Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Chem Res Toxicol ; 34(2): 365-384, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33351593

RESUMEN

Adverse drug reactions (ADRs) are undesired effects of medicines that can harm patients and are a significant source of attrition in drug development. ADRs are anticipated by routinely screening drugs against secondary pharmacology protein panels. However, there is still a lack of quantitative information on the links between these off-target proteins and the reporting of ADRs in humans. Here, we present a systematic analysis of associations between measured and predicted in vitro bioactivities of drugs and adverse events (AEs) in humans from two sources of data: the Side Effect Resource, derived from clinical trials, and the Food and Drug Administration Adverse Event Reporting System, derived from postmarketing surveillance. The ratio of a drug's therapeutic unbound plasma concentration over the drug's in vitro potency against a given protein was used to select proteins most likely to be relevant to in vivo effects. In examining individual target bioactivities as predictors of AEs, we found a trade-off between the positive predictive value and the fraction of drugs with AEs that can be detected. However, considering sets of multiple targets for the same AE can help identify a greater fraction of AE-associated drugs. Of the 45 targets with statistically significant associations to AEs, 30 are included on existing safety target panels. The remaining 15 targets include 9 carbonic anhydrases, of which CA5B is significantly associated with cholestatic jaundice. We include the full quantitative data on associations between measured and predicted in vitro bioactivities and AEs in humans in this work, which can be used to make a more informed selection of safety profiling targets.


Asunto(s)
Preparaciones Farmacéuticas/química , Proteínas/análisis , Ensayos Clínicos como Asunto , Humanos , Estructura Molecular , Preparaciones Farmacéuticas/sangre , Proteínas/antagonistas & inhibidores , Estados Unidos , United States Food and Drug Administration
2.
J Cheminform ; 11(1): 36, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31152262

RESUMEN

Despite the increasing knowledge in both the chemical and biological domains the assimilation and exploration of heterogeneous datasets, encoding information about the chemical, bioactivity and phenotypic properties of compounds, remains a challenge due to requirement for overlap between chemicals assayed across the spaces. Here, we have constructed a novel dataset, larger than we have used in prior work, comprising 579 acute oral toxic compounds and 1427 non-toxic compounds derived from regulatory GHS information, along with their corresponding molecular and protein target descriptors and qHTS in vitro assay readouts from the Tox21 project. We found no clear association between the results of a FAFDrugs4 toxicophore screen and the acute oral toxicity classifications for our compound set; and a screen using a subset of the ToxAlerts toxicophores was also of limited utility, with only slight enrichment toward the toxic set (odds ratio of 1.48). We then investigated to what degree toxic and non-toxic compounds could be separated in each of the spaces, to compare their potential contribution to further analyses. Using an LDA projection, we found the largest degree of separation using chemical descriptors (Cohen's d of 1.95) and the lowest degree of separation between toxicity classes using qHTS descriptors (Cohen's d of 0.67). To compare the predictivity of the feature spaces for the toxicity endpoint, we next trained Random Forest (RF) acute oral toxicity classifiers on either molecular, protein target and qHTS descriptors. RFs trained on molecular and protein target descriptors were most predictive, with ROC AUC values of 0.80-0.92 and 0.70-0.85, respectively, across three test sets. RFs trained on both chemical and protein target descriptors combined exhibited similar predictive performance to the single-domain models (ROC AUC of 0.80-0.91). Model interpretability was improved by the inclusion of protein target descriptors, which allow the identification of specific targets (e.g. Retinal dehydrogenase) with literature links to toxic modes of action (e.g. oxidative stress). The dataset compiled in this study has been made available for future application.

3.
Toxicol Res (Camb) ; 5(3): 883-894, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30090397

RESUMEN

Prediction of compound toxicity is essential because covering the vast chemical space requiring safety assessment using traditional experimentally-based, resource-intensive techniques is impossible. However, such prediction is nontrivial due to the complex causal relationship between compound structure and in vivo harm. Protein target annotations and in vitro experimental outcomes encode relevant bioactivity information complementary to chemicals' structures. This work tests the hypothesis that utilizing three complementary types of data will afford predictive models that outperform traditional models built using fewer data types. A tripartite, heterogeneous descriptor set for 367 compounds was comprised of (a) chemical descriptors, (b) protein target descriptors generated using an algorithm trained on 190 000 ligand-protein interactions from ChEMBL, and (c) descriptors derived from in vitro cell cytotoxicity dose-response data from a panel of human cell lines. 100 random forests classification models for predicting rat LD50 were built using every combination of descriptors. Successive integration of data types improved predictive performance; models built using the full dataset had an average external correct classification rate of 0.82, compared to 0.73-0.80 for models built using two data types and 0.67-0.78 for models built using one. Pairwise comparisons of models trained on the same data showed that including a third data domain on top of chemistry improved average correct classification rate by 1.4-2.4 points, with p-values <0.01. Additionally, the approach enhanced the models' applicability domains and proved useful for generating novel mechanism hypotheses. The use of tripartite heterogeneous bioactivity datasets is a useful technique for improving toxicity prediction. Both protein target descriptors - which have the practical value of being derived in silico - and cytotoxicity descriptors derived from experiment are suitable contributors to such datasets.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA