Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cheminform ; 11(1): 28, 2019 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-30963300

RESUMO

BACKGROUND: Because drug-drug interactions (DDIs) may cause adverse drug reactions or contribute to complex-disease treatments, it is important to identify DDIs before multiple-drug medications are prescribed. As the alternative of high-cost experimental identifications, computational approaches provide a much cheaper screening for potential DDIs on a large scale manner. Nevertheless, most of them only predict whether or not one drug interacts with another, but neglect their enhancive (positive) and depressive (negative) changes of pharmacological effects. Moreover, these comprehensive DDIs do not occur at random, but exhibit a weakly balanced relationship (a structural property when considering the DDI network), which would help understand how high-order DDIs work. RESULTS: This work exploits the intrinsically structural relationship to solve two tasks, including drug community detection as well as comprehensive DDI prediction in the cold-start scenario. Accordingly, we first design a balance regularized semi-nonnegative matrix factorization (BRSNMF) to partition the drugs into communities. Then, to predict enhancive and degressive DDIs in the cold-start scenario, we develop a BRSNMF-based predictive approach, which technically leverages drug-binding proteins (DBP) as features to associate new drugs (having no known DDI) with other drugs (having known DDIs). Our experiments demonstrate that BRSNMF can generate the drug communities, which exhibit more reasonable sizes, the property of weak balance as well as pharmacological significances. Moreover, they demonstrate the superiority of DBP features and the inspiring ability of the BRSNMF-based predictive approach on comprehensive DDI prediction with 94% accuracy among top-50 predicted enhancive and 86% accuracy among bottom-50 predicted degressive DDIs. CONCLUSIONS: Owing to the regularization of the weak balance property of the comprehensive DDI network into semi-nonnegative matrix factorization, our proposed BRSNMF is able to not only generate better drug communities but also provide an inspiring comprehensive DDI prediction in the cold-start scenario.

2.
Comput Methods Programs Biomed ; 168: 1-10, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30527128

RESUMO

BACKGROUND AND OBJECTIVE: Due to the synergistic effects of drugs, drug combination is one of the effective approaches for treating complex diseases. However, the identification of drug combinations by dose-response methods is still costly. It is promising to develop supervised learning-based approaches to predict potential drug combinations on a large scale. Nevertheless, these approaches have the inadequate utilization of heterogeneous features, which causes the loss of information useful to classification. Moreover, they have an intrinsic bias, because they assume unknown drug pairs as non-combinations, of which some could be real drug combinations in practice. METHODS: To address above issues, this work first designs a two-layer multiple classifier system (TLMCS) to effectively integrate heterogeneous features involving anatomical therapeutic chemical codes of drugs, drug-drug interactions, drug-target interactions, gene ontology of drug targets, and side effects. To avoid the bias caused by labelling unknown samples as negative, it then utilizes the one-class support vector machines, (which requires no negative instance and only labels approved drug combinations as positive instances), as the member classifiers in TLMCS. Last, both a 10-fold cross validation (10-CV) and a novel prediction are performed to validate the performance of TLMCS. RESULTS: The comparison with three state-of-the-art approaches under 10-CV exhibits the superiority of TLMCS, which achieves the area under the receiver operating characteristic curve = 0.824 and the area under the precision-recall curve = 0.372. Moreover, the experiment under the novel prediction demonstrates its ability, where 9 out of the top-20 predicted combinative drug pairs are validated by checking the published literature. Furthermore, for each of the newly-validated drug combinations, this work analyses the combining mode of the member drugs and investigates their relationship in terms of drug targeting pathways. CONCLUSIONS: The proposed TLMCS provides an effective framework to integrate those heterogeneous features and is trained by only positive samples such that the bias of taking unknown drug pairs as negative samples can be avoided. Furthermore, its results in the novel prediction reveal five types of drug combinations and three types of drug relationships in terms of pathways.


