320
Views
1
CrossRef citations to date
0
Altmetric
Review

An overview of methodological flaws of real-world studies investigating drug safety in the post-marketing setting

, ORCID Icon, , ORCID Icon &
Pages 373-380 | Received 03 Mar 2023, Accepted 26 May 2023, Published online: 29 May 2023

References

  • Monti S, Grosso V, Todoerti M, et al. Randomized controlled trials and real-world data: differences and similarities to untangle literature data. Rheumatology (Oxford). 2018;57:vii54–8.
  • Bérard A. Pharmacoepidemiology research-real-world evidence for decision making. Front Pharmacol. 2021;12:723427.
  • Spelsberg A, Prugger C, Doshi P, et al. Contribution of industry funded post-marketing studies to drug safety: survey of notifications submitted to regulatory agencies. BMJ. 2017;356:j337.
  • Crisafulli S, Sultana J, Ingrasciotta Y, et al. Role of healthcare databases and registries for surveillance of orphan drugs in the real-world setting: the Italian case study. Expert Opin Drug Saf. 2019;18(6):497–509. DOI:10.1080/14740338.2019.1614165
  • Hemkens LG. How routinely collected data for randomized trials provide long-term randomized real-world evidence. JAMA Netw Open. 2018;1(8):e186014.
  • Crisafulli S, Trifirò G. [The role of real-world evidence for the study of rare diseases epidemiology and the post-marketing evaluation of orphan drugs.]. Recenti Prog Med. 2022;113(7):425–429.
  • Gaasterland CMW, van der Weide MCJ-, du Prie-Olthof MJ, et al. The patient’s view on rare disease trial design – a qualitative study. Orphanet J Rare Dis. 2019;14(1):31. DOI:10.1186/s13023-019-1002-z
  • Mofid S, Bolislis WR, Kühler TC. Real-world data in the postapproval setting as applied by the EMA and the US FDA. Clin Ther. 2022;44(2):306–322.
  • Trifirò G, Crisafulli S. A new era of pharmacovigilance: future challenges and opportunities. Frontiers In Drug Safety And Regulation. 2022;2. DOI:10.3389/fdsfr.2022.866898
  • Dhanda S, Osborne V, Lynn E, et al. Postmarketing studies: can they provide a safety net for COVID-19 vaccines in the UK? BMJ EBM. 2022;27(1):1–6.
  • Food and Drug Administration. Real world evidence; Accessed 2023 Mar 3. Available form https://www.fda.gov/science-research/science-and-research-special-topics/real-world-evidence.
  • Liu F, Panagiotakos D. Real-world data: a brief review of the methods, applications, challenges and opportunities. BMC Med Res Methodol. 2022;22(1):287.
  • Raj N, Fernandes S, Charyulu NR, et al. Postmarket surveillance: a review on key aspects and measures on the effective functioning in the context of the United Kingdom and Canada. Ther Adv Drug Saf. 2019;10:2042098619865413.
  • Lavertu A, Vora B, Giacomini KM, et al. A new era in pharmacovigilance: toward real-world data and digital monitoring. Clin Pharmacol Ther. 2021;109(5):1197–1202.
  • Tan YY, Papez V, Chang WH, et al. Comparing clinical trial population representativeness to real-world populations: an external validity analysis encompassing 43 895 trials and 5 685 738 individuals across 989 unique drugs and 286 conditions in England. Lancet Healthy Longev. 2022;3(10):e674–89.
  • Pinnow E, Amr S, Bentzen SM, et al. Postmarket safety outcomes for new molecular entity (NME) drugs approved by the food and drug administration between 2002 and 2014. Clin Pharmacol Ther. 2018;104(2):390–400. DOI:10.1002/cpt.944
  • Zhang X, Zhang Y, Ye X, et al. Overview of phase IV clinical trials for postmarket drug safety surveillance: a status report from the ClinicalTrials.Gov registry. BMJ Open. 2016;6(11):e010643.
  • V WS, Sreedhara SK, Schneeweiss S. Repeat initiative. reproducibility of real-world evidence studies using clinical practice data to inform regulatory and coverage decisions. Nat Commun. 2022;13(1):5126.
