Publications Search
Explore how scientists all over the world use DrugBank in their research.
Published in 2011
READ PUBLICATION →

Understanding and classifying metabolite space and metabolite-likeness.

Authors: Peironcely JE, Reijmers T, Coulier L, Bender A, Hankemeier T

Abstract: While the entirety of 'Chemical Space' is huge (and assumed to contain between 10(63) and 10(200) 'small molecules'), distinct subsets of this space can nonetheless be defined according to certain structural parameters. An example of such a subspace is the chemical space spanned by endogenous metabolites, defined as 'naturally occurring' products of an organisms' metabolism. In order to understand this part of chemical space in more detail, we analyzed the chemical space populated by human metabolites in two ways. Firstly, in order to understand metabolite space better, we performed Principal Component Analysis (PCA), hierarchical clustering and scaffold analysis of metabolites and non-metabolites in order to analyze which chemical features are characteristic for both classes of compounds. Here we found that heteroatom (both oxygen and nitrogen) content, as well as the presence of particular ring systems was able to distinguish both groups of compounds. Secondly, we established which molecular descriptors and classifiers are capable of distinguishing metabolites from non-metabolites, by assigning a 'metabolite-likeness' score. It was found that the combination of MDL Public Keys and Random Forest exhibited best overall classification performance with an AUC value of 99.13%, a specificity of 99.84% and a selectivity of 88.79%. This performance is slightly better than previous classifiers; and interestingly we found that drugs occupy two distinct areas of metabolite-likeness, the one being more 'synthetic' and the other being more 'metabolite-like'. Also, on a truly prospective dataset of 457 compounds, 95.84% correct classification was achieved. Overall, we are confident that we contributed to the tasks of classifying metabolites, as well as to understanding metabolite chemical space better. This knowledge can now be used in the development of new drugs that need to resemble metabolites, and in our work particularly for assessing the metabolite-likeness of candidate molecules during metabolite identification in the metabolomics field.
Published in 2011
READ PUBLICATION →

Modeling of human prokineticin receptors: interactions with novel small-molecule binders and potential off-target drugs.

Authors: Levit A, Yarnitzky T, Wiener A, Meidan R, Niv MY

Abstract: BACKGROUND AND MOTIVATION: The Prokineticin receptor (PKR) 1 and 2 subtypes are novel members of family A GPCRs, which exhibit an unusually high degree of sequence similarity. Prokineticins (PKs), their cognate ligands, are small secreted proteins of approximately 80 amino acids; however, non-peptidic low-molecular weight antagonists have also been identified. PKs and their receptors play important roles under various physiological conditions such as maintaining circadian rhythm and pain perception, as well as regulating angiogenesis and modulating immunity. Identifying binding sites for known antagonists and for additional potential binders will facilitate studying and regulating these novel receptors. Blocking PKRs may serve as a therapeutic tool for various diseases, including acute pain, inflammation and cancer. METHODS AND RESULTS: Ligand-based pharmacophore models were derived from known antagonists, and virtual screening performed on the DrugBank dataset identified potential human PKR (hPKR) ligands with novel scaffolds. Interestingly, these included several HIV protease inhibitors for which endothelial cell dysfunction is a documented side effect. Our results suggest that the side effects might be due to inhibition of the PKR signaling pathway. Docking of known binders to a 3D homology model of hPKR1 is in agreement with the well-established canonical TM-bundle binding site of family A GPCRs. Furthermore, the docking results highlight residues that may form specific contacts with the ligands. These contacts provide structural explanation for the importance of several chemical features that were obtained from the structure-activity analysis of known binders. With the exception of a single loop residue that might be perused in the future for obtaining subtype-specific regulation, the results suggest an identical TM-bundle binding site for hPKR1 and hPKR2. In addition, analysis of the intracellular regions highlights variable regions that may provide subtype specificity.
Published in 2011
READ PUBLICATION →

Virus interactions with human signal transduction pathways.

Authors: Zhao Z, Xia J, Tastan O, Singh I, Kshirsagar M, Carbonell J, Klein-Seetharaman J

Abstract: Viruses depend on their hosts at every stage of their life cycles and must therefore communicate with them via Protein-Protein Interactions (PPIs). To investigate the mechanisms of communication by different viruses, we overlay reported pairwise human-virus PPIs on human signalling pathways. Of 671 pathways obtained from NCI and Reactome databases, 355 are potentially targeted by at least one virus. The majority of pathways are linked to more than one virus. We find evidence supporting the hypothesis that viruses often interact with different proteins depending on the targeted pathway. Pathway analysis indicates overrepresentation of some pathways targeted by viruses. The merged network of the most statistically significant pathways shows several centrally located proteins, which are also hub proteins. Generally, hub proteins are targeted more frequently by viruses. Numerous proteins in virus-targeted pathways are known drug targets, suggesting that these might be exploited as potential new approaches to treatments against multiple viruses.
Published in 2011
READ PUBLICATION →

Insights into the complex formed by matrix metalloproteinase-2 and alloxan inhibitors: molecular dynamics simulations and free energy calculations.

