1
|
Crisafulli S, Bate A, Brown JS, Candore G, Chandler RE, Hammad TA, Lane S, Maro JC, Norén GN, Pariente A, Russom M, Salas M, Segec A, Shakir S, Spini A, Toh S, Tuccori M, van Puijenbroek E, Trifirò G. Interplay of Spontaneous Reporting and Longitudinal Healthcare Databases for Signal Management: Position Statement from the Real-World Evidence and Big Data Special Interest Group of the International Society of Pharmacovigilance. Drug Saf 2025:10.1007/s40264-025-01548-3. [PMID: 40223041 DOI: 10.1007/s40264-025-01548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Signal management, defined as the set of activities from signal detection to recommendations for action, is conducted using different data sources and leveraging data from spontaneous reporting databases (SRDs), which represent the cornerstone of pharmacovigilance. However, the exponentially increasing generation and availability of real-world data collected in longitudinal healthcare databases (LHDs), along with the rapid evolution of artificial intelligence-based algorithms and other advanced analytical methods, offers a wide range of opportunities to complement SRDs throughout all stages of signal management, especially signal detection. Integrating information derived from SRDs and LHDs may reduce their respective limitations, thus potentially enhancing post-marketing surveillance. The aim of this position statement is to critically evaluate the complementary role of SRDs and LHDs in signal management, exploring the potential benefits and challenges in integrating information coming from these two data sources. Furthermore, we presented successful cases of the interplay between SRDs and LHDs for signal management, along with future opportunities and directions to improve such interplay.
Collapse
Affiliation(s)
- Salvatore Crisafulli
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy
| | - Andrew Bate
- Global Safety, GSK, Brentford, UK
- Department of Non-Communicable Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Jeffrey Stuart Brown
- TriNetX, Cambridge, MA, USA
- Department of Population Medicine, Harvard Medical School, Boston, MA, USA
| | | | | | - Tarek A Hammad
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Samantha Lane
- Drug Safety Research Unit, Southampton, UK
- University of Portsmouth, Portsmouth, UK
| | | | | | - Antoine Pariente
- Université de Bordeaux, INSERM, BPH, Team AHeaD, U1219, 33000, Bordeaux, France
- Service de Pharmacologie Médicale, CHU de Bordeaux, INSERM, U1219, 33000, Bordeaux, France
| | - Mulugeta Russom
- National Medicines and Food Administration, Ministry of Health, Asmara, Eritrea
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maribel Salas
- Bayer Pharmaceuticals Inc., Whippany, NJ, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrej Segec
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Saad Shakir
- Drug Safety Research Unit, Southampton, UK
- University of Portsmouth, Portsmouth, UK
| | - Andrea Spini
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School, Boston, MA, USA
| | - Marco Tuccori
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy
| | - Eugène van Puijenbroek
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
- PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen Research Institute of Pharmacy, Groningen, The Netherlands
| | - Gianluca Trifirò
- Department of Diagnostics and Public Health, University of Verona, P.le L.A. Scuro 10, 37124, Verona, Italy.
| |
Collapse
|
2
|
Ko JS, Mendelsohn AB, Daniels K, Gomez-Lumbreras A, Marshall J, McDermott C, Pawloski PA, Yee GC, Lockhart CM. Patient characteristics and use for bevacizumab in ophthalmology and oncology in a distributed research network. J Manag Care Spec Pharm 2025; 31:157-166. [PMID: 39912810 PMCID: PMC11852795 DOI: 10.18553/jmcp.2025.31.2.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
BACKGROUND Although bevacizumab and its biosimilars are commonly used, there are limited real-world data on bevacizumab use in the United States, especially biosimilar bevacizumab used in ophthalmologic conditions. OBJECTIVE To evaluate use patterns and patient characteristics for the originator bevacizumab relative to its biosimilars for labeled and off-label oncology and ophthalmology conditions and characterize adverse events in patients using bevacizumab for oncologic indications. METHODS We conducted a retrospective cohort study with the Biologics and Biosimilars Collective Intelligence Consortium-distributed database to identify patients aged 21 years and older who received bevacizumab between January 1, 2010, and June 30, 2021. Oncology indications included colon, lung, and gynecologic (cervical, uterine, and ovarian) cancers. Ophthalmologic indications included neovascular age-related macular degeneration (AMD), retinal vein occlusion (RVO), choroidal neovascularization (CNV), and proliferative diabetic retinopathy (PDR). We also captured patients' demographic and clinical characteristics. RESULTS Total bevacizumab product (originator and biosimilars) use increased over time for RVO, CNV, and PDR starting in 2015 but decreased for AMD after 2016. For ophthalmology, bevacizumab product users were primarily male (56.8%), had a mean age of 62.9 years (SD = 0.08), and had a mean Charlson/Elixhauser combined comorbidity score ranging from 0.7 (CNV) to 2.7 (PDR). Bevacizumab users for oncology indications were mostly female (61.8%), had a mean age of 62.9 years (SD = 12.2), and had a mean Charlson/Elixhauser combined comorbidity score of 7.4 (SD = 3.0). Oncologic biosimilar product use increased over time between 2019 and 2020 as follows: colon cancer, 6.2% to 49.4%; lung cancer, 1.9% to 36.2%; and gynecologic cancer, 2.4% to 38.1%. CONCLUSIONS Bevacizumab product use increased across most indications during the study period. Use for biosimilars increased in later years relative to the originator once available on the market. Limited data are available on real-world biosimilar use in the United States; future research should include monitoring for use and adverse events of these products.
Collapse
Affiliation(s)
- Jenice S. Ko
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | - Aaron B. Mendelsohn
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | | | | | - James Marshall
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | - Cara McDermott
- AMCP Biolocgics and Biosimilars Collective Intelligence Consortium, Alexandria, VA
| | | | - Gary C. Yee
- University of Nebraska Medical Center, Omaha
| | | |
Collapse
|
3
|
Shu D, Zou G, Hou L, Petrone AB, Maro JC, Fireman BH, Toh S, Connolly JG. A simple Cox approach to estimating risk ratios without sharing individual-level data in multisite studies. Am J Epidemiol 2025; 194:226-232. [PMID: 38973755 DOI: 10.1093/aje/kwae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 05/06/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024] Open
Abstract
Epidemiologic studies frequently use risk ratios to quantify associations between exposures and binary outcomes. When the data are physically stored at the sites of multiple data partners, it can be challenging to perform individual-level analysis if data cannot be pooled centrally due to privacy constraints. Existing methods either require multiple file transfers between each data partner and an analysis center (eg, distributed regression) or only provide approximate estimation of the risk ratio (eg, meta-analysis). Here we develop a practical method that requires a single transfer of 8 summary-level quantities from each data partner. Our approach leverages an existing risk-set method and software originally developed for Cox regression. Sharing only summary-level information, the proposed method provides risk ratio estimates and 95% CIs identical to those that would be provided-if individual-level data were pooled-by the modified Poisson regression. We justify the method theoretically, confirm its performance using simulated data, and implement it in a distributed analysis of COVID-19 data from the US Food and Drug Administration's Sentinel System. This article is part of a Special Collection on Pharmacoepidemiology.
Collapse
Affiliation(s)
- Di Shu
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, PA 19146, United States
| | - Guangyong Zou
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, ON N6G 2M1, Canada
- Robarts Research Institute, Western University, London, ON N6A 3K7, Canada
| | - Laura Hou
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| | - Andrew B Petrone
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| | - Judith C Maro
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| | - Bruce H Fireman
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, United States
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| | - John G Connolly
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| |
Collapse
|
4
|
Ouazzani K, Ansolabehere X, Journeau F, Vidal A, Jaubourg N, Doublet M, Thollot R, Fabre A, Glatt N. Project Victoria: A pragmatic data model to automate RWE generation from the national French claims database. Health Informatics J 2025; 31:14604582251318250. [PMID: 39913942 DOI: 10.1177/14604582251318250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Objective: This paper describes Victoria, an empirically built data pipeline for SNDS to: - Build an automated, scalable pipeline supporting changes to the data model inherent to the use of large databases, - Deliver a documented pipeline with clear processes, enabling scientific, epidemiological researches, - Ease access to SNDS data in compliance with regulatory requirements. Methods: This paper describes the 2-steps process of the Victoria pipeline and its final output. The initial cleaning step consists in formatting, deleting empty, error or duplicate records and renaming variables without changing their values, accordingly with the official SNDS documentation. The second step consists in creating 2 linearised data models: every line of each table is an event, and each table is indexed with a unique patient identifier, without the need for a central patient or identifier table. These 2 models are: - the epidemiological model, used for answering most of the research questions requiring population phenotyping (demography, diagnosis, procedures characteristics). - the medico-economic model is used for costs and healthcare consumption analyses. It contains more complex information about reimbursements rates and the data quality assessment is focused on costs rather than medico-administrative information. Results: The pipeline was executed on 2 different datasets representing ∼85 000 and ∼870 000 beneficiaries with the following configuration: one master with 4 cores and 16Go of RAM and respectively 4 and 6 workers. The total execution time for the smaller dataset was 25 h and 96 h for the larger one. The longest part of those times is represented by the format conversion to parquet. The cleaning step took only 4 h in both cases. The epidemiological model took 344 min for the smaller dataset and 1934 min for the larger one. The medico-economic model took the longest time with 704 min and 2145 min, respectively. Conclusion: Victoria pipeline is a successfully implemented SNDS pipeline. Compared to previous pipelines, reviewability is part of its design as unit tests and quality assessments can natively be developed to ensure data and analysis quality. The pipeline has been used for 2 published studies. The recent work toward OMOP conversion will be integrated in upcoming versions and, as Victoria is set to run on a CD platform, the potential evolution if SNDS format can be considered.
Collapse
|
5
|
Hoxhaj V, Andaur Navarro CL, Riera‐Arnau J, Elbers RJHJ, Alsina E, Dodd C, Sturkenboom MCJM. INSIGHT: A Tool for Fit-for-Purpose Evaluation and Quality Assessment of Standardized Observational Data Sources for Real World Evidence on Medicine and Vaccine Safety. Pharmacoepidemiol Drug Saf 2025; 34:e70089. [PMID: 39805807 PMCID: PMC11730612 DOI: 10.1002/pds.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE To describe the development of INSIGHT, a real-world data quality tool to assess completeness, consistency, and fitness-for-purpose of observational health data sources. METHODS We designed a three-level pipeline with data quality assessments (DQAs) to be performed in ConcePTION Common Data Model (CDM) instances. The pipeline has been coded using R. RESULTS INSIGHT is an open-source tool that identifies potential data quality issues in CDM-standardized instances through the systematic execution and summary of over 588 configurable DQAs. Level 1 focuses on conformance to the ConcePTION CDM specifications. Level 2 evaluates the temporal plausibility of events and uniqueness of records. Level 3 provides an overview of distributions, outliers, and trends over time to facilitate fit-for-purpose evaluation. Therefore, level 1 and 2 assure a proper data standardization, while level 3 provides information regarding the study population, and potential sub-populations. The DQAs are run locally and assessed centrally by a data quality revisor together with the data access provider's representatives. DISCUSSION Data quality is the sum of several internal and external features of the data. While DQAs can provide reassurance about fitness-for-purpose for secondary-use data sources, improvements in data collection are essential to reduce errors and enhance overall data quality for Real World Evidence. CONCLUSION INSIGHT aims to support clinical and regulatory decision-making for medicines and vaccines by evaluating the quality of observational health data sources to support fit for purpose assessment. Assessing and improving data quality will enhance the reliability and quality of the generated evidence. STUDY REGISTRATION This research was registered in EU PAS registration with number EU50142.
Collapse
Affiliation(s)
- Vjola Hoxhaj
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Constanza L. Andaur Navarro
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Judit Riera‐Arnau
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Clinical Pharmacology ServiceVall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Roel J. H. J. Elbers
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Ema Alsina
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Caitlin Dodd
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Miriam C. J. M. Sturkenboom
- Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
6
|
Ba G, Hao W, Shi Q, Sun H, Jin H, Li M, Cao Y, Qiao J, Xie M, Zhou J, Huang Z, Wang Z, Zhou Y, Jiang G, Zhang J. Epidemiological investigation of adult emergency infusion adverse drug reactions (EIADR) in Nanjing, China: a prospective cross-sectional study (EIADR II). Expert Opin Drug Saf 2024:1-7. [PMID: 39679556 DOI: 10.1080/14740338.2024.2443789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/29/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To analyze the clinical characteristics of adverse drug reactions (ADRs) in adults receiving emergency infusions at a tertiary hospital. METHODS We conducted a prospective observational cohort study involving 585 adult patients who experienced adverse drug reactions (ADRs) between 20 November 2019, and 20 November 2023, during intravenous infusions in the emergency infusion room of a tertiary hospital. The analysis included patients' gender, age, type of drugs involved, organ-system involvement, clinical manifestations of ADRs, severity grading of ADRs, and preventability of ADRs. RESULTS The highest percentage of ADRs occurred in the 30-39 age group. Antimicrobials were the most common cause of ADRs, with skin manifestations being the predominant clinical feature. Approximately 23.93% of ADRs were deemed preventable. CONCLUSION Monitoring ADRs related to antimicrobials is crucial in adult emergency infusions. The 30-39 age group is particularly susceptible to ADRs. Preventive measures and a well-established Electronic Health Record (EHR) system can effectively reduce ADRs incidence.