Assuntos
Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos/métodos , Interações Medicamentosas , Preparações Farmacêuticas/química , Preparações Farmacêuticas/classificação , Farmácia/instrumentação , Algoritmos , Biologia Computacional , Simulação por Computador , Bases de Dados Factuais , Humanos , Farmácia/métodos , Curva ROC , Software
3.
BMC Syst Biol ; 12(Suppl 1): 14, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29671393

RESUMO

BACKGROUND: Drug-drug interactions (DDIs) always cause unexpected and even adverse drug reactions. It is important to identify DDIs before drugs are used in the market. However, preclinical identification of DDIs requires much money and time. Computational approaches have exhibited their abilities to predict potential DDIs on a large scale by utilizing pre-market drug properties (e.g. chemical structure). Nevertheless, none of them can predict two comprehensive types of DDIs, including enhancive and degressive DDIs, which increases and decreases the behaviors of the interacting drugs respectively. There is a lack of systematic analysis on the structural relationship among known DDIs. Revealing such a relationship is very important, because it is able to help understand how DDIs occur. Both the prediction of comprehensive DDIs and the discovery of structural relationship among them play an important guidance when making a co-prescription. RESULTS: In this work, treating a set of comprehensive DDIs as a signed network, we design a novel model (DDINMF) for the prediction of enhancive and degressive DDIs based on semi-nonnegative matrix factorization. Inspiringly, DDINMF achieves the conventional DDI prediction (AUROC = 0.872 and AUPR = 0.605) and the comprehensive DDI prediction (AUROC = 0.796 and AUPR = 0.579). Compared with two state-of-the-art approaches, DDINMF shows it superiority. Finally, representing DDIs as a binary network and a signed network respectively, an analysis based on NMF reveals crucial knowledge hidden among DDIs. CONCLUSIONS: Our approach is able to predict not only conventional binary DDIs but also comprehensive DDIs. More importantly, it reveals several key points about the DDI network: (1) both binary and signed networks show fairly clear clusters, in which both drug degree and the difference between positive degree and negative degree show significant distribution; (2) the drugs having large degrees tend to have a larger difference between positive degree and negative degree; (3) though the binary DDI network contains no information about enhancive and degressive DDIs at all, it implies some of their relationship in the comprehensive DDI matrix; (4) the occurrence of signs indicating enhancive and degressive DDIs is not random because the comprehensive DDI network is equipped with a structural balance.


Assuntos
Biologia Computacional/métodos , Interações Medicamentosas , Algoritmos
4.
BMC Syst Biol ; 12(Suppl 9): 136, 2018 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-30598094

RESUMO

BACKGROUND: During the identification of potential candidates, computational prediction of drug-target interactions (DTIs) is important to subsequent expensive validation in wet-lab. DTI screening considers four scenarios, depending on whether the drug is an existing or a new drug and whether the target is an existing or a new target. However, existing approaches have the following limitations. First, only a few of them can address the most difficult scenario (i.e., predicting interactions between new drugs and new targets). More importantly, none of the existing approaches could provide the explicit information for understanding the mechanism of forming interactions, such as the drug-target feature pairs contributing to the interactions. RESULTS: In this paper, we propose a Triple Matrix Factorization-based model (TMF) to tackle these problems. Compared with former state-of-the-art predictive methods, TMF demonstrates its significant superiority by assessing the predictions on four benchmark datasets over four kinds of screening scenarios. Also, it exhibits its outperformance by validating predicted novel interactions. More importantly, by using PubChem fingerprints of chemical structures as drug features and occurring frequencies of amino acid trimer as protein features, TMF shows its ability to find out the features determining interactions, including dominant feature pairs, frequently occurring substructures, and conserved triplet of amino acids. CONCLUSIONS: Our TMF provides a unified framework of DTI prediction for all the screening scenarios. It also presents a new insight for the underlying mechanism of DTIs by indicating dominant features, which play important roles in the forming of DTI.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas , Benchmarking
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...