British Journal of Clinical Pharmacology

DOI:10.1111/bcp.12622

The potential of translational bioinformatics approaches for pharmacology research Lang Li1,2,3 1

Department of Medical and Molecular Genetics, Indiana University School of Medicine,

Indianapolis, IN, 2Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN and 3Indiana Institute of Personalized Medicine, Indiana University School of Medicine, Indianapolis, IN, USA

Correspondence Professor Lang Li, PhD, Professor of Medical and Molecular Genetics, Clinical Pharmacology, and Biostatistics, Director of Center for Bioinformatics and Computational Biology, Indiana University School of Medicine, 410 West Tenth Street, HITS 5003, Indianapolis, IN, USA. Tel.: +1 317 274 4332 Fax: +1 317 278 9217 E-mail: [email protected] ----------------------------------------------------

Keywords big data, clinical pharmacology, systems pharmacology, translational bioinformatics ----------------------------------------------------

Received 7 October 2014

Accepted 15 February 2015

Accepted Article Published Online 5 March 2015 The field of bioinformatics has allowed the interpretation of massive amounts of biological data, ushering in the era of ‘omics’ to biomedical research. Its potential impact on pharmacology research is enormous and it has shown some emerging successes. A full realization of this potential, however, requires standardized data annotation for large health record databases and molecular data resources. Improved standardization will further stimulate the development of system pharmacology models, using translational bioinformatics methods. This new translational bioinformatics paradigm is highly complementary to current pharmacological research fields, such as personalized medicine, pharmacoepidemiology and drug discovery. In this review, I illustrate the application of transformational bioinformatics to research in numerous pharmacology subdisciplines.

Introduction

Prospects

Bioinformatics is a vibrant research field that has now demonstrated its value in unravelling the novel genomic [1] and proteomic [2] underpinnings of numerous complex diseases. The success of bioinformatics rides with the present day wave of ‘big omics’ datasets associated with rapidly evolving biotechnology advancements [3, 4]. The fundamental driving force of bioinformatics is its powerful nature of discovery. While this discovery approach is not new to clinical pharmacology research, both pharmacogenomics and pharmacoepidemiology research (i.e. pharmacovigilance) [5] are now being increasingly driven by bioinformatics-associated discovery. Moreover, accessibility to enormous data sources presents tremendous opportunities for translational bioinformatics research in pharmacology. Unlike previous review papers, which emphasized either bioinformatics resources [6, 7] or computational methodologies [8], here I will focus on emerging applications of bioinformatics and their impact on clinical pharmacology research.

Knowledge representation, a fundamental step in connecting big data sources

862

/

862–867

/

80:4

/

Br J Clin Pharmacol

Clinical pharmacology research-associated big data sources include observational (e.g. electronic medical/health records, the FDA adverse event reporting system [9], etc.) and drug knowledge (e.g. DrugBank [10], PubMed [11], PharmGKB [12], LINCS [13]) bases and databases. Extensive details of these data sources are well described in Russ Altman’s review paper [7]. Using big data, novel pharmacology research starts with communication between these databases, which in turn depends on standardized data annotation schemes. For example, the observational medical outcomes partnership (OMOP) recently attempted to integrate multiple observational databases through a common data model (CDM) [14] by normalizing the dictionaries of medications, diagnoses and laboratory tests across all its (OMOP) databases. The OMOP has also provided an annotation scheme to characterize the time-dependent nature of medications, laboratory tests and diagnoses. Furthermore, © 2015 The British Pharmacological Society

Translational bioinformatics approaches in pharmacology research

based on the CDM, OMOP has further standardized the implementation and reporting of drug/adverse drug event (ADE) association analyses. Although ADEs have been standardized through MedDRA (a medical terminology database for adverse drug events, www.meddra.org), whose utility in drug labelling has been well accepted, MedDRA still lacks sufficient annotation schemes to link an ADE with its molecular mechanism(s) [15]. Within the informatics research domain, the formulation of a terminology dictionary is now evolving towards the construction of an ontology. Ontology deals with questions concerning what entities exist or can be said to exist, how such entities can be grouped or related within a hierarchy, and how they can be subdivided according to their similarities and differences. In pharmacology research, a properly constructed ontology will facilitate the proper presentation of drugs and ADE data, either from large clinical databases or from the literature. Recently, the Ontology of Adverse Events (OAE, a biomedical ontology in the domain of adverse events) by He et al. [16] further strengthened the link between ADEs and anatomic regions, hence adding the capability of integrating molecular data into ADEs. In addition, the OAE recently defined causal adverse events (i.e. drug-induced ADEs), representing a much-needed improvement over MedDRA. While pharmacokinetic (PK) and pharmacodynamic (PD) studies and models are also essential components of pharmacology research, the interactions between PK/PD and observational studies remain limited. Recent work, however, on PK and drug–drug interaction (DDI) ontology [17] has laid a solid foundation for annotating in vitro, in vivo and clinical terminologies for PK and DDI preclinical and clinical studies. In particular, these ontology advancements have defined PK models and parameters, in vitro PK experimental design and results, clinical PK study design and results, and established criteria for defining DDIs. Thus, ontology is an essential step in connecting the OAE to the OMOP, and consequently, connecting PK to ADEs. All of the above-mentioned data and database standardization efforts are categorized into a field known as knowledge representation, a fundamental system used to connect large data sources. More extensive discussion of biomedical ontology development can be found in a review by Musen et al. [18]. All these newly developed pharmacologyrelated ontologies are publically available for knowledge representation. The primary challenge or roadblock to their utility to the broader pharmacology research community is proper training. The lack of proper tools for researchers, who have limited informatics knowledge to browse these ontologies, is the biggest gap in training.

Data mining of high dimensional drug–ADE associations Traditional pharmacovigilance studies have focused on associating single drugs with single ADEs [5]. However,

more recent successful studies have significantly expanded the dimension of associations. For example, Duke et al. investigated drug interactions, using a local version of the OMOP database at Indiana University [19], to identify successfully six novel drug interaction pairs that significantly increased myopathy risk above a mere additive risk from two single drugs. A notable example was a newly identified interaction between loratadine and simvastatin. In this drug interaction study, myopathy cases and controls were matched by the index time of the cases. This data mining analysis adjusted for covariates such as age, gender and the number of comedications in a logistic regression. In another example of multiple drug–ADE discovery, Tatonetti et al. further expanded association analysis between drugs or drug interactions and adverse events to assess all the FDA approved drugs and ADEs [20]. Using the FDA’s Adverse Event Reporting System (FAERS) as a training set and Stanford’s medical records as the validation set, they identified 47 associations of drugs and drug interaction effects. Notable examples included interactions between antidepressants and thiazide diuretics that resulted in increased incidence of prolonged QT. Additionally, Tatonetti et al.’s data mining algorithm adapted a propensity score approach to adjust for confounding from all co-medications [20]. This paper illustrates the tremendous value of data mining for correcting many false positive associations. To detect associations between any combinations of drugs and any combinations of adverse events, an association rule mining was implemented using the FAERS database. This association rule refers to the frequency of a drug–ADE association in a health record database. It claimed that 67% of associations were clinically validated by domain experts [21]. Computational efficiency of association rule mining was recently further improved by Xiang and Li et al. [22]. The combination of knowledge representation and data mining provides an extremely powerful translational bioinformatics approach for investigating drug– ADE associations. This new approach tremendously scales up association analyses between all FDA approved drugs and all ADEs, and is more discovery driven. It is also complementary to traditional pharmaco-epidemiology research into drug–ADE associations, which rely on stringent epidemiological study design and sensitivity analyses. Similar to the previously described challenges to knowledge representation, training and proper computational tools remain underdeveloped.

The value of text mining in clinical pharmacology research The contributions of text mining to clinical pharmacology research arise mainly from two sources, medical records and research literature. The definitions of diseases or ADE phenotypes using medical records require Br J Clin Pharmacol

/

80:4

/

863

L. Li

specific text mining tools. Wang et al. demonstrated the feasibility of investigating drug-induced ADEs by a natural language processing (NLP) algorithm that could identify and extract ADEs from clinical notes with moderate recall and precision [23]. Additionally, using known ADEs from seven drugs, they demonstrated the feasibility of establishing ADE pharmacovigilance from their NLP algorithm. Another example, by Xu et al., is to develop NLP algorithms to extract medication data (i.e. drug names and dose information) from clinical notes, with significant high performance [24]. Using this NLP system, they successfully reproduced the pharmacogenetic effects of VKORC1 and CYP2C9 on warfarin weekly dosing. Another application of text mining is literature extraction of gene–drug and drug–drug interactions with a two-fold research goal: (1) curating gene–drug and drug–drug interaction data from the literature and (2) discovering novel gene–drug and drug–drug interactions. To that end, Garten & Altman developed a text mining approach that could effectively extract pharmacogenomics relationships from full text articles [25]. More recently, they further expanded gene–drug interaction text mining into drug-drug interaction text mining [26]. To develop ‘gold standards’ and true positive training sets for drug interaction text mining, Li et al. [17] and a Spanish group [27] independently set up a corpora of drug interaction abstracts and sentences from both PubMed and FDA drug labels. Li’s group further developed an ontology of pharmacokinetics and drug interactions, successfully differentiating clinical from in vitro pharmacokinetic drug interaction studies. Using the machine learning algorithms developed from this corpus, Duke et al. mined all the published in vitro DDI studies on the enzyme cytochrome P450 to predict 13 193 novel drug interaction pairs [19]. As diverse disciplines and varied studies are involved [6], pharmacology evidence is often not available across in vitro experiments, clinical pharmacokinetic and clinical pharmacodynamic studies, which creates knowledge gaps. These knowledge gaps are the primary driving force for translational pharmacology research. Text mining, if successfully developed, will be a tremendously powerful approach to identify knowledge gaps for all currently used drugs. As text mining has emerged only recently in pharmacology research, it is limited by its own underdeveloped methodological research. Its theory and application tools need to mature before its further transition to general usage in pharmacology research.

Integrating molecular data with health data The eMERGE (Electronic Medical Records and GEnomics, emerge.mc.vanderbilt.edu) network is the first national consortium developed to investigate and catalogue genomic effects on disease/drug responses [28], as defined using observational databases coupled with biobanks. In a feasibility study, Ritchie et al. investigated associations 864

/

80:4

/

Br J Clin Pharmacol

between five disease phenotypes (atrial fibrillation, Crohn’s disease, multiple sclerosis, rheumatoid arthritis and type 2 diabetes) and various genetic variants [29]. These disease phenotypes were defined by a mixture of coded variables, and their features were extracted from clinical notes using NLP algorithms. Previous genotype– phenotype associations were replicated (P < 0.05) in 8/14 cases, at an odds ratio of > 1.25 [29]. More recently, Denny et al. extended a biobank-based genome-wide association study (GWAS) to 86 reported disease phenotypes in the National Human Genome Research Institute GWAS catalogue, successfully replicating 66% of prior GWAS associations [30]. Using a chemo-informatics approach, Pouliot et al. investigated whether drug-screening data from the publically available PubChem BioAssay database could accurately predict ADEs [31]. Subsequently, in mining the FAERS, they successfully identified nine ADEs, grouped by system organ classes, based on information from the PubChem Bioessay database. Schelleman et al. further demonstrated drug interactions between sulfonylureas and fribrates/statins [32], finding that fenofibrate combined with gemfibrozil increased glyburide-induced hypoglycaemia, based on Medicaid and Medicare claimant data. However, follow-up in vitro experiments predicted that fenofibrate and gemfibrozil only moderately increased glyburide drug exposure, suggesting that CYP inhibition was unlikely to cause this particular adverse drug interaction. In other work, Peng et al. investigated multiple drug targets, using structure-based proteinligand interactome prediction [33]. By scoring the interactions of 1000 FDA-approved drugs docked to 2500 human protein pockets, they predicted even more interactions among them, especially drug ‘off-targets’. Using additional compound physicochemical properties such as flexibility, hydrophobicity and size, they predicted and ranked drugs having similar predicted off-target profiles. In particular, the therapeutic effects of losartan and ergotamine on lung cancer were further evaluated and validated using an electronic medical records database at Indiana University, demonstrating the feasibility of revealing new drugs for lung cancer [33]. The integration of molecular data and health records data is well accepted, and has become widely used in current pharmacology research. The previously described knowledge representation and data mining approaches will bring about the discovery of new genetic risk factors for drug effects. On the other hand, it also currently suffers from the same challenges in training and availability of proper tools.