Authors: Giangreco I, Lattanzi G, Nicolotti O, Catto M, Laghezza A, Leonetti F, Stefanachi A, Carotti A

Abstract: Matrix metalloproteinases (MMP) are well-known biological targets implicated in tumour progression, homeostatic regulation, innate immunity, impaired delivery of pro-apoptotic ligands, and the release and cleavage of cell-surface receptors. Hence, the development of potent and selective inhibitors targeting these enzymes continues to be eagerly sought. In this paper, a number of alloxan-based compounds, initially conceived to bias other therapeutically relevant enzymes, were rationally modified and successfully repurposed to inhibit MMP-2 (also named gelatinase A) in the nanomolar range. Importantly, the alloxan core makes its debut as zinc binding group since it ensures a stable tetrahedral coordination of the catalytic zinc ion in concert with the three histidines of the HExxHxxGxxH metzincin signature motif, further stabilized by a hydrogen bond with the glutamate residue belonging to the same motif. The molecular decoration of the alloxan core with a biphenyl privileged structure allowed to sample the deep S(1)' specificity pocket of MMP-2 and to relate the high affinity towards this enzyme with the chance of forming a hydrogen bond network with the backbone of Leu116 and Asn147 and the side chains of Tyr144, Thr145 and Arg149 at the bottom of the pocket. The effect of even slight structural changes in determining the interaction at the S(1)' subsite of MMP-2 as well as the nature and strength of the binding is elucidated via molecular dynamics simulations and free energy calculations. Among the herein presented compounds, the highest affinity (pIC(50) = 7.06) is found for BAM, a compound exhibiting also selectivity (>20) towards MMP-2, as compared to MMP-9, the other member of the gelatinases.
Published in 2011
READ PUBLICATION →

The BioAssay network and its implications to future therapeutic discovery.

Authors: Zhang J, Lushington GH, Huan J

Abstract: BACKGROUND: Despite intense investment growth and technology development, there is an observed bottleneck in drug discovery and development over the past decade. NIH started the Molecular Libraries Initiative (MLI) in 2003 to enlarge the pool for potential drug targets, especially from the "undruggable" part of human genome, and potential drug candidates from much broader types of drug-like small molecules. All results are being made publicly available in a web portal called PubChem. RESULTS: In this paper we construct a network from bioassay data in PubChem, apply network biology concepts to characterize this bioassay network, integrate information from multiple biological databases (e.g. DrugBank, OMIM, and UniHI), and systematically analyze the potential of bioassay targets being new drug targets in the context of complex biological networks. We propose a model to quantitatively prioritize this druggability of bioassay targets, and literature evidence was found to confirm our prioritization of bioassay targets at a roughly 70% accuracy. CONCLUSIONS: Our analysis provide some measures of the value of the MLI data as a resource for both basic chemical biology research and future therapeutic discovery.
Published in 2011
READ PUBLICATION →

Network neighbors of drug targets contribute to drug side-effect similarity.

Authors: Brouwers L, Iskar M, Zeller G, van Noort V, Bork P

Abstract: In pharmacology, it is essential to identify the molecular mechanisms of drug action in order to understand adverse side effects. These adverse side effects have been used to infer whether two drugs share a target protein. However, side-effect similarity of drugs could also be caused by their target proteins being close in a molecular network, which as such could cause similar downstream effects. In this study, we investigated the proportion of side-effect similarities that is due to targets that are close in the network compared to shared drug targets. We found that only a minor fraction of side-effect similarities (5.8 %) are caused by drugs targeting proteins close in the network, compared to side-effect similarities caused by overlapping drug targets (64%). Moreover, these targets that cause similar side effects are more often in a linear part of the network, having two or less interactions, than drug targets in general. Based on the examples, we gained novel insight into the molecular mechanisms of side effects associated with several drug targets. Looking forward, such analyses will be extremely useful in the process of drug development to better understand adverse side effects.
Published in 2011
READ PUBLICATION →

Rapid analysis of pharmacology for infectious diseases.

