Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Chem Inf Model ; 63(6): 1675-1694, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36926871

RESUMEN

Cytochrome P450 enzymes aid in the elimination of a preponderance of small molecule drugs, but can generate reactive metabolites that may adversely react with protein and DNA and prompt drug candidate attrition or market withdrawal. Previously developed models help understand how these enzymes modify molecule structure by predicting sites of metabolism or characterizing formation of metabolite-biomolecule adducts. However, the majority of reactive metabolites are formed by multiple metabolic steps, and understanding the progenitor molecule's network-level behavior necessitates an integrative approach that blends multiple site of metabolism and structure inference models. Our previously developed tool, XenoNet 1.0, generates metabolic networks, where nodes are molecules and weighted edges are metabolic transformations. We extend XenoNet with a bidirectional message passing neural network that integrates edge feature information and local network structure using edge-conditioned graph convolutions and jumping knowledge to predict the authenticity of inferred Phase I metabolite structures. Our model significantly outperformed prior work and algorithmic baselines on a data set of 311 networks and 6606 intermediates annotated using a chemically diverse set of 20 736 individual in vitro and in vivo reaction records accounting for 92.3% of all human Phase I metabolism in the Accelrys Metabolite Database. Cross-validated predictions resulted in area under the receiver operating characteristic curves of 88.5% and 87.6% for separating experimentally observed and unobserved metabolites at global and network levels, respectively. Further analysis verified robustness to networks of varying depth and breadth, accurate detection of metabolites, such as d,l-methamphetamine, that are experimentally observed or unobserved in different network contexts, extraction of important metabolic subnetworks, and identification of known bioactivation pathways, such as for nimesulide and terbinafine. By exploiting network structures, our approach accurately suggests unreported metabolites for experimental study and may rationalize modifications for avoiding deleterious pathways antecedent to reactive metabolite formation.


Asunto(s)
Redes y Vías Metabólicas , Redes Neurales de la Computación , Humanos , Estructura Molecular , Terbinafina/metabolismo
2.
Curr Opin Nephrol Hypertens ; 31(3): 251-257, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35165248

RESUMEN

PURPOSE OF REVIEW: The field of pathology is currently undergoing a significant transformation from traditional glass slides to a digital format dependent on whole slide imaging. Transitioning from glass to digital has opened the field to development and application of image analysis technology, commonly deep learning methods (artificial intelligence [AI]) to assist pathologists with tissue examination. Nephropathology is poised to leverage this technology to improve precision, accuracy, and efficiency in clinical practice. RECENT FINDINGS: Through a multidisciplinary approach, nephropathologists, and computer scientists have made significant recent advances in developing AI technology to identify histological structures within whole slide images (segmentation), quantification of histologic structures, prediction of clinical outcomes, and classifying disease. Virtual staining of tissue and automation of electron microscopy imaging are emerging applications with particular significance for nephropathology. SUMMARY: AI applied to image analysis in nephropathology has potential to transform the field by improving diagnostic accuracy and reproducibility, efficiency, and prognostic power. Reimbursement, demonstration of clinical utility, and seamless workflow integration are essential to widespread adoption.


Asunto(s)
Inteligencia Artificial , Procesamiento de Imagen Asistido por Computador , Computadores , Humanos , Riñón/diagnóstico por imagen , Reproducibilidad de los Resultados
3.
PLoS Comput Biol ; 17(7): e1009053, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34228716

RESUMEN

Drug-drug interactions account for up to 30% of adverse drug reactions. Increasing prevalence of electronic health records (EHRs) offers a unique opportunity to build machine learning algorithms to identify drug-drug interactions that drive adverse events. In this study, we investigated hospitalizations' data to study drug interactions with non-steroidal anti-inflammatory drugs (NSAIDS) that result in drug-induced liver injury (DILI). We propose a logistic regression based machine learning algorithm that unearths several known interactions from an EHR dataset of about 400,000 hospitalization. Our proposed modeling framework is successful in detecting 87.5% of the positive controls, which are defined by drugs known to interact with diclofenac causing an increased risk of DILI, and correctly ranks aggregate risk of DILI for eight commonly prescribed NSAIDs. We found that our modeling framework is particularly successful in inferring associations of drug-drug interactions from relatively small EHR datasets. Furthermore, we have identified a novel and potentially hepatotoxic interaction that might occur during concomitant use of meloxicam and esomeprazole, which are commonly prescribed together to allay NSAID-induced gastrointestinal (GI) bleeding. Empirically, we validate our approach against prior methods for signal detection on EHR datasets, in which our proposed approach outperforms all the compared methods across most metrics, such as area under the receiver operating characteristic curve (AUROC) and area under the precision-recall curve (AUPRC).


