationships for the effects to Tetrahymena pyriformis of aliphatic, carbonyl-containing, alpha,beta-unsaturated chemicals. Chemical Research in Toxicology 18(2):330341. Schuster D, Langer T (2005). The identification of ligand features essential for PXR activation by pharmacophore modeling. Journal of Chemical Information and Modeling. 45(2):431-439. Schuurmann G, Aptula AO, Kuhne R, Ebert RU (2003). Stepwise discrimination between four modes of toxic action of phenols in the Tetrahymena pyriformis assay. Chemical Research in Toxicology 16(8):974-987. Schwetz BA, Casciano DA (1998). Genetic toxicology: impact on the next generation of toxicology. Environmental and Molecular Mutagenesis 31(1):1-3. Serra JR, Jurs PC, Kaiser KL (2001). Linear regression and computational neural network prediction of tetrahymena acute toxicity for aromatic compounds from molecular structure. Chemical Research in Toxicology 14(11):1535-1545. Serra JR, Thompson ED, Jurs PC (2003). Development of Binary Classification of Structural Chromosome Aberrations for a Diverse Set of Organic Compounds from Molecular Structure. Chemical Research in Toxicology 16:153-163. Shao J, Tu D. (1995). The Jackknife and Bootstrap. New York, NY, USA: Springer. Sheridan RP, SanFeliciano SG, Kearsley SK (2000). Designing targeted libraries with genetic algorithms. Journal of Molecular Graphics & Modelling 18(4-5):320-334, 525. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA, Greene GL (1998). The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell 95(7):927-937. Shiau AK, Barstad D, Radek JT, Meyers MJ, Nettles KW, Katzenellenbogen BS, Katzenellenbogen JA, Agard DA, Greene GL (2002). Structural characterization of a subtype-selective ligand reveals a novel mode of estrogen receptor antagonism. Nature Structural Biology 9(5):359-364. Shoichet BK (2004). Virtual screening of chemical libraries. Nature 432(7019):862-865. Siedlecki W, Sklansky J (1989). A note on genetic algorithms for large--scale feature selection. Pattern Recognition Letters 10:335-347. Simon-Hettich B, Rothfuss A, Steger-Hartmann T (2006). Use of computer-assisted BIBLIOGRAPHY 211 prediction of toxic effects of chemical substances. Toxicology 224(1-2):156-162. Snarey M, Terrett NK, Willett P, Wilton DJ (1997). Comparison of algorithms for dissimilarity-based compound selection. Journal of Molecular Graphics & Modelling 15(6):372-385. Snyder RD, Pearl GS, Mandakas G, Choy WN, Goodsaid F, Rosenblum IY (2004). Assessment of the sensitivity of the computational programs DEREK, TOPKAT, and MCASE in the prediction of the genotoxicity of pharmaceutical molecules. Environmental and molecular mutagenesis. 43(3):143-158. Snyder RD, Green JW (2001). A review of the genotoxicity of marketed pharmaceuticals. Mutation Research-Reviews in Mutation Research 488(2):151-169. Snyder RD, Pearl GS, Mandakas G, Choy WN, Goodsaid F, Rosenblum IY (2004). Assessment of the sensitivity of the computational programs DEREK, TOPKAT, and MCASE in the prediction of the genotoxicity of pharmaceutical molecules. Environmental and Molecular Mutagenesis 43(3):143-158. Somol P, Pudil P. (2000) Oscillating search algorithms for feature selection.; Barcelona. 406-409. Somol P, Pudila p, Novoviova J, Pacli P (1999). Adaptive floating search methods in feature selection. Pattern Recognition Letters 20(11-13):1157-1163. Soto AM, Chung KL, Sonnenschein C (1994). The pesticides endosulfan, toxaphene, and dieldrin have estrogenic effects on human estrogen-sensitive cells. Environmental Health Perspectives 102(4):380-383. Spalding DJ, Harker AJ, Bayliss MK (2000). Combining high-throughput pharmacokinetic screens at the hits-to-leads stage of drug discovery. Drug Discovery Today 5(12 Suppl11):70-76. Specht DF (1990). Probabilistic neural networks. Neural Networks 3(1):109-118. Spycher S, Pellegrini E, Gasteiger J (2005). Use of structure descriptors to discriminate between modes of toxic action of phenols. Journal of Chemical Information and Modeling. 