Network-based system pharmacology Network-based system pharmacology approaches are becoming a new paradigm for drug discovery, as described by Csermely et al. in a comprehensive review of model databases, visualization tools and computational

Translational bioinformatics approaches in pharmacology research

methods [34]. Here, I highlight a few significant recent papers on this topic. Besnard et al. developed a computational scheme for designing drugs against profiles of multiple possible target proteins, demonstrating that an approved acetylcholinesterase inhibitor could evolve into brain-penetrable ligands with selective profiles for G-protein-coupled receptors [35]. Moreover, among 800 predicted ligand-target predictions, 75% were confirmed experimentally [35]. Using another network approach, Li et al. showed strong connections between gene–gene relatedness and specific disease phenotypes, suggesting a method for discovering disease-associated genes [36]. Interestingly, using this network approach, this group further investigated the active compounds of a Chinese herb for treatment of type II diabetes [37]. Almost in parallel, Gottlieb et al. used a similar network approach to decipher drug–drug similarities and drug–disease associations, for eventual drug repurposing and drug response prediction [38, 39]. Most recently, Gottlieb & Altman constructed molecular pharmacology networks for integrative linkage of drug target genes, disease genes and pharmacogenetic genes [40]. Network-based system pharmacology is still in its infancy. Although its impact on pharmacology research is currently limited, its research potential for pharmacodynamic effects and modelling is immense. The current challenge to network-based system pharmacology is that its theory and methodology have not yet met the needs of pharmacology research. In other words, current systems pharmacology models have not well-predicted either in vitro or clinical drug responses.

Emergence of big data sources Social media is emerging as a valuable data source for assessing drug safety. Early work by Benton et al. showed that it was feasible to recapture hormone therapy ADEs using breast cancer medical ‘message boards’ [41]. Subsequently, Mao et al. further demonstrated that discontinuation of hormone therapies could also be captured by medical message boards [42]. Concurrently, White et al. used online health-seeking searches to investigate drug interaction signals, successfully identifying a synergistic hyperglycaemia ADE, based on joint searches for paroxetine and pravastatin [43]. Recently, this line of research was further evaluated by the FDA [44]. Another emerging resource is cell-based perturbation data sources, with one salient example being LINCS (Library of Integrated Cellular Signatures), an NIHsupported research network that has thus far investigated cellular responses (‘signatures’) of nearly 100 cell lines perturbed by 20 000 compounds and 6000 siRNAs [13]. Cellular response signatures are measured using various omics platforms. Such data sources will be extremely valuable to investigate the molecular mechanisms of pharmacodynamic effects.

Although the immense impact of these data resources is on the horizon, the immediate challenge is the lack of user-friendly interfaces, such that a biomedical research investigator with limited informatics skills can browse these data sources.

Conclusions Translational bioinformatics has shown great potential for contributing to two aspects of pharmacology research. First, based on its nature of large scale discovery, it expedites the translational process of pharmacology research. For example, using traditional pharmacology research approaches, it took 8 years of progression from drug metabolism and in vitro interaction studies to clinical pharmacology studies to understand tamoxifen pharmacogenetics and its interaction with selective serotonin receptor inhibitors [45]. By contrast, using translational bioinformatic methods, it took our group only 3 years to establish myopathy-associated drug interaction epidemiology data, combined with molecular mechanisms, by mining numerous drug interaction signals [19]. As text mining and data mining methodologies mature, we believe this process will become even more efficient. Secondly, translational bioinformatics is riding the wave of ‘big data’, and can investigate the same pharmacology problem from many different angles, using different data sources. Our myopathy-associated DDI study is again one such example [19], in which we mined medical records to obtain evidence of pharmacoepidemiological DDIs, while also mining the literature to examine DDI molecular mechanisms. Thus, the strength of translational bioinformatics approaches will only increase, as more and more data sources become more accessible. The emergence of translational bioinformatic approaches will not replace traditional pharmacology research. As a matter of fact, while translational bioinformatics is more discovery driven, it will be highly complimentary to traditional pharmacology research approaches, which are usually hypothesis-driven. For example, data mining medical records for novel drug– drug adverse interactions still need well-designed pharmacoepidemiology studies to validate causal relationships and/or traditional pharmacology experiments, to determine molecular mechanisms. The primary challenge of incorporating translational bioinformatics into clinical pharmacology research is training. Pharmacology research by itself has already faced its own challenges in integrating both clinical and basic sciences, and translational bioinformatics adds yet another dimension to this field. Unlike the PK and PD modelling skills required for pharmacometrics, translational bioinformatics requires much broader skills in data mining, text mining, knowledge representation and omics. These requirements call for broader collaboration among different Br J Clin Pharmacol

