Hostname: page-component-848d4c4894-mwx4w Total loading time: 0 Render date: 2024-06-15T11:47:55.544Z Has data issue: false hasContentIssue false

Advancing regulatory science through real-world data and real-world evidence

Published online by Cambridge University Press:  21 May 2024

Pablo Cure*
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
Christopher M. Hartshorn
Affiliation:
National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 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
*
Corresponding author: P. Cure, Email: pablo.cure@nih.gov
Rights & Permissions [Opens in a new window]

Abstract

Type
Editorial
Creative Commons
Creative Common License - CCCreative Common License - BY
This is a work of the US Government and is not subject to copyright protection within the United States. Published by Cambridge University Press on behalf of 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, 2024

Background and overarching categories for thematic issue

This special-themed issue of the Journal of Clinical and Translational Science will focus on the use of diverse sources of Real-World Data (RWD) and Real-World Evidence (RWE) to Advance Translational Science. The goal is to highlight research, activities, and processes for the translation of RWD from a range of sources into RWE that can enhance the full translational science continuum, ultimately improving the development, approval, adoption, and use of safe and effective medical products.

The manuscripts in this thematic issue have been written for a broad target audience of all those engaged in clinical and translational science. Manuscripts addressing the spectrum of sources and applications of RWD/RWE in translational science were considered, with the following being of particular interest: Use of electronic health records, registries, claims data and other data sources to generate RWE; Opportunities and challenges for Digital Health Technologies to generate RWE; Evolution of novel methods to accelerate the generation and use of RWD/RWE; Policy considerations for the use of RWD/RWE; Use of RWD/RWE to improve clinical trials trial designs; Data access, use, sharing and fit-for-purpose considerations; Emerging use cases for RWD/RWE in translational science; Infrastructure to support the generation and; Use of RWD/RWE in translational science.

Use of RWD to inform trial designs and generate RWE to evaluate safety and efficacy [Reference Dang, Fong and Tarp1Reference Díaz, Lee and Kıcıman12]

Currently, most of the patient-derived RWD used in biomedical research resides in Electronic Health Records (EHRs). Even though their development was not intended for research purposes, the large volume of information, data richness, continuum of care data collection, and ability to be linked and integrated with additional data from medical encounters such as claims data, imaging, and laboratory information can provide a more comprehensive view of an individual and their health journey. It is known that utilization of EHR data can be highly valuable in postmarket safety monitoring and surveillance. RWD leading to RWE has also been used as external control arms in interventional studies where randomization is impractical or unethical, and in different types of non-interventional studies. In this thematic issue, Selker et al. describe how through a more controlled environment utilizing standardized study protocols, the information obtained from a single patient could also help elucidate wanted and unwanted treatment effects that could be used as RWE and inform clinical decision-making at the individual patient level or even more broadly, if data from other similar cases is aggregated [Reference Selker, Dulko, Greenblatt, Palm and Trinquart8]. A series of manuscripts in this thematic issue also describe and apply a causal roadmap that can serve as a guide for studies that generate RWE [Reference Dang, Fong and Tarp1, Reference Dang, Gruber and Lee2, Reference Williamson, Wyss and Stuart11]. In this framework, it is important to consider setting a priori ground rules for the utilization of the data including, fit-for-use, data availability, data quality, study design and analysis, quality of the results, assessment of causality, and grade of evidence to ultimately generate high-quality and relevant information to regulators and other stakeholders for clinical and/or regulatory decision-making.

RWD/RWE to advance health outcomes and address health disparities [Reference Hamer, Sobczak and Whittington13Reference Gonzales, Champieux and Contaxis20]

A goal for RWD and the resulting RWE is to equally benefit patients and populations. Equity in distribution and access to life-saving therapies is also necessary to achieve a broader benefit of RWE. Through collecting RWD from multiple hospitals, medical practices, and health systems, we are now able to capture data from a much larger, widely distributed, and more representative and diverse population sample. The utilization of RWD has the potential to lower the bar for participation in study protocols, making it more convenient for individuals and researchers while increasing data representativeness. However, we must remain vigilant to avoid past mistakes when it comes to inclusion of diverse populations in research and focus on education, community engagement, and returning results to participants in a tailored manner. In this context, the manuscript by Hamer et al, provides an example of how to address barriers to access to care and therapeutics during a public health crisis through multi-level dissemination strategies utilizing RWD [Reference Hamer, Sobczak and Whittington13]. Utilization of multi-prong approaches (including RWD) in primary care settings as described by Krist et al. [Reference Krist, Huffstetler and Villalobos19] and tailoring strategies to community needs can positively impact recruitment of diverse populations. Also, analysis of state-level RWD can help elucidate health disparities and risk factors associated with poor health outcomes such as maternal mortality with resulting evidence incorporated into improving awareness, education, and assisting with resource allocation and policy making [Reference Tran, Jreij, Sistani and Shaya14].

Opportunities for DHTs to generate RWD/RWE [Reference Ostovari, Zhang, Patel and Jurkovitz15, Reference Bautista, Cave, Downey, Bentham and Jayne21Reference Bautista, Kowal, Cave, Downey and Jayne23]

Digital health technologies (DHTs) offer a host of opportunities to change how healthcare is delivered, tracked, and disseminated. DHTs are defined by FDA as integrated systems that use computing platforms, connectivity, software, and/or sensors for healthcare. DHTs are a broad category of data gathering and analysis tools that, in recent years, have paved the way for clinical research, consumer health, and clinical applications. They offer facile and often high-fidelity ways to collect real-world, observational data that is obtainable in real time. Albeit they will continue to be a target as RWD gathering tools moving forward, considerations in their implementation and their ability to deliver rigorous RWE, remain. For example, DHTs that have been leveraged to date for observational data collection are not as rigorously validated as traditional diagnostics; implementation issues remain in terms of those that cannot afford or do not live in regions with appropriate infrastructure; and the technology development pipeline far exceeds the evidence base to support their use. None of these considerations are intractable hurdles to the field writ large yet do involve additional “growing pains” in terms of their general use as RWD sources. Yet there are many examples to point to as to how the field is maturing. For example, in this issue, Bautista et al discuss the state of photoplethysmography, an optical technique that enables measurement of parameters associated with cardiorespiratory function [Reference Bautista, Cave, Downey, Bentham and Jayne21, Reference Bautista, Kowal, Cave, Downey and Jayne23]. They highlight the unique nature of this tool and the methods (contact vs contactless) used currently both in terms of their positive and negative attributes for specific use cases (e.g., validation of measurement) and populations (e.g., neonates vs adults). These types of translational science questions continue to be asked for the plethora of available DHTs and, steadily, are being addressed.

Policy considerations for RWD/RWE [Reference Hendricks-Sturrup and Lu24, Reference Chu, Miller and Mathews25]

Several policy considerations arise in the context of collecting, sharing, and utilizing RWD. While a range of policies and processes have been adopted to help ensure informed consent, data privacy, and the ethical use of individuals’ data, this remains a challenging issue given the evolving nature of the collection and utility of RWD, and potential changing and diverse perspectives on privacy. To provide additional insight on this topic, Hendricks-Sturrup and Lu explore perspectives related to privacy and willingness to share RWD [Reference Hendricks-Sturrup and Lu24]. While participants were generally supportive of sharing prescription history and other types of RWD, there were concerns about sharing other sources of RWD and the potential for use of RWD by third parties without specific consent. This highlights a broader issue of both ensuring needed privacy protections (through policy and technology approaches) and improving education and transparency regarding data collection, use, and privacy practices. Continued efforts in these areas will help ultimately realize the broader benefits of RWD.

Infrastructure to support generating RWE [Reference Chu, Miller and Mathews25Reference Bergquist, Wax and Bennett28]

The global pandemic has taught us that critical infrastructure already needs to be in place when an urgent need arises. The universe of RWD that can result in RWE will continue to expand, and the infrastructure to enable the conversion of various data types (some of which may not even exist yet) into research-ready data will need to be nimble and flexible. In this Thematic Issue, readers will find higher-level perspectives on RWD infrastructure through lens of the Clinical and Translational Science Awards program [Reference Cure, ElShourbagy Ferreira and Fessel26Reference Bergquist, Wax and Bennett28], as well as examples of what can be accomplished when powerful RWD infrastructure is brought together with innovative thinking and scientific inquiry.

Summary

RWD can now be generated from multiple sources and at a much faster pace than traditional and more standardized clinical research information data collection sources (such as traditional case report forms), thus having the potential to, more accurately and in real-time, mimic the health journey of individuals and enrich our data universe in healthcare and research. However, utilization, integration, infrastructure, representativeness, privacy, security, and data sharing remain critical aspects for high quality, equitable, and secure RWD. The amalgamation of manuscripts herein provides a broad picture of sources, utilization, applicability, infrastructure, challenges, and opportunities to obtain meaningful RWD and generate the necessary RWE to ultimately improve the health journey for all.

