1
|
Padhi B, Liu R, Yang Y, Peng X, Li L, Zhang P, Zhang P. Using multiple drug similarity networks to promote adverse drug event detection. Heliyon 2024; 10:e39728. [PMID: 39748955 PMCID: PMC11693886 DOI: 10.1016/j.heliyon.2024.e39728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 01/04/2025] Open
Abstract
The occurrence of an adverse drug event (ADE) has become a serious social concern of public health. Early detection of ADEs can lower the risk of drug safety as well as the expense of the drug. While post-market spontaneous reports of ADEs remain a cornerstone of pharmacovigilance, most existing signal detection algorithms rely on substantial accumulated data, limiting their applicability to early ADE detection when reports are scarce. To address this issue, we propose a label propagation model for generating enhanced drug safety signals using multiple drug features. We first construct multiple drug similarity networks using a range of drug features. We then calculate initial drug safety signals using conventional signal detection algorithms. These original signals are subsequently propagated across each drug similarity network to obtain enhanced drug safety signals. We evaluate our proposed model using two common signal detection algorithms on data from the FDA Adverse Event Reporting System (FAERS). Results demonstrate that enhanced drug safety signals with pre-clinical information outperform the standard safety signal detection algorithms on early ADE detection. In addition, we systematically evaluate the performance of different drug similarities against different types of ADEs. Furthermore, we have developed a web interface (http://drug-drug-sim.aimedlab.net/) to display our multiple drug similarity scores, facilitating access to this valuable resource for drug safety monitoring.
Collapse
Affiliation(s)
- Biswajit Padhi
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Ruoqi Liu
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Yuedi Yang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W. 10th Street HITS 3000, Indianapolis, IN 46202, USA
| | - Xueqiao Peng
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W. 10th Street HITS 3000, Indianapolis, IN 46202, USA
| | - Ping Zhang
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
- Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA
- Translational Data Analytics institute, The Ohio State University, 1760 Neil Ave, Columbus, OH 43210, USA
| |
Collapse
|
2
|
Long W, Li S, He Y, Lin J, Li M, Wen Z. Unraveling Structural Alerts in Marketed Drugs for Improving Adverse Outcome Pathway Framework of Drug-Induced QT Prolongation. Int J Mol Sci 2023; 24:ijms24076771. [PMID: 37047744 PMCID: PMC10095420 DOI: 10.3390/ijms24076771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
In pharmaceutical treatment, many non-cardiac drugs carry the risk of prolonging the QT interval, which can lead to fatal cardiac complications such as torsades de points (TdP). Although the unexpected blockade of ion channels has been widely considered to be one of the main reasons for affecting the repolarization phase of the cardiac action potential and leading to QT interval prolongation, the lack of knowledge regarding chemical structures in drugs that may induce the prolongation of the QT interval remains a barrier to further understanding the underlying mechanism and developing an effective prediction strategy. In this study, we thoroughly investigated the differences in chemical structures between QT-prolonging drugs and drugs with no drug-induced QT prolongation (DIQT) concerns, based on the Drug-Induced QT Prolongation Atlas (DIQTA) dataset. Three categories of structural alerts (SAs), namely amines, ethers, and aromatic compounds, appeared in large quantities in QT-prolonging drugs, but rarely in drugs with no DIQT concerns, indicating a close association between SAs and the risk of DIQT. Moreover, using the molecular descriptors associated with these three categories of SAs as features, the structure–activity relationship (SAR) model for predicting the high risk of inducing QT interval prolongation of marketed drugs achieved recall rates of 72.5% and 80.0% for the DIQTA dataset and the FDA Adverse Event Reporting System (FAERS) dataset, respectively. Our findings may promote a better understanding of the mechanism of DIQT and facilitate research on cardiac adverse drug reactions in drug development.
Collapse
Affiliation(s)
- Wulin Long
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shihai Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yujie He
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Jinzhu Lin
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu 610064, China
- Medical Big Data Center, Sichuan University, Chengdu 610064, China
| |
Collapse
|
3
|
Similarity measures for interval-valued fuzzy sets based on average embeddings and its application to hierarchical clustering. Inf Sci (N Y) 2022. [DOI: 10.1016/j.ins.2022.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4
|
Wei J, Feng G, Lu Z, Han P, Zhu Y, Huang W. Evaluating Drug Risk Using GAN and SMOTE Based on CFDA's Spontaneous Reporting Data. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6033860. [PMID: 34493954 PMCID: PMC8418931 DOI: 10.1155/2021/6033860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/19/2021] [Indexed: 11/17/2022]
Abstract
Adverse drug reactions (ADRs) pose health threats to humans. Therefore, the risk re-evaluation of post-marketing drugs has become an important part of the pharmacovigilance work of various countries. In China, drugs are mainly divided into three categories, from high-risk to low-risk drugs, namely, prescription drugs (Rx), over-the-counter drugs A (OTC-A), and over-the-counter drugs B (OTC-B). Until now, there has been a lack of automated evaluation methods for the three status switch of drugs. Based on China Food and Drug Administration's (CFDA) spontaneous reporting database (CSRD), we proposed a classification model to predict risk level of drugs by using feature enhancement based on Generative Adversarial Networks (GAN) and Synthetic Minority Over-Sampling Technique (SMOTE). A total of 985,960 spontaneous reports from 2011 to 2018 were selected from CSRD in Jiangsu Province as experimental data. After data preprocessing, a class-imbalance data set was obtained, which contained 887 Rx (accounting for 84.72%), 113 OTC-A (10.79%), and 47 OTC-B (4.49%). Taking drugs as the samples, ADRs as the features, and signal detection results obtained by proportional reporting ratio (PRR) method as the feature values, we constructed the original data matrix, where the last column represents the category label of each drug. Our proposed model expands the ADR data from both the sample space and the feature space. In terms of feature space, we use feature selection (FS) to screen ADR symptoms with higher importance scores. Then, we use GAN to generate artificial data, which are added to the feature space to achieve feature enhancement. In terms of sample space, we use SMOTE technology to expand the minority samples to balance three categories of drugs and minimize the classification deviation caused by the gap in the sample size. Finally, we use random forest (RF) algorithm to classify the feature-enhanced and balanced data set. The experimental results show that the accuracy of the proposed classification model reaches 98%. Our proposed model can well evaluate drug risk levels and provide automated methods for status switch of post-marketing drugs.
Collapse
Affiliation(s)
- Jianxiang Wei
- School of Management, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
- Key Research Base of Philosophy and Social Sciences in Jiangsu-Information Industry Integration Innovation and Emergency Management Research Center, Nanjing 210003, China
| | - Guanzhong Feng
- School of Internet of Things, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
| | - Zhiqiang Lu
- School of Internet of Things, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
| | - Pu Han
- School of Management, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
| | - Yunxia Zhu
- School of Internet of Things, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
| | - Weidong Huang
- School of Management, Nanjing University of Posts and Telecommunications, Nanjing 210003, China
- Key Research Base of Philosophy and Social Sciences in Jiangsu-Information Industry Integration Innovation and Emergency Management Research Center, Nanjing 210003, China
| |
Collapse
|
5
|
Zhou Y, Li S, Zhao Y, Guo M, Liu Y, Li M, Wen Z. Quantitative Structure-Activity Relationship (QSAR) Model for the Severity Prediction of Drug-Induced Rhabdomyolysis by Using Random Forest. Chem Res Toxicol 2021; 34:514-521. [PMID: 33393765 DOI: 10.1021/acs.chemrestox.0c00347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Drug-induced rhabdomyolysis (DIR) is a rare and potentially life-threatening muscle injury that is characterized by low incidence and high risk. To our best knowledge, the performance of the current predictive models for the early detection of DIR is suboptimal because of the scarcity and dispersion of DIR cases. Therefore, on the basis of the curated drug information from the Drug-Induced Rhabdomyolysis Atlas (DIRA) database, we proposed a random forest (RF) model to predict the DIR severity of the marketed drugs. Compared with the state-of-art methods, our proposed model outperformed extreme gradient boosting, support vector machine, and logistic regression in distinguishing the Most-DIR concern drugs from the No-DIR concern drugs (Matthews correlation coefficient (MCC) and recall rate of our model were 0.46 and 0.81, respectively). Our model was subsequently applied to predicting the potentially serious DIR for 1402 drugs, which were reported to cause DIR by the postmarketing DIR surveillance data in the FDA Spontaneous Adverse Events Reporting System (FAERS). As a result, 62.7% (94) of drugs ranked in the top 150 drugs with the Most-DIR concerns in FAERS can be identified by our model. The top four drugs (odds ratio >30) including acepromazine, rapacuronium, oxyphenbutazone, and naringenin were correctly predicted by our model. In conclusion, the RF model can well predict the Most-DIR concern drug only based on the chemical structure information and can be a facilitated tool for early DIR detection.
Collapse
Affiliation(s)
- Yifan Zhou
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Shihai Li
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yiru Zhao
- College of Computer Science, Sichuan University, Chengdu, Sichuan 610064, China
| | - Mingkun Guo
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yuan Liu
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China.,Medical Big Data Center, Sichuan University, Chengdu, Sichuan 610064, China
| |
Collapse
|
6
|
Li Y, Jimeno Yepes A, Xiao C. Combining Social Media and FDA Adverse Event Reporting System to Detect Adverse Drug Reactions. Drug Saf 2020; 43:893-903. [PMID: 32385840 PMCID: PMC7434724 DOI: 10.1007/s40264-020-00943-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Adverse drug reactions (ADRs) are unintended reactions caused by a drug or combination of drugs taken by a patient. The current safety surveillance system relies on spontaneous reporting systems (SRSs) and more recently on observational health data; however, ADR detection may be delayed and lack geographic diversity. The broad scope of social media conversations, such as those on Twitter, can include health-related topics. Consequently, these data could be used to detect potentially novel ADRs with less latency. Although research regarding ADR detection using social media has made progress, findings are based on single information sources, and no study has yet integrated drug safety evidence from both an SRS and Twitter. OBJECTIVE The aim of this study was to combine signals from an SRS and Twitter to facilitate the detection of safety signals and compare the performance of the combined system with signals generated by individual data sources. METHODS We extracted potential drug-ADR posts from Twitter, used Monte Carlo expectation maximization to generate drug safety signals from both the US FDA Adverse Event Reporting System and posts from Twitter, and then integrated these signals using a Bayesian hierarchical model. The results from the integrated system and two individual sources were evaluated using a reference standard derived from drug labels. Area under the receiver operating characteristics curve (AUC) was computed to measure performance. RESULTS We observed a significant improvement in the AUC of the combined system when comparing it with Twitter alone, and no improvement when comparing with the SRS alone. The AUCs ranged from 0.587 to 0.637 for the combined SRS and Twitter, from 0.525 to 0.534 for Twitter alone, and from 0.612 to 0.642 for the SRS alone. The results varied because different preprocessing procedures were applied to Twitter. CONCLUSION The accuracy of signal detection using social media can be improved by combining signals with those from SRSs. However, the combined system cannot achieve better AUC performance than data from FAERS alone, which may indicate that Twitter data are not ready to be integrated into a purely data-driven combination system.
Collapse
Affiliation(s)
- Ying Li
- Center for Computational Health, IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd, Yorktown Heights, NY, 10598, USA.
| | | | - Cao Xiao
- Analytics Center of Excellence, IQVIA, Cambridge, MA, USA
| |
Collapse
|
7
|
Dandala B, Joopudi V, Tsou CH, Liang JJ, Suryanarayanan P. Extraction of Information Related to Drug Safety Surveillance From Electronic Health Record Notes: Joint Modeling of Entities and Relations Using Knowledge-Aware Neural Attentive Models. JMIR Med Inform 2020; 8:e18417. [PMID: 32459650 PMCID: PMC7382020 DOI: 10.2196/18417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND An adverse drug event (ADE) is commonly defined as "an injury resulting from medical intervention related to a drug." Providing information related to ADEs and alerting caregivers at the point of care can reduce the risk of prescription and diagnostic errors and improve health outcomes. ADEs captured in structured data in electronic health records (EHRs) as either coded problems or allergies are often incomplete, leading to underreporting. Therefore, it is important to develop capabilities to process unstructured EHR data in the form of clinical notes, which contain a richer documentation of a patient's ADE. Several natural language processing (NLP) systems have been proposed to automatically extract information related to ADEs. However, the results from these systems showed that significant improvement is still required for the automatic extraction of ADEs from clinical notes. OBJECTIVE This study aims to improve the automatic extraction of ADEs and related information such as drugs, their attributes, and reason for administration from the clinical notes of patients. METHODS This research was conducted using discharge summaries from the Medical Information Mart for Intensive Care III (MIMIC-III) database obtained through the 2018 National NLP Clinical Challenges (n2c2) annotated with drugs, drug attributes (ie, strength, form, frequency, route, dosage, duration), ADEs, reasons, and relations between drugs and other entities. We developed a deep learning-based system for extracting these drug-centric concepts and relations simultaneously using a joint method enhanced with contextualized embeddings, a position-attention mechanism, and knowledge representations. The joint method generated different sentence representations for each drug, which were then used to extract related concepts and relations simultaneously. Contextualized representations trained on the MIMIC-III database were used to capture context-sensitive meanings of words. The position-attention mechanism amplified the benefits of the joint method by generating sentence representations that capture long-distance relations. Knowledge representations were obtained from graph embeddings created using the US Food and Drug Administration Adverse Event Reporting System database to improve relation extraction, especially when contextual clues were insufficient. RESULTS Our system achieved new state-of-the-art results on the n2c2 data set, with significant improvements in recognizing crucial drug-reason (F1=0.650 versus F1=0.579) and drug-ADE (F1=0.490 versus F1=0.476) relations. CONCLUSIONS This study presents a system for extracting drug-centric concepts and relations that outperformed current state-of-the-art results and shows that contextualized embeddings, position-attention mechanisms, and knowledge graph embeddings effectively improve deep learning-based concepts and relation extraction. This study demonstrates the potential for deep learning-based methods to help extract real-world evidence from unstructured patient data for drug safety surveillance.
Collapse
|
8
|
Daluwatte C, Schotland P, Strauss DG, Burkhart KK, Racz R. Predicting potential adverse events using safety data from marketed drugs. BMC Bioinformatics 2020; 21:163. [PMID: 32349656 PMCID: PMC7191698 DOI: 10.1186/s12859-020-3509-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/22/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND While clinical trials are considered the gold standard for detecting adverse events, often these trials are not sufficiently powered to detect difficult to observe adverse events. We developed a preliminary approach to predict 135 adverse events using post-market safety data from marketed drugs. Adverse event information available from FDA product labels and scientific literature for drugs that have the same activity at one or more of the same targets, structural and target similarities, and the duration of post market experience were used as features for a classifier algorithm. The proposed method was studied using 54 drugs and a probabilistic approach of performance evaluation using bootstrapping with 10,000 iterations. RESULTS Out of 135 adverse events, 53 had high probability of having high positive predictive value. Cross validation showed that 32% of the model-predicted safety label changes occurred within four to nine years of approval (median: six years). CONCLUSIONS This approach predicts 53 serious adverse events with high positive predictive values where well-characterized target-event relationships exist. Adverse events with well-defined target-event associations were better predicted compared to adverse events that may be idiosyncratic or related to secondary target effects that were poorly captured. Further enhancement of this model with additional features, such as target prediction and drug binding data, may increase accuracy.
Collapse
Affiliation(s)
- Chathuri Daluwatte
- Division of Applied Regulatory Science, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993 USA
| | - Peter Schotland
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD USA
| | - David G. Strauss
- Division of Applied Regulatory Science, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993 USA
| | - Keith K. Burkhart
- Division of Applied Regulatory Science, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993 USA
| | - Rebecca Racz
- Division of Applied Regulatory Science, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993 USA
| |
Collapse
|
9
|
Liu R, Zhang P. Towards early detection of adverse drug reactions: combining pre-clinical drug structures and post-market safety reports. BMC Med Inform Decis Mak 2019; 19:279. [PMID: 31849321 PMCID: PMC6918608 DOI: 10.1186/s12911-019-0999-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 12/04/2019] [Indexed: 01/10/2023] Open
Abstract
Background Adverse drug reaction (ADR) is a major burden for patients and healthcare industry. Early and accurate detection of potential ADRs can help to improve drug safety and reduce financial costs. Post-market spontaneous reports of ADRs remain a cornerstone of pharmacovigilance and a series of drug safety signal detection methods play an important role in providing drug safety insights. However, existing methods require sufficient case reports to generate signals, limiting their usages for newly approved drugs with few (or even no) reports. Methods In this study, we propose a label propagation framework to enhance drug safety signals by combining drug chemical structures with FDA Adverse Event Reporting System (FAERS). First, we compute original drug safety signals via common signal detection algorithms. Then, we construct a drug similarity network based on chemical structures. Finally, we generate enhanced drug safety signals by propagating original signals on the drug similarity network. Our proposed framework enriches post-market safety reports with pre-clinical drug similarity network, effectively alleviating issues of insufficient cases for newly approved drugs. Results We apply the label propagation framework to four popular signal detection algorithms (PRR, ROR, MGPS, BCPNN) and find that our proposed framework generates more accurate drug safety signals than the corresponding baselines. In addition, our framework identifies potential ADRs for newly approved drugs, thus paving the way for early detection of ADRs. Conclusions The proposed label propagation framework combines pre-clinical drug structures with post-market safety reports, generates enhanced drug safety signals, and can potentially help to accurately detect ADRs ahead of time. Availability The source code for this paper is available at: https://github.com/ruoqi-liu/LP-SDA.
