Data-Driven Prediction of Drug Effects and Interactions

Two new databases—one of drug effects and a second of drug-drug interaction side effects—permit identification of drug targets, prediction of drug indications, and discovery of drug class interactions. Avoiding Adversity For some disease-therapy pairs, Francis Bacon was right: “The remedy is worse than the disease.” And when several drugs collide in an individual, the troubling effects can multiply. One goal of the developing field of pharmacogenomics is to make use of clinical data to predict adverse drug events so that future patients can be protected from the sometimes serious consequences. Now, Tatonetti et al. describe two new databases—one of drug effects and a second of drug-drug interaction side effects—that permit the identification of drug targets, prediction of drug indications, and discovery of drug-class interactions. Adverse drug events aren’t merely a nuisance; these toxic interactions can cause debilitating illness and death. The nature of clinical trials doesn’t allow the detection of all serious side effects and drug interactions before approval of the therapy, by regulatory agencies, for use in patients. But these agencies along with pharmaceutical companies, hospitals, and other institutions collect adverse event reports after the drugs are in use in the clinic. When delineated in databases and coupled with the impressive computing power now available, these reports have the potential to permit characterization of drug effects at the population level. However, even with the recent move toward electronic health records, adverse event data often lack crucial information about co-prescribed medications, patient demographics and medical histories, and the reasons that a given drug was prescribed in the first place. One can easily see how that lack of such information thwarts the ability to obtain meaningful analyses of drug side effects and interactions. To address this problem of omission and improve the ability to analyze drug effects, Tatonetti et al. use an adaptive data-driven approach to correct for the lack of such information—the so-called unknown “covariates.” Using this information, the authors developed two comprehensive databases—one of drug effects (Offsides) and another of drug-drug interaction side effects (Twosides)—and then used their new databases to pinpoint drug targets and discover drug-class interactions. Finally, the authors validated 47 of the drug-class interactions in an independent analysis of patient electronic health records. When prescribed together, widely used antidepressant drugs (selective serotonin reuptake inhibitors) and thiazide diuretics were associated with an increase in the incidence of prolonged QT, which indicates a delayed repolarization of the heart after a heartbeat. Prolonged QT can increase a patient’s risk of palpitations, fainting, and even sudden death resulting from ventricular fibrillation. Better than tarot cards or crystal balls, the authors show that intricate analyses of observational clinical data can improve physicians’ ability to predict the future—at least with respect to as yet uncharacterized adverse drug effects and interactions. Adverse drug events remain a leading cause of morbidity and mortality around the world. Many adverse events are not detected during clinical trials before a drug receives approval for use in the clinic. Fortunately, as part of postmarketing surveillance, regulatory agencies and other institutions maintain large collections of adverse event reports, and these databases present an opportunity to study drug effects from patient population data. However, confounding factors such as concomitant medications, patient demographics, patient medical histories, and reasons for prescribing a drug often are uncharacterized in spontaneous reporting systems, and these omissions can limit the use of quantitative signal detection methods used in the analysis of such data. Here, we present an adaptive data-driven approach for correcting these factors in cases for which the covariates are unknown or unmeasured and combine this approach with existing methods to improve analyses of drug effects using three test data sets. We also present a comprehensive database of drug effects (Offsides) and a database of drug-drug interaction side effects (Twosides). To demonstrate the biological use of these new resources, we used them to identify drug targets, predict drug indications, and discover drug class interactions. We then corroborated 47 (P < 0.0001) of the drug class interactions using an independent analysis of electronic medical records. Our analysis suggests that combined treatment with selective serotonin reuptake inhibitors and thiazides is associated with significantly increased incidence of prolonged QT intervals. We conclude that confounding effects from covariates in observational clinical data can be controlled in data analyses and thus improve the detection and prediction of adverse drug effects and interactions.

[1]  C. Kleeman,et al.  Diuretic-induced hyponatremia. , 1971, Annals of internal medicine.

[2]  H. Halkin,et al.  Thiazide-induced hyponatremia. Reproducibility by single dose rechallenge and an analysis of pathogenesis. , 1989, Annals of internal medicine.

[3]  E F Cook,et al.  Performance of tests of significance based on stratification by a multivariate confounder score or by a propensity score. , 1989, Journal of clinical epidemiology.

[4]  N. Laird,et al.  Incidence of Adverse Drug Events and Potential Adverse Drug Events: Implications for Prevention , 1995 .

[5]  A. Bartelink,et al.  Severe hyponatremia during therapy with fluoxetine. , 1996, Archives of internal medicine.

[6]  D. Classen,et al.  Adverse drug events in hospitalized patients. Excess length of stay, extra costs, and attributable mortality. , 1997, JAMA.

[7]  William DuMouchel,et al.  Bayesian Data Mining in Large Frequency Tables, with an Application to the FDA Spontaneous Reporting System , 1999 .

[8]  R. O’Neill,et al.  Use of Screening Algorithms and Computer Systems to Efficiently Signal Higher-Than-Expected Combinations of Drugs and Events in the US FDA’s Spontaneous Reports Database , 2002, Drug safety.

[9]  M Soledad Cepeda,et al.  Comparison of logistic regression versus propensity score when the number of events is low and there are multiple confounders. , 2003, American journal of epidemiology.

[10]  Joseph M. Tonning,et al.  Pharmacovigilance in the 21st Century: New Systematic Tools for an Old Problem , 2004, Pharmacotherapy.

[11]  A. Bate,et al.  Extending the methods used to screen the WHO drug safety database towards analysis of complex associations and improved accuracy for rare events , 2006, Statistics in medicine.

[12]  Johan Hopstadius,et al.  Impact of Stratification on Adverse Drug Reaction Surveillance , 2008, Drug safety.

[13]  Stephen J W Evans Stratification for Spontaneous Report Databases , 2008, Drug safety.

[14]  P. Bork,et al.  Drug Target Identification Using Side-Effect Similarity , 2008, Science.

[15]  R. Sundberg,et al.  A statistical methodology for drug–drug interaction surveillance , 2008, Statistics in medicine.

[16]  Bin Chen,et al.  Gaining Insight into Off-Target Mediated Effects of Drug Candidates with a Comprehensive Systems Chemical Biology Analysis , 2009, J. Chem. Inf. Model..

[17]  Philip E. Bourne,et al.  Drug Discovery Using Chemical Systems Biology: Identification of the Protein-Ligand Binding Network To Explain the Side Effects of CETP Inhibitors , 2009, PLoS Comput. Biol..

[18]  Ravi Iyengar,et al.  Network analyses in systems pharmacology , 2009, Bioinform..

[19]  M. Milik,et al.  Mapping adverse drug reactions in chemical space. , 2009, Journal of medicinal chemistry.

[20]  A. Bate,et al.  Quantitative signal detection using spontaneous ADR reporting , 2009, Pharmacoepidemiology and drug safety.

[21]  Chitta Baral,et al.  Discovering drug–drug interactions: a text-mining and reasoning approach based on properties of drug metabolism , 2010, Bioinform..

[22]  Ugo Moretti,et al.  Identifying Adverse Drug Reactions Associated with Drug-Drug Interactions , 2010, Drug safety.

[23]  Von-Wun Soo,et al.  Analysis of adverse drug reactions using drug and drug target interactions and graph-based methods , 2010, Artif. Intell. Medicine.

[24]  P. Bork,et al.  A side effect resource to capture phenotypic effects of drugs , 2010, Molecular systems biology.

[25]  Carol Friedman,et al.  Mining multi-item drug adverse effect associations in spontaneous reporting systems , 2010, BMC Bioinformatics.

[26]  C. Fletcher Drug Interactions Should Be Evaluated in Patients , 2010, Clinical pharmacology and therapeutics.

[27]  M. Pirmohamed Drug-drug interactions and adverse drug reactions: separating the wheat from the chaff , 2010, Wiener klinische Wochenschrift.

[28]  Pankaj Agarwal,et al.  Systematic Drug Repositioning Based on Clinical Side-Effects , 2011, PloS one.

[29]  Eric Bui,et al.  The importance of drug–drug interactions as a cause of adverse drug reactions: a pharmacovigilance study of serotoninergic reuptake inhibitors in France , 2012, European Journal of Clinical Pharmacology.

[30]  Susumu Goto,et al.  Network-Based Analysis and Characterization of Adverse Drug-Drug Interactions , 2011, J. Chem. Inf. Model..

[31]  O. Kuss,et al.  Treatments effects from randomized trials and propensity score analyses were similar in similar populations in an example from cardiac surgery. , 2011, Journal of clinical epidemiology.

[32]  Ben Y. Reis,et al.  Predicting Adverse Drug Events Using Pharmacological Network Models , 2011, Science Translational Medicine.

[33]  A. Persidis,et al.  Drug repurposing and adverse event prediction using high‐throughput literature analysis , 2011, Wiley interdisciplinary reviews. Systems biology and medicine.

[34]  R. Altman,et al.  Detecting Drug Interactions From Adverse‐Event Reports: Interaction Between Paroxetine and Pravastatin Increases Blood Glucose Levels , 2011, Clinical pharmacology and therapeutics.

[35]  Russ B. Altman,et al.  Discovery and Explanation of Drug-Drug Interactions via Text Mining , 2011, Pacific Symposium on Biocomputing.