Publications Search
Explore how scientists all over the world use DrugBank in their research.
Published on January 26, 2022
READ PUBLICATION →

wSDTNBI: a novel network-based inference method for virtual screening.

Authors: Wu Z, Ma H, Liu Z, Zheng L, Yu Z, Cao S, Fang W, Wu L, Li W, Liu G, Huang J, Tang Y

Abstract: In recent years, the rapid development of network-based methods for the prediction of drug-target interactions (DTIs) provides an opportunity for the emergence of a new type of virtual screening (VS), namely, network-based VS. Herein, we reported a novel network-based inference method named wSDTNBI. Compared with previous network-based methods that use unweighted DTI networks, wSDTNBI uses weighted DTI networks whose edge weights are correlated with binding affinities. A two-pronged approach based on weighted DTI and drug-substructure association networks was employed to calculate prediction scores. To show the practical value of wSDTNBI, we performed network-based VS on retinoid-related orphan receptor gammat (RORgammat), and purchased 72 compounds for experimental validation. Seven of the purchased compounds were confirmed to be novel RORgammat inverse agonists by in vitro experiments, including ursonic acid and oleanonic acid with IC50 values of 10 nM and 0.28 muM, respectively. Moreover, the direct contact between ursonic acid and RORgammat was confirmed using the X-ray crystal structure, and in vivo experiments demonstrated that ursonic acid and oleanonic acid have therapeutic effects on multiple sclerosis. These results indicate that wSDTNBI might be a powerful tool for network-based VS in drug discovery.
Published on January 26, 2022
READ PUBLICATION →

A deep learning approach to predict inter-omics interactions in multi-layer networks.

Authors: Borhani N, Ghaisari J, Abedi M, Kamali M, Gheisari Y

Abstract: BACKGROUND: Despite enormous achievements in the production of high-throughput datasets, constructing comprehensive maps of interactions remains a major challenge. Lack of sufficient experimental evidence on interactions is more significant for heterogeneous molecular types. Hence, developing strategies to predict inter-omics connections is essential to construct holistic maps of disease. RESULTS: Here, as a novel nonlinear deep learning method, Data Integration with Deep Learning (DIDL) was proposed to predict inter-omics interactions. It consisted of an encoder that performs automatic feature extraction for biomolecules according to existing interactions coupled with a predictor that predicts unforeseen interactions. Applicability of DIDL was assessed on different networks, namely drug-target protein, transcription factor-DNA element, and miRNA-mRNA. Also, validity of the novel predictions was evaluated by literature surveys. According to the results, the DIDL outperformed state-of-the-art methods. For all three networks, the areas under the curve and the precision-recall curve exceeded 0.85 and 0.83, respectively. CONCLUSIONS: DIDL offers several advantages like automatic feature extraction from raw data, end-to-end training, and robustness to network sparsity. In addition, reliance solely on existing inter-layer interactions and independence of biochemical features of interacting molecules make this algorithm applicable for a wide variety of networks. DIDL paves the way to understand the underlying mechanisms of complex disorders through constructing integrative networks.
Published on January 26, 2022
READ PUBLICATION →

Network controllability solutions for computational drug repurposing using genetic algorithms.

Authors: Popescu VB, Kanhaiya K, Nastac DI, Czeizler E, Petre I

Abstract: Control theory has seen recently impactful applications in network science, especially in connections with applications in network medicine. A key topic of research is that of finding minimal external interventions that offer control over the dynamics of a given network, a problem known as network controllability. We propose in this article a new solution for this problem based on genetic algorithms. We tailor our solution for applications in computational drug repurposing, seeking to maximize its use of FDA-approved drug targets in a given disease-specific protein-protein interaction network. We demonstrate our algorithm on several cancer networks and on several random networks with their edges distributed according to the Erdos-Renyi, the Scale-Free, and the Small World properties. Overall, we show that our new algorithm is more efficient in identifying relevant drug targets in a disease network, advancing the computational solutions needed for new therapeutic and drug repurposing approaches.
Published on January 25, 2022
READ PUBLICATION →

Bioinformatics screening the novel and promising targets of curcumin in hepatocellular carcinoma chemotherapy and prognosis.

Authors: Yang T, Chen Y, Xu J, Li J, Liu H, Liu N

Abstract: BACKGROUND: The aim of present study was to screen the novel and promising targets of curcumin in hepatocellular carcinoma diagnosis and chemotherapy. METHODS: Potential targets of curcumin were screened from SwissTargetPrediction, ParmMapper and drugbank databases. Potential aberrant genes of hepatocellular carcinoma were screened from Genecards databases. Fifty paired hepatocellular carcinoma patients' gene expression profiles from the GEO database were used to test potential targets of curcumin. Besides, GO analysis, KEGG pathway enrichment analysis and PPI network construction were used to explore the underlying mechanism of candidate hub genes. ROC analysis and Kaplan-Meier analysis were used to evaluate the diagnostic and prognostic value of candidate hub genes, respectively. Real-time PCR was used to verify the results of bioinformatics analysis. RESULTS: Bioinformatics analysis results suggested that AURKA, CDK1, CCNB1, TOP2A, CYP2B6, CYP2C9, and CYP3A4 genes served as candidate hub genes. AURKA, CDK1, CCNB1 and TOP2A were significantly upregulated and correlated with poor prognosis in hepatocellular carcinoma, AUC values of which were 95.7, 96.9, 98.1 and 96.1% respectively. There was not significant correlation between the expression of CYP2B6 and prognosis of hepatocellular carcinoma, while CYP2C9 and CYP3A4 genes were significantly downregulated and correlated with poor prognosis in hepatocellular carcinoma. AUC values of CYP2B6, CYP2C9, and CYP3A4 were 96.0, 97.0 and 88.0% respectively. In vitro, we further confirmed that curcumin significantly downregulated the expression of AURKA, CDK1, and TOP2A genes, while significantly upregulated the expression of CYP2B6, CYP2C9, and CYP3A4 genes. CONCLUSIONS: Our results provided a novel panel of AURKA, CDK1, TOP2A, CYP2C9, and CYP3A4 candidate genes for curcumin related chemotherapy of hepatocellular carcinoma.
Published on January 24, 2022
READ PUBLICATION →

DOTA: Deep Learning Optimal Transport Approach to Advance Drug Repositioning for Alzheimer's Disease.

Authors: Chyr J, Gong H, Zhou X

Abstract: Alzheimer's disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States and incurring a substantial global healthcare cost. Unfortunately, current treatments are only palliative and do not cure AD. There is an urgent need to develop novel anti-AD therapies; however, drug discovery is a time-consuming, expensive, and high-risk process. Drug repositioning, on the other hand, is an attractive approach to identify drugs for AD treatment. Thus, we developed a novel deep learning method called DOTA (Drug repositioning approach using Optimal Transport for Alzheimer's disease) to repurpose effective FDA-approved drugs for AD. Specifically, DOTA consists of two major autoencoders: (1) a multi-modal autoencoder to integrate heterogeneous drug information and (2) a Wasserstein variational autoencoder to identify effective AD drugs. Using our approach, we predict that antipsychotic drugs with circadian effects, such as quetiapine, aripiprazole, risperidone, suvorexant, brexpiprazole, olanzapine, and trazadone, will have efficacious effects in AD patients. These drugs target important brain receptors involved in memory, learning, and cognition, including serotonin 5-HT2A, dopamine D2, and orexin receptors. In summary, DOTA repositions promising drugs that target important biological pathways and are predicted to improve patient cognition, circadian rhythms, and AD pathogenesis.
Published on January 24, 2022
READ PUBLICATION →

Detection of polypharmacy side effects by integrating multiple data sources and convolutional neural networks.

Authors: Lakizadeh A, Babaei M

Abstract: The consumption of drug combinations, named polypharmacy, is commonly used for treating patients with several diseases or those with complex conditions. However, the main drawback of polypharmacy is the increased probability of harmful side effects. The polypharmacy side effects are caused by an interaction between two medications. It means that the drug-drug interaction causes changes in their activities due to interfering in each other's performance. Therefore, discovering these side effects is one of the most challenging and important aspects of drug production and consumption as it is associated with human health. In this paper, a method has been introduced for predicting the polypharmacy side effects, called PSECNN. It is a multi-label multi-class deep learning method that combines various basic features of drugs to predict the polypharmacy side effects. Firstly, PSECNN collects five basic features of drugs, such as individual drug's side effects, drug-protein interactions, chemical substructures, targets, and enzymes in order to create a novel combination of drug features. A feature extraction module creates five feature vectors with the same dimension for each drug based on the Jaccard similarity index. Based on the feature vectors, a unique representative is then created for each drug. These representative vectors are given in pairs as input to the deep neural network to predict the occurrence probability of side effects. According to the experimental evaluations, PSECNN could outperform the state-of-the-art polypharmacy side effects prediction methods up to 74%. It has been found that PSECNN has better performance with polypharmacy side effects with a cause of molecular basis due to the novel combination of basic drug features.
Published on January 24, 2022
READ PUBLICATION →

Unravelling Mechanisms of Doxorubicin-Induced Toxicity in 3D Human Intestinal Organoids.

Authors: Rodrigues D, Coyle L, Fuzi B, Ferreira S, Jo H, Herpers B, Chung SW, Fisher C, Kleinjans JCS, Jennen D, de Kok TM

Abstract: Doxorubicin is widely used in the treatment of different cancers, and its side effects can be severe in many tissues, including the intestines. Symptoms such as diarrhoea and abdominal pain caused by intestinal inflammation lead to the interruption of chemotherapy. Nevertheless, the molecular mechanisms associated with doxorubicin intestinal toxicity have been poorly explored. This study aims to investigate such mechanisms by exposing 3D small intestine and colon organoids to doxorubicin and to evaluate transcriptomic responses in relation to viability and apoptosis as physiological endpoints. The in vitro concentrations and dosing regimens of doxorubicin were selected based on physiologically based pharmacokinetic model simulations of treatment regimens recommended for cancer patients. Cytotoxicity and cell morphology were evaluated as well as gene expression and biological pathways affected by doxorubicin. In both types of organoids, cell cycle, the p53 signalling pathway, and oxidative stress were the most affected pathways. However, significant differences between colon and SI organoids were evident, particularly in essential metabolic pathways. Short time-series expression miner was used to further explore temporal changes in gene profiles, which identified distinct tissue responses. Finally, in silico proteomics revealed important proteins involved in doxorubicin metabolism and cellular processes that were in line with the transcriptomic responses, including cell cycle and senescence, transport of molecules, and mitochondria impairment. This study provides new insight into doxorubicin-induced effects on the gene expression levels in the intestines. Currently, we are exploring the potential use of these data in establishing quantitative systems toxicology models for the prediction of drug-induced gastrointestinal toxicity.
Published on January 20, 2022
READ PUBLICATION →

A novel graph mining approach to predict and evaluate food-drug interactions.

Authors: Rahman MM, Vadrev SM, Magana-Mora A, Levman J, Soufan O

Abstract: Food-drug interactions (FDIs) arise when nutritional dietary consumption regulates biochemical mechanisms involved in drug metabolism. This study proposes FDMine, a novel systematic framework that models the FDI problem as a homogenous graph. Our dataset consists of 788 unique approved small molecule drugs with metabolism-related drug-drug interactions and 320 unique food items, composed of 563 unique compounds. The potential number of interactions is 87,192 and 92,143 for disjoint and joint versions of the graph. We defined several similarity subnetworks comprising food-drug similarity, drug-drug similarity, and food-food similarity networks. A unique part of the graph involves encoding the food composition as a set of nodes and calculating a content contribution score. To predict new FDIs, we considered several link prediction algorithms and various performance metrics, including the precision@top (top 1%, 2%, and 5%) of the newly predicted links. The shortest path-based method has achieved a precision of 84%, 60% and 40% for the top 1%, 2% and 5% of FDIs identified, respectively. We validated the top FDIs predicted using FDMine to demonstrate its applicability, and we relate therapeutic anti-inflammatory effects of food items informed by FDIs. FDMine is publicly available to support clinicians and researchers.
Published on January 20, 2022
READ PUBLICATION →

Drug-target interaction prediction via multiple classification strategies.

Authors: Ye Q, Zhang X, Lin X

Abstract: BACKGROUND: Computational prediction of the interaction between drugs and protein targets is very important for the new drug discovery, as the experimental determination of drug-target interaction (DTI) is expensive and time-consuming. However, different protein targets are with very different numbers of interactions. Specifically, most interactions focus on only a few targets. As a result, targets with larger numbers of interactions could own enough positive samples for predicting their interactions but the positive samples for targets with smaller numbers of interactions could be not enough. Only using a classification strategy may not be able to deal with the above two cases at the same time. To overcome the above problem, in this paper, a drug-target interaction prediction method based on multiple classification strategies (MCSDTI) is proposed. In MCSDTI, targets are firstly divided into two parts according to the number of interactions of the targets, where one part contains targets with smaller numbers of interactions (TWSNI) and another part contains targets with larger numbers of interactions (TWLNI). And then different classification strategies are respectively designed for TWSNI and TWLNI to predict the interaction. Furthermore, TWSNI and TWLNI are evaluated independently, which can overcome the problem that result could be mainly determined by targets with large numbers of interactions when all targets are evaluated together. RESULTS: We propose a new drug-target interaction (MCSDTI) prediction method, which uses multiple classification strategies. MCSDTI is tested on five DTI datasets, such as nuclear receptors (NR), ion channels (IC), G protein coupled receptors (GPCR), enzymes (E), and drug bank (DB). Experiments show that the AUCs of our method are respectively 3.31%, 1.27%, 2.02%, 2.02% and 1.04% higher than that of the second best methods on NR, IC, GPCR and E for TWLNI; And AUCs of our method are respectively 1.00%, 3.20% and 2.70% higher than the second best methods on NR, IC, and E for TWSNI. CONCLUSION: MCSDTI is a competitive method compared to the previous methods for all target parts on most datasets, which administrates that different classification strategies for different target parts is an effective way to improve the effectiveness of DTI prediction.
Published on January 19, 2022
READ PUBLICATION →

In Silico Searching for Alternative Lead Compounds to Treat Type 2 Diabetes through a QSAR and Molecular Dynamics Study.

Authors: Cabrera N, Cuesta SA, Mora JR, Calle L, Marquez EA, Kaunas R, Paz JL

Abstract: Free fatty acid receptor 1 (FFA1) stimulates insulin secretion in pancreatic beta-cells. An advantage of therapies that target FFA1 is their reduced risk of hypoglycemia relative to common type 2 diabetes treatments. In this work, quantitative structure-activity relationship (QSAR) approach was used to construct models to identify possible FFA1 agonists by applying four different machine-learning algorithms. The best model (M2) meets the Tropsha's test requirements and has the statistics parameters R(2) = 0.843, Q(2)CV = 0.785, and Q(2)ext = 0.855. Also, coverage of 100% of the test set based on the applicability domain analysis was obtained. Furthermore, a deep analysis based on the ADME predictions, molecular docking, and molecular dynamics simulations was performed. The lipophilicity and the residue interactions were used as relevant criteria for selecting a candidate from the screening of the DiaNat and DrugBank databases. Finally, the FDA-approved drugs bilastine, bromfenac, and fenofibric acid are suggested as potential and lead FFA1 agonists.