Collapse
Affiliation(s)
- Ruoqi Liu
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, 43210, Ohio, USA
| | - Ping Zhang
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, 43210, Ohio, USA. .,Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Drive, Columbus, 43210, Ohio, USA.
| |
Collapse
|
10
|
Cui X, Liu J, Zhang J, Wu Q, Li X. In silico prediction of drug‐induced rhabdomyolysis with machine‐learning models and structural alerts. J Appl Toxicol 2019; 39:1224-1232. [DOI: 10.1002/jat.3808] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Xueyan Cui
- Department of Pharmacy, Shandong Provincial Qianfoshan HospitalShandong University Jinan China
| | - Juan Liu
- Department of Pharmacy, Shandong Provincial Qianfoshan HospitalShandong University Jinan China
| | - Jinfeng Zhang
- Department of Pharmacy, Shandong Provincial Qianfoshan HospitalShandong University Jinan China
| | - Qiuyun Wu
- Department of Pharmacy, Shandong Provincial Qianfoshan HospitalShandong University Jinan China
| | - Xiao Li
- Department of Pharmacy, Shandong Provincial Qianfoshan HospitalShandong University Jinan China
| |
Collapse
|
11
|
Xie J, Liu X, Dajun Zeng D. Mining e-cigarette adverse events in social media using Bi-LSTM recurrent neural network with word embedding representation. J Am Med Inform Assoc 2019; 25:72-80. [PMID: 28505280 DOI: 10.1093/jamia/ocx045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 04/11/2017] [Indexed: 02/04/2023] Open
Abstract
Objective Recent years have seen increased worldwide popularity of e-cigarette use. However, the risks of e-cigarettes are underexamined. Most e-cigarette adverse event studies have achieved low detection rates due to limited subject sample sizes in the experiments and surveys. Social media provides a large data repository of consumers' e-cigarette feedback and experiences, which are useful for e-cigarette safety surveillance. However, it is difficult to automatically interpret the informal and nontechnical consumer vocabulary about e-cigarettes in social media. This issue hinders the use of social media content for e-cigarette safety surveillance. Recent developments in deep neural network methods have shown promise for named entity extraction from noisy text. Motivated by these observations, we aimed to design a deep neural network approach to extract e-cigarette safety information in social media. Methods Our deep neural language model utilizes word embedding as the representation of text input and recognizes named entity types with the state-of-the-art Bidirectional Long Short-Term Memory (Bi-LSTM) Recurrent Neural Network. Results Our Bi-LSTM model achieved the best performance compared to 3 baseline models, with a precision of 94.10%, a recall of 91.80%, and an F-measure of 92.94%. We identified 1591 unique adverse events and 9930 unique e-cigarette components (ie, chemicals, flavors, and devices) from our research testbed. Conclusion Although the conditional random field baseline model had slightly better precision than our approach, our Bi-LSTM model achieved much higher recall, resulting in the best F-measure. Our method can be generalized to extract medical concepts from social media for other medical applications.
Collapse
Affiliation(s)
- Jiaheng Xie
- Department of Management Information Systems, University of Arizona, Tucson, AZ, USA
| | - Xiao Liu
- Department of Operation and Information Systems, University of Utah, Salt Lake City, UT, USA
| | - Daniel Dajun Zeng
- Department of Management Information Systems, University of Arizona, Tucson, AZ, USA.,State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Wen Z, Liang Y, Hao Y, Delavan B, Huang R, Mikailov M, Tong W, Li M, Liu Z. Drug-Induced Rhabdomyolysis Atlas (DIRA) for idiosyncratic adverse drug reaction management. Drug Discov Today 2019; 24:9-15. [PMID: 29902520 PMCID: PMC7050640 DOI: 10.1016/j.drudis.2018.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/23/2018] [Accepted: 06/06/2018] [Indexed: 01/06/2023]
Abstract
Drug-induced rhabdomyolysis (DIR) is an idiosyncratic and fatal adverse drug reaction (ADR) characterized in severe muscle injuries accompanied by multiple-organ failure. Limited knowledge regarding the pathophysiology of rhabdomyolysis is the main obstacle to developing early biomarkers and prevention strategies. Given the lack of a centralized data resource to curate, organize, and standardize widespread DIR information, here we present a Drug-Induced Rhabdomyolysis Atlas (DIRA) that provides DIR-related information, including: a classification scheme for DIR based on drug labeling information; postmarketing surveillance data of DIR; and DIR drug property information. To elucidate the utility of DIRA, we used precision dosing, concomitant use of DIR drugs, and predictive modeling development to exemplify strategies for idiosyncratic ADR (IADR) management.
Collapse
Affiliation(s)
- Zhining Wen
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yu Liang
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yingyi Hao
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Brian Delavan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72204, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Mike Mikailov
- Office of Science and Engineering Labs, Center for Devices and Radiological Health, US Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD 20993, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China.
| | - Zhichao Liu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
13
|
Vilar S, Friedman C, Hripcsak G. Detection of drug-drug interactions through data mining studies using clinical sources, scientific literature and social media. Brief Bioinform 2018; 19:863-877. [PMID: 28334070 PMCID: PMC6454455 DOI: 10.1093/bib/bbx010] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/28/2016] [Indexed: 11/13/2022] Open
Abstract
Drug-drug interactions (DDIs) constitute an important concern in drug development and postmarketing pharmacovigilance. They are considered the cause of many adverse drug effects exposing patients to higher risks and increasing public health system costs. Methods to follow-up and discover possible DDIs causing harm to the population are a primary aim of drug safety researchers. Here, we review different methodologies and recent advances using data mining to detect DDIs with impact on patients. We focus on data mining of different pharmacovigilance sources, such as the US Food and Drug Administration Adverse Event Reporting System and electronic health records from medical institutions, as well as on the diverse data mining studies that use narrative text available in the scientific biomedical literature and social media. We pay attention to the strengths but also further explain challenges related to these methods. Data mining has important applications in the analysis of DDIs showing the impact of the interactions as a cause of adverse effects, extracting interactions to create knowledge data sets and gold standards and in the discovery of novel and dangerous DDIs.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University, New York, USA
- Department of Organic Chemistry, University of Santiago de Compostela, Spain
| | - Carol Friedman
- Department of Biomedical Informatics, Columbia University, New York, USA
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, USA
| |
Collapse
|
14
|
Harpaz R, DuMouchel W, Schuemie M, Bodenreider O, Friedman C, Horvitz E, Ripple A, Sorbello A, White RW, Winnenburg R, Shah NH. Toward multimodal signal detection of adverse drug reactions. J Biomed Inform 2017; 76:41-49. [PMID: 29081385 PMCID: PMC8502488 DOI: 10.1016/j.jbi.2017.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 10/14/2017] [Accepted: 10/24/2017] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Improving mechanisms to detect adverse drug reactions (ADRs) is key to strengthening post-marketing drug safety surveillance. Signal detection is presently unimodal, relying on a single information source. Multimodal signal detection is based on jointly analyzing multiple information sources. Building on, and expanding the work done in prior studies, the aim of the article is to further research on multimodal signal detection, explore its potential benefits, and propose methods for its construction and evaluation. MATERIAL AND METHODS Four data sources are investigated; FDA's adverse event reporting system, insurance claims, the MEDLINE citation database, and the logs of major Web search engines. Published methods are used to generate and combine signals from each data source. Two distinct reference benchmarks corresponding to well-established and recently labeled ADRs respectively are used to evaluate the performance of multimodal signal detection in terms of area under the ROC curve (AUC) and lead-time-to-detection, with the latter relative to labeling revision dates. RESULTS Limited to our reference benchmarks, multimodal signal detection provides AUC improvements ranging from 0.04 to 0.09 based on a widely used evaluation benchmark, and a comparative added lead-time of 7-22 months relative to labeling revision dates from a time-indexed benchmark. CONCLUSIONS The results support the notion that utilizing and jointly analyzing multiple data sources may lead to improved signal detection. Given certain data and benchmark limitations, the early stage of development, and the complexity of ADRs, it is currently not possible to make definitive statements about the ultimate utility of the concept. Continued development of multimodal signal detection requires a deeper understanding the data sources used, additional benchmarks, and further research on methods to generate and synthesize signals.
Collapse
Affiliation(s)
- Rave Harpaz
- Oracle Health Sciences, Bedford, MA, United States.
| | | | | | | | | | | | - Anna Ripple
- National Library of Medicine, NIH, Bethesda, MD, United States
| | | | | | | | - Nigam H Shah
- Stanford University, Stanford, CA, United States
| |
Collapse
|
15
|
Pardhi T, Vasu K. Identification of dual kinase inhibitors of CK2 and GSK3β: combined qualitative and quantitative pharmacophore modeling approach. J Biomol Struct Dyn 2017; 36:177-194. [DOI: 10.1080/07391102.2016.1270856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Triveni Pardhi
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, C/O B.V. Patel PERD Centre, SG Highway, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Kamala Vasu
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, C/O B.V. Patel PERD Centre, SG Highway, Thaltej, Ahmedabad 380054, Gujarat, India
- Department of Medicinal Chemistry, B. V. Patel Pharmaceutical Education & Research Development (PERD) Centre, Ahmedabad 380054, Gujarat, India
| |
Collapse
|
16
|
Harpaz R, Odgers D, Gaskin G, DuMouchel W, Winnenburg R, Bodenreider O, Ripple A, Szarfman A, Sorbello A, Horvitz E, White RW, Shah NH. A time-indexed reference standard of adverse drug reactions. Sci Data 2016; 1:140043. [PMID: 25632348 PMCID: PMC4306188 DOI: 10.1038/sdata.2014.43] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Undetected adverse drug reactions (ADRs) pose a major burden on the health system. Data mining methodologies designed to identify signals of novel ADRs are of deep importance for drug safety surveillance. The development and evaluation of these methodologies requires proper reference benchmarks. While progress has recently been made in developing such benchmarks, our understanding of the performance characteristics of the data mining methodologies is limited because existing benchmarks do not support prospective performance evaluations. We address this shortcoming by providing a reference standard to support prospective performance evaluations. The reference standard was systematically curated from drug labeling revisions, such as new warnings, which were issued and communicated by the US Food and Drug Administration in 2013. The reference standard includes 62 positive test cases and 75 negative controls, and covers 44 drugs and 38 events. We provide usage guidance and empirical support for the reference standard by applying it to analyze two data sources commonly mined for drug safety surveillance.
Collapse
Affiliation(s)
- Rave Harpaz
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| | - David Odgers
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| | - Greg Gaskin
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| | | | | | | | - Anna Ripple
- National Library of Medicine, NIH, Bethesda, Maryland 20894, USA
| | | | | | - Eric Horvitz
- Microsoft Research, Redmond, Washington 98052, USA
| | - Ryen W White
- Microsoft Research, Redmond, Washington 98052, USA
| | - Nigam H Shah
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
17
|
Banda JM, Evans L, Vanguri RS, Tatonetti NP, Ryan PB, Shah NH. A curated and standardized adverse drug event resource to accelerate drug safety research. Sci Data 2016; 3:160026. [PMID: 27193236 PMCID: PMC4872271 DOI: 10.1038/sdata.2016.26] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/24/2016] [Indexed: 11/08/2022] Open
Abstract
Identification of adverse drug reactions (ADRs) during the post-marketing phase is one of the most important goals of drug safety surveillance. Spontaneous reporting systems (SRS) data, which are the mainstay of traditional drug safety surveillance, are used for hypothesis generation and to validate the newer approaches. The publicly available US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) data requires substantial curation before they can be used appropriately, and applying different strategies for data cleaning and normalization can have material impact on analysis results. We provide a curated and standardized version of FAERS removing duplicate case records, applying standardized vocabularies with drug names mapped to RxNorm concepts and outcomes mapped to SNOMED-CT concepts, and pre-computed summary statistics about drug-outcome relationships for general consumption. This publicly available resource, along with the source code, will accelerate drug safety research by reducing the amount of time spent performing data management on the source FAERS reports, improving the quality of the underlying data, and enabling standardized analyses using common vocabularies.
Collapse
Affiliation(s)
- Juan M. Banda
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| | - Lee Evans
- LTS Computing LLC, West Chester, Pennsylvania 19380, USA
| | - Rami S. Vanguri
- Department of Biomedical Informatics, Columbia University, New York, New York 10032, USA
| | - Nicholas P. Tatonetti
- Department of Biomedical Informatics, Columbia University, New York, New York 10032, USA
| | - Patrick B. Ryan
- Janssen Research & Development, LLC, Titusville, New Jersey 08869, USA
| | - Nigam H. Shah
- Center for Biomedical Informatics Research, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
18
|
White RW, Wang S, Pant A, Harpaz R, Shukla P, Sun W, DuMouchel W, Horvitz E. Early identification of adverse drug reactions from search log data. J Biomed Inform 2016; 59:42-8. [DOI: 10.1016/j.jbi.2015.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/07/2015] [Accepted: 11/12/2015] [Indexed: 01/28/2023]
|
19
|
Ho SS, McLachlan AJ, Chen TF, Hibbs DE, Fois RA. Relationships Between Pharmacovigilance, Molecular, Structural, and Pathway Data: Revealing Mechanisms for Immune-Mediated Drug-Induced Liver Injury. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:426-41. [PMID: 26312166 PMCID: PMC4544056 DOI: 10.1002/psp4.56] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/08/2015] [Indexed: 11/18/2022]
Abstract
Immune-mediated drug-induced liver injury (IMDILI) can be devastating, irreversible, and fatal in the absence of successful transplantation surgery. We present a novel approach that combines the methods of pharmacoepidemiology with in silico molecular modeling to identify specific features in toxic ligands that are associated with clinical features of IMDILI. Specifically, from pharmacovigilance data multivariate logistic regression identified 18 drugs associated with IMDILI (P < 0.00015). Eleven of these drugs, along with their known and proposed metabolites, constituted a training set used to develop a four-point pharmacophore model (sensitivity 75%; specificity 85%). Subsequently, this information was combined with information from immune-pathway reviews and genetic-association studies and complemented with ligand-protein docking simulations to support a hypothesis implicating two putative targets within separate, possibly interacting, immune-system pathways: the major histocompatibility complex within the adaptive immune system and Toll-like receptors (TLRs), in particular TLR-7, which represent pattern recognition receptors of the innate immune system.
Collapse
Affiliation(s)
- S S Ho
- Faculty of Pharmacy (A15), University of Sydney Sydney, NSW, Australia
| | - A J McLachlan
- Faculty of Pharmacy (A15), University of Sydney Sydney, NSW, Australia
| | - T F Chen
- Faculty of Pharmacy (A15), University of Sydney Sydney, NSW, Australia
| | - D E Hibbs
- Faculty of Pharmacy (A15), University of Sydney Sydney, NSW, Australia
| | - R A Fois
- Faculty of Pharmacy (A15), University of Sydney Sydney, NSW, Australia
| |
Collapse
|
20
|
Vilar S, Lorberbaum T, Hripcsak G, Tatonetti NP. Improving Detection of Arrhythmia Drug-Drug Interactions in Pharmacovigilance Data through the Implementation of Similarity-Based Modeling. PLoS One 2015; 10:e0129974. [PMID: 26068584 PMCID: PMC4466327 DOI: 10.1371/journal.pone.0129974] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
Identification of Drug-Drug Interactions (DDIs) is a significant challenge during drug development and clinical practice. DDIs are responsible for many adverse drug effects (ADEs), decreasing patient quality of life and causing higher care expenses. DDIs are not systematically evaluated in pre-clinical or clinical trials and so the FDA U. S. Food and Drug Administration relies on post-marketing surveillance to monitor patient safety. However, existing pharmacovigilance algorithms show poor performance for detecting DDIs exhibiting prohibitively high false positive rates. Alternatively, methods based on chemical structure and pharmacological similarity have shown promise in adverse drug event detection. We hypothesize that the use of chemical biology data in a post hoc analysis of pharmacovigilance results will significantly improve the detection of dangerous interactions. Our model integrates a reference standard of DDIs known to cause arrhythmias with drug similarity data. To compare similarity between drugs we used chemical structure (both 2D and 3D molecular structure), adverse drug side effects, chemogenomic targets, drug indication classes, and known drug-drug interactions. We evaluated the method on external reference standards. Our results showed an enhancement of sensitivity, specificity and precision in different top positions with the use of similarity measures to rank the candidates extracted from pharmacovigilance data. For the top 100 DDI candidates, similarity-based modeling yielded close to twofold precision enhancement compared to the proportional reporting ratio (PRR). Moreover, the method helps in the DDI decision making through the identification of the DDI in the reference standard that generated the candidate.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
- Department of Systems Biology, Columbia University, New York, NY, United States of America
- Observational Health Data Sciences and Informatics (OHDSI), New York, NY, United States of America
- * E-mail:
| | - Tal Lorberbaum
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
- Department of Systems Biology, Columbia University, New York, NY, United States of America
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
- Observational Health Data Sciences and Informatics (OHDSI), New York, NY, United States of America
| | - Nicholas P. Tatonetti
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
- Department of Systems Biology, Columbia University, New York, NY, United States of America
- Observational Health Data Sciences and Informatics (OHDSI), New York, NY, United States of America
- Department of Medicine, Columbia University, New York, NY, United States of America
| |
Collapse
|
21
|
3D pharmacophoric similarity improves multi adverse drug event identification in pharmacovigilance. Sci Rep 2015; 5:8809. [PMID: 25744369 PMCID: PMC4351525 DOI: 10.1038/srep08809] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/30/2015] [Indexed: 11/08/2022] Open
Abstract
Adverse drugs events (ADEs) detection constitutes a considerable concern in patient safety and public health care. For this reason, it is important to develop methods that improve ADE signal detection in pharmacovigilance databases. Our objective is to apply 3D pharmacophoric similarity models to enhance ADE recognition in Offsides, a pharmacovigilance resource with drug-ADE associations extracted from the FDA Adverse Event Reporting System (FAERS). We developed a multi-ADE predictor implementing 3D drug similarity based on a pharmacophoric approach, with an ADE reference standard extracted from the SIDER database. The results showed that the application of our 3D multi-type ADE predictor to the pharmacovigilance data in Offsides improved ADE identification and generated enriched sets of drug-ADE signals. The global ROC curve for the Offsides ADE candidates ranked with the 3D similarity score showed an area of 0.7. The 3D predictor also allows the identification of the most similar drug that causes the ADE under study, which could provide hypotheses about mechanisms of action and ADE etiology. Our method is useful in drug development, screening potential adverse effects in experimental drugs, and in drug safety, applicable to the evaluation of ADE signals selected through pharmacovigilance data mining.
Collapse
|
22
|
Sarker A, Gonzalez G. Portable automatic text classification for adverse drug reaction detection via multi-corpus training. J Biomed Inform 2014; 53:196-207. [PMID: 25451103 DOI: 10.1016/j.jbi.2014.11.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/24/2014] [Accepted: 11/02/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Automatic detection of adverse drug reaction (ADR) mentions from text has recently received significant interest in pharmacovigilance research. Current research focuses on various sources of text-based information, including social media-where enormous amounts of user posted data is available, which have the potential for use in pharmacovigilance if collected and filtered accurately. The aims of this study are: (i) to explore natural language processing (NLP) approaches for generating useful features from text, and utilizing them in optimized machine learning algorithms for automatic classification of ADR assertive text segments; (ii) to present two data sets that we prepared for the task of ADR detection from user posted internet data; and (iii) to investigate if combining training data from distinct corpora can improve automatic classification accuracies. METHODS One of our three data sets contains annotated sentences from clinical reports, and the two other data sets, built in-house, consist of annotated posts from social media. Our text classification approach relies on generating a large set of features, representing semantic properties (e.g., sentiment, polarity, and topic), from short text nuggets. Importantly, using our expanded feature sets, we combine training data from different corpora in attempts to boost classification accuracies. RESULTS Our feature-rich classification approach performs significantly better than previously published approaches with ADR class F-scores of 0.812 (previously reported best: 0.770), 0.538 and 0.678 for the three data sets. Combining training data from multiple compatible corpora further improves the ADR F-scores for the in-house data sets to 0.597 (improvement of 5.9 units) and 0.704 (improvement of 2.6 units) respectively. CONCLUSIONS Our research results indicate that using advanced NLP techniques for generating information rich features from text can significantly improve classification accuracies over existing benchmarks. Our experiments illustrate the benefits of incorporating various semantic features such as topics, concepts, sentiments, and polarities. Finally, we show that integration of information from compatible corpora can significantly improve classification performance. This form of multi-corpus training may be particularly useful in cases where data sets are heavily imbalanced (e.g., social media data), and may reduce the time and costs associated with the annotation of data in the future.
Collapse
Affiliation(s)
- Abeed Sarker
- Department of Biomedical Informatics, Arizona State University, 13212 East Shea Blvd., Scottsdale, AZ 85259, USA.
| | - Graciela Gonzalez
- Department of Biomedical Informatics, Arizona State University, 13212 East Shea Blvd., Scottsdale, AZ 85259, USA.
| |
Collapse
|
23
|
Vilar S, Ryan PB, Madigan D, Stang PE, Schuemie MJ, Friedman C, Tatonetti NP, Hripcsak G. Similarity-based modeling applied to signal detection in pharmacovigilance. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e137. [PMID: 25250527 PMCID: PMC4211266 DOI: 10.1038/psp.2014.35] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/06/2014] [Indexed: 12/31/2022]
Abstract
One of the main objectives in pharmacovigilance is the detection of adverse drug events (ADEs) through mining of healthcare databases, such as electronic health records or administrative claims data. Although different approaches have been shown to be of great value, research is still focusing on the enhancement of signal detection to gain efficiency in further assessment and follow-up. We applied similarity-based modeling techniques, using 2D and 3D molecular structure, ADE, target, and ATC (anatomical therapeutic chemical) similarity measures, to the candidate associations selected previously in a medication-wide association study for four ADE outcomes. Our results showed an improvement in the precision when we ranked the subset of ADE candidates using similarity scorings. This method is simple, useful to strengthen or prioritize signals generated from healthcare databases, and facilitates ADE detection through the identification of the most similar drugs for which ADE information is available.
Collapse
Affiliation(s)
- S Vilar
- 1] Department of Biomedical Informatics, Columbia University, New York, New York, USA [2] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA
| | - P B Ryan
- 1] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA [2] Janssen Research and Development, Titusville, New Jersey, USA
| | - D Madigan
- 1] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA [2] Department of Statistics, Columbia University, New York, New York, USA
| | - P E Stang
- 1] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA [2] Janssen Research and Development, Titusville, New Jersey, USA
| | - M J Schuemie
- 1] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA [2] Janssen Research and Development, Titusville, New Jersey, USA
| | - C Friedman
- 1] Department of Biomedical Informatics, Columbia University, New York, New York, USA [2] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA
| | - N P Tatonetti
- 1] Department of Biomedical Informatics, Columbia University, New York, New York, USA [2] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA [3] Department of Systems Biology, Columbia University Medical Center, New York, New York, USA [4] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - G Hripcsak
- 1] Department of Biomedical Informatics, Columbia University, New York, New York, USA [2] Observational Health Data Sciences and Informatics (OHDSI), New York, New York, USA
| |
Collapse
|
24
|
Vilar S, Uriarte E, Santana L, Lorberbaum T, Hripcsak G, Friedman C, Tatonetti NP. Similarity-based modeling in large-scale prediction of drug-drug interactions. Nat Protoc 2014; 9:2147-63. [PMID: 25122524 PMCID: PMC4422192 DOI: 10.1038/nprot.2014.151] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Drug-drug interactions (DDIs) are a major cause of adverse drug effects and a public health concern, as they increase hospital care expenses and reduce patients' quality of life. DDI detection is, therefore, an important objective in patient safety, one whose pursuit affects drug development and pharmacovigilance. In this article, we describe a protocol applicable on a large scale to predict novel DDIs based on similarity of drug interaction candidates to drugs involved in established DDIs. The method integrates a reference standard database of known DDIs with drug similarity information extracted from different sources, such as 2D and 3D molecular structure, interaction profile, target and side-effect similarities. The method is interpretable in that it generates drug interaction candidates that are traceable to pharmacological or clinical effects. We describe a protocol with applications in patient safety and preclinical toxicity screening. The time frame to implement this protocol is 5-7 h, with additional time potentially necessary, depending on the complexity of the reference standard DDI database and the similarity measures implemented.
Collapse
Affiliation(s)
- Santiago Vilar
- 1] Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA. [2] Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Eugenio Uriarte
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Lourdes Santana
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tal Lorberbaum
- 1] Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA. [2] Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, New York, USA. [3] Department of Systems Biology, Columbia University Medical Center, New York, New York, USA
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA
| | - Carol Friedman
- Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA
| | - Nicholas P Tatonetti
- 1] Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA. [2] Department of Systems Biology, Columbia University Medical Center, New York, New York, USA. [3] Department of Medicine, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
25
|
Wang L, Jiang G, Li D, Liu H. Standardizing adverse drug event reporting data. J Biomed Semantics 2014; 5:36. [PMID: 25157320 PMCID: PMC4142531 DOI: 10.1186/2041-1480-5-36] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/23/2014] [Indexed: 11/16/2022] Open
Abstract
Background The Adverse Event Reporting System (AERS) is an FDA database providing rich information on voluntary reports of adverse drug events (ADEs). Normalizing data in the AERS would improve the mining capacity of the AERS for drug safety signal detection and promote semantic interoperability between the AERS and other data sources. In this study, we normalize the AERS and build a publicly available normalized ADE data source. The drug information in the AERS is normalized to RxNorm, a standard terminology source for medication, using a natural language processing medication extraction tool, MedEx. Drug class information is then obtained from the National Drug File-Reference Terminology (NDF-RT) using a greedy algorithm. Adverse events are aggregated through mapping with the Preferred Term (PT) and System Organ Class (SOC) codes of Medical Dictionary for Regulatory Activities (MedDRA). The performance of MedEx-based annotation was evaluated and case studies were performed to demonstrate the usefulness of our approaches. Results Our study yields an aggregated knowledge-enhanced AERS data mining set (AERS-DM). In total, the AERS-DM contains 37,029,228 Drug-ADE records. Seventy-one percent (10,221/14,490) of normalized drug concepts in the AERS were classified to 9 classes in NDF-RT. The number of unique pairs is 4,639,613 between RxNorm concepts and MedDRA Preferred Term (PT) codes and 205,725 between RxNorm concepts and SOC codes after ADE aggregation. Conclusions We have built an open-source Drug-ADE knowledge resource with data being normalized and aggregated using standard biomedical ontologies. The data resource has the potential to assist the mining of ADE from AERS for the data mining research community.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Medical Informatics, School of Public Health, Jilin University, Jilin, China ; Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Guoqian Jiang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Dingcheng Li
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Hongfang Liu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Polepalli Ramesh B, Belknap SM, Li Z, Frid N, West DP, Yu H. Automatically Recognizing Medication and Adverse Event Information From Food and Drug Administration's Adverse Event Reporting System Narratives. JMIR Med Inform 2014; 2:e10. [PMID: 25600332 PMCID: PMC4288072 DOI: 10.2196/medinform.3022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/10/2013] [Accepted: 12/10/2013] [Indexed: 12/14/2022] Open
Abstract
Background The Food and Drug Administration’s (FDA) Adverse Event Reporting System (FAERS) is a repository of spontaneously-reported adverse drug events (ADEs) for FDA-approved prescription drugs. FAERS reports include both structured reports and unstructured narratives. The narratives often include essential information for evaluation of the severity, causality, and description of ADEs that are not present in the structured data. The timely identification of unknown toxicities of prescription drugs is an important, unsolved problem. Objective The objective of this study was to develop an annotated corpus of FAERS narratives and biomedical named entity tagger to automatically identify ADE related information in the FAERS narratives. Methods We developed an annotation guideline and annotate medication information and adverse event related entities on 122 FAERS narratives comprising approximately 23,000 word tokens. A named entity tagger using supervised machine learning approaches was built for detecting medication information and adverse event entities using various categories of features. Results The annotated corpus had an agreement of over .9 Cohen’s kappa for medication and adverse event entities. The best performing tagger achieves an overall performance of 0.73 F1 score for detection of medication, adverse event and other named entities. Conclusions In this study, we developed an annotated corpus of FAERS narratives and machine learning based models for automatically extracting medication and adverse event information from the FAERS narratives. Our study is an important step towards enriching the FAERS data for postmarketing pharmacovigilance.
Collapse
|
27
|
Toward enhanced pharmacovigilance using patient-generated data on the internet. Clin Pharmacol Ther 2014; 96:239-46. [PMID: 24713590 PMCID: PMC4111778 DOI: 10.1038/clpt.2014.77] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/27/2014] [Indexed: 11/17/2022]
Abstract
The promise of augmenting pharmacovigilance with patient-generated data drawn from the Internet was called out by a scientific committee charged with conducting a review of the current and planned pharmacovigilance practices of the US Food and Drug Administration (FDA). To this end, we present a study on harnessing behavioral data drawn from Internet search logs to detect adverse drug reactions (ADRs). By analyzing search queries collected from 80 million consenting users and by using a widely recognized benchmark of ADRs, we found that the performance of ADR detection via search logs is comparable and complementary to detection based on the FDA’s adverse event reporting system (AERS). We show that by jointly leveraging data from the AERS and search logs, the accuracy of ADR detection can be improved by 19% relative to the use of each data source independently. The results suggest that leveraging nontraditional sources such as online search logs could supplement existing pharmacovigilance approaches.
Collapse
|
28
|
Hur J, Liu Z, Tong W, Laaksonen R, Bai JPF. Drug-induced rhabdomyolysis: from systems pharmacology analysis to biochemical flux. Chem Res Toxicol 2014; 27:421-32. [PMID: 24422454 DOI: 10.1021/tx400409c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The goal of this study was to integrate systems pharmacology and biochemical flux to delineate drug-induced rhabdomyolysis by leveraging prior knowledge and publicly accessible data. A list of 211 rhabdomyolysis-inducing drugs (RIDs) was compiled and curated from multiple sources. Extended pharmacological network analysis revealed that the intermediators directly interacting with the pharmacological targets of RIDs were significantly enriched with functions such as regulation of cell cycle, apoptosis, and ubiquitin-mediated proteolysis. A total of 78 intermediators were shown to be significantly connected to at least five RIDs, including estrogen receptor 1 (ESR1), synuclein gamma (SNCG), and janus kinase 2 (JAK2). Transcriptomic analysis of RIDs profiled in Connectivity Map on the global scale revealed that multiple pathways are perturbed by RIDs, including ErbB signaling and lipid metabolism pathways, and that carnitine palmitoyl transferase 2 (CPT2) was in the top 1 percent of the most differentially perturbed genes. CPT2 was downregulated by nine drugs that perturbed the genes significantly enriched in oxidative phosphorylation and energy-metabolism pathways. With statins as the use case, biochemical pathway analysis on the local scale implicated a role for CPT2 in statin-induced perturbation of energy homeostasis, which is in agreement with reports of statin-CPT2 interaction. Considering the complexity of human biology, an integrative multiple-approach analysis composed of a biochemical flux network, pharmacological on- and off-target networks, and transcriptomic signature is important for understanding drug safety and for providing insight into clinical gene-drug interactions.
Collapse
Affiliation(s)
- Junguk Hur
- Department of Neurology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | | | | | | | | |
Collapse
|
29
|
Low YS, Sedykh AY, Rusyn I, Tropsha A. Integrative approaches for predicting in vivo effects of chemicals from their structural descriptors and the results of short-term biological assays. Curr Top Med Chem 2014; 14:1356-64. [PMID: 24805064 PMCID: PMC5344042 DOI: 10.2174/1568026614666140506121116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 02/05/2014] [Accepted: 02/05/2014] [Indexed: 12/22/2022]
Abstract
Cheminformatics approaches such as Quantitative Structure Activity Relationship (QSAR) modeling have been used traditionally for predicting chemical toxicity. In recent years, high throughput biological assays have been increasingly employed to elucidate mechanisms of chemical toxicity and predict toxic effects of chemicals in vivo. The data generated in such assays can be considered as biological descriptors of chemicals that can be combined with molecular descriptors and employed in QSAR modeling to improve the accuracy of toxicity prediction. In this review, we discuss several approaches for integrating chemical and biological data for predicting biological effects of chemicals in vivo and compare their performance across several data sets. We conclude that while no method consistently shows superior performance, the integrative approaches rank consistently among the best yet offer enriched interpretation of models over those built with either chemical or biological data alone. We discuss the outlook for such interdisciplinary methods and offer recommendations to further improve the accuracy and interpretability of computational models that predict chemical toxicity.
Collapse
Affiliation(s)
| | | | | | - Alexander Tropsha
- 100K Beard Hall, Campus Box 7568, University of North Carolina, Chapel Hill, NC 27599-7568, USA.
| |
Collapse
|
30
|
Liu M, Cai R, Hu Y, Matheny ME, Sun J, Hu J, Xu H. Determining molecular predictors of adverse drug reactions with causality analysis based on structure learning. J Am Med Inform Assoc 2013; 21:245-51. [PMID: 24334612 DOI: 10.1136/amiajnl-2013-002051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE Adverse drug reaction (ADR) can have dire consequences. However, our current understanding of the causes of drug-induced toxicity is still limited. Hence it is of paramount importance to determine molecular factors of adverse drug responses so that safer therapies can be designed. METHODS We propose a causality analysis model based on structure learning (CASTLE) for identifying factors that contribute significantly to ADRs from an integration of chemical and biological properties of drugs. This study aims to address two major limitations of the existing ADR prediction studies. First, ADR prediction is mostly performed by assessing the correlations between the input features and ADRs, and the identified associations may not indicate causal relations. Second, most predictive models lack biological interpretability. RESULTS CASTLE was evaluated in terms of prediction accuracy on 12 organ-specific ADRs using 830 approved drugs. The prediction was carried out by first extracting causal features with structure learning and then applying them to a support vector machine (SVM) for classification. Through rigorous experimental analyses, we observed significant increases in both macro and micro F1 scores compared with the traditional SVM classifier, from 0.88 to 0.89 and 0.74 to 0.81, respectively. Most importantly, identified links between the biological factors and organ-specific drug toxicities were partially supported by evidence in Online Mendelian Inheritance in Man. CONCLUSIONS The proposed CASTLE model not only performed better in prediction than the baseline SVM but also produced more interpretable results (ie, biological factors responsible for ADRs), which is critical to discovering molecular activators of ADRs.
Collapse
Affiliation(s)
- Mei Liu
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Gathering and exploring scientific knowledge in pharmacovigilance. PLoS One 2013; 8:e83016. [PMID: 24349421 PMCID: PMC3859628 DOI: 10.1371/journal.pone.0083016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/08/2013] [Indexed: 11/19/2022] Open
Abstract
Pharmacovigilance plays a key role in the healthcare domain through the assessment, monitoring and discovery of interactions amongst drugs and their effects in the human organism. However, technological advances in this field have been slowing down over the last decade due to miscellaneous legal, ethical and methodological constraints. Pharmaceutical companies started to realize that collaborative and integrative approaches boost current drug research and development processes. Hence, new strategies are required to connect researchers, datasets, biomedical knowledge and analysis algorithms, allowing them to fully exploit the true value behind state-of-the-art pharmacovigilance efforts. This manuscript introduces a new platform directed towards pharmacovigilance knowledge providers. This system, based on a service-oriented architecture, adopts a plugin-based approach to solve fundamental pharmacovigilance software challenges. With the wealth of collected clinical and pharmaceutical data, it is now possible to connect knowledge providers' analysis and exploration algorithms with real data. As a result, new strategies allow a faster identification of high-risk interactions between marketed drugs and adverse events, and enable the automated uncovering of scientific evidence behind them. With this architecture, the pharmacovigilance field has a new platform to coordinate large-scale drug evaluation efforts in a unique ecosystem, publicly available at http://bioinformatics.ua.pt/euadr/.
Collapse
|
32
|
Coloma PM, Trifirò G, Patadia V, Sturkenboom M. Postmarketing safety surveillance : where does signal detection using electronic healthcare records fit into the big picture? Drug Saf 2013; 36:183-97. [PMID: 23377696 DOI: 10.1007/s40264-013-0018-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The safety profile of a drug evolves over its lifetime on the market; there are bound to be changes in the circumstances of a drug's clinical use which may give rise to previously unobserved adverse effects, hence necessitating surveillance postmarketing. Postmarketing surveillance has traditionally been carried out by systematic manual review of spontaneous reports of adverse drug reactions. Vast improvements in computing capabilities have provided opportunities to automate signal detection, and several worldwide initiatives are exploring new approaches to facilitate earlier detection, primarily through mining of routinely-collected data from electronic healthcare records (EHR). This paper provides an overview of ongoing initiatives exploring data from EHR for signal detection vis-à-vis established spontaneous reporting systems (SRS). We describe the role SRS has played in regulatory decision making with respect to safety issues, and evaluate the potential added value of EHR-based signal detection systems to the current practice of drug surveillance. Safety signal detection is both an iterative and dynamic process. It is in the best interest of public health to integrate and understand evidence from all possibly relevant information sources on drug safety. Proper evaluation and communication of potential signals identified remains an imperative and should accompany any signal detection activity.
Collapse
Affiliation(s)
- Preciosa M Coloma
- Ee-2116, Department of Medical Informatics, Erasmus Medical Centre, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
33
|
Shah NH, Tenenbaum JD. The coming age of data-driven medicine: translational bioinformatics' next frontier. J Am Med Inform Assoc 2013; 19:e2-4. [PMID: 22718035 PMCID: PMC3392866 DOI: 10.1136/amiajnl-2012-000969] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Nigam H Shah
- Stanford Center for Biomedical Informatics Research, Stanford University School of Medicine, Stanford, California, USA
| | - Jessica D Tenenbaum
- Duke Translational Medicine Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
34
|
Zhu Q, Freimuth RR, Pathak J, Durski MJ, Chute CG. Disambiguation of PharmGKB drug-disease relations with NDF-RT and SPL. J Biomed Inform 2013; 46:690-6. [PMID: 23727027 DOI: 10.1016/j.jbi.2013.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 04/12/2013] [Accepted: 05/18/2013] [Indexed: 10/26/2022]
Abstract
PharmGKB is a leading resource of high quality pharmacogenomics data that provides information about how genetic variations modulate an individual's response to drugs. PharmGKB contains information about genetic variations, pharmacokinetic and pharmacodynamic pathways, and the effect of variations on drug-related phenotypes. These relationships are represented using very general terms, however, and the precise semantic relationships among drugs, and diseases are not often captured. In this paper we develop a protocol to detect and disambiguate general clinical associations between drugs and diseases using more precise annotation terms from other data sources. PharmGKB provides very detailed clinical associations between genetic variants and drug response, including genotype-specific drug dosing guidelines, and this procedure will armGKB. The availability of more detailed data will help investigators to conduct more precise queries, such as finding particular diseases caused or treated by a specific drug. We first mapped drugs extracted from PharmGKB drug-disease relationships to those in the National Drug File Reference Terminology (NDF-RT) and to Structured Product Labels (SPLs). Specifically, we retrieved drug and disease role relationships describing and defining concepts according to their relationships with other concepts from NDF-RT. We also used the NCBO (National Center for Biomedical Ontology) annotator to annotate disease terms from the free text extracted from five SPL sections (indication, contraindication, ADE, precaution, and warning). Finally, we used the detailed drug and disease relationship information from NDF-RT and the SPLs to annotate and disambiguate the more general PharmGKB drug and disease associations.
Collapse
Affiliation(s)
- Qian Zhu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | |
Collapse
|
35
|
Harpaz R, Vilar S, Dumouchel W, Salmasian H, Haerian K, Shah NH, Chase HS, Friedman C. Combing signals from spontaneous reports and electronic health records for detection of adverse drug reactions. J Am Med Inform Assoc 2013; 20:413-9. [PMID: 23118093 PMCID: PMC3628045 DOI: 10.1136/amiajnl-2012-000930] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 09/24/2012] [Indexed: 11/03/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Rave Harpaz
- Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jiang G, Liu H, Solbrig HR, Chute CG. ADEpedia 2.0: Integration of Normalized Adverse Drug Events (ADEs) Knowledge from the UMLS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2013; 2013:100-4. [PMID: 24303245 PMCID: PMC3845793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A standardized Adverse Drug Events (ADEs) knowledge base that encodes known ADE knowledge can be very useful in improving ADE detection for drug safety surveillance. In our previous study, we developed the ADEpedia that is a standardized knowledge base of ADEs based on drug product labels. The objectives of the present study are 1) to integrate normalized ADE knowledge from the Unified Medical Language System (UMLS) into the ADEpedia; and 2) to enrich the knowledge base with the drug-disorder co-occurrence data from a 51-million-document electronic medical records (EMRs) system. We extracted 266,832 drug-disorder concept pairs from the UMLS, covering 14,256 (1.69%) distinct drug concepts and 19,006 (3.53%) distinct disorder concepts. Of them, 71,626 (26.8%) concept pairs from UMLS co-occurred in the EMRs. We performed a preliminary evaluation on the utility of the UMLS ADE data. In conclusion, we have built an ADEpedia 2.0 framework that intends to integrate known ADE knowledge from disparate sources. The UMLS is a useful source for providing standardized ADE knowledge relevant to indications, contraindications and adverse effects, and complementary to the ADE data from drug product labels. The statistics from EMRs would enable the meaningful use of ADE data for drug safety surveillance.
Collapse
Affiliation(s)
- Guoqian Jiang
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic College of Medicine, Rochester, MN
| | - Hongfang Liu
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic College of Medicine, Rochester, MN
| | - Harold R. Solbrig
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic College of Medicine, Rochester, MN
| | - Christopher G. Chute
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
37
|
Oliveira JL, Lopes P, Nunes T, Campos D, Boyer S, Ahlberg E, van Mulligen EM, Kors JA, Singh B, Furlong LI, Sanz F, Bauer-Mehren A, Carrascosa MC, Mestres J, Avillach P, Diallo G, Díaz Acedo C, van der Lei J. The EU-ADR Web Platform: delivering advanced pharmacovigilance tools. Pharmacoepidemiol Drug Saf 2012. [PMID: 23208789 DOI: 10.1002/pds.3375] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE Pharmacovigilance methods have advanced greatly during the last decades, making post-market drug assessment an essential drug evaluation component. These methods mainly rely on the use of spontaneous reporting systems and health information databases to collect expertise from huge amounts of real-world reports. The EU-ADR Web Platform was built to further facilitate accessing, monitoring and exploring these data, enabling an in-depth analysis of adverse drug reactions risks. METHODS The EU-ADR Web Platform exploits the wealth of data collected within a large-scale European initiative, the EU-ADR project. Millions of electronic health records, provided by national health agencies, are mined for specific drug events, which are correlated with literature, protein and pathway data, resulting in a rich drug-event dataset. Next, advanced distributed computing methods are tailored to coordinate the execution of data-mining and statistical analysis tasks. This permits obtaining a ranked drug-event list, removing spurious entries and highlighting relationships with high risk potential. RESULTS The EU-ADR Web Platform is an open workspace for the integrated analysis of pharmacovigilance datasets. Using this software, researchers can access a variety of tools provided by distinct partners in a single centralized environment. Besides performing standalone drug-event assessments, they can also control the pipeline for an improved batch analysis of custom datasets. Drug-event pairs can be substantiated and statistically analysed within the platform's innovative working environment. CONCLUSIONS A pioneering workspace that helps in explaining the biological path of adverse drug reactions was developed within the EU-ADR project consortium. This tool, targeted at the pharmacovigilance community, is available online at https://bioinformatics.ua.pt/euadr/.
Collapse
|
38
|
Yang C, Srinivasan P, Polgreen PM. Automatic adverse drug events detection using letters to the editor. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2012; 2012:1030-1039. [PMID: 23304379 PMCID: PMC3540506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
We present and test the intuition that letters to the editor in journals carry early signals of adverse drug events (ADEs). Surprisingly these letters have not yet been exploited for automatic ADE detection unlike for example, clinical records and PubMed. Part of the challenge is that it is not easy to access the full-text of letters (for the most part these do not appear in PubMed). Also letters are likely underrated in comparison with full articles. Besides demonstrating that this intuition holds we contribute techniques for post market drug surveillance. Specifically, we test an automatic approach for ADE detection from letters using off-the-shelf machine learning tools. We also involve natural language processing for feature definitions. Overall we achieve high accuracy in our experiments and our method also works well on a second new test set. Our results encourage us to further pursue this line of research.
Collapse
Affiliation(s)
- Chao Yang
- Department of Computer Science, The University of Iowa, Iowa City, IA, USA
| | | | | |
Collapse
|
39
|
Enhancing adverse drug event detection in electronic health records using molecular structure similarity: application to pancreatitis. PLoS One 2012; 7:e41471. [PMID: 22911794 PMCID: PMC3404072 DOI: 10.1371/journal.pone.0041471] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 06/21/2012] [Indexed: 12/14/2022] Open
Abstract
Background Adverse drug events (ADEs) detection and assessment is at the center of pharmacovigilance. Data mining of systems, such as FDA’s Adverse Event Reporting System (AERS) and more recently, Electronic Health Records (EHRs), can aid in the automatic detection and analysis of ADEs. Although different data mining approaches have been shown to be valuable, it is still crucial to improve the quality of the generated signals. Objective To leverage structural similarity by developing molecular fingerprint-based models (MFBMs) to strengthen ADE signals generated from EHR data. Methods A reference standard of drugs known to be causally associated with the adverse event pancreatitis was used to create a MFBM. Electronic Health Records (EHRs) from the New York Presbyterian Hospital were mined to generate structured data. Disproportionality Analysis (DPA) was applied to the data, and 278 possible signals related to the ADE pancreatitis were detected. Candidate drugs associated with these signals were then assessed using the MFBM to find the most promising candidates based on structural similarity. Results The use of MFBM as a means to strengthen or prioritize signals generated from the EHR significantly improved the detection accuracy of ADEs related to pancreatitis. MFBM also highlights the etiology of the ADE by identifying structurally similar drugs, which could follow a similar mechanism of action. Conclusion The method proposed in this paper provides evidence of being a promising adjunct to existing automated ADE detection and analysis approaches.
Collapse
|
40
|
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. [PMID: 22549283 PMCID: PMC3675775 DOI: 10.1038/clpt.2012.50] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
An important goal of the health system is to identify new adverse drug events (ADEs) in the postapproval period. Datamining methods that can transform data into meaningful knowledge to inform patient safety have proven essential for this purpose. New opportunities have emerged to harness data sources that have not been used within the traditional framework. This article provides an overview of recent methodological innovations and data sources used to support ADE discovery and analysis.
Collapse
Affiliation(s)
- R Harpaz
- Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Vilar S, Harpaz R, Uriarte E, Santana L, Rabadan R, Friedman C. Drug-drug interaction through molecular structure similarity analysis. J Am Med Inform Assoc 2012; 19:1066-74. [PMID: 22647690 DOI: 10.1136/amiajnl-2012-000935] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Hohenegger M. Drug induced rhabdomyolysis. Curr Opin Pharmacol 2012; 12:335-9. [PMID: 22560920 PMCID: PMC3387368 DOI: 10.1016/j.coph.2012.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 11/05/2022]
Abstract
Rhabdomyolysis is a clinical condition of potential life threatening destruction of skeletal muscle caused by diverse mechanisms including drugs and toxins. Given the fact that structurally not related compounds cause an identical phenotype pinpoints to common targets or pathways, responsible for executing rhabdomyolysis. A drop in myoplasmic ATP paralleled with sustained elevations in cytosolic Ca2+ concentration represents a common signature of rhabdomyolysis. Interestingly, cardiac tissue is hardly affected or only secondary, as a consequence of imbalance in electrolytes or acid–base equilibrium. This dogma is now impaired by compounds, which show up with combined toxicity in heart and skeletal muscle. In this review, cases of rhabdomyolysis with novel recently approved drugs will be explored for new target mechanisms in the light of previously described pathomechanisms.
Collapse
Affiliation(s)
- Martin Hohenegger
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstrasse 13A, A-1090 Vienna, Austria.
| |
Collapse
|
43
|
Shah NH. Translational bioinformatics embraces big data. Yearb Med Inform 2012; 7:130-134. [PMID: 22890354 PMCID: PMC4370941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
We review the latest trends and major developments in translational bioinformatics in the year 2011-2012. Our emphasis is on highlighting the key events in the field and pointing at promising research areas for the future. The key take-home points are: • Translational informatics is ready to revolutionize human health and healthcare using large-scale measurements on individuals. • Data-centric approaches that compute on massive amounts of data (often called "Big Data") to discover patterns and to make clinically relevant predictions will gain adoption. • Research that bridges the latest multimodal measurement technologies with large amounts of electronic healthcare data is increasing; and is where new breakthroughs will occur.
Collapse
Affiliation(s)
- N H Shah
- Stanford University School of Medicine, 1265 Welch Road, Room X-229, Stanford, CA 94305, USA. E-mail:
| |
Collapse
|