45(1):200-208. Sturk LM, Brock JL, Bagnell CR, Hall JE, Tidwell RR (2004). Distribution and quantitation of the anti-trypanosomal diamidine 2,5-bis(4-amidinophenyl)furan (DB75) and its N-methoxy prodrug DB289 in murine brain tissue. Acta Tropica 91(2):131-143. Susnow RG, Dixon SL (2003a). Use of robust classification techniques for the prediction of human cytochrome P450 2D6 inhibition. Journal of Chemical Information and Computer Sciences 43(4):1308-1315. BIBLIOGRAPHY 212 Susnow RG, Dixon SL (2003b). Use of robust classification techniques for the prediction of human cytochrome P450 2D6 inhibition. Journal of Chemical Information and Computer Sciences 43(4):1308-1315. Sutter JM, Kalivas JH (1993). Comparison of forward selection, backward elimination, and generalized simulated annealing for variable selection. Microchemical Journal 47(1-2):60-66. Tatsumi R, Fujio M, Satoh H, Katayama J, Takanashi S, Hashimoto K, Tanaka H (2005). Discovery of the alpha7 nicotinic acetylcholine receptor agonists. (R)-3'-(5Chlorothiophen-2-yl)spiro-1-azabicyclo[2.2.2]octane-3,5'-[1',3']oxazolidin-2'-one as a novel, potent, selective, and orally bioavailable ligand. Journal of Medicinal Chemistry 48(7):2678-2686. Tentori L, Leonetti C, Scarsella M, Vergati M, Xu W, Calvin D, Morgan L, Tang Z, Woznizk K, Alemu C and others (2005). Brain distribution and efficacy as chemosensitizer of an oral formulation of PARP-1 inhibitor GPI 15427 in experimental models of CNS tumors. International Journal of Oncology 26(2):415-422. Thanikaivelan P, Subramanian V, J. RR, Unni Nair B (2000). Application of quantum chemical descriptors in quantitative structure activity and structure property relationship. Chemical Physics Letters 323(1-2):59-70. Todeschini R, Consonni V. (2000). Handbook of molecular descriptors. Weinheim: Wiley-VCH. Todeschini R, Consonni V, Mauri A, Pavan M. (2005). DRAGON. Milano: TALETE srl. Tong W, Fang H, Hong H, Xie Q, Perkins R, Sheehan D. (2004a). Receptor-mediated toxicity: QSARs for oestrogen receptor binding and priority setting of potential oestrogenic endocrine disruptors. In: Cronin MTD, Livingstone D, editors. Predicting Chemical Toxicity and Fate. Boca Raton, FL: CRC Press. p 285-314. Tong W, Xie Q, Hong H, Shi L, Fang H, Perkins R (2004b). Assessment of prediction confidence and domain extrapolation of two structure-activity relationship models for predicting estrogen receptor binding activity. Environmental Health Perspectives 112(12):1249-1254. Topliss JG, Edwards RP (1979). Chance factors in studies of quantitative structureactivity relationships. Journal of Medicinal Chemistry 22(10):1238-1244. Tropsha A, Gramatica P, Gombar VK (2003). The importance of being earnest: Validation is the absolute essential for successful application and interpretation of QSPR models. QSAR & Combinatorial Science 22(1):69-77. Trotter MWB, Buxton BF, Holden SB (2001). Support vector machines in combinatorial chemistry. Measurement and Control 34(8):235-239. BIBLIOGRAPHY 213 Trotter MWB, Holden SB (2003). Support vector machines for ADME property classification. QSAR & Combinatorial Science 22(5):533-548. Tsai MJ, O'Malley BW (1994). Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annual Review of Biochemistry 63:451-486. Tsodikov OV, Record MTJ, Sergeev YV (2002). Novel computer program for fast exact calculation of accessible and molecular surface areas and average surface curvature. Journal of Computational Chemistry 23:600-609. Turner JV, Maddalena DJ, Cutler DJ (2004). Pharmacokinetic parameter prediction from drug structure using artificial neural networks. International Journal of Pharmaceutics 270(1-2):209-219. Turner JV, Maddalena DJ, Cutler DJ (2005). Pharmacokinetic parameter prediction from drug structure using artificial neural networks. International Journal of Pharmaceutics 270(1-2):209-219. Ung CY, Li H, Yap CW, Chen YZ (2007). In silico prediction of pregnane X receptor activators by machine learning approaches. Molecular Pharmacology 71(1):158168. van de Waterbeemd H, Gifford E (2003). ADMET in silico modelling: towards prediction paradise? Nature reviews. Drug discovery 2:192-204. Vapnik VN. (1995). The nature of statistical learning theory. New York: Springer. Vasilieva S (2002). SOS Chromotest methodology for fundamental genetic research. Research in Microbiology 153(7):435-440. Venkatraman V, Dalby AR, Yang ZR (2004). Evaluation of mutual information and genetic programming for feature selection in QSAR. Journal of Chemical Information and Computer Sciences 44(5):1686-1692. Veropoulos K. (2001). Machine learning approaches to medical decision making: University of Bristol. Votano JR, Parham M, Hall LM, Hall LH, Kier LB, Oloff S, Tropsha A (2006). QSAR modeling of human serum protein binding with several modeling techniques utilizing structure-information representation. Journal of Medicinal Chemistry 49(24):7169-7181. Waldman M, Li H, Hassan M (2000). Novel algorithms for the optimization of molecular diversity of combinatorial libraries. Journal of Molecular Graphics & Modelling 18(4-5):412-426,533-536. Walters WP, Goldman BB (2005). Feature selection in quantitative structure-activity relationships. Current Opinion in Drug Discovery & Development 8(3):329-333. BIBLIOGRAPHY 214 Watari N, Sugiyama Y, Kaneniwa N, Hiura M (1988). Prediction of hepatic first-pass metabolism and plasma levels following intravenous and oral administration of barbiturates in the rabbit based on quantitative structure-pharmacokinetic relationships. Journal of Pharmacokinetics and Biopharmaceutics 16(3):279-301. Watkins RE, Davis-Searles PR, Lambert MH, Redinbo MR (2003a). Coactivator binding promotes the specific interaction between ligand and the pregnane X receptor. Journal of Molecular Biology 331(4):815-828. Watkins RE, Maglich JM, Moore LB, Wisely GB, Noble SM, Davis-Searles PR, Lambert MH, Kliewer SA, Redinbo MR (2003b). 2.1 A crystal structure of human PXR in complex with the St. John's wort compound hyperforin. Biochemistry 42(6):14301438. Watkins RE, Wisely GB, Moore LB, Collins JL, Lambert MH, Williams SP, Willson TM, Kliewer SA, Redinbo MR (2001). The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity. Science 292(5525):2329-2333. Wegner JK. (2005). JOELib/JOELib2: tuebingen.de/software/joelib/index.html. http://www-ra.informatik.uni- Wegner JK, Fröhlich H, Zell A (2004). Feature Selection for Descriptor Based Classification Models. 2. Human Intestinal Absorption (HIA). Journal of Chemical Information and Computer Sciences 44(3):931 -939. Weiss SM, Kulikowski CA. (1991). Computer systems that learn: classification and prediction methods from statistics, neural nets, machine learning, and expert systems. San Francisco, CA, USA: Morgan Kaufmann Publishers Inc. Weston J, Mukherjee S, Chapelle O, Pontil M, Poggio T, Vapnik V. (2001) Feature Selection for SVMs. Weston J, Perez-Cruz F, Bousquet O, Chapelle O, Elisseeff A, Scholkopf B (2003). Feature selection and transduction for prediction of molecular bioactivity for drug design. Bioinformatics 19(6):764-771. White RE (2000). High-throughput screening in drug metabolism and pharmacokinetic support of drug discovery. Annual Review of Pharmacology and Toxicology 40:133-157. Willett P, Barnard JM, Downs GM (1998). Chemical similarity searching. Journal of Chemical Information and Computer Sciences 38:983-996. Wold S, Eriksson L. (1995). Statistical validation of QSAR results. In: Van de Waterbeemd H, editor. Chemometric methods in molecular design. Weinheim; New York; Basel; Cambridge; Tokyo: Wiley-VCH. p 309-318. Wu C, Fry CH, Henry JA (1997). Membrane toxicity of opioids measured by protozoan BIBLIOGRAPHY 215 motility. Toxicology 117(1):35-44. Xie W, Uppal H, Saini SP, Mu Y, Little JM, Radominska-Pandya A, Zemaitis MA (2004). Orphan nuclear receptor-mediated xenobiotic regulation in drug metabolism. Drug