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas , Interacciones Farmacológicas , Registros Electrónicos de Salud/estadística & datos numéricos , Aprendizaje Automático , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Enfermedad Hepática Inducida por Sustancias y Drogas/epidemiología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Biología Computacional , Femenino , Humanos , Hígado/efectos de los fármacos , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Estudios Retrospectivos , Adulto Joven
4.
Drug Metab Dispos ; 49(2): 133-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33239334

RESUMEN

Meclofenamate is a nonsteroidal anti-inflammatory drug used in the treatment of mild-to-moderate pain yet poses a rare risk of hepatotoxicity through an unknown mechanism. Nonsteroidal anti-inflammatory drug (NSAID) bioactivation is a common molecular initiating event for hepatotoxicity. Thus, we hypothesized a similar mechanism for meclofenamate and leveraged computational and experimental approaches to identify and characterize its bioactivation. Analyses employing our XenoNet model indicated possible pathways to meclofenamate bioactivation into 19 reactive metabolites subsequently trapped into glutathione adducts. We describe the first reported bioactivation kinetics for meclofenamate and relative importance of those pathways using human liver microsomes. The findings validated only four of the many bioactivation pathways predicted by modeling. For experimental studies, dansyl glutathione was a critical trap for reactive quinone metabolites and provided a way to characterize adduct structures by mass spectrometry and quantitate yields during reactions. Of the four quinone adducts, we were able to characterize structures for three of them. Based on kinetics, the most efficient bioactivation pathway led to the monohydroxy para-quinone-imine followed by the dechloro-ortho-quinone-imine. Two very inefficient pathways led to the dihydroxy ortho-quinone and a likely multiply adducted quinone. When taken together, bioactivation pathways for meclofenamate accounted for approximately 13% of total metabolism. In sum, XenoNet facilitated prediction of reactive metabolite structures, whereas quantitative experimental studies provided a tractable approach to validate actual bioactivation pathways for meclofenamate. Our results provide a foundation for assessing reactive metabolite load more accurately for future comparative studies with other NSAIDs and drugs in general. SIGNIFICANCE STATEMENT: Meclofenamate bioactivation may initiate hepatotoxicity, yet common risk assessment approaches are often cumbersome and inefficient and yield qualitative insights that do not scale relative bioactivation risks. We developed and applied innovative computational modeling and quantitative kinetics to identify and validate meclofenamate bioactivation pathways and relevance as a function of time and concentration. This strategy yielded novel insights on meclofenamate bioactivation and provides a tractable approach to more accurately and efficiently assess other drug bioactivations and correlate risks to toxicological outcomes.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Ácido Meclofenámico/farmacocinética , Activación Metabólica , Benzoquinonas/metabolismo , Cromatografía Liquida , Glutatión/metabolismo , Humanos , Espectrometría de Masas , Microsomas Hepáticos/metabolismo , Modelos Químicos , Espectrometría de Fluorescencia
5.
Chem Res Toxicol ; 34(2): 584-600, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33496184

RESUMEN

Electrophilically reactive drug metabolites are implicated in many adverse drug reactions. In this mechanism-termed bioactivation-metabolic enzymes convert drugs into reactive metabolites that often conjugate to nucleophilic sites within biological macromolecules like proteins. Toxic metabolite-product adducts induce severe immune responses that can cause sometimes fatal disorders, most commonly in the form of liver injury, blood dyscrasia, or the dermatologic conditions toxic epidermal necrolysis and Stevens-Johnson syndrome. This study models four of the most common metabolic transformations that result in bioactivation: quinone formation, epoxidation, thiophene sulfur-oxidation, and nitroaromatic reduction, by synthesizing models of metabolism and reactivity. First, the metabolism models predict the formation probabilities of all possible metabolites among the pathways studied. Second, the exact structures of these metabolites are enumerated. Third, using these structures, the reactivity model predicts the reactivity of each metabolite. Finally, a feedfoward neural network converts the metabolism and reactivity predictions to a bioactivation prediction for each possible metabolite. These bioactivation predictions represent the joint probability that a metabolite forms and that this metabolite subsequently conjugates to protein or glutathione. Among molecules bioactivated by these pathways, we predicted the correct pathway with an AUC accuracy of 89.98%. Furthermore, the model predicts whether molecules will be bioactivated, distinguishing bioactivated and nonbioactivated molecules with 81.06% AUC. We applied this algorithm to withdrawn drugs. The known bioactivation pathways of alclofenac and benzbromarone were identified by the algorithm, and high probability bioactivation pathways not yet confirmed were identified for safrazine, zimelidine, and astemizole. This bioactivation model-the first of its kind that jointly considers both metabolism and reactivity-enables drug candidates to be quickly evaluated for a toxicity risk that often evades detection during preclinical trials. The XenoSite bioactivation model is available at http://swami.wustl.edu/xenosite/p/bioactivation.


Asunto(s)
Compuestos Epoxi/metabolismo , Modelos Biológicos , Nitrobencenos/metabolismo , Quinonas/metabolismo , Azufre/metabolismo , Tiofenos/metabolismo , Compuestos Epoxi/química , Humanos , Estructura Molecular , Nitrobencenos/química , Oxidación-Reducción , Quinonas/química , Azufre/química , Tiofenos/química
6.
J Phys Chem A ; 125(40): 8978-8986, 2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34609871

RESUMEN

Computing quantum chemical properties of small molecules and polymers can provide insights valuable into physicists, chemists, and biologists when designing new materials, catalysts, biological probes, and drugs. Deep learning can compute quantum chemical properties accurately in a fraction of time required by commonly used methods such as density functional theory. Most current approaches to deep learning in quantum chemistry begin with geometric information from experimentally derived molecular structures or pre-calculated atom coordinates. These approaches have many useful applications, but they can be costly in time and computational resources. In this study, we demonstrate that accurate quantum chemical computations can be performed without geometric information by operating in the coordinate-free domain using deep learning on graph encodings. Coordinate-free methods rely only on molecular graphs, the connectivity of atoms and bonds, without atom coordinates or bond distances. We also find that the choice of graph-encoding architecture substantially affects the performance of these methods. The structures of these graph-encoding architectures provide an opportunity to probe an important, outstanding question in quantum mechanics: what types of quantum chemical properties can be represented by local variable models? We find that Wave, a local variable model, accurately calculates the quantum chemical properties, while graph convolutional architectures require global variables. Furthermore, local variable Wave models outperform global variable graph convolution models on complex molecules with large, correlated systems.

7.
J Chem Inf Model ; 60(7): 3431-3449, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32525671

RESUMEN

Drug metabolism is a common cause of adverse drug reactions. Drug molecules can be metabolized into reactive metabolites, which can conjugate to biomolecules, like protein and DNA, in a process termed bioactivation. To mitigate adverse reactions caused by bioactivation, both experimental and computational screening assays are utilized. Experimental assays for assessing the formation of reactive metabolites are low throughput and expensive to perform, so they are often reserved until later stages of the drug development pipeline when the drug candidate pools are already significantly narrowed. In contrast, computational methods are high throughput and cheap to perform to screen thousands to millions of compounds for potentially toxic molecules during the early stages of the drug development pipeline. Commonly used computational methods focus on detecting and structurally characterizing reactive metabolite-biomolecule adducts or predicting sites on a drug molecule that are liable to form reactive metabolites. However, such methods are often only concerned with the structure of the initial drug molecule or of the adduct formed when a biomolecule conjugates to a reactive metabolite. Thus, these methods are likely to miss intermediate metabolites that may lead to subsequent reactive metabolite formation. To address these shortcomings, we create XenoNet, a metabolic network predictor, that can take a pair of a substrate and a target product as input and (1) enumerate pathways, or sequences of intermediate metabolite structures, between the pair, and (2) compute the likelihood of those pathways and intermediate metabolites. We validate XenoNet on a large, chemically diverse data set of 17 054 metabolic networks built from a literature-derived reaction database. Each metabolic network has a defined substrate molecule that has been experimentally observed to undergo metabolism into a defined product metabolite. XenoNet can predict experimentally observed pathways and intermediate metabolites linking the input substrate and product pair with a recall of 88 and 46%, respectively. Using likelihood scoring, XenoNet also achieves a top-one pathway and intermediate metabolite accuracy of 93.6 and 51.9%, respectively. We further validate XenoNet against prior methods for metabolite prediction. XenoNet significantly outperforms all prior methods across multiple metrics. XenoNet is available at https://swami.wustl.edu/xenonet.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Redes y Vías Metabólicas
8.
J Chem Inf Model ; 60(3): 1146-1164, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32040319

