Santiago Vilar
University of Santiago de Compostela
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Santiago Vilar.
Current Topics in Medicinal Chemistry | 2008
Santiago Vilar; Giorgio Cozza; Stefano Moro
The search for new compounds with a given biological activity requires enormous effort in terms of manpower and cost. This effort arises from the large number of compounds that need to be synthesized and subsequently biologically evaluated. For this reason the pharmaceutical industry has shown great interest in theoretical methods that enable the rational design of pharmaceutical agents. In the last years bioinformatics has experienced a great evolution due to the development of specialized software and to the increasing computer power. The codification of the structural information of molecules through molecular descriptors and the subsequent data analysis allow establishing QSAR models (Quantitative Structure-Activity Relationship) that can be applied to the design and the virtual screening of new drugs. The development of sophisticated Docking methodologies also allows a more accurate predict of the biological activity of molecules. Moreover, through this type of computational techniques and theoretical approaches, it is possible to develop explanatory hypothesis on the mechanism of action of drugs. This work provides a brief description of a series of studies implemented in the software MOE (Molecular Operating Environment) with particular attention to the medicinal chemistry aspects.
Journal of the American Medical Informatics Association | 2013
Rave Harpaz; Santiago Vilar; William DuMouchel; Hojjat Salmasian; Krystl Haerian; Nigam H. Shah; Herbert S. Chase; Carol Friedman
OBJECTIVE Data-mining algorithms that can produce accurate signals of potentially novel adverse drug reactions (ADRs) are a central component of pharmacovigilance. We propose a signal-detection strategy that combines the adverse event reporting system (AERS) of the Food and Drug Administration and electronic health records (EHRs) by requiring signaling in both sources. We claim that this approach leads to improved accuracy of signal detection when the goal is to produce a highly selective ranked set of candidate ADRs. MATERIALS AND METHODS Our investigation was based on over 4 million AERS reports and information extracted from 1.2 million EHR narratives. Well-established methodologies were used to generate signals from each source. The study focused on ADRs related to three high-profile serious adverse reactions. A reference standard of over 600 established and plausible ADRs was created and used to evaluate the proposed approach against a comparator. RESULTS The combined signaling system achieved a statistically significant large improvement over AERS (baseline) in the precision of top ranked signals. The average improvement ranged from 31% to almost threefold for different evaluation categories. Using this system, we identified a new association between the agent, rasburicase, and the adverse event, acute pancreatitis, which was supported by clinical review. CONCLUSIONS The results provide promising initial evidence that combining AERS with EHRs via the framework of replicated signaling can improve the accuracy of signal detection for certain operating scenarios. The use of additional EHR data is required to further evaluate the capacity and limits of this system and to extend the generalizability of these results.
Journal of Chemical Information and Modeling | 2002
Santiago Vilar; Ernesto Estrada; Eugenio Uriarte; Lourdes Santana; Yaquelin Gutierrez
The TOPological Substructural MOlecular DEsign (TOPS-MODE) approach has been used to predict the anti-HIV activity in MT-4 assays (Estrada et al., 2002) of a diverse range of purine-based nucleosides. A database of 206 nucleosides has been selected from the literature and a theoretical virtual screening model has been developed. The model is able of discriminating between compounds that have anti-HIV activity and those that do not, with a good classification level of 85% in the training and 82.8% in the cross-validation series. On the basis of the information generated by the model, the correct classification of practically 80% of compounds from an external prediction set has been achieved using the theoretical model. Furthermore, the contribution of a range of molecular fragments to the pharmacological action has been calculated and this could provide a powerful tool in the design of nucleoside analogues that show activity against the HIV. Finally, a QSAR model has been developed that allows quantitative data to be obtained regarding the pharmacological potency shown by this type of compound.
Science Translational Medicine | 2013
Xiaochen Sun; Santiago Vilar; Nicholas P. Tatonetti
Combinatorial drug discovery will rely on new approaches to analyzing highly dimensional data resources. Human disease is a complex network of interacting pathways. As such, it may be necessary to treat the system with a combination of drugs to boost efficacy and reduce side effects. In this Review, the authors discuss the current landscape of methodologies for using highly dimensional data resources and high-throughput biological measurements for combinatorial drug discovery. A more nuanced approach to drug design is to use multiple drugs in combination to target interacting or complementary pathways. Drug combination treatments have shown higher efficacy, fewer side effects, and less toxicity compared to single-drug treatment. In this Review, we focus on the use of high-throughput biological measurements (genetics, transcripts, and chemogenetic interactions) and the computational methods they necessitate to further combinatorial drug design (CDD). We highlight the state-of-the-art analytical methods, including network analysis, integrative informatics, and dynamic molecular modeling, that have been used successfully in CDD. Finally, we present an exhaustive list of the publicly available data and methodological resources available to the community. Such next-generation technologies that enable the measurement of millions of cellular data points simultaneously may usher in a new paradigm in drug discovery, where medicine is viewed as a system of interacting genes and pathways rather than the result of an individual protein or gene.