/

80:4

/

865

L. Li

academic units, such as medicine, pharmacology and informatics. Joint training programmes will be absolutely necessary to prepare young scientists to enter this field. We believe, however, that these challenges will be met with innovative solutions, similar to our long history of successful adoption of emerging scientific pursuits. Proper bioinformatics training for pharmacology research investigators requires user-friendly bioinformatics tools, which will allow them to mine large medical databases and literature text, build network-based systems models to predict drug responses and explore large databases. As many of these translational bioinformatics areas remain in development and many tools are not necessarily available at the moment, an alternative strategy is to seek synergistic collaborations with informaticians. As the research goals and approaches between informaticians and those of traditional clinical pharmacologists and molecular pharmacologists are rather complementary, I fully expect successful synergistic collaborations.

Competing Interests The author has completed the Unified Competing Interest form at www.icmje.org/coi_disclosure.pdf and declares no support from any organization for the submitted work [LL], no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years [LL] and no other relationships or activities that could appear to have influenced the submitted work [LL]. This work was supported by DK102694, GM10448301 and LM011945.

REFERENCES 1 Taking pan-cancer analysis global. Nat Genet 2013; 45: 1263. 2 Zhang B, Wang J, Wang X, Zhu J, Liu Q, Shi Z, Chambers MC, Zimmerman LJ, Shaddox KF, Kim S, Davies SR, Wang S, Wang P, Kinsinger CR, Rivers RC, Rodriguez H, Townsend RR, Ellis MJ, Carr SA, Tabb DL, Coffey RJ, Slebos RJ, Liebler DC, Nci C. Proteogenomic characterization of human colon and rectal cancer. Nature 2014; 513: 382–7. 3 Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet 2010; 11: 685–96.

6 Altman RB. Personal genomic measurements: the opportunity for information integration. Clin Pharmacol Ther 2013; 93: 21–3. 7 Altman RB. Translational bioinformatics: linking the molecular world to the clinical world. Clin Pharmacol Ther 2012; 91: 994–1000. 8 Harpaz R, DuMouchel W, Shah NH, Madigan D, Ryan P, Friedman C. Novel data-mining methodologies for adverse drug event discovery and analysis. Clin Pharmacol Ther 2012; 91: 1010–21. 9 Available at http://www.fda.gov/Drugs/ GuidanceComplianceRegulatoryInformation/Surveillance/ AdverseDrugEffects/ (last accessed 30 September 2014). 10 Available at http://www.drugbank.ca (last accessed 30 September 2014). 11 Available at http://www.pubmed.org (last accessed 30 September 2014). 12 Available at http://www.pharmgkb.org (last accessed 30 September 2014). 13 Available at http://www.lincscloud.org (last accessed 30 September 2014). 14 Stang PE, Ryan PB, Racoosin JA, Overhage JM, Hartzema AG, Reich C, Welebob E, Scarnecchia T, Woodcock J. Advancing the science for active surveillance: rationale and design for the Observational Medical Outcomes Partnership. Ann Intern Med 2010; 153: 600–6. 15 Zhichkin PE, Athey BD, Avigan MI, Abernethy DR. Needs for an expanded ontology-based classification of adverse drug reactions and related mechanisms. Clin Pharmacol Ther 2012; 91: 963–5. 16 He Y, Sarntivijai S, Lin Y, Xiang Z, Guo A, Zhang S, Jagannathan D, Toldo L, Tao C, Smith B. OAE: The Ontology of Adverse Events. J Biomed Semant 2014; 5: 29. 17 Wu HY, Karnik S, Subhadarshini A, Wang Z, Philips S, Han X, Chiang C, Liu L, Boustani M, Rocha LM, Quinney SK, Flockhart D, Li L. An integrated pharmacokinetics ontology and corpus for text mining. BMC Bioinformatics 2013; 14: 35. 18 Musen MA, Noy NF, Shah NH, Whetzel PL, Chute CG, Story MA, Smith B, Team N. The National Center for Biomedical Ontology. J Am Med Inform Assoc 2012; 19: 190–5. 19 Duke JD, Han X, Wang Z, Subhadarshini A, Karnik SD, Li X, Hall SD, Jin Y, Callaghan JT, Overhage MJ, Flockhart DA, Strother RM, Quinney SK, Li L. Literature based drug interaction prediction with clinical assessment using electronic medical records: novel myopathy associated drug interactions. PLoS Comput Biol 2012; 8: e1002614.