RESUMEN

Metabolism of drugs affects their absorption, distribution, efficacy, excretion, and toxicity profiles. Metabolism is routinely assessed experimentally using recombinant enzymes, human liver microsome, and animal models. Unfortunately, these experiments are expensive, time-consuming, and often extrapolate poorly to humans because they fail to capture the full breadth of metabolic reactions observed in vivo. As a result, metabolic pathways leading to the formation of toxic metabolites are often missed during drug development, giving rise to costly failures. To address some of these limitations, computational metabolism models can rapidly and cost-effectively predict sites of metabolism-the atoms or bonds which undergo enzymatic modifications-on thousands of drug candidates, thereby improving the likelihood of discovering metabolic transformations forming toxic metabolites. However, current computational metabolism models are often unable to predict the specific metabolites formed by metabolism at certain sites. Identification of reaction type is a key step toward metabolite prediction. Phase I enzymes, which are responsible for the metabolism of more than 90% of FDA approved drugs, catalyze highly diverse types of reactions and produce metabolites with substantial structural variability. Without knowledge of potential metabolite structures, medicinal chemists cannot differentiate harmful metabolic transformations from beneficial ones. To address this shortcoming, we propose a system for simultaneously labeling sites of metabolism and reaction types, by classifying them into five key reaction classes: stable and unstable oxidations, dehydrogenation, hydrolysis, and reduction. These classes unambiguously identify 21 types of phase I reactions, which cover 92.3% of known reactions in our database. We used this labeling system to train a neural network model of phase I metabolism on a literature-derived data set encompassing 20 736 human phase I metabolic reactions. Our model, Rainbow XenoSite, was able to identify reaction-type specific sites of metabolism with a cross-validated accuracy of 97.1% area under the receiver operator curve. Rainbow XenoSite with five-color and combined output is available for use free and online through our secure server at http://swami.wustl.edu/xenosite/p/phase1_rainbow.


Asunto(s)
Aprendizaje Profundo , Animales , Color , Humanos , Redes y Vías Metabólicas , Microsomas Hepáticos , Redes Neurales de la Computación
9.
J Chem Inf Model ; 60(10): 4702-4716, 2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-32881497

RESUMEN

Adverse drug metabolism often severely impacts patient morbidity and mortality. Unfortunately, drug metabolism experimental assays are costly, inefficient, and slow. Instead, computational modeling could rapidly flag potentially toxic molecules across thousands of candidates in the early stages of drug development. Most metabolism models focus on predicting sites of metabolism (SOMs): the specific substrate atoms targeted by metabolic enzymes. However, SOMs are merely a proxy for metabolic structures: knowledge of an SOM does not explicitly provide the actual metabolite structure. Without an explicit metabolite structure, computational systems cannot evaluate the new molecule's properties. For example, the metabolite's reactivity cannot be automatically predicted, a crucial limitation because reactive drug metabolites are a key driver of adverse drug reactions (ADRs). Additionally, further metabolic events cannot be forecast, even though the metabolic path of the majority of substrates includes two or more sequential steps. To overcome the myopia of the SOM paradigm, this study constructs a well-defined system-termed the metabolic forest-for generating exact metabolite structures. We validate the metabolic forest with the substrate and product structures from a large, chemically diverse, literature-derived dataset of 20 736 records. The metabolic forest finds a pathway linking each substrate and product for 79.42% of these records. By performing a breadth-first search of depth two or three, we improve performance to 88.43 and 88.77%, respectively. The metabolic forest includes a specialized algorithm for producing accurate quinone structures, the most common type of reactive metabolite. To our knowledge, this quinone structure algorithm is the first of its kind, as the diverse mechanisms of quinone formation are difficult to systematically reproduce. We validate the metabolic forest on a previously published dataset of 576 quinone reactions, predicting their structures with a depth three performance of 91.84%. The metabolic forest accurately enumerates metabolite structures, enabling promising new directions such as joint metabolism and reactivity modeling.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas , Bosques , Humanos
10.
J Phys Chem A ; 124(44): 9194-9202, 2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33084331

RESUMEN

Atom- or bond-level chemical properties of interest in medicinal chemistry, such as drug metabolism and electrophilic reactivity, are important to understand and predict across arbitrary new molecules. Deep learning can be used to map molecular structures to their chemical properties, but the data sets for these tasks are relatively small, which can limit accuracy and generalizability. To overcome this limitation, it would be preferable to model these properties on the basis of the underlying quantum chemical characteristics of small molecules. However, it is difficult to learn higher level chemical properties from lower level quantum calculations. To overcome this challenge, we pretrained deep learning models to compute quantum chemical properties and then reused the intermediate representations constructed by the pretrained network. Transfer learning, in this way, substantially outperformed models based on chemical graphs alone or quantum chemical properties alone. This result was robust, observable in five prediction tasks: identifying sites of epoxidation by metabolic enzymes and identifying sites of covalent reactivity with cyanide, glutathione, DNA and protein. We see that this approach may substantially improve the accuracy of deep learning models for specific chemical structures, such as aromatic systems.


