Hostname: page-component-76fb5796d-vfjqv Total loading time: 0 Render date: 2024-04-27T23:24:29.251Z Has data issue: false hasContentIssue false

Real-world data for 21st-century medicine: The clinical and translational science awards program perspective

Published online by Cambridge University Press:  11 September 2023

Pablo Cure*
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
Sanae ElShourbagy Ferreira
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
Joshua P. Fessel
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
Deborah Ossip
Affiliation:
Center for Leading Innovation and Collaboration (CLIC), Clinical and Translational Science Program National Coordinating Center, University of Rochester Medical Center, Rochester, NY, USA Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
Martin S. Zand
Affiliation:
Center for Leading Innovation and Collaboration (CLIC), Clinical and Translational Science Program National Coordinating Center, University of Rochester Medical Center, Rochester, NY, USA Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
Scott J. Steele
Affiliation:
Center for Leading Innovation and Collaboration (CLIC), Clinical and Translational Science Program National Coordinating Center, University of Rochester Medical Center, Rochester, NY, USA Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
Kenneth Gersing
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
Christopher M. Hartshorn
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
*
Corresponding author: P. Cure, MD, MPH; Email: pablo.cure@nih.gov
Rights & Permissions [Opens in a new window]

Abstract

Type
Perspective
Creative Commons
Creative Common License - CCCreative Common License - BY
To the extent this is a work of the US Government, it is not subject to copyright protection within the United States. Published by Cambridge University Press on behalf of The Association for Clinical and Translational Science.
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© National Institutes of Health (NIH), 2023.

Background

Real-world data (RWD) refers to data related to patient health status and/or the delivery of healthcare routinely collected from a variety of sources. The clinical evidence derived from analysis of RWD regarding the utilization and potential benefits or risks associated with a medical product or intervention is known as real-world evidence (RWE) [1,Reference Concato and Corrigan-Curay2]. Real-world data includes a variety of data sources such as electronic health records (EHR), laboratory and imaging, claims/billing, vital records, digital health technologies (DHT), and other modes of remote data collection that can be obtained retrospectively and/or prospectively during the patient’s continuum of care. In addition, other types of data sources such as environmental exposures, pollution levels (indoor and outdoor), geolocation, text messaging, social media, economic measures, and other sources have the potential to “enrich” health-related data for patients and populations. In the last decade, several advances have brought the potential for RWD applications to a new level: an exponential increase in the sheer quantity of data that can be collected from single and multiple sources; data integration and “ingesting” capabilities from multiple Common Data Models (CDMs); new analytical tools and novel methods such as artificial intelligence (AI) and machine learning (ML); and higher capacity to store and maintain information locally and/or centrally within secure cloud environments.

To keep up with evolving types and uses for RWD, the Food and Drug Administration (FDA) has been developing a series of RWD guidance documents [3] and has launched an Advancing Real-World Evidence Program [4]. In addition, the National Institutes of Health (NIH), the National Cancer Institute (NCI), the National Center for Advancing Translational Sciences (NCATS), and the National Library of Medicine have been addressing the need for standardization around RWD collection and use. For example, since 2017, the FDA-led Common Data Models Harmonization and Open Standards for Evidence Generation project [5] has worked to ensure infrastructure is in place to support standardized data generated across the translational spectrum. These CDMs allow for a unified database model to help in the integration of various RWD sources according to the same standard, facilitating the interoperability of the data from multiple data sources for the conversion of RWD to RWE. During the COVID-19 pandemic, these efforts further materialized in the development of the National COVID-19 Cohort Collaborative (N3C) [6], one of the largest repositories of HIPAA-defined limited data sets in the country that, as of March 2023, includes data from over 77 sites in the U.S. and > 7 million COVID + cases and > 11 million controls, “ingesting” data from a variety of CDMs [6]. Through partnerships among many organizations that provide clinical data, and by securely making data accessible to more than 3,000 researchers, N3C has helped to answer critical questions about COVID-19 biology, clinical behavior, and treatment strategies. For example, one of the first RWD-driven characterizations of the post-acute sequelae of COVID-19 (“long COVID”) was accomplished using N3C [Reference Pfaff, Girvin and Bennett7]. N3C offers one model of how to create a large de-identified database through collection, curation, and analysis of multisite data in a single protected data enclave that can facilitate rapid response to a public health emergency. Recently, to further the potential of these types of RWD approaches, NCATS launched the N3C Public Health Answers to Speed Tractable Results to deliver fast, actionable analyses on pressing COVID-19 questions [8]. Efforts such as N3C can continue to model how institutions, government, and other stakeholders can work together in developing large-scale RWD. In addition, combining clinical/health-related data from EHR-based datasets such as N3C with non-clinical data (e.g., sociodemographic, environmental, geolocation, etc.) can provide a more comprehensive picture of the health and well-being of individuals and communities.

Through its Clinical and Translational Science Awards (CTSA) Program, NCATS supports a national network of biomedical research institutions that accelerate the translation of scientific observations into innovative health solutions [9]. Advancing translational science and improving the efficiency and effectiveness of translation requires a coordinated, collaborative effort. Incorporating RWD into translational science requires combining lessons learned, processes, and know-how from experts and from the communities directly impacted by advances in RWD. As the field rapidly evolves and more data from a wide variety of sources are incorporated into clinical and translational science (CTS) research and day-to-day patient care, the challenges and opportunities presented herein (Table 1) represent a possible blueprint for how the CTSA program could make RWD and RWE a reality to help transform 21st-century medicine.

Table 1. Challenges and opportunities in the development and implementation of RWD for RWE in the CTSA program

CDMs = common data models; CTSA = clinical and translational science awards; N3C = national covid cohort collaborative; RWD = real-world data; RWE = real-world evidence.

Infrastructure, Sources, and Quality

Data infrastructure, sources, quality, and reliability can be highly variable between individual data ecosystems [Reference Spengler, Lang, Mahapatra, Gatz, Kuhn and Prasser10Reference Fowe, Wallace and Rissi12]. Further, ethical aspects of use of patient-derived data for research and protection of privacy and confidentiality continue to pose challenges [Reference Faden, Kass, Goodman, Pronovost, Tunis and Beauchamp13]. Informed consent, de-identification, data ownership, linkages, and sharing of data are issues inherent to any RWD effort [Reference Kushida, Nichols, Jadrnicek, Miller, Walsh and Griffin14]. Methods to store data and control levels of access for researchers and analysts to specific datasets are as important as the data itself. These challenges are further compounded by continuous and rapid advances in data science and adjacent fields. Together, the challenges require data experts and institutions to constantly re-visit their policies and procedures to maintain the highest standards of quality, reliability, privacy, and security of data and their sources. In addition, data governance and provenance are important parts of the data ecosystem to ensure accuracy and quality of the data collected [Reference Haendel, Chute and Bennett15].

Capabilities to simultaneously collect/aggregate, securely link, and analyze data from several RWD sources are in great demand to provide a more complete assessment of patients’ and communities’ health (Fig. 1). Currently, EHRs are one of the main data sources for RWD; however, challenges with the quality, reliability, heterogeneity, and utility of the data collected in the EHR continue to be an issue [Reference Ta and Weng16Reference Weiner and Embi18]. Indeed, EHR systems were developed for clinical documentation, administrative, and billing purposes, so their “re-purposing” as RWD sources for CTS research requires adaptations in data collection, integration, validation, and analysis. Furthermore, overlaying “traditional” sources of RWD with other contextual information can potentially impact data robustness and privacy. Environmental exposures, geolocation, economic measures, and other data extracted from publicly and non-publicly available data sets can enrich analyses. Yet, sources not typically thought of as health-related have different regulations and policies governing their use that must be considered.

Figure 1. Overall data infrastructure, sources, and integration. CMS = centers for medicare & medicaid services claims data; DHT = digital health technologies; EDW = enterprise data warehouse; HER = electronic health records; Labs = laboratory values; X-ray images; RWD = real-world data.

Some data sources are well-suited to RWD/RWE applications. For example, disease or specialty-specific registries and databases have successfully supported new indications for existing drugs [19Reference Fitzsimmons, Erdman and Wolfram21]. These registries require strategic planning – incorporating adequate governance, infrastructure, resources, and expertise beyond the registry itself – and flexibility, allowing for dynamic evolution to bring in novel RWD data sources and methods that bolster the registry’s utility. One example is the NCI Surveillance, Epidemiology, and End Results cancer registry [Reference Cronin, Ries and Edwards22,Reference Penberthy, Rivera, Lund, Bruno and Meyer23], which provides a high-quality population base (state or metropolitan area) from central cancer registries that enable monitoring of disease prevalence, cancer population needs, and health disparities and their impact in these communities. Bringing RWD together from various sources is only the first step. Generating RWE requires making RWD research-ready and employing adequate and sometimes sophisticated analytical methods.