4 Shapiro E, Biezuner T, Linnarsson S. Single-cell sequencingbased technologies will revolutionize whole-organism science. Nat Rev Genet 2013; 14: 618–30.

20 Tatonetti NP, Fernald GH, Altman RB. A novel signal detection algorithm for identifying hidden drug-drug interactions in adverse event reports. J Am Med Inform Assoc 2012; 19: 79–85.

5 Ryan PB, Madigan D, Stang PE, Overhage JM, Racoosin JA, Hartzema AG. Empirical assessment of methods for risk identification in healthcare data: results from the experiments of the Observational Medical Outcomes Partnership. Stat Med 2012; 31: 4401–15.

21 Harpaz R, Haerian K, Chase HS, Friedman C. Statistical Mining of Potential Drug Interaction Adverse Effects in FDA’s Spontaneous Reporting System. AMIA Annu Symp Proc 2010; 2010: 281–5.

866

/

80:4

/

Br J Clin Pharmacol

Translational bioinformatics approaches in pharmacology research 22 Xiang Y, Albin A, Ren K, Zhang P, Etter JP, Lin S, Li L. Efficiently mining Adverse Event Reporting System for multiple drug interactions. AMIA Summit on Translational Science Proceeding 2014, 120–125. 23 Wang X, Hripcsak G, Markatou M, Friedman C. Active computerized pharmacovigilance using natural language processing, statistics, and electronic health records: a feasibility study. J Am Med Inform Assoc 2009; 16: 328–37. 24 Xu H, Jiang M, Oetjens M, Bowton EA, Ramirez AH, Jeff JM, Basford MA, Pulley JM, Cowan JD, Wang X, Ritchie MD, Masys DR, Roden DM, Crawford DC, Denny JC. Facilitating pharmacogenetic studies using electronic health records and natural-language processing: a case study of warfarin. J Am Med Inform Assoc 2011; 18: 387–91. 25 Garten Y, Altman RB. Pharmspresso: a text mining tool for extraction of pharmacogenomic concepts and relationships from full text. BMC Bioinformatics 2009; 10: S6. 26 Percha B, Garten Y, Altman RB. Discovery and explanation of drug-drug interactions via text mining. Pac Symp Biocomput 2012: 410–21. 27 Available at http://www.mavir.net/conf/137ddiextraction2013 (last accessed 30 September 2014). 28 Gottesman O, Kuivaniemi H, Tromp G, Faucett WA, Li R, Manolio TA, Sanderson SC, Kannry J, Zinberg R, Basford MA, Brilliant M, Carey DJ, Chisholm RL, Chute CG, Connolly JJ, Crosslin D, Denny JC, Gallego CJ, Haines JL, Hakonarson H, Harley J, Jarvik GP, Kohane I, Kullo IJ, Larson EB, McCarty C, Ritchie MD, Roden DM, Smith ME, Bottinger EP, Williams MS, The e MERGE Network. The Electronic Medical Records and Genomics (eMERGE) Network: past, present, and future. Genet Med 2013; 15: 761–71. 29 Ritchie MD, Denny JC, Crawford DC, Ramirez AH, Weiner JB, Pulley JM, Basford MA, Brown-Gentry K, Balser JR, Masys DR, Haines JL, Roden DM. Robust replication of genotypephenotype associations across multiple diseases in an electronic medical record. Am J Hum Genet 2010; 86: 560–72. 30 Denny JC, Bastarache L, Ritchie MD, Carroll RJ, Zink R, Mosley JD, Field JR, Pulley JM, Ramirez AH, Bowton E, Basford MA, Carrell DS, Peissig PL, Kho AN, Pacheco JA, Rasmussen LV, Crosslin DR, Crane PK, Pathak J, Bielinski SJ, Pendergrass SA, Xu H, Hindorff LA, Li R, Manolio TA, Chute CG, Chisholm RL, Larson EB, Jarvik GP, Brilliant MH, McCarty CA, Kullo IJ, Haines JL, Crawford DC, Masys DR, Roden DM. Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data. Nat Biotechnol 2013; 31: 1102–10. 31 Pouliot Y, Chiang AP, Butte AJ. Predicting adverse drug reactions using publicly available PubChem BioAssay data. Clin Pharmacol Ther 2011; 90: 90–9. 32 Schelleman H, Han X, Brensinger CM, Quinney SK, Bilker WB, Flockhart DA, Li L, Hennessy S. Pharmacoepidemiologic and in vitro evaluation of potential drug–drug interactions of

sulfonylureas with fibrates and statins. Br J Clin Pharmacol 2014; 78: 639–48. 33 Peng X, Wang F, Li L, Bum-Erdene K, Xu D, Wang B, Sinn AA, Pollok KE, Sandusky GE, Li L, Turchi JJ, Jalal SI, Meroueh SO. Exploring a structural protein-drug interactome for new therapeutics in lung cancer. Mol Biosyst 2014; 10: 581–91. 34 Csermely P, Korcsmaros T, Kiss HJ, London G, Nussinov R. Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol Ther 2013; 138: 333–408. 35 Besnard J, Ruda GF, Setola V, Abecassis K, Rodriguiz RM, Huang XP, Norval S, Sassano MF, Shin AI, Webster LA, Simeons FR, Stojanovski L, Prat A, Seidah NG, Constam DB, Bickerton GR, Read KD, Wetsel WC, Gilbert IH, Roth BL, Hopkins AL. Automated design of ligands to polypharmacological profiles. Nature 2012; 492: 215–20. 36 Wu X, Jiang R, Zhang MQ, Li S. Network-based global inference of human disease genes. Mol Syst Biol 2008; 4: 189. 37 Li H, Zhao L, Zhang B, Jiang Y, Wang X, Guo Y, Liu H, Li S, Tong X. A network pharmacology approach to determine active compounds and action mechanisms of ge-gen-qinlian decoction for treatment of type 2 diabetes. Evid base Compl Alternative Med 2014; 2014: 495840. 38 Gottlieb A, Magger O, Berman I, Ruppin E, Sharan R. PRINCIPLE: a tool for associating genes with diseases via network propagation. Bioinformatics 2011; 27: 3325–6. 39 Silberberg Y, Gottlieb A, Kupiec M, Ruppin E, Sharan R. Large-scale elucidation of drug response pathways in humans. J Comput Biol 2012; 19: 163–74. 40 Gottlieb A, Altman RB. Integrating systems biology sources illuminates drug action. Clin Pharmacol Ther 2014; 95: 663–9. 41 Benton A, Ungar L, Hill S, Hennessy S, Mao J, Chung A, Leonard CE, Holmes JH. Identifying potential adverse effects using the web: a new approach to medical hypothesis generation. J Biomed Inform 2011; 44: 989–96. 42 Mao JJ, Chung A, Benton A, Hill S, Ungar L, Leonard CE, Hennessy S, Holmes JH. Online discussion of drug side effects and discontinuation among breast cancer survivors. Pharmacoepidemiol Drug Saf 2013; 22: 256–62. 43 White RW, Tatonetti NP, Shah NH, Altman RB, Horvitz E. Web-scale pharmacovigilance: listening to signals from the crowd. J Am Med Inform Assoc 2013; 20: 404–8. 44 Sarntivijai S, Abernethy DR. Use of internet search logs to evaluate potential drug adverse events. Clin Pharmacol Ther 2014; 96: 149–50. 45 Goetz MP, Rae JM, Suman VJ, Safgren SL, Ames MM, Visscher DW, Reynolds C, Couch FJ, Lingle WL, Flockhart DA, Desta Z, Perez EA, Ingle JN. Pharmacogenetics of tamoxifen biotransformation is associated with clinical outcomes of efficacy and hot flashes. J Clin Oncol 2005; 23: 9312–8.

Br J Clin Pharmacol

/

80:4

/

867

The potential of translational bioinformatics approaches for pharmacology research.

The field of bioinformatics has allowed the interpretation of massive amounts of biological data, ushering in the era of 'omics' to biomedical researc...
241KB Sizes 3 Downloads 7 Views