Author contributions

All authors contributed equally to this manuscript.

Funding statement

None.

Competing interests

This work is solely the responsibility of the authors and does not represent the official views or policies of the NIH or the FDA.

Footnotes

*

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

References

Dang, LE, Fong, E, Tarp, JM, et al. Case study of semaglutide and cardiovascular outcomes: an application of the Causal roadmap to a hybrid design for augmenting an RCT control arm with real-world data. J Clin Transl Sci. 2023;7(1):e231. doi: 10.1017/cts.2023.656.CrossRefGoogle ScholarPubMed
Dang, LE, Gruber, S, Lee, H, et al. A causal roadmap for generating high-quality real-world evidence. J Clin Transl Sci. 2023;7(1):e212. doi: 10.1017/cts.2023.635.CrossRefGoogle ScholarPubMed
Elkin, PL, Brown, SH, Resendez, S, et al. COVID-19 vaccination and venous thromboembolism risk in older veterans. J Clin Transl Sci. 2023;7(1):e55. doi: 10.1017/cts.2022.527.CrossRefGoogle ScholarPubMed
Gilmore, CM, Lee, GC, Schmidt, S, Frei, CR. Antibiotic prescribing by age, sex, race, and ethnicity for patients admitted to the hospital with community-acquired bacterial pneumonia (CABP) in the all of us database. J Clin Transl Sci. 2023;7(1):e13226. doi: 10.1017/cts.2023.567.Google Scholar
Guarino, SH, Williams, KD, Caplan, RJ, Fawcett, M, Lanzkron, S. COVID-19 in hospitalized adult patients with sickle cell disease: a 2020 US cohort using cerner real-world data(TM) (CRWD). J Clin Transl Sci. 2023;7(1):e152. doi: 10.1017/cts.2023.577.CrossRefGoogle Scholar
López, JE, Kyle, A, Hosseini, AJ, Wilson, M, Soares, S. Educational video while “waiting-to-be-seen” in a cardiology outpatient clinic promotes opt-in self-consent for biobanking of remnant clinical biospecimens: a randomized-controlled trial. J Clin Transl Sci. 2023;7(1):e103. doi: 10.1017/cts.2023.518.CrossRefGoogle Scholar
Mohamed, Y, Song, X, McMahon, TM, et al. Electronic health record data quality variability across a multistate clinical research network. J Clin Transl Sci. 2023;7(1):e130. doi: 10.1017/cts.2023.548.CrossRefGoogle ScholarPubMed
Selker, HP, Dulko, D, Greenblatt, DJ, Palm, M, Trinquart, L. The use of N-of-1 trials to generate real-world evidence for optimal treatment of individuals and populations. J Clin Transl Sci. 2023;7(1):e203. doi: 10.1017/cts.2023.604.CrossRefGoogle ScholarPubMed
St Sauver, J, Fu, S, Sohn, S, et al. Identification of delirium from real-world electronic health record clinical notes. J Clin Transl Sci. 2023;7(1):e187. doi: 10.1017/cts.2023.610.CrossRefGoogle ScholarPubMed
Wasser, JS, Greenblatt, DJ. Applying real-world data from expanded-access (“compassionate use”) patients to drug development. J Clin Transl Sci. 2023;7(1):e181. doi: 10.1017/cts.2023.606.CrossRefGoogle ScholarPubMed
Williamson, BD, Wyss, R, Stuart, EA, et al. An application of the Causal roadmap in two safety monitoring case studies: causal inference and outcome prediction using electronic health record data. J Clin Transl Sci. 2023;7(1):e208. doi: 10.1017/cts.2023.632.CrossRefGoogle ScholarPubMed
Díaz, I, Lee, H, Kıcıman, E, et al. Sensitivity analysis for causality in observational studies for regulatory science. Journal of Clinical and Translational Science 2023;7(1):e267. doi: 10.1017/cts.2023.688.CrossRefGoogle ScholarPubMed
Hamer, MK, Sobczak, C, Whittington, L, et al. Real-world data to evaluate effects of a multi-level dissemination strategy on access, outcomes, and equity of monoclonal antibodies for COVID-19. J Clin Transl Sci. 2023;7(1):e258. doi: 10.1017/cts.2023.679.CrossRefGoogle ScholarPubMed
Tran, P, Jreij, B, Sistani, F, Shaya, FT. Disparities in maternal mortality. J Clin Transl Sci. 2023;7(1):e192. doi: 10.1017/cts.2023.520.CrossRefGoogle ScholarPubMed
Ostovari, M, Zhang, Z, Patel, V, Jurkovitz, C. Telemedicine and health disparities: association between the area deprivation index and primary care telemedicine utilization during the COVID-19 pandemic. J Clin Transl Sci. 2023;7(1):e168. doi: 10.1017/cts.2023.580.CrossRefGoogle ScholarPubMed
Kolba, NK, Lee, B, Tannous, HJ, Bilfinger, TV, Shroyer, AL. Preoperative mental illness and postoperative atrial fibrillation in cardiac surgery patients: identifying a vulnerable population. J Clin Transl Sci. 2023;7(1):e15. doi: 10.1017/cts.2022.493.CrossRefGoogle ScholarPubMed
Harris, DR, Anthony, N, Quesinberry, D, Delcher, C. Evidence of housing instability identified by addresses, clinical notes, and diagnostic codes in a real-world population with substance use disorders. J Clin Transl Sci. 2023;7(1):e196. doi: 10.1017/cts.2023.626.CrossRefGoogle Scholar
Adekunle, TB, Arreola, A, Sembian, S, et al. Feasibility and anticipated acceptability of community health worker-facilitated HPV self-sampling for cervical cancer screening around Lake County, Indiana. J Clin Transl Sci 2023;7(1):e157. doi: 10.1017/cts.2023.578.CrossRefGoogle ScholarPubMed
Krist, AH, Huffstetler, AN, Villalobos, G, et al. Use of population health data to promote equitable recruitment for a primary care practice implementation trial addressing unhealthy alcohol use. J Clin Transl Sci. 2023;7(1):e110. doi: 10.1017/cts.2023.530.CrossRefGoogle ScholarPubMed
Gonzales, S, Champieux, R, Contaxis, N, et al. Clinical and translational science personas: expansion and use cases. J Clin Transl Sci. 2023;7(1):e147. doi: 10.1017/cts.2023.572.CrossRefGoogle ScholarPubMed
Bautista, M, Cave, D, Downey, C, Bentham, JR, Jayne, D Clinical applications of contactless photoplethysmography for vital signs monitoring in pediatrics: A systematic review and meta-analysis. J Clin Transl Sci 2023;7(1): e144. doi: 10.1017/cts.2023.557.Google Scholar
Merkel, CA, Brady, KM, Votava-Smith, JK, Tran, NN. A pilot study: comparing a novel noninvasive measure of cerebrovascular stability index with an invasive measure of cerebral autoregulation in neonates with congenital heart disease. J Clin Transl Sci. 2023;7(1):e165. doi: 10.1017/cts.2023.581.CrossRefGoogle ScholarPubMed
Bautista, MJ, Kowal, M, Cave, DGW, Downey, C, Jayne, DG. Clinical applications of contactless photoplethysmography for monitoring in adults: A systematic review and meta-analysis. J Clin Transl Sci 2023;7(1):e129. doi: 10.1017/cts.2023.547.CrossRefGoogle ScholarPubMed
Hendricks-Sturrup, RM, Lu, CY. A survey of United States adult privacy perspectives and willingness to share real-world data. J Clin Transl Sci. 2023;7(1):e64. doi: 10.1017/cts.2023.4.CrossRefGoogle ScholarPubMed
Chu, I, Miller, R, Mathews, I, et al. FAIR Enough: Building an Academic Data Ecosystem to Make Real-World Data Available for Translational Research. Journal of Clinical and Translational Science 2024:1-23. doi: 10.1017/cts.2024.530.CrossRefGoogle Scholar
Cure, P, ElShourbagy Ferreira, S, Fessel, JP, et al. Real-world data for 21(st)-century medicine: the clinical and translational science awards program perspective. J Clin Transl Sci. 2023;7(1):e201. doi: 10.1017/cts.2023.588.CrossRefGoogle ScholarPubMed
Morrato, EH, Lennox, LA, Dearing, JW, et al. The evolve to next-gen ACT network: an evolving open-access, real-world data resource primed for real-world evidence research across the clinical and translational science award consortium. J Clin Transl Sci. 2023;7(1):e224. doi: 10.1017/cts.2023.617.CrossRefGoogle ScholarPubMed
Bergquist, T, Wax, M, Bennett, TD, et al. A framework for future national pediatric pandemic respiratory disease severity triage: the HHS pediatric COVID-19 data challenge. J Clin Transl Sci. 2023;7(1):e175. doi: 10.1017/cts.2023.549.CrossRefGoogle ScholarPubMed