Journal of Molecular Graphics & Modelling | 2011
Santiago Vilar; Giulio Ferino; Sharangdhar S. Phatak; Barkin Berk; Claudio N. Cavasotto; Stefano Costanzi
G protein-coupled receptors (GPCRs) regulate a wide range of physiological functions and hold great pharmaceutical interest. Using the β(2)-adrenergic receptor as a case study, this article explores the applicability of docking-based virtual screening to the discovery of GPCR ligands and defines methods intended to improve the screening performance. Our controlled computational experiments were performed on a compound dataset containing known agonists and blockers of the receptor as well as a large number of decoys. The screening based on the structure of the receptor crystallized in complex with its inverse agonist carazolol yielded excellent results, with a clearly delineated prioritization of ligands over decoys. Blockers generally were preferred over agonists; however, agonists were also well distinguished from decoys. A method was devised to increase the screening yields by generating an ensemble of alternative conformations of the receptor that accounts for its flexibility. Moreover, a method was devised to improve the retrieval of agonists, based on the optimization of the receptor around a known agonist. Finally, the applicability of docking-based virtual screening also to homology models endowed with different levels of accuracy was proved. This last point is of uttermost importance, since crystal structures are available only for a limited number of GPCRs, and extends our conclusions to the entire superfamily. The outcome of this analysis definitely supports the application of computer-aided techniques to the discovery of novel GPCR ligands, especially in light of the fact that, in the near future, experimental structures are expected to be solved and become available for an ever increasing number of GPCRs.
Journal of the American Medical Informatics Association | 2012
Santiago Vilar; Rave Harpaz; Eugenio Uriarte; Lourdes Santana; Raul Rabadan; Carol Friedman
BACKGROUND Drug-drug interactions (DDIs) are responsible for many serious adverse events; their detection is crucial for patient safety but is very challenging. Currently, the US Food and Drug Administration and pharmaceutical companies are showing great interest in the development of improved tools for identifying DDIs. METHODS We present a new methodology applicable on a large scale that identifies novel DDIs based on molecular structural similarity to drugs involved in established DDIs. The underlying assumption is that if drug A and drug B interact to produce a specific biological effect, then drugs similar to drug A (or drug B) are likely to interact with drug B (or drug A) to produce the same effect. DrugBank was used as a resource for collecting 9454 established DDIs. The structural similarity of all pairs of drugs in DrugBank was computed to identify DDI candidates. RESULTS The methodology was evaluated using as a gold standard the interactions retrieved from the initial DrugBank database. Results demonstrated an overall sensitivity of 0.68, specificity of 0.96, and precision of 0.26. Additionally, the methodology was also evaluated in an independent test using the Micromedex/Drugdex database. CONCLUSION The proposed methodology is simple, efficient, allows the investigation of large numbers of drugs, and helps highlight the etiology of DDI. A database of 58 403 predicted DDIs with structural evidence is provided as an open resource for investigators seeking to analyze DDIs.
Journal of Theoretical Biology | 2009
Santiago Vilar; Humberto González-Díaz; Lourdes Santana; Eugenio Uriarte
The combination of the network theory and the calculation of topological indices (TIs) allow establishing relationships between the molecular structure of large molecules like the genes and proteins and their properties at a biological level. This type of models can be considered quantitative structure-activity relationships (QSAR) for biopolymers. In the present work a QSAR model is reported for proteins, related to human colorectal cancer (HCC) and codified by different genes that have been identified experimentally by Sjöblom et al. [2006. The consensus coding sequences of human breast and colorectal cancers. Science 314, 268-274] among more than 10000 human genes. The 69 proteins related to human colorectal cancer (HCCp) and a control group of 200 proteins not related to HCC (no-HCCp) were represented through an HP Lattice type Network. Starting from the generated graphs we calculate a set of descriptors of electrostatic potential type (xi(k)) that allow to establish, through a linear discriminant analysis (LDA), a QSAR model of relatively high percentage of good classification (higher than 80%) to differentiate between HCCp and no-HCCp proteins. The purpose of this study is helping to predict the possible implication of a certain gene and/or protein (biomarker) in the colorectal cancer. Different procedures of validation of the obtained model have been carried out in order to corroborate its stability, including cross-validation series (CV) and evaluation of an additional series of 200 no-HCCp. This biostatistic methodology could be applied to predict human colorectal cancer biomarkers and to understand much better the biological aspects of this disease.