Data collected from patients and/or populations requires the highest ethical, confidentiality, and privacy standards. Techniques such as de-identification, aggregation, data pooling, and other methods to maintain privacy should be in place both locally (at institutions) and centrally (within a central enclave environment). Further advances in obtaining informed consent, such as e-consent, should be up to speed with the pace of technologies used to collect participant data and their willingness to be contacted for future participation in prospective trials.

Integration, Harmonization, and Analysis

Combining data from multiple sources and systems requires harmonization of the data being collected to facilitate future evaluations and analyses. As the ability to integrate data from multiple sources improves, evidence from single data sources may become less preferable. However, the process of data harmonization and curation is currently resource-intensive. Harmonizing CDMs of various networks or data sources also allows researchers to ask specific data questions of potentially much larger (combined) sources of RWD. It is tempting to combine as much data as possible, but it is critical to first identify the purpose, timepoints, and desired impact of data and collect it in a way that minimally burdens patients, healthcare providers, and others at the data generation source. Further, it is important to adhere to the highest possible data quality standards. RWD data from multiple sources often improves data richness, but not all data will be equally useable without proper integration and harmonization.

When combining data from multiple data sources, maintaining privacy and security must be a focus of RWD/RWE efforts. Privacy-Preserving Record Linkage (PPRL) is one way of connecting records that refer to the same individual across different data sources using secure, pseudonymization processes while maintaining the individual’s privacy [Reference Vatsalan, Christen and Verykios24]. NCATS is piloting PPRL technology in the N3C to determine if linking multiple data sets enhances usability of COVID-19 RWD. Identifying high-quality data and mapping their provenance allows for selection of the best and most representative data for multisite analyses.

Despite rapid advances, there are important remaining challenges for broader use of multimodal RWD. For example, errors – inherent to each source and/or arising during harmonization – could confound the process of RWD integration and must be addressed. More robust interoperability for CDMs and codes of conduct to reduce misuse of research findings from RWD and research data in general is necessary. Additionally, newly formed federated or centrally kept data environments must incorporate tools to enable use of completely de-identified or HIPAA-limited data for additional flexibility and adaptation to both institutional and multisite data aggregation needs. Efficient and meaningful integration of data from multiple sources and institutions could help create the next generation of data systems for evidence-based medicine and real-time clinical support [Reference Subbiah25Reference Wynia, Beaty and Bennett27].

RWD can also come from more specific – disease-based – fully consented registries which include strict policies and procedures for data linkages and patient-identified data requests in order to protect patient privacy [Reference Leppke, Leighton and Zaun28]. In these cases, data harmonization is mostly unnecessary with all data points and variables specified from the outset allowing for more specific questions and outcomes of interest (such as mortality, disease burden, laboratory results, and other variables) to be followed over time. However, as with well-designed clinical trials, research with registries must include a priori definition of meaningful outcomes to evaluate and pre-defined analytical methods to prevent or diminish bias.

Novel analytical techniques utilizing AI and ML can provide additional capacity to help analyze large amounts of data and identify “subtle” risk factors that may not be easily identified through more conventional analysis. At the same time, utilizing both automated and manual data review techniques can be used for data quality control and assurance in large multisite data efforts to improve RWD obtained locally and in aggregate with automatically maintained data provenance as a requirement [Reference Pfaff, Girvin and Gabriel29].

Diversity, Equity, Inclusion, and Accessibility (DEIA)

Insufficient diversity, equity, inclusion, and accessibility (DEIA) considerations in planning, analyzing, and collecting data can introduce bias and limit the ability of RWD to develop meaningful RWE. Gichoya et al., developed an AI algorithm that accurately identified self-reported race from imaging data only, a phenomenon that could not be replicated by human radiologists and that was not readily explainable [Reference Gichoya, Banerjee and Bhimireddy30]. This deliberate example highlights the fact that AI algorithms can make race-specific conclusions based on factors invisible to human evaluators. If those cryptic conclusions are erroneous, or worse, perpetuate biases, they could lead to healthcare decisions that perpetuate health disparities. Of course, these kinds of models may also accelerate beneficial discoveries that would be otherwise difficult to achieve. To ensure the most beneficial patient and public health outcomes, the potential for introduction and amplification of biases or structural inequities must be reckoned with proactively.

Equitable RWE begins with high-quality, representative RWD. Addressing the “digital divide” and fair access to DHTs is also central to discussions of DEIA in RWD. Technologies are becoming more widely used to assess health parameters – e.g., continuous glucose levels, vital signs, and physical activity. Such strategies can adapt to users across the lifespan and expand testing of interventions or therapeutics to more diverse or rural communities or even monitor activity and health in space [Reference Crucian, Valentine and Calaway31]. Low (e.g. text messaging)- and high-tech (e.g. smart apps) options need to be available and tailored to users to promote equitable access to and representativeness of RWD for all populations intended to benefit.

To advance DHTs, other DEIA-related roadblocks and access questions must be addressed. Services and support tools built and validated to promote inclusivity and equity in data collection and analytical technologies are crucial. Building trust in the use of novel technologies for the benefit of patients and communities is part of the providers’ role in the development and implementation process. At the same time, patient/user input in the development and optimization of new technologies plays an important role in improving utilization, user satisfaction, and adherence. The million-dollar question and challenge for CTS researchers remains: how do we align 21st-century RWD with 21st-century medicine for the benefit of all? The answer requires the enterprise to focus on integrated approaches intentionally attuned to DEIA considerations.

Training, Education, and Career Development in RWD/RWE

Training the next generation of scientists is one of the main goals of NIH. For example, the CTSA program supports trainees and scholars through a number of funding opportunities such as the KL2 program (now K12), TL1 program (now T32 pre-doctoral and T32 post-doctoral), as well as other programs such as diversity, reentry, and reintegration supplements [32]. Each program is geared towards a specific phase in the training and career development of the trainee/scholar. Identifying training opportunities, activities and resources tailored to the specific trainee/scholar needs in data management, analytics, and reporting can be crucial in the development of a highly skilled/data-driven workforce of the future. Recently, the CTSA program diversity was published showing areas where we can improve to attract a more diverse workforce in clinical and translational science [Reference Rubio, Baker and Doyle33]. Specialized pieces of training in data management and novel analytical methods using AI and ML can also help in the development and retention of professionals focused on health and related RWD in this fast-growing field. Facilitating an environment where cross-communication between clinicians, data managers, data analysis experts, regulatory authorities, and the community can provide the catalyzing force to maximize efficiency, impact, and return on investment of RWD-oriented projects. As in the device development “world” where clinicians, bioengineers, technology transfer, and marketing experts come together to solve unmet medical needs, we need to develop a similar team science approach where access to specialized resources and expertise in RWD can bring data managers, experts in data analysis (including AI and ML data experts) and others, closer to their clinical/scientific counterparts to help answer meaningful health questions. All needs to be done in parallel with identifying pressing community and population health needs and questions that if answered through RWD could help to significantly improve individual and public health locally and nationally.

Discussion

To make RWD and RWE a reality within the CTSA Program, investigators, research participants, clinicians, patient advocates, funding agencies, regulatory agencies, industry, and many others need to collaboratively identify strategic priorities that maximize impact of data on scientific knowledge and health outcomes. Developing generalized, intervention-agnostic approaches driven by translational science could be an area for consideration by the CTSA consortium. Several key areas already align with CTSA infrastructure and resources (Fig. 2).

Figure 2. Key areas in real-world data within CTSA programs. D&I = dissemination and implementation; FAIR = findable, accessible, interoperable, reproducible; HER = electronic health records; HS = human subjects; PPRL = privacy-preserving record linkages.

Furthermore overlaying “traditional” sources of RWD with other information that can potentially impact health and health outcomes, such as environmental exposures, geolocation, place of residence, economic measures, and other data extracted from publicly and non-publicly available data sets (e.g., from social networking sites such as Facebook, LinkedIn, Instagram, etc., or even data from the “Internet of Things”) can add richness (and further complexity) to the data. Yet, these datasets derive from sources not typically thought of as being health-related and that may have different regulations and policies governing their use could create additional challenges in the successfully translating RWD into RWE. Integration of RWD from smaller, well-designed, fit-for-use disease registries as well as other prospective hypothesis-generating types of observational datasets requires a priori definition of meaningful outcomes to evaluate using pre-defined analytical methods to minimize bias.

Several limitations must be taken into consideration when utilizing electronic health records data from efforts such as N3C, including overrepresentation of certain populations such as patients with more access to health services, high utilizers of health care, patients with more severe symptoms, and conditions and inpatients. In addition, clinical follow-ups outside of the health system, as it happens in community hospitals or other outside-of-the-health-system settings (e.g. private doctors’ offices) can also be missing/not recorded and therefore outcomes are limited to data within the enclave [Reference Pfaff, Girvin and Bennett7]. It is therefore important to acknowledge the limitations of these large RWD sources before making broad population-based conclusions. Data privacy, security, and consent for future contact remain cornerstones to make sure the research performed using RWD through large and broad datasets or smaller and more controlled datasets (e.g., registries) can have all the necessary safeguards to allow for an ethical and scientifically rigorous process.

Federated or centralized approaches both provide advantages and disadvantages when it comes to data sharing, integration, harmonization, and quality [Reference Pfaff, Girvin and Gabriel29]. For example, efforts to improve data quality in N3C and providing those data to institutions are now part of the feedback received by institutions sharing data in the N3C data enclave. Using federated and centralized approaches to collect RWD can also help with both data quality, by providing feedback to institutions contributing data in a centralized platform/data enclave, or by performing participant screening for future trials in a federated environment, under the right participant consent for future contact. Data standardization at the collection source also represents a major deficiency of large, integrated datasets. Establishing standards for data entry at the outset (during the clinical encounter or shortly after) as well as quality control methods while maintaining data provenance remains critical.

Training the next generation of data managers, statisticians, clinical informaticians, data clinicians and other experts within the CTSA collaborative/team science approach can greatly enhance the capability of the consortium to timely respond to current and future public health needs using RWD. Well-developed, customizable, complementary, and competency-based training programs may represent one of the biggest opportunities for developing the field of RWD and data science within the CTSA consortium. In addition, basic principles of data management and novel methods training could bring great added value to current and future clinical researchers.

Combining these CTSA assets and developing innovative approaches to improve the quality, utilization, and reproducibility of RWD findings under FAIR (Findable, Accessible, Interoperable, and Reproducible) guiding principles [Reference Wilkinson, Dumontier and Aalbersberg34], can bring new and critical scientifically sound programmatic activities to fruition. Designing approaches to validate and implement new technologies and analytical tools can accelerate RWE advances. Issues of DEIA, including equitable access to technologies and representativeness of data, need to be addressed from the start and not as an afterthought. Further, applying an equity lens to securely source, integrate, and harmonize clinically relevant, high quality, representative RWD could result in RWE-based approaches that transform healthcare and enhance patient health – as the ultimate goal of all RWE is to answer real-world questions and deliver real-world returns, to all.

Acknowledgments

Some of the work presented herein came out of the workshop and Un-Meeting held on June 8-9, 2022, to address RWD/RWE challenges and opportunities from a CTSA Program vantage point. The authors would like to acknowledge the CTSA Coordinating CLIC at the University of Rochester for their work in the coordination of the meeting, the meeting presenters, and all the participants for their time, contributions, and the lively meeting discussions. Special thanks to the meeting Steering Committee, CLIC meeting Coordinator Judy Giordano, and the Breakout session meeting catalysts.

Funding statement

This work was funded in part by the University of Rochester CLIC under Grant U24TR002260. CLIC was the coordinating center for the CTSA program funded by the NCATS at the NIH. This work is solely the responsibility of the authors and does not necessarily represent the official views of the NIH, NCATS, FDA, or CLIC. For more information about Un-meetings: https://doi.org/10.5281/zenodo.6983868

Competing interests

This work is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or the FDA. The findings, knowledge gaps, challenges and opportunities described in this manuscript represent a summary of individual opinions and ideas expressed during the workshop. The summary does not represent any sort of consensus or directive, nor does it represent an expression of research priorities or intended research directions for NCATS or NIH.

Footnotes

Current address: Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA

References

FDA. Real-World Evidence: Food and Drug Administration; 2022. [FDA RWE Definitions and Selected Guidance]. Available from: https://www.fda.gov/science-research/science-and-research-special-topics/real-world-evidence. Accessed December 22, 2022.Google Scholar
Concato, J, Corrigan-Curay, J. Real-world evidence - where are we now? New Engl J Med. 2022;386(18):16801682.CrossRefGoogle ScholarPubMed
FDA. FDA Issues Draft Guidances on Real-World Evidence, Prepares to Publish More in Future: Food and Drug Administration; 2022. Available from: https://www.fda.gov/drugs/news-events-human-drugs/fda-issues-draft-guidances-real-world-evidence-prepares-publish-more-future. Accessed December 22, 2022.Google Scholar
FDA. Advancing Real-World Evidence Program: Food and Drug Administration 2022. Available from: https://www.fda.gov/drugs/development-resources/advancing-real-world-evidence-program. Accessed December 22, 2022.Google Scholar
HL7 International. Common Data Models Harmonization - Biomedical Research and Regulation Work Group 2019. Available from: https://build.fhir.org/ig/HL7/cdmh/#guidance-to-the-readers. Accessed December 22, 2022.Google Scholar
NCATS. National Center for Advancing Translational Sciences, National Institutes of Health 2023. About the National COVID Cohort Collaborative N3C. 2023. Available from: https://ncats.nih.gov/n3c/about. Accessed March 23, 2023.Google Scholar
Pfaff, ER, Girvin, AT, Bennett, TD, et al. Identifying who has long COVID in the USA: a machine learning approach using N3C data. Lancet Digit Health. 2022;4(7):E532E41.CrossRefGoogle Scholar
N3C. N3C Public Health Answers to Speed Tractable Results (PHASTR): National COVID Cohort Collaborative, NCATS, DHHS; 2023. Available from: https://covid.cd2h.org/phastr. Accessed March 15, 2023.Google Scholar
NCATS. Clinical and Translational Science Awards (CTSA) Program: National Center for Advancing Translational Sciences, National Institutes of Health 2022. Available from: https://ncats.nih.gov/ctsa. Accessed December 22, 2022.Google Scholar
Spengler, H, Lang, C, Mahapatra, T, Gatz, I, Kuhn, KA, Prasser, F. Enabling agile clinical and translational data warehousing: platform development and evaluation. JMIR Med Inform. 2020;8(7):e15918.CrossRefGoogle ScholarPubMed
Schneeweiss, S. Learning from big health care data. N Engl J Med. 2014;370(23):21612163.CrossRefGoogle ScholarPubMed
Fowe, IE, Wallace, NT, Rissi, JJ. The evolution and typology of learning health system hospitals and other health information interested hospitals in the US. Perspect Health Inf Manag. 2022;19(3):1b.Google ScholarPubMed
Faden, RR, Kass, NE, Goodman, SN, Pronovost, P, Tunis, S, Beauchamp, TL. An ethics framework for a learning health care system: a departure from traditional research ethics and clinical ethics. Hastings Cent Rep. 2013;43(s1):S16S27.CrossRefGoogle Scholar
Kushida, CA, Nichols, DA, Jadrnicek, R, Miller, R, Walsh, JK, Griffin, K. Strategies for de-identification and anonymization of electronic health record data for use in multicenter research studies. Med Care. 2012;50(Suppl):S82101.CrossRefGoogle ScholarPubMed
Haendel, MA, Chute, CG, Bennett, TD, et al. The national COVID cohort collaborative (N3C): rationale, design, infrastructure, and deployment. J Am Med Inform Assn. 2021;28(3):427443.CrossRefGoogle ScholarPubMed
Ta, CN, Weng, C. Detecting systemic data quality issues in electronic health records. Stud Health Technol Inform. 2019;264:383387.Google ScholarPubMed
Jetley, G, Zhang, H. Electronic health records in IS research: quality issues, essential thresholds and remedial actions. Decis Support Syst. 2019;126:113137.CrossRefGoogle Scholar
Weiner, MG, Embi, PJ. Toward reuse of clinical data for research and quality improvement: the end of the beginning? Ann Intern Med. 2009;151(5):359360.CrossRefGoogle ScholarPubMed
FDA. FDA approves new use of transplant drug based on real-world evidence: Food and Drug Administration; 2021. Available from: https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-new-use-transplant-drug-based-real-world-evidence. Accessed December 22, 2022.Google Scholar
Erdman, J, Wolfram, J, Nimke, D, et al. Lung transplant outcomes based on immunosuppressive regimen at discharge: data from the US scientific registry of transplant recipients (SRTR). J Heart Lung Transpl. 2021;40(4):S165S.CrossRefGoogle Scholar
Fitzsimmons, W, Erdman, J, Wolfram, J, et al. Lung transplant outcomes based on immunosuppressive regimen at discharge: data from the US scientific registry of transplant recipients (SRTR). Am J Transplant. 2021;21:800801.Google Scholar
Cronin, KA, Ries, LAG, Edwards, BK. Preface. Cancer. 2014;120(S23):37553757.CrossRefGoogle Scholar
Penberthy, LT, Rivera, DR, Lund, JL, Bruno, MA, Meyer, AM. An overview of real-world data sources for oncology and considerations for research. CA Cancer J Clin. 2022;72(3):287300.CrossRefGoogle ScholarPubMed
Vatsalan, D, Christen, P, Verykios, VS. A taxonomy of privacy-preserving record linkage techniques. Information Systems. 2013;38(6):946969.CrossRefGoogle Scholar
Subbiah, V. The next generation of evidence-based medicine. Nature Medicine. 2023;29(1):4958.CrossRefGoogle ScholarPubMed
Califf, RM. Now is the time to fix the evidence generation system. Clin Trials. 2023;20(1):17407745221147689–12.CrossRefGoogle ScholarPubMed
Wynia, M, Beaty, L, Bennett, T, et al. Real-world evidence of neutralizing monoclonal antibodies for preventing hospitalization and mortality in COVID-19 outpatients. Chest. 2022;163(5):10611070.CrossRefGoogle ScholarPubMed
Leppke, S, Leighton, T, Zaun, D, et al. Scientific registry of transplant recipients: collecting, analyzing, and reporting data on transplantation in the United States. Transplantation Reviews. 2013;27(2):5056.CrossRefGoogle ScholarPubMed
Pfaff, ER, Girvin, AT, Gabriel, DL, et al. Synergies between centralized and federated approaches to data quality: a report from the national COVID cohort collaborative. J Am Med Inform Assoc. 2022;29(4):609618.CrossRefGoogle ScholarPubMed
Gichoya, JW, Banerjee, I, Bhimireddy, AR, et al. Al recognition of patient race in medical imaging: a modelling study. Lancet Digit Health. 2022;4(6):E406E14.CrossRefGoogle Scholar
Crucian, B, Valentine, R, Calaway, K, et al. Spaceflight validation of technology for point-of-care monitoring of peripheral blood WBC and differential in astronauts during space missions. Life Sciences in Space Research. 2021;31:2933.CrossRefGoogle ScholarPubMed
NCATS. National Center for Advancing Translational Sciences, National Institutes of Health. Training and Career Development Programs 2018. https://ncats.nih.gov/ctsa/about/training/programs. Accessed March 23, 2023.Google Scholar
Rubio, M, Baker, HL, Doyle, JM. Demographic composition of national institutes of health clinical and translational science awards (CTSA) program principal investigators, scholars, and trainees. J Clin Transl Sci. 2023;7(1):e19.CrossRefGoogle ScholarPubMed
Wilkinson, MD, Dumontier, M, Aalbersberg, IJ, et al. The FAIR guiding principles for scientific data management and stewardship. Scientific Data. 2016;3(1):160018.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Challenges and opportunities in the development and implementation of RWD for RWE in the CTSA program

Figure 1

Figure 1. Overall data infrastructure, sources, and integration. CMS = centers for medicare & medicaid services claims data; DHT = digital health technologies; EDW = enterprise data warehouse; HER = electronic health records; Labs = laboratory values; X-ray images; RWD = real-world data.

Figure 2

Figure 2. Key areas in real-world data within CTSA programs. D&I = dissemination and implementation; FAIR = findable, accessible, interoperable, reproducible; HER = electronic health records; HS = human subjects; PPRL = privacy-preserving record linkages.