  • Hiramatsu K, Barrett A, Miyata Y, et al. Current status, challenges, and future perspectives of real-world data and real-world evidence in Japan. Drugs Real World Outcomes. 2021;8(4):459–480. DOI:10.1007/s40801-021-00266-3
  • Burns L, Le RN, Kalesnik-Orszulak R, et al. Real-world evidence for regulatory decision-making: guidance from around the world. Clin Ther. 2022;44(3):420–437. DOI:10.1016/j.clinthera.2022.01.012
  • Flynn R, Plueschke K, Quinten C, et al. Marketing authorization applications made to the European medicines agency in 2018–2019: what was the contribution of real-world evidence? Clin Pharmacol Ther. 2022;111(1):90–97. DOI:10.1002/cpt.2461
  • Purpura CA, Garry EM, Honig N, et al. The role of real‐world evidence in FDA‐approved new drug and biologics license applications. Clin Pharmacol Ther. 2022;111(1):135–144.
  • Concato J, Corrigan-Curay J. Real-world evidence — where are we now? N Engl J Med. 2022;386(18):1680–1682.
  • Hall GC, Sauer B, Bourke A, et al. Guidelines for good database selection and use in pharmacoepidemiology research. Pharmacoepidemiol Drug Saf. 2012;21(1):1–10.
  • Food and Drug Administration, Sentinel Operations Center. Standardization and querying of data quality metrics and characteristics for electronic health data: data quality metrics system final report; 2023. https://www.sentinelinitiative.org/sites/default/files/Methods/Standardization_and_Querying_of_Data_Quality_Metrics.pdf
  • European Network of Centres for Pharmacoepidemiology and Pharmacovigilance(ENCePP). Guide on methodological standards in pharmacoepidemiology; 2022 [Accessed 2023 Mar 3], ( Revision 10). t: http://www.encepp.eu/standards_and_guidance
  • Ingrasciotta Y, Sultana J, Giorgianni F, et al. Association of individual non-steroidal anti-inflammatory drugs and chronic kidney disease: a population-based case control study. PLoS ONE. 2015;10(4):e0122899. DOI:10.1371/journal.pone.0122899
  • Crisafulli S, Ientile V, L’Abbate L, et al. COVID-19 patient management in outpatient setting: a population-based study from southern Italy. JCM. 2021;11(1):11. DOI:10.3390/jcm11010051
  • Belleudi V, Trotta F, Pinnarelli L, et al. Neonatal outcomes following new reimbursement limitations on palivizumab in Italy. Arch Dis Child. 2018;103(12):1163–1167.
  • Sultana J, Giorgianni F, Rea F, et al. All-cause mortality and antipsychotic use among elderly persons with high baseline cardiovascular and cerebrovascular risk: a multi-center retrospective cohort study in Italy. Expert Opin Drug Metab Toxicol. 2019;15(2):179–188. DOI:10.1080/17425255.2019.1561860
  • Ingrasciotta Y, Bertuccio MP, Crisafulli S, et al. Real world use of antidiabetic drugs in the years 2011–2017: a population-based study from Southern Italy. IJERPH. 2020;17(24):17. DOI:10.3390/ijerph17249514
  • Rea F, Calusi G, Franchi M, et al. Adherence of elderly patients with cardiovascular disease to statins and the risk of exacerbation of chronic obstructive pulmonary disease: evidence from an Italian real-world investigation. Drugs Aging. 2018;35(12):1099–1108. DOI:10.1007/s40266-018-0600-0
  • Trifirò G, Gini R, Barone-Adesi F, et al. The role of European healthcare databases for post-marketing drug effectiveness, safety and value evaluation: where does Italy stand? Drug Saf. 2019;42(3):347–363. DOI:10.1007/s40264-018-0732-5
  • Sultana J, Fontana A, Giorgianni F, et al. Measuring the effectiveness of safety warnings on the risk of stroke in older antipsychotic users: a nationwide cohort study in two large electronic medical records databases in the United Kingdom and Italy. Drug Saf. 2019;42(12):1471–1485. DOI:10.1007/s40264-019-00860-z
  • Lapi F, Capogrosso Sansone A, Mantarro S, et al. Hepatitis C virus infection: opportunities for an earlier detection in primary care. Eur J Gastroenterol Hepatol. 2017;29(3):271–276. DOI:10.1097/MEG.0000000000000785
  • Ferrajolo C, Verhamme KMC, Trifirò G, ’T Jong GW, Picelli G, Giaquinto C, et al. Antibiotic-induced liver injury in paediatric outpatients: a case-control study in primary care databases. Drug Saf. 2017;40(4):305–315. DOI:10.1007/s40264-016-0493-y
  • Traversa G, Sagliocca L, Magrini N, et al. A perspective for the role of drug registries in the post-marketing surveillance. Recenti Prog Med. 2013;104(6):280–287.
  • Khan MAA, Hamid S, Z-U-D B. Pharmacovigilance in high-income countries: current developments and a review of literature. Pharmacy (Basel). 2023;11(1):11.
  • Coloma PM, Trifirò G, Patadia V, et al. Postmarketing safety surveillance: where does signal detection using electronic healthcare records fit into the big picture? Drug Saf. 2013;36(3):183–197.
  • Pal SN, Duncombe C, Falzon D, et al. WHO strategy for collecting safety data in public health programmes: complementing spontaneous reporting systems. Drug Saf. 2013;36(2):75–81.
  • World Health Organization. Protocol template to be used as template for observational study protocols: cohort event monitoring (‎CEM)‎ for safety signal detection after vaccination with COVID-19 vaccines; 2021 [Accessed 2023 Mar 3]. Available from: https://apps.who.int/iris/handle/10665/342193
  • Barbieri MA, Cicala G, Cutroneo PM, et al. Safety profile of biologics used in rheumatology: an Italian prospective pharmacovigilance study. JCM. 2020;9(4):1227. DOI:10.3390/jcm9041227
  • Rachlis B, Karwa R, Chema C, et al. Targeted spontaneous reporting: assessing opportunities to conduct routine pharmacovigilance for antiretroviral treatment on an international scale. Drug Saf. 2016;39(10):959–976. DOI:10.1007/s40264-016-0434-9
  • Crisafulli S, Cutroneo PM, Verhamme K, et al. Drug-induced urinary retention: an analysis of a national spontaneous adverse drug reaction reporting database. Eur Urol Focus. 2022;8(5):1424–1432. DOI:10.1016/j.euf.2021.07.001
  • Crisafulli S, Cutroneo PM, Luxi N, et al. Is pegylation of drugs associated with hypersensitivity reactions? an analysis of the Italian national spontaneous adverse drug reaction reporting system. Drug Saf. 2023;46(4):343–355. DOI:10.1007/s40264-023-01277-5
  • Faillie J-L. Case–non-case studies: principle, methods, bias and interpretation. Therapie. 2019;74(2):225–232.
  • Franco V, Barbieri MA, Cutroneo PM, et al. Pediatric adverse reactions to antiseizure medications – an analysis of data from the Italian spontaneous reporting system (2001–2019). Epilepsy Behav. 2021;119:107989.
  • Cicala G, Barbieri MA, Santoro V, et al. Safety and tolerability of antipsychotic drugs in pediatric patients: data from a 1-year naturalistic study. Front Psychiatry. 2020;11:11.
  • Ventola CL. Big data and pharmacovigilance: data mining for adverse drug events and interactions. P T. 2018;43(6):340–351.
  • Penberthy LT, Rivera DR, Lund JL, et al. An overview of real-world data sources for oncology and considerations for research. CA Cancer J Clin. 2022;72(3):287–300.
  • Ho Y-F, Hu F-C, Lee P-I. The advantages and challenges of using real-world data for patient care. Clin Transl Sci. 2020;13(1):4–7.
  • Cea Soriano L, Soriano-Gabarró M, García Rodríguez LA. Validation of low-dose aspirin prescription data in the health improvement network: how much misclassification due to over-the-counter use? Pharmacoepidemiol Drug Saf. 2016;25(4):392–398.
  • Bower JK, Patel S, Rudy JE, et al. Addressing bias in electronic health record-based surveillance of cardiovascular disease risk: finding the signal through the noise. Curr Epidemiol Rep. 2017;4(4):346–352.
  • Hazell L, Shakir SAW. Under-reporting of adverse drug reactions: a systematic review. Drug Saf. 2006;29(5):385–396.
  • Palleria C, Leporini C, Chimirri S, et al. Limitations and obstacles of the spontaneous adverse drugs reactions reporting: two “challenging” case reports. J Pharmacol Pharmacother. 2013;4(1_suppl):S66–72. DOI:10.4103/0976-500X.120955
  • Cutroneo PM, Giardina C, Ientile V, et al. Overview of the safety of anti-VEGF drugs: analysis of the Italian spontaneous reporting system. Drug Saf. 2017;40(11):1131–1140. DOI:10.1007/s40264-017-0553-y
  • Arora A, Jalali RK, Vohora D. Relevance of the weber effect in contemporary pharmacovigilance of oncology drugs. TCRM. 2017;13:1195–1203.
  • Hung E, Hauben M, Essex H, et al. More extreme duplication in FDA adverse event reporting system detected by literature reference normalization and fuzzy string matching. Pharmacoepidemiol Drug Saf. 2023;32(3):387–391.
  • Stürmer T, Wang T, Golightly YM, et al. Methodological considerations when analysing and interpreting real-world data. Rheumatology (Oxford). 2020;59(1):14–25.
  • Danaei G, Tavakkoli M, Hernán MA. Bias in observational studies of prevalent users: lessons for comparative effectiveness research from a meta-analysis of statins. Am J Epidemiol. 2012;175(4):250–262.
  • Moride Y, Abenhaim L. Evidence of the depletion of susceptibles effect in non-experimental pharmacoepidemiologic research. J Clin Epidemiol. 1994;47(7):731–737.
  • Althubaiti A. Information bias in health research: definition, pitfalls, and adjustment methods. JMDH. 2016;9:211–217.
  • Coughlin SS. Recall bias in epidemiologic studies. J Clin Epidemiol. 1990;43(1):87–91.
  • Horwitz RI, Feinstein AR. The problem of “protopathic bias” in case-control studies. Am j med. 1980;68(2):255–258.
  • Abrahami D, Pradhan R, Yin H, et al. Proton pump inhibitors and the risk of inflammatory bowel disease: population-based cohort study. Gut. 2023. DOI:10.1136/gutjnl-2022-328866
  • Suissa S. Immortal time bias in pharmaco-epidemiology. Am J Epidemiol. 2008;167(4):492–499.
  • Suissa S. Immortal time bias in observational studies of drug effects. Pharmacoepidem Drug Safe. 2007;16(3):241–249.
  • Suissa S, Azoulay L. Metformin and the risk of cancer: time-related biases in observational studies. Diabetes Care. 2012;35(12):2665–2673.
  • Gini R, Sturkenboom MCJ, Sultana J, et al. Different strategies to execute multi-database studies for medicines surveillance in real-world setting: a reflection on the European model. Clin Pharmacol Ther. 2020;108(2):228–235. DOI:10.1002/cpt.1833
  • Kent S, Burn E, Dawoud D, et al. Common problems, common data model solutions. Evidence Generation For Health Technology Assessment Pharmacoeconomics. 2021;39(3):275–285. DOI:10.1007/s40273-020-00981-9
  • Madigan D, Ryan PB, Schuemie M. Does design matter? Systematic evaluation of the impact of analytical choices on effect estimates in observational studies. Ther Adv Drug Saf. 2013;4(2):53–62.
  • Madigan D, Ryan PB, Schuemie M, et al. Evaluating the impact of database heterogeneity on observational study results. Am J Epidemiol. 2013;178(4):645–651. DOI:10.1093/aje/kwt010
  • Bykov K, Patorno E, D’Andrea E, et al. Prevalence of avoidable and bias-inflicting methodological pitfalls in real-world studies of medication safety and effectiveness. Clin Pharmacol Ther. 2022;111(1):209–217. DOI:10.1002/cpt.2364
  • Heads of Medicines Agencies, European Medicines Agency. HMA-EMA joint big data taskforce phase ii report: “evolving data-driven regulation.“ 2018 [Accessed 2023 Mar 3]. https://www.ema.europa.eu/en/documents/other/hma-ema-joint-big-datataskforce-phase-ii-report-evolving-data-drivenregulation_en.pdf
  • Food and Drug Administration. Framework for FDA’s real-world evidence program; 2018 [Accessed 2023 March 3]. https://www.fda.gov/media/120060/download
  • Desai RJ, Franklin JM. Alternative approaches for confounding adjustment in observational studies using weighting based on the propensity score: a primer for practitioners. BMJ. 2019;367:l5657.
  • Rosenbaum PR, Rubin DB. The central role of the propensity score in observational studies for causal effects. Biometrika. 1983;70(1):41–55.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.