Journal of Molecular Graphics & Modelling | 2010
Santiago Vilar; Mayukh Chakrabarti; Stefano Costanzi
The distribution of compounds between blood and brain is a very important consideration for new candidate drug molecules. In this paper, we describe the derivation of two linear discriminant analysis (LDA) models for the prediction of passive blood-brain partitioning, expressed in terms of logBB values. The models are based on computationally derived physicochemical descriptors, namely the octanol/water partition coefficient (logP), the topological polar surface area (TPSA) and the total number of acidic and basic atoms, and were obtained using a homogeneous training set of 307 compounds, for all of which the published experimental logBB data had been determined in vivo. In particular, since molecules with logBB>0.3 cross the blood-brain barrier (BBB) readily while molecules with logBB<-1 are poorly distributed to the brain, on the basis of these thresholds we derived two distinct models, both of which show a percentage of good classification of about 80%. Notably, the predictive power of our models was confirmed by the analysis of a large external dataset of compounds with reported activity on the central nervous system (CNS) or lack thereof. The calculation of straightforward physicochemical descriptors is the only requirement for the prediction of the logBB of novel compounds through our models, which can be conveniently applied in conjunction with drug design and virtual screenings.
Journal of the American Medical Informatics Association | 2011
Santiago Vilar; Rave Harpaz; Herbert S. Chase; Stefano Costanzi; Raul Rabadan; Carol Friedman
BACKGROUND Adverse drug events (ADE) cause considerable harm to patients, and consequently their detection is critical for patient safety. The US Food and Drug Administration maintains an adverse event reporting system (AERS) to facilitate the detection of ADE in drugs. Various data mining approaches have been developed that use AERS to detect signals identifying associations between drugs and ADE. The signals must then be monitored further by domain experts, which is a time-consuming task. OBJECTIVE To develop a new methodology that combines existing data mining algorithms with chemical information by analysis of molecular fingerprints to enhance initial ADE signals generated from AERS, and to provide a decision support mechanism to facilitate the identification of novel adverse events. RESULTS The method achieved a significant improvement in precision in identifying known ADE, and a more than twofold signal enhancement when applied to the ADE rhabdomyolysis. The simplicity of the method assists in highlighting the etiology of the ADE by identifying structurally similar drugs. A set of drugs with strong evidence from both AERS and molecular fingerprint-based modeling is constructed for further analysis. CONCLUSION The results demonstrate that the proposed methodology could be used as a pharmacovigilance decision support tool to facilitate ADE detection.
Journal of Biomedical Informatics | 2015
Serkan Ayvaz; John R. Horn; Oktie Hassanzadeh; Qian Zhu; Johann Stan; Nicholas P. Tatonetti; Santiago Vilar; Mathias Brochhausen; Matthias Samwald; Majid Rastegar-Mojarad; Michel Dumontier; Richard D. Boyce
Although potential drug-drug interactions (PDDIs) are a significant source of preventable drug-related harm, there is currently no single complete source of PDDI information. In the current study, all publically available sources of PDDI information that could be identified using a comprehensive and broad search were combined into a single dataset. The combined dataset merged fourteen different sources including 5 clinically-oriented information sources, 4 Natural Language Processing (NLP) Corpora, and 5 Bioinformatics/Pharmacovigilance information sources. As a comprehensive PDDI source, the merged dataset might benefit the pharmacovigilance text mining community by making it possible to compare the representativeness of NLP corpora for PDDI text extraction tasks, and specifying elements that can be useful for future PDDI extraction purposes. An analysis of the overlap between and across the data sources showed that there was little overlap. Even comprehensive PDDI lists such as DrugBank, KEGG, and the NDF-RT had less than 50% overlap with each other. Moreover, all of the comprehensive lists had incomplete coverage of two data sources that focus on PDDIs of interest in most clinical settings. Based on this information, we think that systems that provide access to the comprehensive lists, such as APIs into RxNorm, should be careful to inform users that the lists may be incomplete with respect to PDDIs that drug experts suggest clinicians be aware of. In spite of the low degree of overlap, several dozen cases were identified where PDDI information provided in drug product labeling might be augmented by the merged dataset. Moreover, the combined dataset was also shown to improve the performance of an existing PDDI NLP pipeline and a recently published PDDI pharmacovigilance protocol. Future work will focus on improvement of the methods for mapping between PDDI information sources, identifying methods to improve the use of the merged dataset in PDDI NLP algorithms, integrating high-quality PDDI information from the merged dataset into Wikidata, and making the combined dataset accessible as Semantic Web Linked Data.