Collapse
Affiliation(s)
- Gen Ba
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
- Institute of Poisoning, Nanjing Medical University, Nanjing, China
| | - Weiwen Hao
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Qifang Shi
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
- Institute of Poisoning, Nanjing Medical University, Nanjing, China
| | - Hao Sun
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
- Institute of Poisoning, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, China
| | - Hua Jin
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Meng Li
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
- Institute of Poisoning, Nanjing Medical University, Nanjing, China
| | - Yun Cao
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Jie Qiao
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Min Xie
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Juan Zhou
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Zhenzhen Huang
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Zihao Wang
- Department of Pharmacy, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Ying Zhou
- Department of Pharmacy, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Guiping Jiang
- Department of Pharmacy, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
| | - Jinsong Zhang
- Emergency Medical Center, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, China
- Institute of Poisoning, Nanjing Medical University, Nanjing, China
- The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Maro JC, Platt R, Toh S. Considerations Regarding Association of Semaglutide and Nonarteritic Anterior Ischemic Optic Neuropathy. JAMA Ophthalmol 2024; 142:1176. [PMID: 39480446 DOI: 10.1001/jamaophthalmol.2024.4527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Affiliation(s)
- Judith C Maro
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Richard Platt
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| |
Collapse
|
8
|
Walsh CG, Wilimitis D, Chen Q, Wright A, Kolli J, Robinson K, Ripperger MA, Johnson KB, Carrell D, Desai RJ, Mosholder A, Dharmarajan S, Adimadhyam S, Fabbri D, Stojanovic D, Matheny ME, Bejan CA. Scalable incident detection via natural language processing and probabilistic language models. Sci Rep 2024; 14:23429. [PMID: 39379449 PMCID: PMC11461638 DOI: 10.1038/s41598-024-72756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Post marketing safety surveillance depends in part on the ability to detect concerning clinical events at scale. Spontaneous reporting might be an effective component of safety surveillance, but it requires awareness and understanding among healthcare professionals to achieve its potential. Reliance on readily available structured data such as diagnostic codes risks under-coding and imprecision. Clinical textual data might bridge these gaps, and natural language processing (NLP) has been shown to aid in scalable phenotyping across healthcare records in multiple clinical domains. In this study, we developed and validated a novel incident phenotyping approach using unstructured clinical textual data agnostic to Electronic Health Record (EHR) and note type. It's based on a published, validated approach (PheRe) used to ascertain social determinants of health and suicidality across entire healthcare records. To demonstrate generalizability, we validated this approach on two separate phenotypes that share common challenges with respect to accurate ascertainment: (1) suicide attempt; (2) sleep-related behaviors. With samples of 89,428 records and 35,863 records for suicide attempt and sleep-related behaviors, respectively, we conducted silver standard (diagnostic coding) and gold standard (manual chart review) validation. We showed Area Under the Precision-Recall Curve of ~ 0.77 (95% CI 0.75-0.78) for suicide attempt and AUPR ~ 0.31 (95% CI 0.28-0.34) for sleep-related behaviors. We also evaluated performance by coded race and demonstrated differences in performance by race differed across phenotypes. Scalable phenotyping models, like most healthcare AI, require algorithmovigilance and debiasing prior to implementation.
Collapse
Affiliation(s)
- Colin G Walsh
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt University Medical Center, Nashville, USA.
| | - Drew Wilimitis
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qingxia Chen
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aileen Wright
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jhansi Kolli
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katelyn Robinson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Ripperger
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kevin B Johnson
- Department of Biostatistics, Epidemiology and Informatics, and Pediatrics, University of Pennsylvania, Pennsylvania, USA
- Department of Computer and Information Science, Bioengineering, University of Pennsylvania, Pennsylvania, USA
- Department of Science Communication, University of Pennsylvania, Pennsylvania, USA
| | - David Carrell
- Washington Health Research Institute, , Kaiser Permanente Washington, Washington, USA
| | - Rishi J Desai
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Andrew Mosholder
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Maryland, USA
- Office of Surveillance and Epidemiology, United States Food and Drug Administration, Maryland, USA
| | - Sai Dharmarajan
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Maryland, USA
- Office of Translational Science, United States Food and Drug Administration, Maryland, USA
| | - Sruthi Adimadhyam
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, USA
| | - Daniel Fabbri
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Danijela Stojanovic
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Maryland, USA
- Office of Surveillance and Epidemiology, United States Food and Drug Administration, Maryland, USA
| | - Michael E Matheny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
9
|
Mai X, Mendelsohn AB, Marshall J, Lin ND, McDermott CL, Ko JS, Pawloski PA, Jamal-Allial A, Daniels K, McMahill-Walraven CN, Djibo DA, Lockhart CM. Utilization and patient characteristics for the trastuzumab reference and biosimilars, and other human epidermal growth factor receptor 2 inhibitors in the United States. J Manag Care Spec Pharm 2024; 30:1160-1166. [PMID: 39321121 PMCID: PMC11424920 DOI: 10.18553/jmcp.2024.30.10.1160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
BACKGROUND Trastuzumab is an antihuman epidermal growth factor receptor 2 monoclonal antibody used to treat breast and other cancers. Trastuzumab biosimilars were approved in the United States beginning in 2017. Utilization information on these biosimilars is limited. OBJECTIVE To examine utilization patterns and characteristics of patients treated with trastuzumab (biosimilars and reference) and other human epidermal growth factor receptor 2 products. METHODS We evaluated health care claims data from the Biologics and Biosimilars Collective Intelligence Consortium distributed research network, representing a large, geographically diverse US population of commercially insured individuals. We queried 4 distributed research network health plan partners to capture product usage data and patient information from October 1, 2016, to October 31, 2022. Patients were required to be continuously enrolled in their health plan for at least 365 days before their first observed trastuzumab utilization date in this study period. Data were aggregated across data partners. RESULTS More than 16 million eligible health plan members representing more than 31 million person-years of data were evaluated. We identified 5,984 incident treatment episodes; 3,878 (64.8%) episodes were with the reference trastuzumab. The mean ages were consistent across trastuzumab products (60.2 to 65.1 years) and at least 80% of the episodes were among female patients. The mean comorbidity index score was 1.2 (SD = 1.9) among users of the reference vs the biosimilars (range 1.2-2.5). Other clinical characteristics (eg, diabetes, hypertension) were comparable across products. The proportion of total incident episodes of the reference trastuzumab decreased substantially over time (96% in 2016 vs 28% in 2021) as utilization of the biosimilars increased (eg, use of trastuzumab-anns increased from 2% [2019] to 36% [2021]). Similar utilization trends were seen among patients with and without metastatic breast cancer. CONCLUSIONS Trastuzumab biosimilars utilization has grown since their introduction to the US market. Exploration of these biosimilars' comparative effectiveness and safety to their reference product is warranted.
Collapse
Affiliation(s)
- Xiaodan Mai
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | - Aaron B Mendelsohn
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | - James Marshall
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | | | - Cara L McDermott
- AMCP Biologics and Biosimilars Collective Intelligence Consortium, Alexandria, VA
| | - Jenice S Ko
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
| | | | | | | | | | | | - Catherine M Lockhart
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA
- AMCP Biologics and Biosimilars Collective Intelligence Consortium, Alexandria, VA
| |
Collapse
|
10
|
Lyons JG, Shinde MU, Maro JC, Petrone A, Cosgrove A, Kempner ME, Andrade SE, Mwidau J, Stojanovic D, Hernández-Muñoz JJ, Toh S. Use of the Sentinel System to Examine Medical Product Use and Outcomes During Pregnancy. Drug Saf 2024; 47:931-940. [PMID: 38940904 DOI: 10.1007/s40264-024-01447-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/29/2024]
Abstract
While many pregnant individuals use prescription medications, evidence supporting product safety during pregnancy is often inadequate. Existing electronic healthcare data sources provide large, diverse samples of health plan members to allow for the study of medical product utilization during pregnancy, as well as pregnancy, maternal, and infant outcomes. The Sentinel System is a national medical product surveillance system that includes administrative claims and electronic health record databases from large national and regional health insurers. In addition to these data sources, Sentinel develops and maintains a sizeable selection of analytic tools to facilitate epidemiologic analyses in a way that protects patient privacy and health system autonomy. In this article, we provide an overview of Sentinel System infrastructure, including the Mother-Infant Linkage Table, parameterizable analytic tools, and algorithms to estimate gestational age and identify pregnancy outcomes. We also describe past and future Sentinel work that contributes to our understanding of the way medical products are used and the safety of these products during pregnancy.
Collapse
Affiliation(s)
- Jennifer G Lyons
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA.
| | - Mayura U Shinde
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Judith C Maro
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Andrew Petrone
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Austin Cosgrove
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Maria E Kempner
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Susan E Andrade
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| | - Jamila Mwidau
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Danijela Stojanovic
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - José J Hernández-Muñoz
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Sengwee Toh
- Division of Therapeutics Research and Infectious Disease Epidemiology, Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA, 02215, USA
| |
Collapse
|
11
|
Desai RJ, Marsolo K, Smith J, Carrell D, Penfold R, Pillai HS, Lii J, Ngan K, Winter R, Adgent M, Ramaprasan A, Driscoll MR, Scarnecchia D, Kiernan D, Draper C, Lyons JG, Khurshid A, Maro JC, Zimmerman R, Brown J, Bright P, Hernández-Muñoz JJ, Matheny ME, Schneeweiss S. The FDA Sentinel Real World Evidence Data Enterprise (RWE-DE). Pharmacoepidemiol Drug Saf 2024; 33:e70028. [PMID: 39385712 DOI: 10.1002/pds.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE The US Food and Drug Administration's Sentinel Innovation Center aimed to establish a query-ready, quality-checked distributed data network containing electronic health records (EHRs) linked with insurance claims data for at least 10 million individuals to expand the utility of real-world data for regulatory decision-making. METHODS In this report, we describe the resulting network, the Real-World Evidence Data Enterprise (RWE-DE), including data from two commercial EHR-claims linked assets collectively termed the Commercial Network covering 21 million lives, and four academic partner institutions collectively termed the Development Network covering 4.5 million lives. RESULTS We discuss provenance and completeness of the data converted in the Sentinel Common Data Model (SCDM), describe patient populations, and report on EHR-claims linkage characterization for all contributing data sources. Further, we introduce a standardized process to store free-text notes in the Development Network for efficient retrieval as needed. CONCLUSIONS Finally, we outline typical use cases for the RWE-DE where it can broaden the reach of the types of questions that can be addressed by the Sentinel system.
Collapse
Affiliation(s)
- Rishi J Desai
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Marsolo
- Department of Population Health Sciences, Duke University, Durham, North Carolina, USA
| | - Joshua Smith
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington State, USA
| | - Robert Penfold
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington State, USA
| | - Haritha S Pillai
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joyce Lii
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kerry Ngan
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Winter
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Margaret Adgent
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Arvind Ramaprasan
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington State, USA
| | - Meighan Rogers Driscoll
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Scarnecchia
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Kiernan
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Christine Draper
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer G Lyons
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Anjum Khurshid
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Judith C Maro
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Patricia Bright
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland, USA
| | - José J Hernández-Muñoz
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland, USA
| | - Michael E Matheny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Geriatrics Research Education and Clinical Care Center, Tennessee Valley Healthcare System VA, Nashville, Tennessee, USA
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Chen C, Eworuke E, Rai A, Hou L, Ko JS, Southworth MR, Hernández-Muñoz JJ, Zhang M. Use of Hydrochlorothiazide in the United States Following Label Update About Skin Cancer Risk. Pharmacoepidemiol Drug Saf 2024; 33:e70040. [PMID: 39397256 DOI: 10.1002/pds.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE On August 20, 2020, the United States (U.S.) Food and Drug Administration (FDA) issued a Drug Safety Communication (DSC) along with labeling updates to inform the public about a small increased risk of non-melanoma skin cancer (NMSC) associated with hydrochlorothiazide (HCTZ) use. This study aims to assess whether the DSC impacted HCTZ use in the U.S. METHODS We conducted a trend analysis in the Sentinel Distributed Database using national healthcare administrative data from January 2017 to November 2022. We identified two cohorts each month: An overall cohort of all enrollees and a skin cancer cohort of those with a history of NMSC. For each cohort, we plotted the monthly proportion of patients receiving HCTZ-containing products among those receiving any thiazide diuretics. We performed interrupted time series analyses to quantify the impact of the DSC on these monthly proportions. Secondary analyses were conducted on the proportion of HCTZ users among patients receiving any antihypertensives. RESULTS In the overall cohort, the DSC was only associated with a statistically significant but clinically negligible trend change of monthly HCTZ proportion within this cohort (0.018%; 95% CI, 0.012%-0.025%). Similar results were observed in the skin cancer cohort. The secondary analysis found no significant level change or trend change in the monthly proportion of HCTZ use among antihypertensive users. CONCLUSIONS We did not observe significant changes in HCTZ use following the DSC about its NMSC risk, among the overall population and those with a history of NMSC. Our findings were in accordance with the DSC recommendation.
Collapse
Affiliation(s)
- Cheng Chen
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Efe Eworuke
- Epidemiology and Drug Safety, IQVIA Real World Solutions, Washington, DC, USA
| | - Ashish Rai
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Hou
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jenice S Ko
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary Ross Southworth
- Division of Cardiology and Nephrology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - José J Hernández-Muñoz
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mingfeng Zhang
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
13
|
Carrell DS, Floyd JS, Gruber S, Hazlehurst BL, Heagerty PJ, Nelson JC, Williamson BD, Ball R. A general framework for developing computable clinical phenotype algorithms. J Am Med Inform Assoc 2024; 31:1785-1796. [PMID: 38748991 PMCID: PMC11258420 DOI: 10.1093/jamia/ocae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 07/20/2024] Open
Abstract
OBJECTIVE To present a general framework providing high-level guidance to developers of computable algorithms for identifying patients with specific clinical conditions (phenotypes) through a variety of approaches, including but not limited to machine learning and natural language processing methods to incorporate rich electronic health record data. MATERIALS AND METHODS Drawing on extensive prior phenotyping experiences and insights derived from 3 algorithm development projects conducted specifically for this purpose, our team with expertise in clinical medicine, statistics, informatics, pharmacoepidemiology, and healthcare data science methods conceptualized stages of development and corresponding sets of principles, strategies, and practical guidelines for improving the algorithm development process. RESULTS We propose 5 stages of algorithm development and corresponding principles, strategies, and guidelines: (1) assessing fitness-for-purpose, (2) creating gold standard data, (3) feature engineering, (4) model development, and (5) model evaluation. DISCUSSION AND CONCLUSION This framework is intended to provide practical guidance and serve as a basis for future elaboration and extension.
Collapse
Affiliation(s)
- David S Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - James S Floyd
- Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195, United States
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98195, United States
| | - Susan Gruber
- Putnam Data Sciences, LLC, Cambridge, MA 02139, United States
| | - Brian L Hazlehurst
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR 97227, United States
| | - Patrick J Heagerty
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA 98195, United States
| | - Jennifer C Nelson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Brian D Williamson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Robert Ball
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, United States
| |
Collapse
|
14
|
Fleurence RL, Kent S, Adamson B, Tcheng J, Balicer R, Ross JS, Haynes K, Muller P, Campbell J, Bouée-Benhamiche E, García Martí S, Ramsey S. Assessing Real-World Data From Electronic Health Records for Health Technology Assessment: The SUITABILITY Checklist: A Good Practices Report of an ISPOR Task Force. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:692-701. [PMID: 38871437 PMCID: PMC11182651 DOI: 10.1016/j.jval.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/23/2024] [Indexed: 06/15/2024]
Abstract
This ISPOR Good Practices report provides a framework for assessing the suitability of electronic health records data for use in health technology assessments (HTAs). Although electronic health record (EHR) data can fill evidence gaps and improve decisions, several important limitations can affect its validity and relevance. The ISPOR framework includes 2 components: data delineation and data fitness for purpose. Data delineation provides a complete understanding of the data and an assessment of its trustworthiness by describing (1) data characteristics; (2) data provenance; and (3) data governance. Fitness for purpose comprises (1) data reliability items, ie, how accurate and complete the estimates are for answering the question at hand and (2) data relevance items, which assess how well the data are suited to answer the particular question from a decision-making perspective. The report includes a checklist specific to EHR data reporting: the ISPOR SUITABILITY Checklist. It also provides recommendations for HTA agencies and policy makers to improve the use of EHR-derived data over time. The report concludes with a discussion of limitations and future directions in the field, including the potential impact from the substantial and rapid advances in the diffusion and capabilities of large language models and generative artificial intelligence. The report's immediate audiences are HTA evidence developers and users. We anticipate that it will also be useful to other stakeholders, particularly regulators and manufacturers, in the future.
Collapse
Affiliation(s)
| | - Seamus Kent
- Erasmus School of Health & Policy Management, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | - Joseph S Ross
- Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kevin Haynes
- Janssen Research and Development, Titusville, NJ, USA
| | - Patrick Muller
- Centre for Guidelines, National Institute for Health and Care Excellence, Manchester or London, England, UK
| | - Jon Campbell
- National Pharmaceutical Council, Washington, DC, USA
| | - Elsa Bouée-Benhamiche
- Public Health and Healthcare Division, Institut National du Cancer, Boulogne-Billancourt, France
| | - Sebastián García Martí
- Health Technology Assessment and Health Economics Department, Institute for Clinical Effectiveness and Health Policy, Buenos Aires, Argentina
| | - Scott Ramsey
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
15
|
Rai A, Maro JC, Dutcher S, Bright P, Toh S. Transparency, reproducibility, and replicability of pharmacoepidemiology studies in a distributed network environment. Pharmacoepidemiol Drug Saf 2024; 33:e5820. [PMID: 38783407 DOI: 10.1002/pds.5820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Our objective is to describe how the U.S. Food and Drug Administration (FDA)'s Sentinel System implements best practices to ensure trust in drug safety studies using real-world data from disparate sources. METHODS We present a stepwise schematic for Sentinel's data harmonization, data quality check, query design and implementation, and reporting practices, and describe approaches to enhancing the transparency, reproducibility, and replicability of studies at each step. CONCLUSIONS Each Sentinel data partner converts its source data into the Sentinel Common Data Model. The transformed data undergoes rigorous quality checks before it can be used for Sentinel queries. The Sentinel Common Data Model framework, data transformation codes for several data sources, and data quality assurance packages are publicly available. Designed to run against the Sentinel Common Data Model, Sentinel's querying system comprises a suite of pre-tested, parametrizable computer programs that allow users to perform sophisticated descriptive and inferential analysis without having to exchange individual-level data across sites. Detailed documentation of capabilities of the programs as well as the codes and information required to execute them are publicly available on the Sentinel website. Sentinel also provides public trainings and online resources to facilitate use of its data model and querying system. Its study specifications conform to established reporting frameworks aimed at facilitating reproducibility and replicability of real-world data studies. Reports from Sentinel queries and associated design and analytic specifications are available for download on the Sentinel website. Sentinel is an example of how real-world data can be used to generate regulatory-grade evidence at scale using a transparent, reproducible, and replicable process.
Collapse
Affiliation(s)
- Ashish Rai
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Judith C Maro
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Sarah Dutcher
- U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Patricia Bright
- U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Dumiaty Y, Underwood BM, Phy-Lim J, Chee MJ. Neurocircuitry underlying the actions of glucagon-like peptide 1 and peptide YY 3-36 in the suppression of food, drug-seeking, and anxiogenesis. Neuropeptides 2024; 105:102427. [PMID: 38579490 DOI: 10.1016/j.npep.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.
Collapse
Affiliation(s)
- Yasmina Dumiaty
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Brett M Underwood
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Jenny Phy-Lim
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
17
|
Verkerk K, Voest EE. Generating and using real-world data: A worthwhile uphill battle. Cell 2024; 187:1636-1650. [PMID: 38552611 DOI: 10.1016/j.cell.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 04/02/2024]
Abstract
The precision oncology paradigm challenges the feasibility and data generalizability of traditional clinical trials. Consequently, an unmet need exists for practical approaches to test many subgroups, evaluate real-world drug value, and gather comprehensive, accessible datasets to validate novel biomarkers. Real-world data (RWD) are increasingly recognized to have the potential to fill this gap in research methodology. Established applications of RWD include informing disease epidemiology, pharmacovigilance, and healthcare quality assessment. Currently, concerns regarding RWD quality and comprehensiveness, privacy, and biases hamper their broader application. Nonetheless, RWD may play a pivotal role in supplementing clinical trials, enabling conditional reimbursement and accelerated drug access, and innovating trial conduct. Moreover, purpose-built RWD repositories may support the extension or refinement of drug indications and facilitate the discovery and validation of new biomarkers. This perspective explores the potential of leveraging RWD to advance oncology, highlights its benefits and challenges, and suggests a path forward in this evolving field.
Collapse
Affiliation(s)
- K Verkerk
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - E E Voest
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands.
| |
Collapse
|
18
|
Muntner P, Hernandez RK, Kent ST, Browning JE, Gilbertson DT, Hurwitz KE, Jick SS, Lai EC, Lash TL, Monda KL, Rothman KJ, Bradbury BD, Brookhart MA. Staging and clean room: Constructs designed to facilitate transparency and reduce bias in comparative analyses of real-world data. Pharmacoepidemiol Drug Saf 2024; 33:e5770. [PMID: 38419140 DOI: 10.1002/pds.5770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE We describe constructs designed to protect the integrity of the results from comparative analyses using real-world data (RWD): staging and clean room. METHODS Staging involves performing sequential preliminary analyses and evaluating the population size available and potential bias before conducting comparative analyses. A clean room involves restricted access to data and preliminary results, policies governing exploratory analyses and protocol deviations, and audit trail. These constructs are intended to allow decisions about protocol deviations, such as changes to design or model specification, to be made without knowledge of how they might affect subsequent analyses. We describe an example for implementing staging with a clean room. RESULTS Stage 1 may involve selecting a data source, developing and registering a protocol, establishing a clean room, and applying inclusion/exclusion criteria. Stage 2 may involve attempting to achieve covariate balance, often through propensity score models. Stage 3 may involve evaluating the presence of residual confounding using negative control outcomes. After each stage, check points may be implemented when a team of statisticians, epidemiologists and clinicians masked to how their decisions may affect study outcomes, reviews the results. This review team may be tasked with making recommendations for protocol deviations to address study precision or bias. They may recommend proceeding to the next stage, conducting additional analyses to address bias, or terminating the study. Stage 4 may involve conducting the comparative analyses. CONCLUSIONS The staging and clean room constructs are intended to protect the integrity and enhance confidence in the results of analyses of RWD.
Collapse
Affiliation(s)
- Paul Muntner
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rohini K Hernandez
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - Shia T Kent
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - James E Browning
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - David T Gilbertson
- Chronic Disease Research Group, Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
| | | | - Susan S Jick
- Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Edward C Lai
- School of Pharmacy, Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Keri L Monda
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - Kenneth J Rothman
- RTI Health Solutions, Research Triangle Institute, Research Triangle Park, North Carolina, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Brian D Bradbury
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - M Alan Brookhart
- Department of Population Health Sciences, Duke University, Durham, North Carolina, USA
| |
Collapse
|
19
|
Smith JC, Williamson BD, Cronkite DJ, Park D, Whitaker JM, McLemore MF, Osmanski JT, Winter R, Ramaprasan A, Kelley A, Shea M, Wittayanukorn S, Stojanovic D, Zhao Y, Toh S, Johnson KB, Aronoff DM, Carrell DS. Data-driven automated classification algorithms for acute health conditions: applying PheNorm to COVID-19 disease. J Am Med Inform Assoc 2024; 31:574-582. [PMID: 38109888 PMCID: PMC10873852 DOI: 10.1093/jamia/ocad241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/19/2023] [Accepted: 11/27/2023] [Indexed: 12/20/2023] Open
Abstract
OBJECTIVES Automated phenotyping algorithms can reduce development time and operator dependence compared to manually developed algorithms. One such approach, PheNorm, has performed well for identifying chronic health conditions, but its performance for acute conditions is largely unknown. Herein, we implement and evaluate PheNorm applied to symptomatic COVID-19 disease to investigate its potential feasibility for rapid phenotyping of acute health conditions. MATERIALS AND METHODS PheNorm is a general-purpose automated approach to creating computable phenotype algorithms based on natural language processing, machine learning, and (low cost) silver-standard training labels. We applied PheNorm to cohorts of potential COVID-19 patients from 2 institutions and used gold-standard manual chart review data to investigate the impact on performance of alternative feature engineering options and implementing externally trained models without local retraining. RESULTS Models at each institution achieved AUC, sensitivity, and positive predictive value of 0.853, 0.879, 0.851 and 0.804, 0.976, and 0.885, respectively, at quantiles of model-predicted risk that maximize F1. We report performance metrics for all combinations of silver labels, feature engineering options, and models trained internally versus externally. DISCUSSION Phenotyping algorithms developed using PheNorm performed well at both institutions. Performance varied with different silver-standard labels and feature engineering options. Models developed locally at one site also worked well when implemented externally at the other site. CONCLUSION PheNorm models successfully identified an acute health condition, symptomatic COVID-19. The simplicity of the PheNorm approach allows it to be applied at multiple study sites with substantially reduced overhead compared to traditional approaches.
Collapse
Affiliation(s)
- Joshua C Smith
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Brian D Williamson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - David J Cronkite
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Daniel Park
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Jill M Whitaker
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Michael F McLemore
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Joshua T Osmanski
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Robert Winter
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Arvind Ramaprasan
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Ann Kelley
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Mary Shea
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| | - Saranrat Wittayanukorn
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20903, United States
| | - Danijela Stojanovic
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20903, United States
| | - Yueqin Zhao
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20903, United States
| | - Sengwee Toh
- Harvard Pilgrim Health Care Institute, Boston, MA 02215, United States
| | - Kevin B Johnson
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - David M Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - David S Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101, United States
| |
Collapse
|
20
|
Desai RJ, Wang SV, Sreedhara SK, Zabotka L, Khosrow-Khavar F, Nelson JC, Shi X, Toh S, Wyss R, Patorno E, Dutcher S, Li J, Lee H, Ball R, Dal Pan G, Segal JB, Suissa S, Rothman KJ, Greenland S, Hernán MA, Heagerty PJ, Schneeweiss S. Process guide for inferential studies using healthcare data from routine clinical practice to evaluate causal effects of drugs (PRINCIPLED): considerations from the FDA Sentinel Innovation Center. BMJ 2024; 384:e076460. [PMID: 38346815 DOI: 10.1136/bmj-2023-076460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Affiliation(s)
- Rishi J Desai
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Sushama Kattinakere Sreedhara
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Luke Zabotka
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Farzin Khosrow-Khavar
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Jennifer C Nelson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Xu Shi
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Richard Wyss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Elisabetta Patorno
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| | - Sarah Dutcher
- US Food and Drug Administration, Silver Spring, MD, USA
| | - Jie Li
- US Food and Drug Administration, Silver Spring, MD, USA
| | - Hana Lee
- US Food and Drug Administration, Silver Spring, MD, USA
| | - Robert Ball
- US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Jodi B Segal
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samy Suissa
- Departments of Epidemiology and Biostatistics, and Medicine, McGill University, Montreal, QC, Canada
| | | | - Sander Greenland
- Department of Epidemiology and Department of Statistics, University of California, Los Angeles, CA, USA
| | - Miguel A Hernán
- CAUSALab and Departments of Epidemiology and Biostatistics, Harvard T H Chan School of Public Health, Boston, MA, USA
| | | | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
21
|
Heyard R, Held L, Schneeweiss S, Wang SV. Design differences and variation in results between randomised trials and non-randomised emulations: meta-analysis of RCT-DUPLICATE data. BMJ MEDICINE 2024; 3:e000709. [PMID: 38348308 PMCID: PMC10860009 DOI: 10.1136/bmjmed-2023-000709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/27/2023] [Indexed: 02/15/2024]
Abstract
Objective To explore how design emulation and population differences relate to variation in results between randomised controlled trials (RCT) and non-randomised real world evidence (RWE) studies, based on the RCT-DUPLICATE initiative (Randomised, Controlled Trials Duplicated Using Prospective Longitudinal Insurance Claims: Applying Techniques of Epidemiology). Design Meta-analysis of RCT-DUPLICATE data. Data sources Trials included in RCT-DUPLICATE, a demonstration project that emulated 32 randomised controlled trials using three real world data sources: Optum Clinformatics Data Mart, 2004-19; IBM MarketScan, 2003-17; and subsets of Medicare parts A, B, and D, 2009-17. Eligibility criteria for selecting studies Trials where the primary analysis resulted in a hazard ratio; 29 RCT-RWE study pairs from RCT-DUPLICATE. Results Differences and variation in effect sizes between the results from randomised controlled trials and real world evidence studies were investigated. Most of the heterogeneity in effect estimates between the RCT-RWE study pairs in this sample could be explained by three emulation differences in the meta-regression model: treatment started in hospital (which does not appear in health insurance claims data), discontinuation of some baseline treatments at randomisation (which would have been an unusual care decision in clinical practice), and delayed onset of drug effects (which would be under-reported in real world clinical practice because of the relatively short persistence of the treatment). Adding the three emulation differences to the meta-regression reduced heterogeneity from 1.9 to almost 1 (absence of heterogeneity). Conclusions This analysis suggests that a substantial proportion of the observed variation between results from randomised controlled trials and real world evidence studies can be attributed to differences in design emulation.
Collapse
Affiliation(s)
- Rachel Heyard
- Center for Reproducible Science, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Leonhard Held
- Center for Reproducible Science, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology, Brigham and Womems Hospital Harvard Medical School, Boston, Massachusetts, USA
| | - Shirley V Wang
- Division of Pharmacoepidemiology, Brigham and Womems Hospital Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Walsh CG, Wilimitis D, Chen Q, Wright A, Kolli J, Robinson K, Ripperger MA, Johnson KB, Carrell D, Desai RJ, Mosholder A, Dharmarajan S, Adimadhyam S, Fabbri D, Stojanovic D, Matheny ME, Bejan CA. Scalable Incident Detection via Natural Language Processing and Probabilistic Language Models. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.30.23299249. [PMID: 38076830 PMCID: PMC10705655 DOI: 10.1101/2023.11.30.23299249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Abstract
Post marketing safety surveillance depends in part on the ability to detect concerning clinical events at scale. Spontaneous reporting might be an effective component of safety surveillance, but it requires awareness and understanding among healthcare professionals to achieve its potential. Reliance on readily available structured data such as diagnostic codes risk under-coding and imprecision. Clinical textual data might bridge these gaps, and natural language processing (NLP) has been shown to aid in scalable phenotyping across healthcare records in multiple clinical domains. In this study, we developed and validated a novel incident phenotyping approach using unstructured clinical textual data agnostic to Electronic Health Record (EHR) and note type. It's based on a published, validated approach (PheRe) used to ascertain social determinants of health and suicidality across entire healthcare records. To demonstrate generalizability, we validated this approach on two separate phenotypes that share common challenges with respect to accurate ascertainment: 1) suicide attempt; 2) sleep-related behaviors. With samples of 89,428 records and 35,863 records for suicide attempt and sleep-related behaviors, respectively, we conducted silver standard (diagnostic coding) and gold standard (manual chart review) validation. We showed Area Under the Precision-Recall Curve of ∼ 0.77 (95% CI 0.75-0.78) for suicide attempt and AUPR ∼ 0.31 (95% CI 0.28-0.34) for sleep-related behaviors. We also evaluated performance by coded race and demonstrated differences in performance by race were dissimilar across phenotypes and require algorithmovigilance and debiasing prior to implementation.
Collapse
|
23
|
Lee YS, Lee YJ, Ha IH. Real-world data analysis on effectiveness of integrative therapies: A practical guide to study design and data analysis using healthcare databases. Integr Med Res 2023; 12:101000. [PMID: 37953753 PMCID: PMC10637915 DOI: 10.1016/j.imr.2023.101000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
Real world data (RWD) is increasingly used to investigate health outcomes and treatment efficacy in the field of integrative medicine. Due to the fact that the majority of RWDs are not intended for research, their secondary use in research necessitates complex study designs to account for bias and confounding. To conduct a robust analysis of RWD in integrative medicine, a comprehensive study design process that reflects the characteristics of integrative therapies is necessary. In this paper, we present a guide for designing comparative effectiveness RWE research in integrative medicine. We discuss key factors to consider when selecting RWDs for research on integrative medicine. We provide practical steps for developing a research question, formulating the PICOT objectives (population, intervention, comparator, outcome, and time horizon), and selecting and defining covariates with a summary table. Specific study designs are depicted with corresponding diagrams. Finally, data analysis procedures are introduced. We hope this article clarifies the importance of RWE research design and related processes in order to improve the rigor of RWD studies in the field of integrative medicine research.
Collapse
Affiliation(s)
- Ye-Seul Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Korea
| | - Yoon Jae Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Korea
| | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, Korea
| |
Collapse
|
24
|
Gonzalez NR, Amin-Hanjani S, Bang OY, Coffey C, Du R, Fierstra J, Fraser JF, Kuroda S, Tietjen GE, Yaghi S. Adult Moyamoya Disease and Syndrome: Current Perspectives and Future Directions: A Scientific Statement From the American Heart Association/American Stroke Association. Stroke 2023; 54:e465-e479. [PMID: 37609846 DOI: 10.1161/str.0000000000000443] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Adult moyamoya disease and syndrome are rare disorders with significant morbidity and mortality. A writing group of experts was selected to conduct a literature search, summarize the current knowledge on the topic, and provide a road map for future investigation. The document presents an update in the definitions of moyamoya disease and syndrome, modern methods for diagnosis, and updated information on pathophysiology, epidemiology, and both medical and surgical treatment. Despite recent advancements, there are still many unresolved questions about moyamoya disease and syndrome, including lack of unified diagnostic criteria, reliable biomarkers, better understanding of the underlying pathophysiology, and stronger evidence for treatment guidelines. To advance progress in this area, it is crucial to acknowledge the limitations and weaknesses of current studies and explore new approaches, which are outlined in this scientific statement for future research strategies.
Collapse
|
25
|
Maro JC, Nguyen MD, Kolonoski J, Schoeplein R, Huang TY, Dutcher SK, Dal Pan GJ, Ball R. Six Years of the US Food and Drug Administration's Postmarket Active Risk Identification and Analysis System in the Sentinel Initiative: Implications for Real World Evidence Generation. Clin Pharmacol Ther 2023; 114:815-824. [PMID: 37391385 DOI: 10.1002/cpt.2979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/25/2023] [Indexed: 07/02/2023]
Abstract
Congress mandated the creation of a postmarket Active Risk Identification and Analysis (ARIA) system containing data on 100 million individuals for monitoring risks associated with drug and biologic products using data from disparate sources to complement the US Food and Drug Administration's (FDA's) existing postmarket capabilities. We report on the first 6 years of ARIA utilization in the Sentinel System (2016-2021). The FDA has used the ARIA system to evaluate 133 safety concerns; 54 of these evaluations have closed with regulatory determinations, whereas the rest remain in progress. If the ARIA system and the FDA's Adverse Event Reporting System are deemed insufficient to address a safety concern, then the FDA may issue a postmarket requirement to a product's manufacturer. One hundred ninety-seven ARIA insufficiency determinations have been made. The most common situation for which ARIA was found to be insufficient is the evaluation of adverse pregnancy and fetal outcomes following in utero drug exposure, followed by neoplasms and death. ARIA was most likely to be sufficient for thromboembolic events, which have high positive predictive value in claims data alone and do not require supplemental clinical data. The lessons learned from this experience illustrate the continued challenges using administrative claims data, especially to define novel clinical outcomes. This analysis can help to identify where more granular clinical data are needed to fill gaps to improve the use of real-world data for drug safety analyses and provide insights into what is needed to efficiently generate high-quality real-world evidence for efficacy.
Collapse
Affiliation(s)
- Judith C Maro
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael D Nguyen
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Joy Kolonoski
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan Schoeplein
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Ting-Ying Huang
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah K Dutcher
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gerald J Dal Pan
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Robert Ball
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
26
|
Dhingra LS, Shen M, Mangla A, Khera R. Cardiovascular Care Innovation through Data-Driven Discoveries in the Electronic Health Record. Am J Cardiol 2023; 203:136-148. [PMID: 37499593 PMCID: PMC10865722 DOI: 10.1016/j.amjcard.2023.06.104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/24/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
The electronic health record (EHR) represents a rich source of patient information, increasingly being leveraged for cardiovascular research. Although its primary use remains the seamless delivery of health care, the various longitudinally aggregated structured and unstructured data elements for each patient within the EHR can define the computational phenotypes of disease and care signatures and their association with outcomes. Although structured data elements, such as demographic characteristics, laboratory measurements, problem lists, and medications, are easily extracted, unstructured data are underused. The latter include free text in clinical narratives, documentation of procedures, and reports of imaging and pathology. Rapid scaling up of data storage and rapid innovation in natural language processing and computer vision can power insights from unstructured data streams. However, despite an array of opportunities for research using the EHR, specific expertise is necessary to adequately address confidentiality, accuracy, completeness, and heterogeneity challenges in EHR-based research. These often require methodological innovation and best practices to design and conduct successful research studies. Our review discusses these challenges and their proposed solutions. In addition, we highlight the ongoing innovations in federated learning in the EHR through a greater focus on common data models and discuss ongoing work that defines such an approach to large-scale, multicenter, federated studies. Such parallel improvements in technology and research methods enable innovative care and optimization of patient outcomes.
Collapse
Affiliation(s)
| | - Miles Shen
- Section of Cardiovascular Medicine, Department of Internal Medicine; Department of Internal Medicine
| | - Anjali Mangla
- Section of Cardiovascular Medicine, Department of Internal Medicine; Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine; Center for Outcomes Research and Evaluation (CORE), Yale New Haven Hospital, New Haven, Connecticut; Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut.; Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
27
|
Shu D, Li X, Her Q, Wong J, Li D, Wang R, Toh S. Combining meta-analysis with multiple imputation for one-step, privacy-protecting estimation of causal treatment effects in multi-site studies. Res Synth Methods 2023; 14:742-763. [PMID: 37527843 DOI: 10.1002/jrsm.1660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/10/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
Missing data complicates statistical analyses in multi-site studies, especially when it is not feasible to centrally pool individual-level data across sites. We combined meta-analysis with within-site multiple imputation for one-step estimation of the average causal effect (ACE) of a target population comprised of all individuals from all data-contributing sites within a multi-site distributed data network, without the need for sharing individual-level data to handle missing data. We considered two orders of combination and three choices of weights for meta-analysis, resulting in six approaches. The first three approaches, denoted as RR + metaF, RR + metaR and RR + std, first combined results from imputed data sets within each site using Rubin's rules and then meta-analyzed the combined results across sites using fixed-effect, random-effects and sample-standardization weights, respectively. The last three approaches, denoted as metaF + RR, metaR + RR and std + RR, first meta-analyzed results across sites separately for each imputation and then combined the meta-analysis results using Rubin's rules. Simulation results confirmed very good performance of RR + std and std + RR under various missing completely at random and missing at random settings. A direct application of the inverse-variance weighted meta-analysis based on site-specific ACEs can lead to biased results for the targeted network-wide ACE in the presence of treatment effect heterogeneity by site, demonstrating the need to clearly specify the target population and estimand and properly account for potential site heterogeneity in meta-analyses seeking to draw causal interpretations. An illustration using a large administrative claims database is presented.
Collapse
Affiliation(s)
- Di Shu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Xiaojuan Li
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Qoua Her
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Jenna Wong
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Dongdong Li
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Rui Wang
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Heyard R, Held L, Schneeweiss S, Wang SV. DESIGN DIFFERENCES EXPLAIN VARIATION IN RESULTS BETWEEN RANDOMIZED TRIALS AND THEIR NON-RANDOMIZED EMULATIONS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.13.23292601. [PMID: 37502999 PMCID: PMC10370236 DOI: 10.1101/2023.07.13.23292601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Objectives While randomized controlled trials (RCTs) are considered a standard for evidence on the efficacy of medical treatments, non-randomized real-world evidence (RWE) studies using data from health insurance claims or electronic health records can provide important complementary evidence. The use of RWE to inform decision-making has been questioned because of concerns regarding confounding in non-randomized studies and the use of secondary data. RCT-DUPLICATE was a demonstration project that emulated the design of 32 RCTs with non-randomized RWE studies. We sought to explore how emulation differences relate to variation in results between the RCT-RWE study pairs. Methods We include all RCT-RWE study pairs from RCT-DUPLICATE where the measure of effect was a hazard ratio and use exploratory meta-regression methods to explain differences and variation in the effect sizes between the results from the RCT and the RWE study. The considered explanatory variables are related to design and population differences. Results Most of the observed variation in effect estimates between RCT-RWE study pairs in this sample could be explained by three emulation differences in the meta-regression model: (i) in-hospital start of treatment (not observed in claims data), (ii) discontinuation of certain baseline therapies at randomization (not part of clinical practice), (iii) delayed onset of drug effects (missed by short medication persistence in clinical practice). Conclusions This analysis suggests that a substantial proportion of the observed variation between results from RCTs and RWE studies can be attributed to design emulation differences. (238 words).
Collapse
Affiliation(s)
- Rachel Heyard
- Center for Reproducible Science, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, 8001 Zurich, Switzerland
| | - Leonhard Held
- Center for Reproducible Science, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, 8001 Zurich, Switzerland
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 1620 Tremon St, Boston MA 02120
| | - Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 1620 Tremon St, Boston MA 02120
| |
Collapse
|
29
|
Fuller CC, Cosgrove A, Shinde M, Rosen E, Haffenreffer K, Hague C, McLean LE, Perlin J, Poland RE, Sands KE, Pratt N, Bright P, Platt R, Cocoros NM, Dutcher SK. Treatment and care received by children hospitalized with COVID-19 in a large hospital network in the United States, February 2020 to September 2021. PLoS One 2023; 18:e0288284. [PMID: 37432951 DOI: 10.1371/journal.pone.0288284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
We described care received by hospitalized children with COVID-19 or multi-system inflammatory syndrome (MIS-C) prior to the 2021 COVID-19 Omicron variant surge in the US. We identified hospitalized children <18 years of age with a COVID-19 or MIS-C diagnosis (COVID-19 not required), separately, from February 2020-September 2021 (n = 126 hospitals). We described high-risk conditions, inpatient treatments, and complications among these groups. Among 383,083 pediatric hospitalizations, 2,186 had COVID-19 and 395 had MIS-C diagnosis. Less than 1% had both COVID-19 and MIS-C diagnosis (n = 154). Over half were >6 years old (54% COVID-19, 70% MIS-C). High-risk conditions included asthma (14% COVID-19, 11% MIS-C), and obesity (9% COVID-19, 10% MIS-C). Pulmonary complications in children with COVID-19 included viral pneumonia (24%) and acute respiratory failure (11%). In reference to children with COVID-19, those with MIS-C had more hematological disorders (62% vs 34%), sepsis (16% vs 6%), pericarditis (13% vs 2%), myocarditis (8% vs 1%). Few were ventilated or died, but some required oxygen support (38% COVID-19, 45% MIS-C) or intensive care (42% COVID-19, 69% MIS-C). Treatments included: methylprednisolone (34% COVID-19, 75% MIS-C), dexamethasone (25% COVID-19, 15% MIS-C), remdesivir (13% COVID-19, 5% MIS-C). Antibiotics (50% COVID-19, 68% MIS-C) and low-molecular weight heparin (17% COVID-19, 34% MIS-C) were frequently administered. Markers of illness severity among hospitalized children with COVID-19 prior to the 2021 Omicron surge are consistent with previous studies. We report important trends on treatments in hospitalized children with COVID-19 to improve the understanding of real-world treatment patterns in this population.
Collapse
Affiliation(s)
- Candace C Fuller
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Austin Cosgrove
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Mayura Shinde
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Edward Rosen
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Katie Haffenreffer
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Christian Hague
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Laura E McLean
- HCA Healthcare, Nashville, Tennessee, United States of America
| | - Jonathan Perlin
- HCA Healthcare, Nashville, Tennessee, United States of America
| | - Russell E Poland
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
- HCA Healthcare, Nashville, Tennessee, United States of America
| | - Kenneth E Sands
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
- HCA Healthcare, Nashville, Tennessee, United States of America
| | - Natasha Pratt
- US Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Patricia Bright
- US Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Richard Platt
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Noelle M Cocoros
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, United States of America
| | - Sarah K Dutcher
- US Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
30
|
Muzaffar AF, Abdul-Massih S, Stevenson JM, Alvarez-Arango S. Use of the Electronic Health Record for Monitoring Adverse Drug Reactions. Curr Allergy Asthma Rep 2023; 23:417-426. [PMID: 37191903 DOI: 10.1007/s11882-023-01087-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE OF REVIEW Adverse drug reactions (ADRs) are a significant cause of morbidity and mortality. The electronic health record (EHR) provides an opportunity to monitor ADRs, mainly through the utilization of drug allergy data and pharmacogenomics. This review article explores the current use of the EHR for ADR monitoring and highlights areas that require improvement. RECENT FINDINGS Recent research has identified several issues with using EHR for ADR monitoring. These include the lack of standardization between EHR systems, specificity in data entry options, incomplete and inaccurate documentation, and alert fatigue. These issues can limit the effectiveness of ADR monitoring and compromise patient safety. The EHR has great potential for monitoring ADR but needs significant updates to improve patient safety and optimize care. Future research should concentrate on developing standardized documentation and clinical decision support systems within EHRs. Healthcare professionals should also be educated on the significance of accurate and complete ADR monitoring.
Collapse
Affiliation(s)
- Anum F Muzaffar
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Abdul-Massih
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James M Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Santiago Alvarez-Arango
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Hopkins Bayview Circle, 5501, MD, 21224, Baltimore, USA.
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
31
|
Heo S, Yu JY, Kang EA, Shin H, Ryu K, Kim C, Chegal Y, Jung H, Lee S, Park RW, Kim K, Hwangbo Y, Lee JH, Park YR. Development and Verification of Time-Series Deep Learning for Drug-Induced Liver Injury Detection in Patients Taking Angiotensin II Receptor Blockers: A Multicenter Distributed Research Network Approach. Healthc Inform Res 2023; 29:246-255. [PMID: 37591680 PMCID: PMC10440200 DOI: 10.4258/hir.2023.29.3.246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/19/2023] Open
Abstract
OBJECTIVES The objective of this study was to develop and validate a multicenter-based, multi-model, time-series deep learning model for predicting drug-induced liver injury (DILI) in patients taking angiotensin receptor blockers (ARBs). The study leveraged a national-level multicenter approach, utilizing electronic health records (EHRs) from six hospitals in Korea. METHODS A retrospective cohort analysis was conducted using EHRs from six hospitals in Korea, comprising a total of 10,852 patients whose data were converted to the Common Data Model. The study assessed the incidence rate of DILI among patients taking ARBs and compared it to a control group. Temporal patterns of important variables were analyzed using an interpretable timeseries model. RESULTS The overall incidence rate of DILI among patients taking ARBs was found to be 1.09%. The incidence rates varied for each specific ARB drug and institution, with valsartan having the highest rate (1.24%) and olmesartan having the lowest rate (0.83%). The DILI prediction models showed varying performance, measured by the average area under the receiver operating characteristic curve, with telmisartan (0.93), losartan (0.92), and irbesartan (0.90) exhibiting higher classification performance. The aggregated attention scores from the models highlighted the importance of variables such as hematocrit, albumin, prothrombin time, and lymphocytes in predicting DILI. CONCLUSIONS Implementing a multicenter-based timeseries classification model provided evidence that could be valuable to clinicians regarding temporal patterns associated with DILI in ARB users. This information supports informed decisions regarding appropriate drug use and treatment strategies.
Collapse
Affiliation(s)
- Suncheol Heo
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul,
Korea
| | - Jae Yong Yu
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul,
Korea
| | - Eun Ae Kang
- Medical Informatics Collaborative Unit, Department of Research Affairs, Yonsei University College of Medicine, Seoul,
Korea
| | - Hyunah Shin
- Healthcare Data Science Center, Konyang University Hospital, Daejeon,
Korea
| | - Kyeongmin Ryu
- Healthcare Data Science Center, Konyang University Hospital, Daejeon,
Korea
| | - Chungsoo Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Seoul,
Korea
| | - Yebin Chegal
- Department of Statistics, Korea University, Suwon,
Korea
| | - Hyojung Jung
- Healthcare AI Team, National Cancer Center, Goyang,
Korea
| | - Suehyun Lee
- Healthcare Data Science Center, Konyang University Hospital, Daejeon,
Korea
| | - Rae Woong Park
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Seoul,
Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul,
Korea
| | - Yul Hwangbo
- Healthcare AI Team, National Cancer Center, Goyang,
Korea
| | - Jae-Hyun Lee
- Division of Allergy and Immunology, Department of Internal Medicine, Institute of Allergy, Yonsei University College of Medicine, Seoul,
Korea
| | - Yu Rang Park
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul,
Korea
| |
Collapse
|
32
|
Jackson DB, Racz R, Kim S, Brock S, Burkhart K. Rewiring Drug Research and Development through Human Data-Driven Discovery (HD 3). Pharmaceutics 2023; 15:1673. [PMID: 37376121 PMCID: PMC10303279 DOI: 10.3390/pharmaceutics15061673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
In an era of unparalleled technical advancement, the pharmaceutical industry is struggling to transform data into increased research and development efficiency, and, as a corollary, new drugs for patients. Here, we briefly review some of the commonly discussed issues around this counterintuitive innovation crisis. Looking at both industry- and science-related factors, we posit that traditional preclinical research is front-loading the development pipeline with data and drug candidates that are unlikely to succeed in patients. Applying a first principles analysis, we highlight the critical culprits and provide suggestions as to how these issues can be rectified through the pursuit of a Human Data-driven Discovery (HD3) paradigm. Consistent with other examples of disruptive innovation, we propose that new levels of success are not dependent on new inventions, but rather on the strategic integration of existing data and technology assets. In support of these suggestions, we highlight the power of HD3, through recently published proof-of-concept applications in the areas of drug safety analysis and prediction, drug repositioning, the rational design of combination therapies and the global response to the COVID-19 pandemic. We conclude that innovators must play a key role in expediting the path to a largely human-focused, systems-based approach to drug discovery and research.
Collapse
Affiliation(s)
| | - Rebecca Racz
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA; (R.R.); (K.B.)
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL 32827, USA;
| | | | - Keith Burkhart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA; (R.R.); (K.B.)
| |
Collapse
|
33
|
Lovis C, Siebel J, Fuhrmann S, Fischer A, Sedlmayr M, Weidner J, Bathelt F. Assessment and Improvement of Drug Data Structuredness From Electronic Health Records: Algorithm Development and Validation. JMIR Med Inform 2023; 11:e40312. [PMID: 36696159 PMCID: PMC9909518 DOI: 10.2196/40312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Digitization offers a multitude of opportunities to gain insights into current diagnostics and therapies from retrospective data. In this context, real-world data and their accessibility are of increasing importance to support unbiased and reliable research on big data. However, routinely collected data are not readily usable for research owing to the unstructured nature of health care systems and a lack of interoperability between these systems. This challenge is evident in drug data. OBJECTIVE This study aimed to present an approach that identifies and increases the structuredness of drug data while ensuring standardization according to Anatomical Therapeutic Chemical (ATC) classification. METHODS Our approach was based on available drug prescriptions and a drug catalog and consisted of 4 steps. First, we performed an initial analysis of the structuredness of local drug data to define a point of comparison for the effectiveness of the overall approach. Second, we applied 3 algorithms to unstructured data that translated text into ATC codes based on string comparisons in terms of ingredients and product names and performed similarity comparisons based on Levenshtein distance. Third, we validated the results of the 3 algorithms with expert knowledge based on the 1000 most frequently used prescription texts. Fourth, we performed a final validation to determine the increased degree of structuredness. RESULTS Initially, 47.73% (n=843,980) of 1,768,153 drug prescriptions were classified as structured. With the application of the 3 algorithms, we were able to increase the degree of structuredness to 85.18% (n=1,506,059) based on the 1000 most frequent medication prescriptions. In this regard, the combination of algorithms 1, 2, and 3 resulted in a correctness level of 100% (with 57,264 ATC codes identified), algorithms 1 and 3 resulted in 99.6% (with 152,404 codes identified), and algorithms 1 and 2 resulted in 95.9% (with 39,472 codes identified). CONCLUSIONS As shown in the first analysis steps of our approach, the availability of a product catalog to select during the documentation process is not sufficient to generate structured data. Our 4-step approach reduces the problems and reliably increases the structuredness automatically. Similarity matching shows promising results, particularly for entries with no connection to a product catalog. However, further enhancement of the correctness of such a similarity matching algorithm needs to be investigated in future work.
Collapse
Affiliation(s)
| | - Joscha Siebel
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Saskia Fuhrmann
- Center for Evidence-Based Healthcare, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Hospital Pharmacy, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Andreas Fischer
- Hospital Pharmacy, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Sedlmayr
- Center for Evidence-Based Healthcare, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jens Weidner
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Franziska Bathelt
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
34
|
Wang SV, Pottegård A, Crown W, Arlett P, Ashcroft DM, Benchimol EI, Berger ML, Crane G, Goettsch W, Hua W, Kabadi S, Kern DM, Kurz X, Langan S, Nonaka T, Orsini L, Perez-Gutthann S, Pinheiro S, Pratt N, Schneeweiss S, Toussi M, Williams RJ. HARmonized Protocol Template to Enhance Reproducibility of hypothesis evaluating real-world evidence studies on treatment effects: A good practices report of a joint ISPE/ISPOR task force. Pharmacoepidemiol Drug Saf 2023; 32:44-55. [PMID: 36215113 PMCID: PMC9771861 DOI: 10.1002/pds.5507] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023]
Abstract
PROBLEM Ambiguity in communication of key study parameters limits the utility of real-world evidence (RWE) studies in healthcare decision-making. Clear communication about data provenance, design, analysis, and implementation is needed. This would facilitate reproducibility, replication in independent data, and assessment of potential sources of bias. WHAT WE DID The International Society for Pharmacoepidemiology (ISPE) and ISPOR-The Professional Society for Health Economics and Outcomes Research (ISPOR) convened a joint task force, including representation from key international stakeholders, to create a harmonized protocol template for RWE studies that evaluate a treatment effect and are intended to inform decision-making. The template builds on existing efforts to improve transparency and incorporates recent insights regarding the level of detail needed to enable RWE study reproducibility. The overarching principle was to reach for sufficient clarity regarding data, design, analysis, and implementation to achieve 3 main goals. One, to help investigators thoroughly consider, then document their choices and rationale for key study parameters that define the causal question (e.g., target estimand), two, to facilitate decision-making by enabling reviewers to readily assess potential for biases related to these choices, and three, to facilitate reproducibility. STRATEGIES TO DISSEMINATE AND FACILITATE USE Recognizing that the impact of this harmonized template relies on uptake, we have outlined a plan to introduce and pilot the template with key international stakeholders over the next 2 years. CONCLUSION The HARmonized Protocol Template to Enhance Reproducibility (HARPER) helps to create a shared understanding of intended scientific decisions through a common text, tabular and visual structure. The template provides a set of core recommendations for clear and reproducible RWE study protocols and is intended to be used as a backbone throughout the research process from developing a valid study protocol, to registration, through implementation and reporting on those implementation decisions.
Collapse
Affiliation(s)
| | | | | | | | | | - Eric I Benchimol
- 1. Department of Paediatrics and Institute of Health Policy, Management and Evaluation, The Hospital for Sick Children, University of Toronto, Toronto, Canada,2. Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Canada,3. ICES, Toronto, Canada
| | | | | | - Wim Goettsch
- The National Health Care Institute, Diemen, and Utrecht University, Utrecht, the Netherlands
| | - Wei Hua
- US Food and Drug Administration
| | | | | | | | | | | | | | | | | | - Nicole Pratt
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia
| | | | | | | |
Collapse
|
35
|
Lu J, Wang G, Ying X, Li Z. A novel drug selection decision support model based on real-world medical data by the hybrid entropic weight TOPSIS method. Technol Health Care 2023; 31:691-703. [PMID: 36278366 DOI: 10.3233/thc-220355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
BACKGROUND The medicine selection method is a critical and challenging issue in medical insurance decision-making. OBJECTIVES This study proposed a real-world data-based multi-criteria decision analysis (MCDA) model with a hybrid entropic weight Technique for Order of Preference by Similarity to Ideal Solution (TOPSIS) algorithms to select satisfactory drugs. METHODS The evaluation index includes two levels: primary criteria and sub-criteria. Firstly, we proposed six primary criteria to form the value health framework. The primary criteria's weights were derived from the policymakers' questionnaire. Meanwhile, clinically relevant sub-criteria were derived from high-quality (screened by GRADE scores) clinical-research literature. Their weights are determined by the entropy weight (EW) algorithm. Secondly, we split the primary criteria into six mini-EW-TOPSIS models. Then, we obtained six ideal closeness degree scores (ICDS) for each candidate drug. Thirdly, we get the total utility score by linear weighting the ICDS. The higher the utility score, the higher the ranking. RESULTS A national multicenter real-world case study of the ranking of four generic antibiotics validated the proposed model. This model is verified by comparative experiments and sensitivity analysis. The whole ranking model was consistent and reliable. Based on these results, medical policymakers can intuitively and easily understand the characteristics of each drug to facilitate follow-up drug policy-making. CONCLUSION The ranking algorithm combines the objective characteristics of medicine and policy makers' opinions, which can improve the applicability of the results. This model can help decision-makers, clinicians, and related researchers better understand the drug assessment process.
Collapse
Affiliation(s)
- Jinmiao Lu
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| | - Guangfei Wang
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaohua Ying
- NHC Key Laboratory of Health Technology Assessment, Department of Health Economics, School of Public Health, Fudan University, Shanghai, China
| | - Zhiping Li
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
36
|
Wang T, Yan Y, Xiang S, Tan J, Yang C, Zhao W. A comparative study of antihypertensive drugs prediction models for the elderly based on machine learning algorithms. Front Cardiovasc Med 2022; 9:1056263. [PMID: 36531716 PMCID: PMC9753549 DOI: 10.3389/fcvm.2022.1056263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/17/2022] [Indexed: 11/04/2023] Open
Abstract
Background Globally, blood pressure management strategies were ineffective, and a low percentage of patients receiving hypertension treatment had their blood pressure controlled. In this study, we aimed to build a medication prediction model by correlating patient attributes with medications to help physicians quickly and rationally match appropriate medications. Methods We collected clinical data from elderly hypertensive patients during hospitalization and combined statistical methods and machine learning (ML) algorithms to filter out typical indicators. We constructed five ML models to evaluate all datasets using 5-fold cross-validation. Include random forest (RF), support vector machine (SVM), light gradient boosting machine (LightGBM), artificial neural network (ANN), and naive Bayes (NB) models. And the performance of the models was evaluated using the micro-F1 score. Results Our experiments showed that by statistical methods and ML algorithms for feature selection, we finally selected Age, SBP, DBP, Lymph, RBC, HCT, MCHC, PLT, AST, TBIL, Cr, UA, Urea, K, Na, Ga, TP, GLU, TC, TG, γ-GT, Gender, HTN CAD, and RI as feature metrics of the models. LightGBM had the best prediction performance with the micro-F1 of 78.45%, which was higher than the other four models. Conclusion LightGBM model has good results in predicting antihypertensive medication regimens, and the model can be beneficial in improving the personalization of hypertension treatment.
Collapse
Affiliation(s)
- Tiantian Wang
- School of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Yongjie Yan
- Medical Records and Statistics Office, The Third Affiliated Hospital of Army Medical University, Chongqing, China
| | - Shoushu Xiang
- Medical Records and Statistics Room, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Juntao Tan
- Operation Management Office, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Yang
- School of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Wenlong Zhao
- School of Medical Informatics, Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Brown JS, Mendelsohn AB, Nam YH, Maro JC, Cocoros NM, Rodriguez-Watson C, Lockhart CM, Platt R, Ball R, Dal Pan GJ, Toh S. The US Food and Drug Administration Sentinel System: a national resource for a learning health system. J Am Med Inform Assoc 2022; 29:2191-2200. [PMID: 36094070 PMCID: PMC9667154 DOI: 10.1093/jamia/ocac153] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 07/23/2023] Open
Abstract
The US Food and Drug Administration (FDA) created the Sentinel System in response to a requirement in the FDA Amendments Act of 2007 that the agency establish a system for monitoring risks associated with drug and biologic products using data from disparate sources. The Sentinel System has completed hundreds of analyses, including many that have directly informed regulatory decisions. The Sentinel System also was designed to support a national infrastructure for a learning health system. Sentinel governance and guiding principles were designed to facilitate Sentinel's role as a national resource. The Sentinel System infrastructure now supports multiple non-FDA projects for stakeholders ranging from regulated industry to other federal agencies, international regulators, and academics. The Sentinel System is a working example of a learning health system that is expanding with the potential to create a global learning health system that can support medical product safety assessments and other research.
Collapse
Affiliation(s)
- Jeffrey S Brown
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron B Mendelsohn
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Young Hee Nam
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Judith C Maro
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Noelle M Cocoros
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Carla Rodriguez-Watson
- Reagan-Udall Foundation for the Food and Drug Administration, Washington, District of Columbia, USA
| | - Catherine M Lockhart
- Biologics and Biosimilars Collective Intelligence Consortium, Alexandria, Virginia, USA
| | - Richard Platt
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Ball
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gerald J Dal Pan
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sengwee Toh
- Corresponding Author: Sengwee Toh, ScD, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, 401 Park Drive, Suite 401 East, Boston, MA 02215, USA;
| |
Collapse
|
38
|
Wang SV, Sreedhara SK, Bessette LG, Schneeweiss S. Understanding variation in the results of real-world evidence studies that seem to address the same question. J Clin Epidemiol 2022; 151:161-170. [PMID: 36075314 DOI: 10.1016/j.jclinepi.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Multiple database studies on the same question, conducted by different investigators using different approaches or different data sources, can be considered sensitivity analyses for the same causal treatment effect question. We evaluated the contribution of alternative study design parameters and analysis choices to variation in estimates of the risk of major bleeding with dabigatran compared with warfarin. STUDY DESIGN AND SETTING We followed a 7-step process: (1) identify published studies asking the same question, (2) independently reproduce selected studies in the same data sources as the original authors, (3) contact original authors, (4) evaluate validity, (5) document critical study parameter specifications, (6) implement a designed matrix of variations in study parameters based on the original studies, and (7) evaluate contributors to variation in results. RESULTS Most variation remained unexplained (60-88%). Of the explained variation, two-thirds were related to data and population differences, and one-third were related to the use of alternative study design and analysis parameters. Among these, the most prominent were differences in outcome algorithms and criteria used to define follow-up. CONCLUSION When making policy decisions based on database study findings, it is important to evaluate the validity, consistency, and robustness of results to alternative design and analysis decisions.
Collapse
Affiliation(s)
- Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, 1630 Tremont St Suite 303, Boston, MA 02120, USA; Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | | | - Lily G Bessette
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Boston, MA, USA
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital; Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Wang SV, Pottegård A, Crown W, Arlett P, Ashcroft DM, Benchimol EI, Berger ML, Crane G, Goettsch W, Hua W, Kabadi S, Kern DM, Kurz X, Langan S, Nonaka T, Orsini L, Perez-Gutthann S, Pinheiro S, Pratt N, Schneeweiss S, Toussi M, Williams RJ. HARmonized Protocol Template to Enhance Reproducibility of Hypothesis Evaluating Real-World Evidence Studies on Treatment Effects: A Good Practices Report of a Joint ISPE/ISPOR Task Force. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:1663-1672. [PMID: 36241338 DOI: 10.1016/j.jval.2022.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Ambiguity in communication of key study parameters limits the utility of real-world evidence (RWE) studies in healthcare decision-making. Clear communication about data provenance, design, analysis, and implementation is needed. This would facilitate reproducibility, replication in independent data, and assessment of potential sources of bias. METHODS The International Society for Pharmacoepidemiology (ISPE) and ISPOR-The Professional Society for Health Economics and Outcomes Research (ISPOR) convened a joint task force, including representation from key international stakeholders, to create a harmonized protocol template for RWE studies that evaluate a treatment effect and are intended to inform decision-making. The template builds on existing efforts to improve transparency and incorporates recent insights regarding the level of detail needed to enable RWE study reproducibility. The over-arching principle was to reach for sufficient clarity regarding data, design, analysis, and implementation to achieve 3 main goals. One, to help investigators thoroughly consider, then document their choices and rationale for key study parameters that define the causal question (e.g., target estimand), two, to facilitate decision-making by enabling reviewers to readily assess potential for biases related to these choices, and three, to facilitate reproducibility. STRATEGIES TO DISSEMINATE AND FACILITATE USE Recognizing that the impact of this harmonized template relies on uptake, we have outlined a plan to introduce and pilot the template with key international stakeholders over the next 2 years. CONCLUSION The HARmonized Protocol Template to Enhance Reproducibility (HARPER) helps to create a shared understanding of intended scientific decisions through a common text, tabular and visual structure. The template provides a set of core recommendations for clear and reproducible RWE study protocols and is intended to be used as a backbone throughout the research process from developing a valid study protocol, to registration, through implementation and reporting on those implementation decisions.
Collapse
Affiliation(s)
- Shirley V Wang
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | - Eric I Benchimol
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada; ICES, Toronto, Ontario, Canada; Department of Paediatrics and Institute of Health Policy, Management and Evaluation, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Wim Goettsch
- The National Health Care Institute, Diemen, The Netherlands; Utrecht University, Utrecht, The Netherlands
| | - Wei Hua
- US Food and Drug Administration, Silver Springs, Maryland, USA
| | - Shaum Kabadi
- Sanofi-Aventis US LLC, North Potomac, Maryland, USA
| | - David M Kern
- Janssen Research & Development, LLC, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | - Simone Pinheiro
- US Food and Drug Administration, Silver Springs, Maryland, USA
| | - Nicole Pratt
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, South Australia, Australia
| | | | | | | |
Collapse
|
40
|
Kumar S, Arnold M, James G, Padman R. Developing a common data model approach for DISCOVER CKD: A retrospective, global cohort of real-world patients with chronic kidney disease. PLoS One 2022; 17:e0274131. [PMID: 36173958 PMCID: PMC9521926 DOI: 10.1371/journal.pone.0274131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To describe a flexible common data model (CDM) approach that can be efficiently tailored to study-specific needs to facilitate pooled patient-level analysis and aggregated/meta-analysis of routinely collected retrospective patient data from disparate data sources; and to detail the application of this CDM approach to the DISCOVER CKD retrospective cohort, a longitudinal database of routinely collected (secondary) patient data of individuals with chronic kidney disease (CKD). METHODS The flexible CDM approach incorporated three independent, exchangeable components that preceded data mapping and data model implementation: (1) standardized code lists (unifying medical events from different coding systems); (2) laboratory unit harmonization tables; and (3) base cohort definitions. Events between different coding vocabularies were not mapped code-to-code; for each data source, code lists of labels were curated at the entity/event level. A study team of epidemiologists, clinicians, informaticists, and data scientists were included within the validation of each component. RESULTS Applying the CDM to the DISCOVER CKD retrospective cohort, secondary data from 1,857,593 patients with CKD were harmonized from five data sources, across three countries, into a discrete database for rapid real-world evidence generation. CONCLUSIONS This flexible CDM approach facilitates evidence generation from real-world data within the DISCOVER CKD retrospective cohort, providing novel insights into the epidemiology of CKD that may expedite improvements in diagnosis, prognosis, early intervention, and disease management. The adaptable architecture of this CDM approach ensures scalable, fast, and efficient application within other therapy areas to facilitate the combined analysis of different types of secondary data from multiple, heterogeneous sources.
Collapse
Affiliation(s)
- Supriya Kumar
- Real World Evidence Data and Analytics, BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, MD, United States of America
| | - Matthew Arnold
- Real World Evidence Data and Analytics, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom
| | - Glen James
- Formerly Cardiovascular, Renal, Metabolism & Epidemiology, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom
| | - Rema Padman
- Heinz College of Information Systems and Public Policy, Carnegie Mellon University, Pittsburgh, PA, United States of America
| |
Collapse
|
41
|
Shinde M, Rodriguez-Watson C, Zhang TC, Carrell DS, Mendelsohn AB, Nam YH, Carruth A, Petronis KR, McMahill-Walraven CN, Jamal-Allial A, Nair V, Pawloski PA, Hickman A, Brown MT, Francis J, Hornbuckle K, Brown JS, Mo J. Patient characteristics, pain treatment patterns, and incidence of total joint replacement in a US population with osteoarthritis. BMC Musculoskelet Disord 2022; 23:883. [PMID: 36151530 PMCID: PMC9502954 DOI: 10.1186/s12891-022-05823-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Currently available medications for chronic osteoarthritis pain are only moderately effective, and their use is limited in many patients because of serious adverse effects and contraindications. The primary surgical option for osteoarthritis is total joint replacement (TJR). The objectives of this study were to describe the treatment history of patients with osteoarthritis receiving prescription pain medications and/or intra-articular corticosteroid injections, and to estimate the incidence of TJR in these patients. METHODS This retrospective, multicenter, cohort study utilized health plan administrative claims data (January 1, 2013, through December 31, 2019) of adult patients with osteoarthritis in the Innovation in Medical Evidence Development and Surveillance Distributed Database, a subset of the US FDA Sentinel Distributed Database. Patients were analyzed in two cohorts: those with prevalent use of "any pain medication" (prescription non-steroidal anti-inflammatory drugs [NSAIDs], opioids, and/or intra-articular corticosteroid injections) using only the first qualifying dispensing (index date); and those with prevalent use of "each specific pain medication class" with all qualifying treatment episodes identified. RESULTS Among 1 992 670 prevalent users of "any pain medication", pain medications prescribed on the index date were NSAIDs (596 624 [29.9%] patients), opioids (1 161 806 [58.3%]), and intra-articular corticosteroids (323 459 [16.2%]). Further, 92 026 patients received multiple pain medications on the index date, including 71 632 (3.6%) receiving both NSAIDs and opioids. Altogether, 20.6% of patients used an NSAID at any time following an opioid index dispensing and 17.2% used an opioid following an NSAID index dispensing. The TJR incidence rates per 100 person-years (95% confidence interval [CI]) were 3.21 (95% CI: 3.20-3.23) in the "any pain medication" user cohort, and among those receiving "each specific pain medication class" were NSAIDs, 4.63 (95% CI: 4.58-4.67); opioids, 7.45 (95% CI: 7.40-7.49); and intra-articular corticosteroids, 8.05 (95% CI: 7.97-8.13). CONCLUSIONS In patients treated with prescription medications for osteoarthritis pain, opioids were more commonly prescribed at index than NSAIDs and intra-articular corticosteroid injections. Of the pain medication classes examined, the incidence of TJR was highest in patients receiving intra-articular corticosteroids and lowest in patients receiving NSAIDs.
Collapse
Affiliation(s)
- Mayura Shinde
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA.
| | | | - Tancy C Zhang
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - David S Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Aaron B Mendelsohn
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - Young Hee Nam
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - Amanda Carruth
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Vinit Nair
- Humana Healthcare Research Inc, Louisville, KY, USA
| | | | | | | | | | | | - Jeffrey S Brown
- Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
42
|
Wang SV, Sreedhara SK, Schneeweiss S. Reproducibility of real-world evidence studies using clinical practice data to inform regulatory and coverage decisions. Nat Commun 2022; 13:5126. [PMID: 36045130 PMCID: PMC9430007 DOI: 10.1038/s41467-022-32310-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 07/26/2022] [Indexed: 11/26/2022] Open
Abstract
Studies that generate real-world evidence on the effects of medical products through analysis of digital data collected in clinical practice provide key insights for regulators, payers, and other healthcare decision-makers. Ensuring reproducibility of such findings is fundamental to effective evidence-based decision-making. We reproduce results for 150 studies published in peer-reviewed journals using the same healthcare databases as original investigators and evaluate the completeness of reporting for 250. Original and reproduction effect sizes were positively correlated (Pearson's correlation = 0.85), a strong relationship with some room for improvement. The median and interquartile range for the relative magnitude of effect (e.g., hazard ratiooriginal/hazard ratioreproduction) is 1.0 [0.9, 1.1], range [0.3, 2.1]. While the majority of results are closely reproduced, a subset are not. The latter can be explained by incomplete reporting and updated data. Greater methodological transparency aligned with new guidance may further improve reproducibility and validity assessment, thus facilitating evidence-based decision-making. Study registration number: EUPAS19636.
Collapse
Affiliation(s)
- Shirley V Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | | | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Zhang D, Song J, Dharmarajan S, Jung TH, Lee H, Ma Y, Zhang R, Levenson M. The Use of Machine Learning in Regulatory Drug Safety Evaluation. Stat Biopharm Res 2022. [DOI: 10.1080/19466315.2022.2108135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Di Zhang
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Jaejoon Song
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Sai Dharmarajan
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Tae Hyun Jung
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Hana Lee
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Yong Ma
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Rongmei Zhang
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Mark Levenson
- Division of Biometrics VII, Office of Biostatistics, Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| |
Collapse
|
44
|
Lewis JT, Stephens J, Musick B, Brown S, Malateste K, Ostinelli CHD, Maxwell N, Jayathilake K, Shi Q, Brazier E, Kariminia A, Hogan B, Duda SN. The IeDEA harmonist data toolkit: A data quality and data sharing solution for a global HIV research consortium. J Biomed Inform 2022; 131:104110. [PMID: 35680074 PMCID: PMC9893518 DOI: 10.1016/j.jbi.2022.104110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 02/04/2023]
Abstract
We describe the design, implementation, and impact of a data harmonization, data quality checking, and dynamic report generation application in an international observational HIV research network. The IeDEA Harmonist Data Toolkit is a web-based application written in the open source programming language R, employs the R/Shiny and RMarkdown packages, and leverages the REDCap data collection platform for data model definition and user authentication. The Toolkit performs data quality checks on uploaded datasets, checks for conformance with the network's common data model, displays the results both interactively and in downloadable reports, and stores approved datasets in secure cloud storage for retrieval by the requesting investigator. Including stakeholders and users in the design process was key to the successful adoption of the application. A survey of regional data managers as well as initial usage metrics indicate that the Toolkit saves time and results in improved data quality, with a 61% mean reduction in the number of error records in a dataset. The generalized application design allows the Toolkit to be easily adapted to other research networks.
Collapse
Affiliation(s)
- Judith T Lewis
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN USA,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jeremy Stephens
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Beverly Musick
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Steven Brown
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Karen Malateste
- French National Research Institute for Sustainable Development (IRD), Inserm, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Cam Ha Dao Ostinelli
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Nicola Maxwell
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Karu Jayathilake
- Department of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qiuhu Shi
- Department of Public Health, New York Medical College, Valhalla, NY, USA
| | - Ellen Brazier
- Institute for Implementation Science in Population Health, City University of New York, New York, New York, USA,Graduate School of Public Health and Health Policy, City University of New York, New York, New York, USA
| | | | - Brenna Hogan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Stephany N Duda
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN USA,Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
45
|
Edmondson MJ, Luo C, Nazmul Islam M, Sheils NE, Buresh J, Chen Z, Bian J, Chen Y. Distributed Quasi-Poisson regression algorithm for modeling multi-site count outcomes in distributed data networks. J Biomed Inform 2022; 131:104097. [PMID: 35643272 PMCID: PMC11874216 DOI: 10.1016/j.jbi.2022.104097] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Observational studies incorporating real-world data from multiple institutions facilitate study of rare outcomes or exposures and improve generalizability of results. Due to privacy concerns surrounding patient-level data sharing across institutions, methods for performing regression analyses distributively are desirable. Meta-analysis of institution-specific estimates is commonly used, but has been shown to produce biased estimates in certain settings. While distributed regression methods are increasingly available, methods for analyzing count outcomes are currently limited. Count data in practice are commonly subject to overdispersion, exhibiting greater variability than expected under a given statistical model. OBJECTIVE We propose a novel computational method, a one-shot distributed algorithm for quasi-Poisson regression (ODAP), to distributively model count outcomes while accounting for overdispersion. METHODS ODAP incorporates a surrogate likelihood approach to perform distributed quasi-Poisson regression without requiring patient-level data sharing, only requiring sharing of aggregate data from each participating institution. ODAP requires at most three rounds of non-iterative communication among institutions to generate coefficient estimates and corresponding standard errors. In simulations, we evaluate ODAP under several data scenarios possible in multi-site analyses, comparing ODAP and meta-analysis estimates in terms of error relative to pooled regression estimates, considered the gold standard. In a proof-of-concept real-world data analysis, we similarly compare ODAP and meta-analysis in terms of relative error to pooled estimatation using data from the OneFlorida Clinical Research Consortium, modeling length of stay in COVID-19 patients as a function of various patient characteristics. In a second proof-of-concept analysis, using the same outcome and covariates, we incorporate data from the UnitedHealth Group Clinical Discovery Database together with the OneFlorida data in a distributed analysis to compare estimates produced by ODAP and meta-analysis. RESULTS In simulations, ODAP exhibited negligible error relative to pooled regression estimates across all settings explored. Meta-analysis estimates, while largely unbiased, were increasingly variable as heterogeneity in the outcome increased across institutions. When baseline expected count was 0.2, relative error for meta-analysis was above 5% in 25% of iterations (250/1000), while the largest relative error for ODAP in any iteration was 3.59%. In our proof-of-concept analysis using only OneFlorida data, ODAP estimates were closer to pooled regression estimates than those produced by meta-analysis for all 15 covariates. In our distributed analysis incorporating data from both OneFlorida and the UnitedHealth Group Clinical Discovery Database, ODAP and meta-analysis estimates were largely similar, while some differences in estimates (as large as 13.8%) could be indicative of bias in meta-analytic estimates. CONCLUSIONS ODAP performs privacy-preserving, communication-efficient distributed quasi-Poisson regression to analyze count outcomes using data stored within multiple institutions. Our method produces estimates nearly matching pooled regression estimates and sometimes more accurate than meta-analysis estimates, most notably in settings with relatively low counts and high outcome heterogeneity across institutions.
Collapse
Affiliation(s)
- Mackenzie J Edmondson
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Chongliang Luo
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - John Buresh
- Optum Labs at UnitedHealth Group, Minnetonka, MN, USA
| | - Zhaoyi Chen
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA; Cancer Informatics Shared Resource, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA; Cancer Informatics Shared Resource, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Li X, Lo Re V, Toh S. Profiling Real-World Data Sources for Pharmacoepidemiologic Research: A Call for Papers. Pharmacoepidemiol Drug Saf 2022; 31:929-931. [PMID: 35611675 DOI: 10.1002/pds.5481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Xiaojuan Li
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Vincent Lo Re
- Center for Pharmacoepidemiology Research and Training, Center for Clinical Epidemiology and Biostatistics, and Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| |
Collapse
|
47
|
Soldatos TG, Kim S, Schmidt S, Lesko LJ, Jackson DB. Advancing drug safety science by integrating molecular knowledge with post-marketing adverse event reports. CPT Pharmacometrics Syst Pharmacol 2022; 11:540-555. [PMID: 35143713 PMCID: PMC9124355 DOI: 10.1002/psp4.12765] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022] Open
Abstract
Promising drug development efforts may frequently fail due to unintended adverse reactions. Several methods have been developed to analyze such data, aiming to improve pharmacovigilance and drug safety. In this work, we provide a brief review of key directions to quantitatively analyzing adverse events and explore the potential of augmenting these methods using additional molecular data descriptors. We argue that molecular expansion of adverse event data may provide a path to improving the insights gained through more traditional pharmacovigilance approaches. Examples include the ability to assess statistical relevance with respect to underlying biomolecular mechanisms, the ability to generate plausible causative hypotheses and/or confirmation where possible, the ability to computationally study potential clinical trial designs and/or results, as well as the further provision of advanced features incorporated in innovative methods, such as machine learning. In summary, molecular data expansion provides an elegant way to extend mechanistic modeling, systems pharmacology, and patient‐centered approaches for the assessment of drug safety. We anticipate that such advances in real‐world data informatics and outcome analytics will help to better inform public health, via the improved ability to prospectively understand and predict various types of drug‐induced molecular perturbations and adverse events.
Collapse
Affiliation(s)
| | - Sarah Kim
- Department of PharmaceuticsCenter for Pharmacometrics and Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | - Stephan Schmidt
- Department of PharmaceuticsCenter for Pharmacometrics and Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | - Lawrence J. Lesko
- Department of PharmaceuticsCenter for Pharmacometrics and Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | | |
Collapse
|
48
|
Wang SV, Schneeweiss S. A Framework for Visualizing Study Designs and Data Observability in Electronic Health Record Data. Clin Epidemiol 2022; 14:601-608. [PMID: 35520277 PMCID: PMC9063805 DOI: 10.2147/clep.s358583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/18/2022] [Indexed: 01/07/2023] Open
Abstract
Background There is growing interest in using evidence generated from clinical practice data to support regulatory, coverage and other healthcare decision-making. A graphical framework for depicting longitudinal study designs to mitigate this barrier was introduced and has found wide acceptance. We sought to enhance the framework to contain information that helps readers assess the appropriateness of the source data in which the study design was applied. Methods For the enhanced graphical framework, we added a simple visualization of data type and observability to capture differences between electronic health record (EHR) and other registry data that may have limited data continuity and insurance claims data that have enrollment files. Results We illustrate the revised graphical framework with 2 example studies conducted using different data sources, including administrative claims only, EHR only, linked claims and EHR, as well as specialty community based EHRs with and without external linkages. Conclusion The enhanced visualization framework is important because evaluation of study validity needs to consider the triad of study question, design, and data together. Any given data source or study design may be appropriate for some questions but not others.
Collapse
Affiliation(s)
- Shirley V Wang
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
49
|
Sarntivijai S, Blomberg N, Lauer KB, Briggs K, Steger-Hartmann T, van der Lei J, Sauer JM, Liwski R, Mourby M, Camprubi M. eTRANSAFE: Building a sustainable framework to share reproducible drug safety knowledge with the public domain. F1000Res 2022; 11. [PMID: 35602243 PMCID: PMC9096149 DOI: 10.12688/f1000research.74024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/20/2022] Open
Abstract
Integrative drug safety research in translational health informatics has rapidly evolved and included data that are drawn in from many resources, combining diverse data that are either reused from (curated) repositories, or newly generated at source. Each resource is mandated by different sets of metadata rules that are imposed on the incoming data. Combination of the data cannot be readily achieved without interference of data stewardship and the top-down policy guidelines that supervise and inform the process for data combination to aid meaningful interpretation and analysis of such data. The eTRANSAFE Consortium's effort to drive integrative drug safety research at a large scale hereby present the lessons learnt and the proposal of solution at the guidelines in practice at this Innovative Medicines Initiative (IMI) project. Recommendations in these guidelines were compiled from feedback received from key stakeholders in regulatory agencies, EFPIA companies, and academic partners. The research reproducibility guidelines presented in this study lay the foundation for a comprehensive data sharing and knowledge management plans accounting for research data management in the drug safety space - FAIR data sharing guidelines, and the model verification guidelines as generic deliverables that best practices that can be reused by other scientific community members at large. FAIR data sharing is a dynamic landscape that rapidly evolves with fast-paced technology advancements. The research reproducibility in drug safety guidelines introduced in this study provides a reusable framework that can be adopted by other research communities that aim to integrate public and private data in biomedical research space.
Collapse
Affiliation(s)
| | - Niklas Blomberg
- ELIXIR Hub, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | | | - Katharine Briggs
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Leeds, LS11 5PS, UK
| | - Thomas Steger-Hartmann
- Bayer AG, Research & Development, Pharmaceuticals, Investigational Toxicology, 13342 Berlin, Germany
| | - Johan van der Lei
- Department of Medical Informatics, Erasmus University Rotterdam, EUR - Erasmus Medical Center (MC), Rotterdam, The Netherlands
| | - John-Michael Sauer
- Predictive Safety Testing Consortium, Critical Path Institute, Tucson, Arizona, 85718, USA
| | - Richard Liwski
- Predictive Safety Testing Consortium, Critical Path Institute, Tucson, Arizona, 85718, USA
| | - Miranda Mourby
- Centre for Health, Law and Emerging Technologies (HeLEX), Faculty of Law, University of Oxford, Oxford, OX2 7DD, UK
| | - Montse Camprubi
- Synapse Research Management Partners S.L., C. Diputació 237, Àtic 3a, 08007, Barcelona, Spain
| | | |
Collapse
|
50
|
Fuller CC, Cosgrove A, Sands K, Miller KM, Poland RE, Rosen E, Sorbello A, Francis H, Orr R, Dutcher SK, Measer GT, Cocoros NM. Using inpatient electronic medical records to study influenza for pandemic preparedness. Influenza Other Respir Viruses 2022; 16:265-275. [PMID: 34697904 PMCID: PMC8818824 DOI: 10.1111/irv.12921] [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: 09/10/2021] [Accepted: 09/25/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND We assessed the ability to identify key data relevant to influenza and other respiratory virus surveillance in a large-scale US-based hospital electronic medical record (EMR) dataset using seasonal influenza as a use case. We describe characteristics and outcomes of hospitalized influenza cases across three seasons. METHODS We identified patients with an influenza diagnosis between March 2017 and March 2020 in 140 US hospitals as part of the US FDA's Sentinel System. We calculated descriptive statistics on the presence of high-risk conditions, influenza antiviral administrations, and severity endpoints. RESULTS Among 5.1 million hospitalizations, we identified 29,520 hospitalizations with an influenza diagnosis; 64% were treated with an influenza antiviral within 2 days of admission, and 25% were treated >2 days after admission. Patients treated >2 days after admission had more comorbidities than patients treated within 2 days of admission. Patients never treated during hospitalization had more documentation of cardiovascular and other diseases than treated patients. We observed more severe endpoints in patients never treated (death = 3%, mechanical ventilation [MV] = 9%, intensive care unit [ICU] = 26%) or patients treated >2 days after admission (death = 2%, MV = 14%, ICU = 32%) than in patients treated earlier (treated on admission: death = 1%, MV = 5%, ICU = 23%, treated within 2 days of admission: death = 1%, MV = 7%, ICU = 27%). CONCLUSIONS We identified important trends in influenza severity related to treatment timing in a large inpatient dataset, laying the groundwork for the use of this and other inpatient EMR data for influenza and other respiratory virus surveillance.
Collapse
Affiliation(s)
- Candace C. Fuller
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Austin Cosgrove
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Kenneth Sands
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
- HCA HealthcareNashvilleTennesseeUSA
| | | | - Russell E. Poland
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
- HCA HealthcareNashvilleTennesseeUSA
| | - Edward Rosen
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Alfred Sorbello
- United States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Henry Francis
- United States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Robert Orr
- United States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Sarah K. Dutcher
- United States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Gregory T. Measer
- At the time of the project, Gregory Measer was with the United States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Noelle M. Cocoros
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| |
Collapse
|