Next Article in Journal
eHealth Literacy in German Skin Cancer Patients
Next Article in Special Issue
Development and Evaluation of Machine Learning-Based High-Cost Prediction Model Using Health Check-Up Data by the National Health Insurance Service of Korea
Previous Article in Journal
In Silico Approach in the Evaluation of Pro-Inflammatory Potential of Polycyclic Aromatic Hydrocarbons and Volatile Organic Compounds through Binding Affinity to the Human Toll-Like Receptor 4
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers

by
Teresa Magalhães
1,2,3,4,*,
Ricardo Jorge Dinis-Oliveira
1,3,4,5,* and
Tiago Taveira-Gomes
2,3,6,7,*
1
Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
2
Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal
3
MTG Research and Development Lab, 4200-604 Porto, Portugal
4
TOXRUN—Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), CRL, 4585-116 Gandra, Portugal
5
UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
6
Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
7
Faculty of Health Sciences, University Fernando Pessoa (FCS-UFP), 4249-004 Porto, Portugal
*
Authors to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2022, 19(14), 8364; https://doi.org/10.3390/ijerph19148364
Submission received: 28 June 2022 / Accepted: 7 July 2022 / Published: 8 July 2022

Abstract

:
Real world data (RWD) and real-world evidence (RWE) plays an increasingly important role in clinical research since scientific knowledge is obtained during routine clinical large-scale practice and not experimentally as occurs in the highly controlled traditional clinical trials. Particularly, the electronic health records (EHRs) are a relevant source of data. Nevertheless, there are also significant challenges in the correct use and interpretation of EHRs data, such as bias, heterogeneity of the population, and missing or non-standardized data formats. Despite the RWD and RWE recognized difficulties, these are easily outweighed by the benefits of ensuring the efficacy, safety, and cost-effectiveness in complement to the gold standards of the randomized controlled trial (RCT), namely by providing a complete picture regarding factors and variables that can guide robust clinical decisions. Their relevance can be even further evident as healthcare units develop more accurate EHRs always in the respect for the privacy of patient data. This editorial is an overview of the RWD and RWE major aspects of the state of the art and supports the Special Issue on “Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers” aimed to explore all the potential and the utility of RWD and RWE in offering insights on diseases in a broad spectrum.

Real-world data (RWD) are currently collected from a variety of sources, namely electronic medical records (EMRs), claims and billing databases, product and disease registries, patient-generated data, and home medical devices for monitoring patients, such as the smartwatches. From RWD and through robust analytics, real-world evidence (RWE) can be produced with clear potential benefits for the health and outcomes of patients [1,2,3]. In other words, RWE offers a real difference between what is expected to happen and what is really happening specially in comparison to traditional clinical trials, whose well-known limitations of more homogeneous populations, make it difficult to generalize findings to larger scales. Examples of real-world health and medicine data include EMRs regarding patient demographics, family history, comorbidities, treatments, outcomes, and other information, the National Health Insurance claims data, cancer registry data, and reports on adverse drug reactions. Automated data abstraction (e.g., from EHRs) has also been shown to be highly accurate and faster than manual abstraction and its value may increase if more structured EHRs, high-quality datasets and terms accepted as being the gold standard definition are used; in other words this evolution can project RWE studies to an higher scale [4,5]. Indeed, the successful implementation of EHRs in institutions across the globe has opened a novel research era with new challenges for the scientific medical research community. Even in comparison to randomized controlled clinical trials (RCTs), RWE studies are emerging as a post-market surveillance approach to confer valuable complementing evidence specially aiming to identify rare adverse events or long-term effects of existing drugs due to their potential to analyze larger patient populations across longer timelines [6,7,8]. Recently our group has been exploring the usefulness of RWD and RWE in several fields of medicine, namely in studying the prevalence of heart failure, type 2 diabetes mellitus, chronic kidney disease, to name a few [9,10]. The Sentinel System [11] is an example of the extensive use of RWD to uncover adverse drug reactions. Recognizing the potential for additional use of RWD and RWE for regulatory and funding purposes, the United States Congress included in the 21st Century Cures legislation (Cures) (Public Law 114–225, 13 December 2016) the direction for Food and Drug Administration (FDA) to develop a program to evaluate the potential use of RWE to support the approval of a new indication of an approved drug or to support or satisfy post-approval study requirements. The potential benefits of leveraging RWD, stored in EHRs and other sources, have become even more evident with the COVID-19 pandemic either by evaluating the efficacy of vaccine, the risk of diabetes following COVID-19 or the course of COVID-19 in patients with lysosomal storage disorders [12,13,14].
In another emergent perspective, and as complement to the modern techniques available to forensic sciences to solve many cases, by incorporating big data analytics and RWE, a reshaping the world of forensics is expected, for example by developing predictive models of violence, such as domestic violence and accidents. Despite the huge amount of data generated in forensic cases, as recently highlighted, in the field of forensics, big data is still waiting for a comprehensive outline of its contribution to the field [15]. Additionally, insurance medicine is an importance source of RWD regarding diseases and accidents, not only in the traditional view of health but also in the social medicine perspective [1].
In the Special Issue entitled “Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers” (https://www.mdpi.com/journal/ijerph/special_issues/digital_big_data), we are interested in receiving original articles, reviews, technical notes, protocols, guidelines, etc., with no restriction on the length of the papers, exploring the usefulness of RWD and RWE in offering insights on diseases, regarding the pathophysiological aspects, new techniques for diagnosis, more safe treatments, novel preventive, and predictive models of diseases identification, evaluate the effectiveness of clinical guidelines, study the morbidity, mortality, and socioeconomic impact. However, much more can be generated from RWE to contribute to the medical knowledge from the regional up to the global level, ultimately leading to changes in healthcare policies and contribute to the sustainability of healthcare systems, especially if performed and respecting specific criteria of robustness and transparency to achieve high-quality evidence avoiding incorrect or unreliable conclusions. Despite RWE can virtually be generated from every healthcare institution around the globe, to reach the RWE expected outcomes, the setting in which evidence is generated and the methodologic approach used to conduct the surveillance or research are two critical dimensions to be considered.

Author Contributions

All authors contributed for the study conception and design, selection of bibliography and revision and final approval of the final version for submission. R.J.D.-O. prepared first draft. All attests that listed authors meet authorship criteria and that no others meeting the criteria have been omitted. All authors have read and agreed to the published version of the manuscript.

Funding

The author has no relevant affiliation or financial involvement with any organisation or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

Conflicts of Interest

T.M. and T.T.-G. hold shares in MTG. R.J.D.-O. declares no conflict of interest.