Authors: Hopkins AL, Bickerton GR, Carruthers IM, Boyer SK, Rubin H, Overington JP

Abstract: Pandemic, epidemic and endemic infectious diseases are united by a common problem: how do we rapidly and cost-effectively identify potential pharmacological interventions to treat infections? Given the large number of emerging and neglected infectious diseases and the fact that they disproportionately afflict the poorest members of the global society, new ways of thinking are required to developed high productivity discovery systems that can be applied to a larger number of pathogens. The growing availability of parasite genome data provides the basis for developing methods to prioritize, a priori, the potential drug target and pharmacological landscape of an infectious disease. Thus the overall objective of infectious disease informatics is to enable the rapid generation of plausible, novel medical hypotheses of testable pharmacological experiments, by uncovering undiscovered relationships in the wealth of biomedical literature and databases that were collected for other purposes. In particular our goal is to identify potential drug targets present in a pathogen genome and prioritize which pharmacological experiments are most likely to discover drug-like lead compounds rapidly against a pathogen (i.e. which specific compounds and drug targets should be screened, in which assays and where they can be sourced). An integral part of the challenge is the development and integration of methods to predict druggability, essentiality, synthetic lethality and polypharmacology in pathogen genomes, while simultaneously integrating the inevitable issues of chemical tractability and the potential for acquired drug resistance from the start.
Published in December 2011
READ PUBLICATION →

Prediction of drug combinations by integrating molecular and pharmacological data.

Authors: Zhao XM, Iskar M, Zeller G, Kuhn M, van Noort V, Bork P

Abstract: Combinatorial therapy is a promising strategy for combating complex disorders due to improved efficacy and reduced side effects. However, screening new drug combinations exhaustively is impractical considering all possible combinations between drugs. Here, we present a novel computational approach to predict drug combinations by integrating molecular and pharmacological data. Specifically, drugs are represented by a set of their properties, such as their targets or indications. By integrating several of these features, we show that feature patterns enriched in approved drug combinations are not only predictive for new drug combinations but also provide insights into mechanisms underlying combinatorial therapy. Further analysis confirmed that among our top ranked predictions of effective combinations, 69% are supported by literature, while the others represent novel potential drug combinations. We believe that our proposed approach can help to limit the search space of drug combinations and provide a new way to effectively utilize existing drugs for new purposes.
Published on December 23, 2011
READ PUBLICATION →

Predicting adverse side effects of drugs.

Authors: Huang LC, Wu X, Chen JY

Abstract: BACKGROUND: Studies of toxicity and unintended side effects can lead to improved drug safety and efficacy. One promising form of study comes from molecular systems biology in the form of "systems pharmacology". Systems pharmacology combines data from clinical observation and molecular biology. This approach is new, however, and there are few examples of how it can practically predict adverse reactions (ADRs) from an experimental drug with acceptable accuracy. RESULTS: We have developed a new and practical computational framework to accurately predict ADRs of trial drugs. We combine clinical observation data with drug target data, protein-protein interaction (PPI) networks, and gene ontology (GO) annotations. We use cardiotoxicity, one of the major causes for drug withdrawals, as a case study to demonstrate the power of the framework. Our results show that an in silico model built on this framework can achieve a satisfactory cardiotoxicity ADR prediction performance (median AUC = 0.771, Accuracy = 0.675, Sensitivity = 0.632, and Specificity = 0.789). Our results also demonstrate the significance of incorporating prior knowledge, including gene networks and gene annotations, to improve future ADR assessments. CONCLUSIONS: Biomolecular network and gene annotation information can significantly improve the predictive accuracy of ADR of drugs under development. The use of PPI networks can increase prediction specificity and the use of GO annotations can increase prediction sensitivity. Using cardiotoxicity as an example, we are able to further identify cardiotoxicity-related proteins among drug target expanding PPI networks. The systems pharmacology approach that we developed in this study can be generally applicable to all future developmental drug ADR assessments and predictions.
Published on December 1, 2011
READ PUBLICATION →

Pharmacophore-based discovery of FXR agonists. Part I: Model development and experimental validation.

Authors: Schuster D, Markt P, Grienke U, Mihaly-Bison J, Binder M, Noha SM, Rollinger JM, Stuppner H, Bochkov VN, Wolber G

Abstract: The farnesoid X receptor (FXR) is involved in glucose and lipid metabolism regulation, which makes it an attractive target for the metabolic syndrome, dyslipidemia, atherosclerosis, and type 2 diabetes. In order to find novel FXR agonists, a structure-based pharmacophore model collection was developed and theoretically evaluated against virtual databases including the ChEMBL database. The most suitable models were used to screen the National Cancer Institute (NCI) database. Biological evaluation of virtual hits led to the discovery of a novel FXR agonist with a piperazine scaffold (compound 19) that shows comparable activity as the endogenous FXR agonist chenodeoxycholic acid (CDCA, compound 2).