Discovery Today 9(10):442-449. Xu L, Zhang WJ (2001). Comparison of different methods for variable selection. Analytica Chimica Acta 446:475-481. Xue CX, Zhang RS, Liu HX, Yao XJ, Liu MC, Hu ZD, Fan BT (2004a). QSAR models for the prediction of binding affinities to human serum albumin using the heuristic method and a support vector machine. Journal of Chemical Information and Computer Sciences 44(5):1693-1700. Xue Y, Li H, Ung CY, Yap CW, Chen YZ (2006). Classification of a diverse set of Tetrahymena pyriformis toxicity chemical compounds from molecular descriptors by statistical learning methods. Chemical Research in Toxicology 19(8):10301039. Xue Y, Li ZR, Yap CW, Sun LZ, Chen X, Chen YZ (2004b). Effect of Molecular Descriptor Feature Selection in Support Vector Machine Classification of Pharmacokinetic and Toxicological Properties of Chemical Agents. Journal of Chemical Information and Computer Sciences 44(5):1630-1638. Xue Y, Yap CW, Sun LZ, Cao ZW, Wang JF, Chen YZ (2004c). Prediction of pglycoprotein substrates by support vector machine approach. Journal of Chemical Information and Computer Sciences 44(4):1497-1505. Yamazaki K, Kanaoka M (2004). Computational prediction of the plasma protein-binding percent of diverse pharmaceutical compounds. Journal of Pharmaceutical Sciences 93(6):1480-1494. Yao X, Liu H, Zhang R, Liu M, Hu Z, Panaye A, Doucet JP, Fan B (2005). QSAR and classification study of 1,4-dihydropyridine calcium channel antagonists based on least squares support vector machines. Molecular Pharmaceutics 2(5):348-356. Yap CW, Cai CZ, Xue Y, Chen YZ (2004a). Prediction of torsade-causing potential of drugs by support vector machine approach. Toxicological Sciences 79(1):170-177. Yap CW, Chen YZ (2004b). Quantitative structure-pharmacokinetic relationships for drug distribution properties by using general regression neural network. Journal of Pharmaceutical Sciences 94(1):153-168. Yap CW, Chen YZ (2005). Prediction of cytochrome P450 3A4, 2D6, and 2C9 inhibitors and substrates by using support vector machines. Journal of Chemical Information and Modeling. 45(4):982-992. Young RC, Mitchell RC, Brown TH, Ganellin CR, Griffith R, Jones M, Rana KK, BIBLIOGRAPHY 216 Saundesr D, Smith IR, Sore NE and others (1988). Development of a new physicochemical model for brain penetration and its application to the design of centrally acting H2 receptor histamine antagonists. Journal of Medicinal Chemistry 31:656-671. Yu H, Yang J, Wang W, Han J. (2003) Discovering Compact and Highly Discriminative Features or Feature Combinations of Drug Activities Using Support Vector Machines.; August 11 - 14; Stanford, California. 220-228. Zernov VV, Balakin KV, Ivaschenko AA, Savchuk NP, Pletnev IV (2003). Drug discovery using support vector machines. The case studies of drug-likeness, agrochemical-likeness, and enzyme inhibition predictions. Journal of Chemical Information and Computer Sciences 43(6):2048-2056. Zuegge J, Fechner U, Roche O, Parrott NJ, Engkvist O, Schneider G (2002). A fast virtual screening filter for cytochrome P450 3A4 inhibition liability of compound libraries. Quantitative Structure-Activity Relationships 21(3):249-256. [...]... part of the efforts for developing such tools, computational methods have been explored for predicting various pharmacological properties of pharmaceutical agents SLMs are the computational approaches that are increasingly used for in silico HTS of compounds with diverse structures in early drug discovery stage 1.2 Statistical learning methods for characterization pharmacological properties of pharmaceutical. .. a decision tree process for the prediction of pharmaceutical agents of a particular pharmacological property from their structure by using a statistical learning method – C4.5 decision tree 46 Figure 2.4 Schematic diagrams illustrating the process of the prediction of pharmaceutical agents with a particular pharmacological property from its structure by using