Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Brief Bioinform ; 23(3)2022 05 13.
Article in English | MEDLINE | ID: covidwho-1806277

ABSTRACT

Target prediction and virtual screening are two powerful tools of computer-aided drug design. Target identification is of great significance for hit discovery, lead optimization, drug repurposing and elucidation of the mechanism. Virtual screening can improve the hit rate of drug screening to shorten the cycle of drug discovery and development. Therefore, target prediction and virtual screening are of great importance for developing highly effective drugs against COVID-19. Here we present D3AI-CoV, a platform for target prediction and virtual screening for the discovery of anti-COVID-19 drugs. The platform is composed of three newly developed deep learning-based models i.e., MultiDTI, MPNNs-CNN and MPNNs-CNN-R models. To compare the predictive performance of D3AI-CoV with other methods, an external test set, named Test-78, was prepared, which consists of 39 newly published independent active compounds and 39 inactive compounds from DrugBank. For target prediction, the areas under the receiver operating characteristic curves (AUCs) of MultiDTI and MPNNs-CNN models are 0.93 and 0.91, respectively, whereas the AUCs of the other reported approaches range from 0.51 to 0.74. For virtual screening, the hit rate of D3AI-CoV is also better than other methods. D3AI-CoV is available for free as a web application at http://www.d3pharma.com/D3Targets-2019-nCoV/D3AI-CoV/index.php, which can serve as a rapid online tool for predicting potential targets for active compounds and for identifying active molecules against a specific target protein for COVID-19 treatment.


Subject(s)
COVID-19 Drug Treatment , Deep Learning , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Repositioning , Humans , Molecular Docking Simulation , SARS-CoV-2
2.
Methods ; 198: 11-18, 2022 02.
Article in English | MEDLINE | ID: covidwho-1721112

ABSTRACT

Coronavirus Disease-19 (COVID-19) has lead global epidemics with high morbidity and mortality. However, there are currently no proven effective drugs targeting COVID-19. Identifying drug-virus associations can not only provide insights into the understanding of drug-virus interaction mechanism, but also guide and facilitate the screening of compound candidates for antiviral drug discovery. Since conventional experiment methods are time-consuming, laborious and expensive, computational methods to identify potential drug candidates for viruses (e.g., COVID-19) provide an alternative strategy. In this work, we propose a novel framework of Heterogeneous Graph Attention Networks for Drug-Virus Association predictions, named HGATDVA. First, we fully incorporate multiple sources of biomedical data, e.g., drug chemical information, virus genome sequences and viral protein sequences, to construct abundant features for drugs and viruses. Second, we construct two drug-virus heterogeneous graphs. For each graph, we design a self-enhanced graph attention network (SGAT) to explicitly model the dependency between a node and its local neighbors and derive the graph-specific representations for nodes. Third, we further develop a neural network architecture with tri-aggregator to aggregate the graph-specific representations to generate the final node representations. Extensive experiments were conducted on two datasets, i.e., DrugVirus and MDAD, and the results demonstrated that our model outperformed 7 state-of-the-art methods. Case study on SARS-CoV-2 validated the effectiveness of our model in identifying potential drugs for viruses.


Subject(s)
COVID-19 , Pharmaceutical Preparations , Drug Interactions , Humans , Neural Networks, Computer , SARS-CoV-2
3.
Interdiscip Sci ; 14(1): 15-21, 2022 Mar.
Article in English | MEDLINE | ID: covidwho-1641016

ABSTRACT

The coronavirus disease (COVID-19) has led to an rush to repurpose existing drugs, although the underlying evidence base is of variable quality. Drug repurposing is a technique by taking advantage of existing known drugs or drug combinations to be explored in an unexpected medical scenario. Drug repurposing, hence, plays a vital role in accelerating the pre-clinical process of designing novel drugs by saving time and cost compared to the traditional de novo drug discovery processes. Since drug repurposing depends on massive observed data from existing drugs and diseases, the tremendous growth of publicly available large-scale machine learning methods supplies the state-of-the-art application of data science to signaling disease, medicine, therapeutics, and identifying targets with the least error. In this article, we introduce guidelines on strategies and options of utilizing machine learning approaches for accelerating drug repurposing. We discuss how to employ machine learning methods in studying precision medicine, and as an instance, how machine learning approaches can accelerate COVID-19 drug repurposing by developing Chinese traditional medicine therapy. This article provides a strong reasonableness for employing machine learning methods for drug repurposing, including during fighting for COVID-19 pandemic.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , Drug Repositioning/methods , Humans , Machine Learning , Pandemics , SARS-CoV-2
4.
Pathogens ; 10(10)2021 Oct 06.
Article in English | MEDLINE | ID: covidwho-1526854

ABSTRACT

The genome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) encodes 16 non-structural (Nsp) and 4 structural proteins. Among the Nsps, Nsp1 inhibits host gene expression and also evades the immune system. This protein has been proposed as a target for vaccine development and also for drug design. Owing to its important role, the current study aimed to identify mutations in Nsp1 and their effect on protein stability and flexibility. This is the first comprehensive study in which 295,000 complete genomes have been screened for mutations after alignment with the Wuhan-Hu-1 reference genome (Accession NC_045512), using the CoVsurver app. The sequences harbored 933 mutations in the entire coding region of Nsp1. The most frequently occurring mutation in the 180-amino-acid Nsp1 protein was R24C (n = 1122), followed by D75E (n = 890), D48G (n = 881), H110Y (n = 860), and D144A (n = 648). Among the 933 non-synonymous mutations, 529 exhibited a destabilizing effect. Similarly, a gain in flexibility was observed in 542 mutations. The majority of the most frequent mutations were detected in the loop regions. These findings imply that Nsp1 mutations might be useful to exploit SARS-CoV-2's pathogenicity. Genomic sequencing of SARS-CoV-2 on a regular basis will further assist in analyzing variations among the drug targets and to test the diagnostic accuracy. This wide range of mutations and their effect on Nsp1's stability may have some consequences for the host's innate immune response to SARS-CoV-2 infection and also for the vaccines' efficacy. Based on this mutational information, geographically strain-specific drugs, vaccines, and antibody combinations could be a useful strategy against SARS-CoV-2 infection.

5.
IEEE/ACM Trans Comput Biol Bioinform ; 18(4): 1290-1298, 2021.
Article in English | MEDLINE | ID: covidwho-1349906

ABSTRACT

An outbreak of COVID-19 that began in late 2019 was caused by a novel coronavirus(SARS-CoV-2). It has become a global pandemic. As of June 9, 2020, it has infected nearly 7 million people and killed more than 400,000, but there is no specific drug. Therefore, there is an urgent need to find or develop more drugs to suppress the virus. Here, we propose a new nonlinear end-to-end model called LUNAR. It uses graph convolutional neural networks to automatically learn the neighborhood information of complex heterogeneous relational networks and combines the attention mechanism to reflect the importance of the sum of different types of neighborhood information to obtain the representation characteristics of each node. Finally, through the topology reconstruction process, the feature representations of drugs and targets are forcibly extracted to match the observed network as much as possible. Through this reconstruction process, we obtain the strength of the relationship between different nodes and predict drug candidates that may affect the treatment of COVID-19 based on the known targets of COVID-19. These selected candidate drugs can be used as a reference for experimental scientists and accelerate the speed of drug development. LUNAR can well integrate various topological structure information in heterogeneous networks, and skillfully combine attention mechanisms to reflect the importance of neighborhood information of different types of nodes, improving the interpretability of the model. The area under the curve(AUC) of the model is 0.949 and the accurate recall curve (AUPR) is 0.866 using 10-fold cross-validation. These two performance indexes show that the model has superior predictive performance. Besides, some of the drugs screened out by our model have appeared in some clinical studies to further illustrate the effectiveness of the model.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19/virology , Drug Evaluation, Preclinical/methods , Neural Networks, Computer , SARS-CoV-2/drug effects , COVID-19/epidemiology , Computational Biology , Databases, Pharmaceutical/statistics & numerical data , Drug Development/methods , Drug Development/statistics & numerical data , Drug Evaluation, Preclinical/statistics & numerical data , Drug Repositioning/methods , Drug Repositioning/statistics & numerical data , Host Microbial Interactions/drug effects , Humans , Nonlinear Dynamics , Pandemics
7.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: covidwho-1266105

ABSTRACT

Recent studies have demonstrated that the excessive inflammatory response is an important factor of death in coronavirus disease 2019 (COVID-19) patients. In this study, we propose a deep representation on heterogeneous drug networks, termed DeepR2cov, to discover potential agents for treating the excessive inflammatory response in COVID-19 patients. This work explores the multi-hub characteristic of a heterogeneous drug network integrating eight unique networks. Inspired by the multi-hub characteristic, we design 3 billion special meta paths to train a deep representation model for learning low-dimensional vectors that integrate long-range structure dependency and complex semantic relation among network nodes. Based on the representation vectors and transcriptomics data, we predict 22 drugs that bind to tumor necrosis factor-α or interleukin-6, whose therapeutic associations with the inflammation storm in COVID-19 patients, and molecular binding model are further validated via data from PubMed publications, ongoing clinical trials and a docking program. In addition, the results on five biomedical applications suggest that DeepR2cov significantly outperforms five existing representation approaches. In summary, DeepR2cov is a powerful network representation approach and holds the potential to accelerate treatment of the inflammatory responses in COVID-19 patients. The source code and data can be downloaded from https://github.com/pengsl-lab/DeepR2cov.git.


Subject(s)
COVID-19 Drug Treatment , Drug Repositioning , Inflammation/drug therapy , SARS-CoV-2/drug effects , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , COVID-19/complications , COVID-19/genetics , COVID-19/virology , Computational Biology , Deep Learning , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/virology , Neural Networks, Computer , SARS-CoV-2/pathogenicity , Software , Transcriptome/drug effects , Transcriptome/genetics
8.
Interdiscip Sci ; 13(1): 61-72, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1012252

ABSTRACT

Assessing pulmonary lesions using computed tomography (CT) images is of great significance to the severity diagnosis and treatment of coronavirus disease 2019 (COVID-19)-infected patients. Such assessment mainly depends on radiologists' subjective judgment, which is inefficient and presents difficulty for those with low levels of experience, especially in rural areas. This work focuses on developing a radiomics signature to quantitatively analyze whether COVID-19-infected pulmonary lesions are mild (Grade I) or moderate/severe (Grade II). We retrospectively analyzed 1160 COVID-19-infected pulmonary lesions from 16 hospitals. First, texture features were extracted from the pulmonary lesion regions of CT images. Then, feature preselection was performed and a radiomics signature was built using a stepwise logistic regression. The stepwise logistic regression also calculated the correlation between the radiomics signature and the grade of a pulmonary lesion. Finally, a logistic regression model was trained to classify the grades of pulmonary lesions. Given a significance level of α = 0.001, the stepwise logistic regression achieved an R (multiple correlation coefficient) of 0.70, which is much larger than Rα = 0.18 (the critical value of R). In the classification, the logistic regression model achieved an AUC of 0.87 on an independent test set. Overall, the radiomics signature is significantly correlated with the grade of a pulmonary lesion in COVID-19 infection. The classification model is interpretable and can assist radiologists in quickly and efficiently diagnosing pulmonary lesions. This work aims to develop a CT-based radiomics signature to quantitatively analyze whether COVID-19-infected pulmonary lesions are mild (Grade I) or moderate/severe (Grade II). The logistic regression model established based on this radiomics signature can assist radiologists to quickly and efficiently diagnose the grades of pulmonary lesions. The model calculates a radiomics score for a lesion and is interpretable and appropriate for clinical use.


Subject(s)
COVID-19/diagnostic imaging , COVID-19/pathology , Lung/diagnostic imaging , Lung/virology , Adult , Aged , Algorithms , Area Under Curve , Calibration , Female , Humans , Logistic Models , Lung/pathology , Male , Middle Aged , ROC Curve , SARS-CoV-2/physiology , Tomography, X-Ray Computed , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL