87 research outputs found

    Training and assessing classification rules with unbalanced data

    Get PDF
    The problem of modeling binary responses by using cross-sectional data has been addressed with a number of satisfying solutions that draw on both parametric and nonparametric methods. However, there exist many real situations where one of the two responses (usually the most interesting for the analysis) is rare. It has been largely reported that this class imbalance heavily compromises the process of learning, because the model tends to focus on the prevalent class and to ignore the rare events. However, not only the estimation of the classification model is affected by a skewed distribution of the classes, but also the evaluation of its accuracy is jeopardized, because the scarcity of data leads to poor estimates of the model’s accuracy. In this work, the effects of class imbalance on model training and model assessing are discussed. Moreover, a unified and systematic framework for dealing with both the problems is proposed, based on a smoothed bootstrap re-sampling technique

    Asymmetric bagging and feature selection for activities prediction of drug molecules

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Activities of drug molecules can be predicted by QSAR (quantitative structure activity relationship) models, which overcomes the disadvantages of high cost and long cycle by employing the traditional experimental method. With the fact that the number of drug molecules with positive activity is rather fewer than that of negatives, it is important to predict molecular activities considering such an unbalanced situation.</p> <p>Results</p> <p>Here, asymmetric bagging and feature selection are introduced into the problem and asymmetric bagging of support vector machines (asBagging) is proposed on predicting drug activities to treat the unbalanced problem. At the same time, the features extracted from the structures of drug molecules affect prediction accuracy of QSAR models. Therefore, a novel algorithm named PRIFEAB is proposed, which applies an embedded feature selection method to remove redundant and irrelevant features for asBagging. Numerical experimental results on a data set of molecular activities show that asBagging improve the AUC and sensitivity values of molecular activities and PRIFEAB with feature selection further helps to improve the prediction ability.</p> <p>Conclusion</p> <p>Asymmetric bagging can help to improve prediction accuracy of activities of drug molecules, which can be furthermore improved by performing feature selection to select relevant features from the drug molecules data sets.</p

    Supervised Methods for Biomarker Detection from Microarray Experiments

    Get PDF
    Biomarkers are valuable indicators of the state of a biological system. Microarray technology has been extensively used to identify biomarkers and build computational predictive models for disease prognosis, drug sensitivity and toxicity evaluations. Activation biomarkers can be used to understand the underlying signaling cascades, mechanisms of action and biological cross talk. Biomarker detection from microarray data requires several considerations both from the biological and computational points of view. In this chapter, we describe the main methodology used in biomarkers discovery and predictive modeling and we address some of the related challenges. Moreover, we discuss biomarker validation and give some insights into multiomics strategies for biomarker detection.Non peer reviewe

    Prediction of drug-drug interaction potential using machine learning approaches

    Get PDF
    Drug discovery is a long, expensive, and complex, yet crucial process for the benefit of society. Selecting potential drug candidates requires an understanding of how well a compound will perform at its task, and more importantly, how safe the compound will act in patients. A key safety insight is understanding a molecule\u27s potential for drug-drug interactions. The metabolism of many drugs is mediated by members of the cytochrome P450 superfamily, notably, the CYP3A4 enzyme. Inhibition of these enzymes can alter the bioavailability of other drugs, potentially increasing their levels to toxic amounts. Four models were developed to predict CYP3A4 inhibition: logistic regression, random forests, support vector machine, and neural network. Two novel convolutional approaches were explored for data featurization: SMILES string auto-extraction and 2D structure auto-extraction. The logistic regression model achieved an accuracy of 83.2%, the random forests model, 83.4%, the support vector machine model, 81.9%, and the neural network model, 82.3%. Additionally, the model built with SMILE string auto-extraction had an accuracy of 82.3%, and the model with 2D structure auto-extraction, 76.4%. The advantages of the novel featurization methods are their ability to learn relevant features from compound SMILE strings, eliminating feature engineering. The developed methodologies can be extended towards predicting any structure-activity relationship and fitted for other areas of drug discovery and development

    Computational prediction of metabolism: sites, products, SAR, P450 enzyme dynamics, and mechanisms.

    Get PDF
    Metabolism of xenobiotics remains a central challenge for the discovery and development of drugs, cosmetics, nutritional supplements, and agrochemicals. Metabolic transformations are frequently related to the incidence of toxic effects that may result from the emergence of reactive species, the systemic accumulation of metabolites, or by induction of metabolic pathways. Experimental investigation of the metabolism of small organic molecules is particularly resource demanding; hence, computational methods are of considerable interest to complement experimental approaches. This review provides a broad overview of structure- and ligand-based computational methods for the prediction of xenobiotic metabolism. Current computational approaches to address xenobiotic metabolism are discussed from three major perspectives: (i) prediction of sites of metabolism (SOMs), (ii) elucidation of potential metabolites and their chemical structures, and (iii) prediction of direct and indirect effects of xenobiotics on metabolizing enzymes, where the focus is on the cytochrome P450 (CYP) superfamily of enzymes, the cardinal xenobiotics metabolizing enzymes. For each of these domains, a variety of approaches and their applications are systematically reviewed, including expert systems, data mining approaches, quantitative structure-activity relationships (QSARs), and machine learning-based methods, pharmacophore-based algorithms, shape-focused techniques, molecular interaction fields (MIFs), reactivity-focused techniques, protein-ligand docking, molecular dynamics (MD) simulations, and combinations of methods. Predictive metabolism is a developing area, and there is still enormous potential for improvement. However, it is clear that the combination of rapidly increasing amounts of available ligand- and structure-related experimental data (in particular, quantitative data) with novel and diverse simulation and modeling approaches is accelerating the development of effective tools for prediction of in vivo metabolism, which is reflected by the diverse and comprehensive data sources and methods for metabolism prediction reviewed here. This review attempts to survey the range and scope of computational methods applied to metabolism prediction and also to compare and contrast their applicability and performance.JK, MJW, JT, PJB, AB and RCG thank Unilever for funding

    Machine Learning for Modelling Tissue Distribution of Drugs and the Impact of Transporters

    Get PDF
    The ability to predict human pharmacokinetics in early stages of drug development is of paramount importance to prevent late stage attrition as well as in managing toxicity. This thesis explores the machine learning modelling of one of the main pharmacokinetics parameters that determines the therapeutic success of a drug - volume of distribution. In order to do so, a variety of physiological phenomena with known mechanisms of impact on drug distribution were considered as input features during the modelling of volume of distribution namely, Solute Carriers-mediated uptake and ATP-binding Cassette-mediated efflux, drug-induced phospholipidosis and plasma protein binding. These were paired with molecular descriptors to provide both chemical and biological information to the building of the predictive models. Since biological data used as input is limited, prior to modelling volume of distribution, the various types of physiological descriptors were also modelled. Here, a focus was placed on harnessing the information contained in correlations within the two transporter families, which was done by using multi-label classification. The application of such approach to transporter data is very recent and its use to model Solute Carriers data, for example, is reported here for the first time. On both transporter families, there was evidence that accounting for correlations between transporters offers useful information that is not portrayed by molecular descriptors. This effort also allowed uncovering new potential links between members of the Solute Carriers family, which are not obvious from a purely physiological standpoint. The models created for the different physiological parameters were then used to predict these parameters and fill in the gaps in the available experimental data, and the resulting merging of experimental and predicted data was used to model volume of distribution. This exercise improved the accuracy of volume of distribution models, and the generated models incorporated a wide variety of the different physiological descriptors supplied along with molecular features. The use of most of these physiological descriptors in the modelling of distribution is unprecedented, which is one of the main novelty points of this thesis. Additionally, as a parallel complementary work, a new method to characterize the predictive reliability of machine learning classification model was proposed, and an in depth analysis of mispredictions, their trends and causes was carried out, using one of the transporter models as example. This is an important complement to the main body of work in this thesis, as predictive performance is necessarily tied to prediction reliability

    Data mining methods for the prediction of intestinal absorption using QSAR

    Get PDF
    Oral administration is the most common route for administration of drugs. With the growing cost of drug discovery, the development of Quantitative Structure-Activity Relationships (QSAR) as computational methods to predict oral absorption is highly desirable for cost effective reasons. The aim of this research was to develop QSAR models that are highly accurate and interpretable for the prediction of oral absorption. In this investigation the problems addressed were datasets with unbalanced class distributions, feature selection and the effects of solubility and permeability towards oral absorption prediction. Firstly, oral absorption models were obtained by overcoming the problem of unbalanced class distributions in datasets using two techniques, under-sampling of compounds belonging to the majority class and the use of different misclassification costs for different types of misclassifications. Using these methods, models with higher accuracy were produced using regression and linear/non-linear classification techniques. Secondly, the use of several pre-processing feature selection methods in tandem with decision tree classification analysis – including misclassification costs – were found to produce models with better interpretability and higher predictive accuracy. These methods were successful to select the most important molecular descriptors and to overcome the problem of unbalanced classes. Thirdly, the roles of solubility and permeability in oral absorption were also investigated. This involved expansion of oral absorption datasets and collection of in vitro and aqueous solubility data. This work found that the inclusion of predicted and experimental solubility in permeability models can improve model accuracy. However, the impact of solubility on oral absorption prediction was not as influential as expected. Finally, predictive models of permeability and solubility were built to predict a provisional Biopharmaceutic Classification System (BCS) class using two multi-label classification techniques, binary relevance and classifier chain. The classifier chain method was shown to have higher predictive accuracy by using predicted solubility as a molecular descriptor for permeability models, and hence better final provisional BCS prediction. Overall, this research has resulted in predictive and interpretable models that could be useful in a drug discovery context

    Machine Learning Applications for Drug Repurposing

    Full text link
    The cost of bringing a drug to market is astounding and the failure rate is intimidating. Drug discovery has been of limited success under the conventional reductionist model of one-drug-one-gene-one-disease paradigm, where a single disease-associated gene is identified and a molecular binder to the specific target is subsequently designed. Under the simplistic paradigm of drug discovery, a drug molecule is assumed to interact only with the intended on-target. However, small molecular drugs often interact with multiple targets, and those off-target interactions are not considered under the conventional paradigm. As a result, drug-induced side effects and adverse reactions are often neglected until a very late stage of the drug discovery, where the discovery of drug-induced side effects and potential drug resistance can decrease the value of the drug and even completely invalidate the use of the drug. Thus, a new paradigm in drug discovery is needed. Structural systems pharmacology is a new paradigm in drug discovery that the drug activities are studied by data-driven large-scale models with considerations of the structures and drugs. Structural systems pharmacology will model, on a genome scale, the energetic and dynamic modifications of protein targets by drug molecules as well as the subsequent collective effects of drug-target interactions on the phenotypic drug responses. To date, however, few experimental and computational methods can determine genome-wide protein-ligand interaction networks and the clinical outcomes mediated by them. As a result, the majority of proteins have not been charted for their small molecular ligands; we have a limited understanding of drug actions. To address the challenge, this dissertation seeks to develop and experimentally validate innovative computational methods to infer genome-wide protein-ligand interactions and multi-scale drug-phenotype associations, including drug-induced side effects. The hypothesis is that the integration of data-driven bioinformatics tools with structure-and-mechanism-based molecular modeling methods will lead to an optimal tool for accurately predicting drug actions and drug associated phenotypic responses, such as side effects. This dissertation starts by reviewing the current status of computational drug discovery for complex diseases in Chapter 1. In Chapter 2, we present REMAP, a one-class collaborative filtering method to predict off-target interactions from protein-ligand interaction network. In our later work, REMAP was integrated with structural genomics and statistical machine learning methods to design a dual-indication polypharmacological anticancer therapy. In Chapter 3, we extend REMAP, the core method in Chapter 2, into a multi-ranked collaborative filtering algorithm, WINTF, and present relevant mathematical justifications. Chapter 4 is an application of WINTF to repurpose an FDA-approved drug diazoxide as a potential treatment for triple negative breast cancer, a deadly subtype of breast cancer. In Chapter 5, we present a multilayer extension of REMAP, applied to predict drug-induced side effects and the associated biological pathways. In Chapter 6, we close this dissertation by presenting a deep learning application to learn biochemical features from protein sequence representation using a natural language processing method
    corecore