a statistical learning method — k-nearest... process of using feature selection method for selecting molecular descriptors most appropriate in the prediction of compounds of a particular pharmacological property by statistical learning methods 42 Figure 2.2 Schematic diagrams illustrating the process of the prediction of pharmaceutical agents with a particular pharmacological property from its structure by using a statistical learning. .. illustrating the process of the prediction of pharmaceutical agents with a particular pharmacological property from its structure by using a statistical learning method — probabilistic neural networks (PNN) 50 Figure 2.6 PNN four layers architecture 51 Figure 2.7 Schematic diagram illustrating the process of the prediction of pharmaceutical agents with a particular pharmacological property... INTRODUCTION 5 al 2004] have been the primary reasons for the failure of drug candidates in later stages of drug development Tools for predicting pharmacokinetic and pharmacodynamic properties as well as toxicological properties in the early drug design stages are needed for fast elimination of agents with undesirable properties so that development efforts can be focused on the most promising candidates... classification of genotoxic (GT+) and non-genotoxic (GT-) agents 141 Table 5.4 Overview of the prediction accuracies of genotoxic (GT+) and non-genotoxic (GT-) agents from this work as with those from other studies 145 xii Table 5.5 Comparison of the prediction accuracy of Tetrahymena pyriformis toxic (TPT) and non-toxic (non-TPT) agents by different statistical learning methods 161 Table 5.6 Performance... development of additional molecular descriptors for representing agents of more complex structures and for modeling more extensive sets of properties Detailed description of molecular descriptors will be presented in Chapter 2 1.4 Feature selection methods In practice, not all of those molecular descriptors are needed for representing features of a particular class of agents Among the descriptors some are irrelevant... Performance of support vector machines (SVM) and support vector machines with recursive feature elimination (SVM+RFE) for predicting Tetrahymena pyriformis toxic (TPT) and non-toxic (non-TPT) agents as evaluated by 5-fold cross validation 162 Table 5.7 Comparison of the prediction accuracy of Tetrahymena pyriformis toxic (TPT) and non-toxic (non-TPT) agents by different statistical learning. .. Li, C.W Yap, Y Xue, Z.R Li, C.Y Ung, L.Y Han, and Y.Z Chen Statistical Learning Approach for Predicting Specific Pharmacodynamic, Pharmacokinetic or Toxicological Properties of Pharmaceutical Agents Drug Dev Res 2006; 66 (4):245-259 5 H Li, C.W Yap, C.Y Ung, Y Xue, Z.R Li, L.Y Han, H.H Lin and Y.Z Chen Machine Learning Approaches for Predicting Compounds That Interact with Therapeutic and ADMET Related... capability of these statistical models The computed r2 values of these regression-based SLMs listed in Table 1.1 are in the range from 0.51 to 0.95 [Ertl et al 2000; Yamazaki et al 2004], which are at a level useful for predicting the activity values of compounds of particular pharmacological properties In an attempt to develop pharmacological property prediction models that cover more diverse ranges of structures . pharmacological properties of pharmaceutical agents 2 1.2 Statistical learning methods in characterization of pharmacological properties of pharmaceutical agents 5 1.3 Describing molecular properties using. STATISTICAL LEARNING APPROACHES FOR PREDICTING PHARMACOLOGICAL PROPERTIES OF PHARMACEUTICAL AGENTS LI HU (B.Sc, M.Sc, Jilin University) A THESIS SUBMITTED FOR THE. been explored for predicting various pharmacological properties of pharmaceutical agents. In particular, statistical learning methods (SLMs) have shown promise for these tasks by statistically

Ngày đăng: 14/09/2015, 12:43

TỪ KHÓA LIÊN QUAN

TÀI LIỆU CÙNG NGƯỜI DÙNG

TÀI LIỆU LIÊN QUAN