Asunto(s)
Aprendizaje Profundo , Teoría Cuántica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
11.
Genet Med ; 21(4): 972-981, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287923

RESUMEN

PURPOSE: Following automated variant calling, manual review of aligned read sequences is required to identify a high-quality list of somatic variants. Despite widespread use in analyzing sequence data, methods to standardize manual review have not been described, resulting in high inter- and intralab variability. METHODS: This manual review standard operating procedure (SOP) consists of methods to annotate variants with four different calls and 19 tags. The calls indicate a reviewer's confidence in each variant and the tags indicate commonly observed sequencing patterns and artifacts that inform the manual review call. Four individuals were asked to classify variants prior to, and after, reading the SOP and accuracy was assessed by comparing reviewer calls with orthogonal validation sequencing. RESULTS: After reading the SOP, average accuracy in somatic variant identification increased by 16.7% (p value = 0.0298) and average interreviewer agreement increased by 12.7% (p value < 0.001). Manual review conducted after reading the SOP did not significantly increase reviewer time. CONCLUSION: This SOP supports and enhances manual somatic variant detection by improving reviewer accuracy while reducing the interreviewer variability for variant calling and annotation.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/normas , Mutación/genética , Neoplasias/genética , Programas Informáticos , Algoritmos , Humanos , Neoplasias/patología , Polimorfismo de Nucleótido Simple/genética , Alineación de Secuencia
12.
Chem Res Toxicol ; 32(8): 1707-1721, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31304741

RESUMEN

Pediatric patients are at elevated risk of adverse drug reactions, and there is insufficient information on drug safety in children. Complicating risk assessment in children, there are numerous age-dependent changes in the absorption, distribution, metabolism, and elimination of drugs. A key contributor to age-dependent drug toxicity risk is the ontogeny of drug metabolism enzymes, the changes in both abundance and type throughout development from the fetal period through adulthood. Critically, these changes affect not only the overall clearance of drugs but also exposure to individual metabolites. In this study, we introduce time-embedding neural networks in order to model population-level variation in metabolism enzyme expression as a function of age. We use a time-embedding network to model the ontogeny of 23 drug metabolism enzymes. The time-embedding network recapitulates known demographic factors impacting 3A5 expression. The time-embedding network also effectively models the nonlinear dynamics of 2D6 expression, enabling a better fit to clinical data than prior work. In contrast, a standard neural network fails to model these features of 3A5 and 2D6 expression. Finally, we combine the time-embedding model of ontogeny with additional information to estimate age-dependent changes in reactive metabolite exposure. This simple approach identifies age-dependent changes in exposure to valproic acid and dextromethorphan metabolites and suggests potential mechanisms of valproic acid toxicity. This approach may help researchers evaluate the risk of drug toxicity in pediatric populations.


Asunto(s)
Neoplasias Hepáticas/metabolismo , Redes Neurales de la Computación , Adolescente , Carboxilesterasa/metabolismo , Niño , Preescolar , Sistema Enzimático del Citocromo P-450/metabolismo , Glucuronosiltransferasa/metabolismo , Glutatión Transferasa/metabolismo , Humanos , Inactivación Metabólica , Lactante , Oxigenasas/metabolismo , Análisis de Componente Principal , Sulfurtransferasas/metabolismo , Factores de Tiempo
13.
Chem Res Toxicol ; 32(6): 1151-1164, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-30925039

RESUMEN

Lamisil (terbinafine) is an effective, widely prescribed antifungal drug that causes rare idiosyncratic hepatotoxicity. The proposed toxic mechanism involves a reactive metabolite, 6,6-dimethyl-2-hepten-4-ynal (TBF-A), formed through three N-dealkylation pathways. We were the first to characterize them using in vitro studies with human liver microsomes and modeling approaches, yet knowledge of the individual enzymes catalyzing reactions remained unknown. Herein, we employed experimental and computational tools to assess terbinafine metabolism by specific cytochrome P450 isozymes. In vitro inhibitor phenotyping studies revealed six isozymes were involved in one or more N-dealkylation pathways. CYP2C19 and 3A4 contributed to all pathways, and so, we targeted them for steady-state analyses with recombinant isozymes. N-Dealkylation yielding TBF-A directly was catalyzed by CYP2C19 and 3A4 similarly. Nevertheless, CYP2C19 was more efficient than CYP3A4 at N-demethylation and other steps leading to TBF-A. Unlike microsomal reactions, N-denaphthylation was surprisingly efficient for CYP2C19 and 3A4, which was validated by controls. CYP2C19 was the most efficient among all reactions. Nonetheless, CYP3A4 was more selective at steps leading to TBF-A, making it more effective in terbinafine bioactivation based on metabolic split ratios for competing pathways. Model predictions did not extrapolate to quantitative kinetic constants, yet some results for CYP3A4 and CYP2C19 agreed qualitatively with preferred reaction steps and pathways. Clinical data on drug interactions support the CYP3A4 role in terbinafine metabolism, while CYP2C19 remains understudied. Taken together, knowledge of P450s responsible for terbinafine metabolism and TBF-A formation provides a foundation for investigating and mitigating the impact of P450 variations in toxic risks posed to patients.


Asunto(s)
Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inhibidores Enzimáticos/farmacología , Terbinafina/farmacología , Biocatálisis , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Cinética , Modelos Moleculares , Estructura Molecular , Terbinafina/química , Terbinafina/metabolismo
14.
Brief Bioinform ; 17(1): 2-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25832646

RESUMEN

Computational drug repositioning or repurposing is a promising and efficient tool for discovering new uses from existing drugs and holds the great potential for precision medicine in the age of big data. The explosive growth of large-scale genomic and phenotypic data, as well as data of small molecular compounds with granted regulatory approval, is enabling new developments for computational repositioning. To achieve the shortest path toward new drug indications, advanced data processing and analysis strategies are critical for making sense of these heterogeneous molecular measurements. In this review, we show recent advancements in the critical areas of computational drug repositioning from multiple aspects. First, we summarize available data sources and the corresponding computational repositioning strategies. Second, we characterize the commonly used computational techniques. Third, we discuss validation strategies for repositioning studies, including both computational and experimental methods. Finally, we highlight potential opportunities and use-cases, including a few target areas such as cancers. We conclude with a brief discussion of the remaining challenges in computational drug repositioning.


Asunto(s)
Reposicionamiento de Medicamentos/tendencias , Biología Computacional/tendencias , Minería de Datos , Combinación de Medicamentos , Reposicionamiento de Medicamentos/estadística & datos numéricos , Genómica , Humanos , Aprendizaje Automático , Estructura Molecular , Fenotipo , Encuestas y Cuestionarios
15.
Bioinformatics ; 33(15): 2288-2295, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28379348

RESUMEN

MOTIVATION: Characterizing the binding specificities of transcription factors (TFs) is crucial to the study of gene expression regulation. Recently developed high-throughput experimental methods, including protein binding microarrays (PBM) and high-throughput SELEX (HT-SELEX), have enabled rapid measurements of the specificities for hundreds of TFs. However, few studies have developed efficient algorithms for estimating binding motifs based on HT-SELEX data. Also the simple method of constructing a position weight matrix (PWM) by comparing the frequency of the preferred sequence with single-nucleotide variants has the risk of generating motifs with higher information content than the true binding specificity. RESULTS: We developed an algorithm called BEESEM that builds on a comprehensive biophysical model of protein-DNA interactions, which is trained using the expectation maximization method. BEESEM is capable of selecting the optimal motif length and calculating the confidence intervals of estimated parameters. By comparing BEESEM with the published motifs estimated using the same HT-SELEX data, we demonstrate that BEESEM provides significant improvements. We also evaluate several motif discovery algorithms on independent PBM and ChIP-seq data. BEESEM provides significantly better fits to in vitro data, but its performance is similar to some other methods on in vivo data under the criterion of the area under the receiver operating characteristic curve (AUROC). This highlights the limitations of the purely rank-based AUROC criterion. Using quantitative binding data to assess models, however, demonstrates that BEESEM improves on prior models. AVAILABILITY AND IMPLEMENTATION: Freely available on the web at http://stormo.wustl.edu/resources.html . CONTACT: stormo@wustl.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , ADN/metabolismo , Análisis por Matrices de Proteínas/métodos , Programas Informáticos , Termodinámica , Factores de Transcripción/metabolismo , Algoritmos , Animales , Sitios de Unión , ADN/química , Humanos , Ratones , Posición Específica de Matrices de Puntuación , Unión Proteica , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/química
16.
Chem Res Toxicol ; 31(2): 68-80, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29355304

RESUMEN

Cytochromes P450 (CYPs) oxidize alkylated amines commonly found in drugs and other biologically active molecules, cleaving them into an amine and an aldehyde. Metabolic studies usually neglect to report or investigate aldehydes, even though they can be toxic. It is assumed that they are efficiently detoxified into carboxylic acids and alcohols. Nevertheless, some aldehydes are reactive and escape detoxification pathways to cause adverse events by forming DNA and protein adducts. Herein, we modeled N-dealkylations that produce both amine and aldehyde metabolites and then predicted the reactivity of the aldehyde. This model used a deep learning approach previously developed by our group to predict other types of drug metabolism. In this study, we trained the model to predict N-dealkylation by human liver microsomes (HLM), finding that including isozyme-specific metabolism data alongside HLM data significantly improved results. The final HLM model accurately predicted the site of N-dealkylation within metabolized substrates (97% top-two and 94% area under the ROC curve). Next, we combined the metabolism, metabolite structure prediction, and previously published reactivity models into a bioactivation model. This combined model predicted the structure of the most likely reactive metabolite of a small validation set of drug-like molecules known to be bioactivated by N-dealkylation. Applying this model to approved and withdrawn medicines, we found that aldehyde metabolites produced from N-dealkylation may explain the hepatotoxicity of several drugs: indinavir, piperacillin, verapamil, and ziprasidone. Our results suggest that N-dealkylation may be an under-appreciated bioactivation pathway, especially in clinical contexts where aldehyde detoxification pathways are inhibited. Moreover, this is the first report of a bioactivation model constructed by combining a metabolism and reactivity model. These results raise hope that more comprehensive models of bioactivation are possible. The model developed in this study is available at http://swami.wustl.edu/xenosite/ .


Asunto(s)
Indinavir/metabolismo , Hígado/metabolismo , Microsomas Hepáticos/metabolismo , Piperacilina/metabolismo , Piperazinas/metabolismo , Tiazoles/metabolismo , Verapamilo/metabolismo , Aldehídos/química , Aldehídos/metabolismo , Aminas/química , Aminas/metabolismo , Remoción de Radical Alquila , Humanos , Indinavir/farmacología , Hígado/efectos de los fármacos , Microsomas Hepáticos/química , Microsomas Hepáticos/efectos de los fármacos , Modelos Moleculares , Estructura Molecular , Piperacilina/farmacología , Piperazinas/farmacología , Tiazoles/farmacología , Verapamilo/farmacología
17.
J Chem Inf Model ; 58(8): 1483-1500, 2018 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-29990427

RESUMEN

Scientists rely on high-throughput screening tools to identify promising small-molecule compounds for the development of biochemical probes and drugs. This study focuses on the identification of promiscuous bioactive compounds, which are compounds that appear active in many high-throughput screening experiments against diverse targets but are often false-positives which may not be easily developed into successful probes. These compounds can exhibit bioactivity due to nonspecific, intractable mechanisms of action and/or by interference with specific assay technology readouts. Such "frequent hitters" are now commonly identified using substructure filters, including pan assay interference compounds (PAINS). Herein, we show that mechanistic modeling of small-molecule reactivity using deep learning can improve upon PAINS filters when modeling promiscuous bioactivity in PubChem assays. Without training on high-throughput screening data, a deep learning model of small-molecule reactivity achieves a sensitivity and specificity of 18.5% and 95.5%, respectively, in identifying promiscuous bioactive compounds. This performance is similar to PAINS filters, which achieve a sensitivity of 20.3% at the same specificity. Importantly, such reactivity modeling is complementary to PAINS filters. When PAINS filters and reactivity models are combined, the resulting model outperforms either method alone, achieving a sensitivity of 24% at the same specificity. However, as a probabilistic model, the sensitivity and specificity of the deep learning model can be tuned by adjusting the threshold. Moreover, for a subset of PAINS filters, this reactivity model can help discriminate between promiscuous and nonpromiscuous bioactive compounds even among compounds matching those filters. Critically, the reactivity model provides mechanistic hypotheses for assay interference by predicting the precise atoms involved in compound reactivity. Overall, our analysis suggests that deep learning approaches to modeling promiscuous compound bioactivity may provide a complementary approach to current methods for identifying promiscuous compounds.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Simulación por Computador , Bases de Datos Factuales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Modelos Biológicos , Redes Neurales de la Computación
18.
Bioinformatics ; 32(20): 3183-3189, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27324196

RESUMEN

MOTIVATION: Uridine diphosphate glucunosyltransferases (UGTs) metabolize 15% of FDA approved drugs. Lead optimization efforts benefit from knowing how candidate drugs are metabolized by UGTs. This paper describes a computational method for predicting sites of UGT-mediated metabolism on drug-like molecules. RESULTS: XenoSite correctly predicts test molecule's sites of glucoronidation in the Top-1 or Top-2 predictions at a rate of 86 and 97%, respectively. In addition to predicting common sites of UGT conjugation, like hydroxyl groups, it can also accurately predict the glucoronidation of atypical sites, such as carbons. We also describe a simple heuristic model for predicting UGT-mediated sites of metabolism that performs nearly as well (with, respectively, 80 and 91% Top-1 and Top-2 accuracy), and can identify the most challenging molecules to predict on which to assess more complex models. Compared with prior studies, this model is more generally applicable, more accurate and simpler (not requiring expensive quantum modeling). AVAILABILITY AND IMPLEMENTATION: The UGT metabolism predictor developed in this study is available at http://swami.wustl.edu/xenosite/p/ugt CONTACT: : swamidass@wustl.eduSupplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Preparaciones Farmacéuticas/metabolismo , Interacciones Farmacológicas , Humanos
19.
Chem Res Toxicol ; 30(2): 642-656, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28099803

RESUMEN

Many adverse drug reactions are thought to be caused by electrophilically reactive drug metabolites that conjugate to nucleophilic sites within DNA and proteins, causing cancer or toxic immune responses. Quinone species, including quinone-imines, quinone-methides, and imine-methides, are electrophilic Michael acceptors that are often highly reactive and comprise over 40% of all known reactive metabolites. Quinone metabolites are created by cytochromes P450 and peroxidases. For example, cytochromes P450 oxidize acetaminophen to N-acetyl-p-benzoquinone imine, which is electrophilically reactive and covalently binds to nucleophilic sites within proteins. This reactive quinone metabolite elicits a toxic immune response when acetaminophen exceeds a safe dose. Using a deep learning approach, this study reports the first published method for predicting quinone formation: the formation of a quinone species by metabolic oxidation. We model both one- and two-step quinone formation, enabling accurate quinone formation predictions in nonobvious cases. We predict atom pairs that form quinones with an AUC accuracy of 97.6%, and we identify molecules that form quinones with 88.2% AUC. By modeling the formation of quinones, one of the most common types of reactive metabolites, our method provides a rapid screening tool for a key drug toxicity risk. The XenoSite quinone formation model is available at http://swami.wustl.edu/xenosite/p/quinone .


Asunto(s)
Quinonas/metabolismo , Área Bajo la Curva , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Oxidación-Reducción , Teoría Cuántica , Quinonas/química
20.
Chem Res Toxicol ; 30(4): 1046-1059, 2017 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-28256829

RESUMEN

Structural alerts are commonly used in drug discovery to identify molecules likely to form reactive metabolites and thereby become toxic. Unfortunately, as useful as structural alerts are, they do not effectively model if, when, and why metabolism renders safe molecules toxic. Toxicity due to a specific structural alert is highly conditional, depending on the metabolism of the alert, the reactivity of its metabolites, dosage, and competing detoxification pathways. A systems approach, which explicitly models these pathways, could more effectively assess the toxicity risk of drug candidates. In this study, we demonstrated that mathematical models of P450 metabolism can predict the context-specific probability that a structural alert will be bioactivated in a given molecule. This study focuses on the furan, phenol, nitroaromatic, and thiophene alerts. Each of these structural alerts can produce reactive metabolites through certain metabolic pathways but not always. We tested whether our metabolism modeling approach, XenoSite, can predict when a given molecule's alerts will be bioactivated. Specifically, we used models of epoxidation, quinone formation, reduction, and sulfur-oxidation to predict the bioactivation of furan-, phenol-, nitroaromatic-, and thiophene-containing drugs. Our models separated bioactivated and not-bioactivated furan-, phenol-, nitroaromatic-, and thiophene-containing drugs with AUC performances of 100%, 73%, 93%, and 88%, respectively. Metabolism models accurately predict whether alerts are bioactivated and thus serve as a practical approach to improve the interpretability and usefulness of structural alerts. We expect that this same computational approach can be extended to most other structural alerts and later integrated into toxicity risk models. This advance is one necessary step toward our long-term goal of building comprehensive metabolic models of bioactivation and detoxification to guide assessment and design of new therapeutic molecules.


Asunto(s)
Furanos/química , Modelos Químicos , Fenoles/química , Tiofenos/química , Animales , Área Bajo la Curva , Benzoquinonas/química , Benzoquinonas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Furanos/metabolismo , Furanos/toxicidad , Hígado/efectos de los fármacos , Oxidación-Reducción , Fenoles/metabolismo , Fenoles/toxicidad , Curva ROC , Tiofenos/metabolismo , Tiofenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA