Publications Search
Explore how scientists all over the world use DrugBank in their research.
Published on June 24, 2021
READ PUBLICATION →

A Systems Approach to Brain Tumor Treatment.

Authors: Park JH, de Lomana ALG, Marzese DM, Juarez T, Feroze A, Hothi P, Cobbs C, Patel AP, Kesari S, Huang S, Baliga NS

Abstract: Brain tumors are among the most lethal tumors. Glioblastoma, the most frequent primary brain tumor in adults, has a median survival time of approximately 15 months after diagnosis or a five-year survival rate of 10%; the recurrence rate is nearly 90%. Unfortunately, this prognosis has not improved for several decades. The lack of progress in the treatment of brain tumors has been attributed to their high rate of primary therapy resistance. Challenges such as pronounced inter-patient variability, intratumoral heterogeneity, and drug delivery across the blood-brain barrier hinder progress. A comprehensive, multiscale understanding of the disease, from the molecular to the whole tumor level, is needed to address the intratumor heterogeneity resulting from the coexistence of a diversity of neoplastic and non-neoplastic cell types in the tumor tissue. By contrast, inter-patient variability must be addressed by subtyping brain tumors to stratify patients and identify the best-matched drug(s) and therapies for a particular patient or cohort of patients. Accomplishing these diverse tasks will require a new framework, one involving a systems perspective in assessing the immense complexity of brain tumors. This would in turn entail a shift in how clinical medicine interfaces with the rapidly advancing high-throughput (HTP) technologies that have enabled the omics-scale profiling of molecular features of brain tumors from the single-cell to the tissue level. However, several gaps must be closed before such a framework can fulfill the promise of precision and personalized medicine for brain tumors. Ultimately, the goal is to integrate seamlessly multiscale systems analyses of patient tumors and clinical medicine. Accomplishing this goal would facilitate the rational design of therapeutic strategies matched to the characteristics of patients and their tumors. Here, we discuss some of the technologies, methodologies, and computational tools that will facilitate the realization of this vision to practice.
Published on June 24, 2021
READ PUBLICATION →

Molecular screening of glycyrrhizin-based inhibitors against ACE2 host receptor of SARS-CoV-2.

Authors: Ahmad S, Waheed Y, Abro A, Abbasi SW, Ismail S

Abstract: The interaction between SARS-CoV-2 Spike protein and angiotensin-converting enzyme 2 (ACE2) is essential to viral attachment and the subsequent fusion process. Interfering with this event represents an attractive avenue for the development of therapeutics and vaccine development. Here, a hybrid approach of ligand- and structure-based virtual screening techniques were employed to disclose similar analogues of a reported antiviral phytochemical, glycyrrhizin, targeting the blockade of ACE2 interaction with the SARS-CoV-2 Spike. A ligand-based similarity search using a stringent cut-off revealed 40 FDA-approved compounds in DrugBank. These filtered hits were screened against ACE2 using a blind docking approach to determine the natural binding tendency of the compounds with ACE2. Three compounds, deslanoside, digitoxin, and digoxin, were reported to show strong binding with ACE2. These compounds bind at the H1-H2 binding pocket, in a manner similar to that of glycyrrhizin which was used as a control. To achieve consistency in the docking results, docking calculations were performed via two sets of docking software that predicted binding energy as ACE2-Deslanoside (AutoDock, -10.3 kcal/mol and DockThor, -9.53 kcal/mol), ACE2-Digitoxin (AutoDock, -10.6 kcal/mol and DockThor, -8.84 kcal/mol), and ACE2-Digoxin (AutoDock, -10.6 kcal/mol and DockThor, -8.81 kcal/mol). The docking results were validated by running molecular simulations in aqueous solution that demonstrated the stability of ACE2 with no major conformational changes in the ligand original binding mode (~ 2 A average RMSD). Binding interactions remained quite stable with an increased potential for getting stronger as the simulation proceeded. MMGB/PBSA binding free energies were also estimated and these supported the high stability of the complexes compared to the control (~ -50 kcal/mol net MMGB/PBSA binding energy versus ~ -30 kcal/mol). Collectively, the data demonstrated that the compounds shortlisted in this study might be subjected to experimental evaluation to uncover their real blockade capacity of SARS-CoV-2 host ACE2 receptor.
Published on June 24, 2021
READ PUBLICATION →

Drug-Drug Interaction Predictions via Knowledge Graph and Text Embedding: Instrument Validation Study.

Authors: Wang M, Wang H, Liu X, Ma X, Wang B

Abstract: BACKGROUND: Minimizing adverse reactions caused by drug-drug interactions (DDIs) has always been a prominent research topic in clinical pharmacology. Detecting all possible interactions through clinical studies before a drug is released to the market is a demanding task. The power of big data is opening up new approaches to discovering various DDIs. However, these data contain a huge amount of noise and provide knowledge bases that are far from being complete or used with reliability. Most existing studies focus on predicting binary DDIs between drug pairs and ignore other interactions. OBJECTIVE: Leveraging both drug knowledge graphs and biomedical text is a promising pathway for rich and comprehensive DDI prediction, but it is not without issues. Our proposed model seeks to address the following challenges: data noise and incompleteness, data sparsity, and computational complexity. METHODS: We propose a novel framework, Predicting Rich DDI, to predict DDIs. The framework uses graph embedding to overcome data incompleteness and sparsity issues to make multiple DDI label predictions. First, a large-scale drug knowledge graph is generated from different sources. The knowledge graph is then embedded with comprehensive biomedical text into a common low-dimensional space. Finally, the learned embeddings are used to efficiently compute rich DDI information through a link prediction process. RESULTS: To validate the effectiveness of the proposed framework, extensive experiments were conducted on real-world data sets. The results demonstrate that our model outperforms several state-of-the-art baseline methods in terms of capability and accuracy. CONCLUSIONS: We propose a novel framework, Predicting Rich DDI, to predict DDIs. Using rich DDI information, it can competently predict multiple labels for a pair of drugs across numerous domains, ranging from pharmacological mechanisms to side effects. To the best of our knowledge, this framework is the first to provide a joint translation-based embedding model that learns DDIs by integrating drug knowledge graphs and biomedical text simultaneously in a common low-dimensional space. The model also predicts DDIs using multiple labels rather than single or binary labels. Extensive experiments were conducted on real-world data sets to demonstrate the effectiveness and efficiency of the model. The results show our proposed framework outperforms several state-of-the-art baselines.
Published on June 24, 2021
READ PUBLICATION →

Bioactivity descriptors for uncharacterized chemical compounds.

Authors: Bertoni M, Duran-Frigola M, Badia-I-Mompel P, Pauls E, Orozco-Ruiz M, Guitart-Pla O, Alcalde V, Diaz VM, Berenguer-Llergo A, Brun-Heath I, Villegas N, de Herreros AG, Aloy P

Abstract: Chemical descriptors encode the physicochemical and structural properties of small molecules, and they are at the core of chemoinformatics. The broad release of bioactivity data has prompted enriched representations of compounds, reaching beyond chemical structures and capturing their known biological properties. Unfortunately, bioactivity descriptors are not available for most small molecules, which limits their applicability to a few thousand well characterized compounds. Here we present a collection of deep neural networks able to infer bioactivity signatures for any compound of interest, even when little or no experimental information is available for them. Our signaturizers relate to bioactivities of 25 different types (including target profiles, cellular response and clinical outcomes) and can be used as drop-in replacements for chemical descriptors in day-to-day chemoinformatics tasks. Indeed, we illustrate how inferred bioactivity signatures are useful to navigate the chemical space in a biologically relevant manner, unveiling higher-order organization in natural product collections, and to enrich mostly uncharacterized chemical libraries for activity against the drug-orphan target Snail1. Moreover, we implement a battery of signature-activity relationship (SigAR) models and show a substantial improvement in performance, with respect to chemistry-based classifiers, across a series of biophysics and physiology activity prediction benchmarks.
Published on June 24, 2021
READ PUBLICATION →

Repurposing of FDA-approved drugs against active site and potential allosteric drug-binding sites of COVID-19 main protease.

Authors: Yuce M, Cicek E, Inan T, Dag AB, Kurkcuoglu O, Sungur FA

Abstract: The novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) still has serious negative effects on health, social life, and economics. Recently, vaccines from various companies have been urgently approved to control SARS-CoV-2 infections. However, any specific antiviral drug has not been confirmed so far for regular treatment. An important target is the main protease (M(pro) ), which plays a major role in replication of the virus. In this study, Gaussian and residue network models are employed to reveal two distinct potential allosteric sites on M(pro) that can be evaluated as drug targets besides the active site. Then, Food and Drug Administration (FDA)-approved drugs are docked to three distinct sites with flexible docking using AutoDock Vina to identify potential drug candidates. Fourteen best molecule hits for the active site of M(pro) are determined. Six of these also exhibit high docking scores for the potential allosteric regions. Full-atom molecular dynamics simulations with MM-GBSA method indicate that compounds docked to active and potential allosteric sites form stable interactions with high binding free energy (Gbind ) values. Gbind values reach -52.06 kcal/mol for the active site, -51.08 kcal/mol for the potential allosteric site 1, and - 42.93 kcal/mol for the potential allosteric site 2. Energy decomposition calculations per residue elucidate key binding residues stabilizing the ligands that can further serve to design pharmacophores. This systematic and efficient computational analysis successfully determines ivermectine, diosmin, and selinexor currently subjected to clinical trials, and further proposes bromocriptine, elbasvir as M(pro) inhibitor candidates to be evaluated against SARS-CoV-2 infections.
Published on June 24, 2021
READ PUBLICATION →

Ligands of Adrenergic Receptors: A Structural Point of View.

Authors: Wu Y, Zeng L, Zhao S

Abstract: Adrenergic receptors are G protein-coupled receptors for epinephrine and norepinephrine. They are targets of many drugs for various conditions, including treatment of hypertension, hypotension, and asthma. Adrenergic receptors are intensively studied in structural biology, displayed for binding poses of different types of ligands. Here, we summarized molecular mechanisms of ligand recognition and receptor activation exhibited by structure. We also reviewed recent advances in structure-based ligand discovery against adrenergic receptors.
Published on June 23, 2021
READ PUBLICATION →

Evolving scenario of big data and Artificial Intelligence (AI) in drug discovery.

Authors: Tripathi MK, Nath A, Singh TP, Ethayathulla AS, Kaur P

Abstract: The accumulation of massive data in the plethora of Cheminformatics databases has made the role of big data and artificial intelligence (AI) indispensable in drug design. This has necessitated the development of newer algorithms and architectures to mine these databases and fulfil the specific needs of various drug discovery processes such as virtual drug screening, de novo molecule design and discovery in this big data era. The development of deep learning neural networks and their variants with the corresponding increase in chemical data has resulted in a paradigm shift in information mining pertaining to the chemical space. The present review summarizes the role of big data and AI techniques currently being implemented to satisfy the ever-increasing research demands in drug discovery pipelines.
Published on June 23, 2021
READ PUBLICATION →

DNA-Encoded Chemistry: Drug Discovery from a Few Good Reactions.

Authors: Fitzgerald PR, Paegel BM

Abstract: Click chemistry, proposed nearly 20 years ago, promised access to novel chemical space by empowering combinatorial library synthesis with a "few good reactions". These click reactions fulfilled key criteria (broad scope, quantitative yield, abundant starting material, mild reaction conditions, and high chemoselectivity), keeping the focus on molecules that would be easy to make, yet structurally diverse. This philosophy bears a striking resemblance to DNA-encoded library (DEL) technology, the now-dominant combinatorial chemistry paradigm. This review highlights the similarities between click and DEL reaction design and deployment in combinatorial library settings, providing a framework for the design of new DEL synthesis technologies to enable next-generation drug discovery.
Published on June 23, 2021
READ PUBLICATION →

Integrated network analysis identifying potential novel drug candidates and targets for Parkinson's disease.

Authors: Quan P, Wang K, Yan S, Wen S, Wei C, Zhang X, Cao J, Yao L

Abstract: This study aimed to identify potential novel drug candidates and targets for Parkinson's disease. First, 970 genes that have been reported to be related to PD were collected from five databases, and functional enrichment analysis of these genes was conducted to investigate their potential mechanisms. Then, we collected drugs and related targets from DrugBank, narrowed the list by proximity scores and Inverted Gene Set Enrichment analysis of drug targets, and identified potential drug candidates for PD treatment. Finally, we compared the expression distribution of the candidate drug-target genes between the PD group and the control group in the public dataset with the largest sample size (GSE99039) in Gene Expression Omnibus. Ten drugs with an FDR < 0.1 and their corresponding targets were identified. Some target genes of the ten drugs significantly overlapped with PD-related genes or already known therapeutic targets for PD. Nine differentially expressed drug-target genes with p < 0.05 were screened. This work will facilitate further research into the possible efficacy of new drugs for PD and will provide valuable clues for drug design.
Published on June 21, 2021
READ PUBLICATION →

Side effect prediction based on drug-induced gene expression profiles and random forest with iterative feature selection.

Authors: Cakir A, Tuncer M, Taymaz-Nikerel H, Ulucan O

Abstract: One in every ten drug candidates fail in clinical trials mainly due to efficacy and safety related issues, despite in-depth preclinical testing. Even some of the approved drugs such as chemotherapeutics are notorious for their side effects that are burdensome on patients. In order to pave the way for new therapeutics with more tolerable side effects, the mechanisms underlying side effects need to be fully elucidated. In this work, we addressed the common side effects of chemotherapeutics, namely alopecia, diarrhea and edema. A strategy based on Random Forest algorithm unveiled an expression signature involving 40 genes that predicted these side effects with an accuracy of 89%. We further characterized the resulting signature and its association with the side effects using functional enrichment analysis and protein-protein interaction networks. This work contributes to the ongoing efforts in drug development for early identification of side effects to use the resources more effectively.