References

  1. Ahn, E.K. A brief introduction to research based on real-world evidence: Considering the Korean National Health Insurance Service database. Integr. Med. Res. 2022, 11, 100797. [Google Scholar] [CrossRef] [PubMed]
  2. Sherman, R.E.; Anderson, S.A.; Dal Pan, G.J.; Gray, G.W.; Gross, T.; Hunter, N.L.; LaVange, L.; Marinac-Dabic, D.; Marks, P.W.; Robb, M.A.; et al. Real-World Evidence—What Is It and What Can It Tell Us? N. Engl. J. Med. 2016, 375, 2293–2297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Levenson, M.S. Regulatory-grade clinical trial design using real-world data. Clin. Trials 2020, 17, 377–382. [Google Scholar] [CrossRef] [PubMed]
  4. Gauthier, M.P.; Law, J.H.; Le, L.W.; Li, J.J.N.; Zahir, S.; Nirmalakumar, S.; Sung, M.; Pettengell, C.; Aviv, S.; Chu, R.; et al. Automating Access to Real-World Evidence. JTO Clin. Res. Rep. 2022, 3, 100340. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, J.; Symons, J.; Agapow, P.; Teo, J.T.; Paxton, C.A.; Abdi, J.; Mattie, H.; Davie, C.; Torres, A.Z.; Folarin, A.; et al. Best practices in the real-world data life cycle. PLoS Digit. Health 2022, 1, e0000003. [Google Scholar] [CrossRef]
  6. Beaulieu-Jones, B.K.; Finlayson, S.G.; Yuan, W.; Altman, R.B.; Kohane, I.S.; Prasad, V.; Yu, K.H. Examining the Use of Real-World Evidence in the Regulatory Process. Clin. Pharmacol. Ther. 2020, 107, 843–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Monti, S.; Grosso, V.; Todoerti, M.; Caporali, R. Randomized controlled trials and real-world data: Differences and similarities to untangle literature data. Rheumatology 2018, 57, vii54–vii58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Chen, D. Real-world studies: Bridging the gap between trial-assessed efficacy and routine care. J. Biomed. Res. 2022, 36, 147–154. [Google Scholar] [CrossRef] [PubMed]
  9. Gavina, C.; Carvalho, D.S.; Dias, D.M.; Bernardo, F.; Martinho, H.; Couceiro, J.; Santos-Araújo, C.; Dinis-Oliveira, R.J.; Taveira-Gomes, T. Premature Mortality in Type 2 Diabetes Mellitus Associated with Heart Failure and Chronic Kidney Disease: 20 Years of Real-World Data. J. Clin. Med. 2022, 11, 2131. [Google Scholar] [CrossRef] [PubMed]
  10. Gavina, C.; Carvalho, D.S.; Valente, F.; Bernardo, F.; Dinis-Oliveira, R.J.; Santos-Araújo, C.; Taveira-Gomes, T. 20 Years of Real-World Data to Estimate the Prevalence of Heart Failure and Its Subtypes in an Unselected Population of Integrated Care Units. J. Cardiovasc. Dev. Dis. 2022, 9, 149. [Google Scholar] [CrossRef] [PubMed]
  11. Platt, R.; Wilson, M.; Chan, K.A.; Benner, J.S.; Marchibroda, J.; McClellan, M. The new Sentinel Network—Improving the evidence of medical-product safety. N. Engl. J. Med. 2009, 361, 645–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zaccardi, F.; Khunti, K. Risk of diabetes following COVID-19: Translating evidence into clinical and public health actions. J. Clin. Endocrinol. Metab. 2022, dgac384. [Google Scholar] [CrossRef] [PubMed]
  13. Perrella, A.; Bisogno, M.; D’Argenzio, A.; Trama, U.; Coscioni, E.; Orlando, V. Risk of SARS-CoV-2 Infection Breakthrough among the Non-Vaccinated and Vaccinated Population in Italy: A Real-World Evidence Study Based on Big Data. Healthcare 2022, 10, 1085. [Google Scholar] [CrossRef] [PubMed]
  14. Kilavuz, S.; Kor, D.; Bulut, F.D.; Serbes, M.; Karagoz, D.; Altıntas, D.U.; Bisgin, A.; Seydaoğlu, G.; Mungan, H.N.O. Real-world patient data on immunity and COVID-19 status of patients with MPS, Gaucher, and Pompe diseases from Turkey. Arch. Pediatr. 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  15. Lefèvre, T. Big data in forensic science and medicine. J. Forensic Leg. Med. 2018, 57, 1–6. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Magalhães, T.; Dinis-Oliveira, R.J.; Taveira-Gomes, T. Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers. Int. J. Environ. Res. Public Health 2022, 19, 8364. https://doi.org/10.3390/ijerph19148364

AMA Style

Magalhães T, Dinis-Oliveira RJ, Taveira-Gomes T. Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers. International Journal of Environmental Research and Public Health. 2022; 19(14):8364. https://doi.org/10.3390/ijerph19148364

Chicago/Turabian Style

Magalhães, Teresa, Ricardo Jorge Dinis-Oliveira, and Tiago Taveira-Gomes. 2022. "Digital Health and Big Data Analytics: Implications of Real-World Evidence for Clinicians and Policymakers" International Journal of Environmental Research and Public Health 19, no. 14: 8364. https://doi.org/10.3390/ijerph19148364

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop