445
Views
3
CrossRef citations to date
0
Altmetric
Special Section: A Collection of Articles on Opportunities and Challenges in Utilizing Real-World Data for Clinical Trials and Medical Product Development

Applying Quantitative Approaches in the Use of RWE in Clinical Development and Life-Cycle Management

, , &
Pages 57-68 | Received 02 Feb 2021, Accepted 04 May 2021, Published online: 06 Jul 2021

References

  • Aetion (2020), “eBook: The Role of Real-World Evidence in FDA Approvals,” available at https://aetion.com/evidence-hub/ebook-the-role-of-real-world-evidence-in-fda-approvals/.
  • Alexander, M., Loomis, A. K., Fairburn-Beech, J., van der Lei, J., Duarte-Salles, T., Prieto-Alhambra, D., Ansell, D., Pasqua, A., Lapi, F., Rijnbeek, P., Mosseveld, M., Avillach, P., Egger, P., Kendrick, S., Waterworth, D. M., Sattar, N., and Alazawi, W. (2018), “Real-world Data Reveal a Diagnostic Gap in Non-Alcoholic Fatty Liver Disease,” BMC Medicine, 16, 130. DOI: 10.1186/s12916-018-1103-x.
  • Austin, P. C. (2011), “An Introduction to Propensity Score Methods for Reducing the Effects of Confounding in Observational Studies,” Multivariate Behavioral Research, 46, 399–424. DOI: 10.1080/00273171.2011.568786.
  • Baumfeld Andre, E., Reynolds, R., Caubel, P., Azoulay, L., and Dreyer, N. A. (2020), “Trial Designs Using Real-world Data: The Changing Landscape of the Regulatory Approval Process,” Pharmacoepidemiology and Drug Safety, 29, 1201–1212. DOI: 10.1002/pds.4932.
  • Berger, M. L., Sox, H., Willke, R. J., Brixner, D. L., Eichler, H., Goettsch, W., Madigan, D., Makady, A., Schneeweiss, S., Tarricone, R., Wang, S. V., Watkins, J., and Mullins, C. D. (2017), “Good Practices for Real-World Data Studies of Treatment and/or Comparative Effectiveness: Recommendations from the Joint ISPOR-ISPE Special Task Force on Real-World Evidence in Health Care Decision Making,” Value in Health, 20, 1003–1008. DOI: 10.1016/j.jval.2017.08.3019.
  • BIO (2019), “Incorporating Real-world Evidence Within the Label of an FDA-approved Drug: Perspectives from BIO Membership,” available at https://www.bio.org/sites/default/files/legacy/bioorg/docs/BIO/%20White/%20paper/%20/%20-/%20Incorporating/%20RWE/%20Within/%20the/%20Label/_FINAL/%202019.pdf.
  • Bolislis, W. R., Fay, M., and Kühler, T. C. (2020), “Use of Real-World Data for New Drug Applications and Line Extensions,” Clinical Therapeutics, 42, 926–938. DOI: 10.1016/j.clinthera.2020.03.006.
  • Burcu, M., Dreyer, N. A., Franklin, J. M., Blum, M. D., Critchlow, C. W., Perfetto, E. M., and Zhou, W. (2020), “Real-World Evidence to Support Regulatory Decision-Making for Medicines: Considerations for External Control Arms,” Pharmacoepidemiology and Drug Safety, 29, 1228–1235. DOI: 10.1002/pds.4975.
  • Chen, J., Ho, M., Lee, K., Song, Y., Fang, Y., Goldstein, B. A., He, W., Irony, T., Jiang, Q., van der Laan, M., Lee, H., Lin, X., Meng, Z., Mishra-Kalyani, P., Rockhold, F., Wang, H., and White, R. (2021), “The Current Landscape in Biostatistics of Real-World Data and Evidence: Use of RWD/RWE to Inform Clinical Study Design and Analysis,” Statistics in Biopharmaceutical Research, 1–14.
  • Darrow, J. J., Avorn, J., and Kesselheim, A. S. (2017), “Speed, Safety, and Industry Funding — From PDUFA I to PDUFA VI,” New England Journal of Medicine, 377, 2278–2286. DOI: 10.1056/NEJMhle1710706.
  • Dreyer, N. A., Velentgas, P., Westrich, K., and Dubois, R. (2014), “The GRACE Checklist for Rating the Quality of Observational Studies of Comparative Effectiveness: A Tale of Hope and Caution,” Journal of Managed Care Pharmacy, 20, 301–308. DOI: 10.18553/jmcp.2014.20.3.301.
  • Duke Margolis (2017), “A Framework for Regulatory Use of Real World Evidence,” Duke-Margolis Center for Health Policy.
  • Duke Margolis (2018), “Characterizing RWD Quality and Relevancy for Regulatory Purposes,” Duke-Margolis Center for Health Policy.
  • Duke Margolis (2019a), “Determining Real-World Data’s Fitness for Use and the Role of Reliability. Duke-Margolis Center for Health Policy,” Duke-Margolis Center for Health Policy.
  • Duke Margolis (2019b), “Understanding the Need for Non-Interventional Studies Using Secondary Data to Generate Real-World Evidence for Regulatory Decision Making, and Demonstrating Their Credibility,” Duke-Margolis Center for Health Policy.
  • Duke Margolis (2020), “A Roadmap for Developing Study Endpoints in Real-World Settings,” Duke-Margolis Center for Health Policy.
  • EHDEN (2017), “European Health Data & Evidence Network,” available at https://www.ehden.eu/.
  • EMA (2018), “Post-Authorisation Safety Studies (PASS),” European Medicines Agency, available at https://www.ema.europa.eu/en/human-regulatory/post-authorisation/pharmacovigilance/post-authorisation-safety-studies-pass-0.
  • EMA (2020), “Guideline on Registry-based Studies,” European Medicines Agency, available at https://www.ema.europa.eu/en/guideline-registry-based-studies.
  • Fang, Y., He, W., Hu, X., and Wang, H. (2021), “A Method for Sample Size Calculation via E-Value in the Planning of Observational Studies,” Pharmaceutical Statistics, 20, 163–174. DOI: 10.1002/pst.2064.
  • Fang, Y., He, W., Wang, H., and Wu, M. (2020a), “Key Considerations in the Design of Real-World Studies,” Contemporary Clinical Trials, 96, 106091. DOI: 10.1016/j.cct.2020.106091.
  • Fang, Y., Wang, H., and He, W. (2020b), “A Statistical Roadmap for Journey from Real-World Data to Real-World Evidence,” Therapeutic Innovation & Regulatory Science, 54, 749–757. DOI: 10.1007/s43441-019-00008-2.
  • FDA (2010), “Regulations 21CFR314.126 – Adequate and Well-controlled Studies,” available at https://www.govinfo.gov/app/details/CFR-2010-title21-vol5/CFR-2010-title21-vol5-sec314-126/summary.
  • FDA (2018), “Framework for FDA’s Real-World Evidence Program,” available at https://www.fda.gov/media/120060/download.
  • FDA (2019a), “Submitting Documents Using Real-World Data and Real-World Evidence to FDA for Drugs and Biologics Guidance for Industry,” U.S. Food and Drug Administration, FDA, available at https://www.fda.gov/regulatory-information/search-fda-guidance-documents/submitting-documents-using-real-world-data-and-real-world-evidence-fda-drugs-and-biologics-guidance.
  • FDA (2019b), “Demonstrating Substandtial Evidence of Effectiveness for Human Drug and Biological Products Guidance for Industry,” U.S. Food and Drug Administration, FDA, available at https://www.fda.gov/regulatory-information/search-fda-guidance-documents/demonstrating-substantial-evidence-effectiveness-human-drug-and-biological-products.
  • FDA (2020a), “Exploring the Use of Real-World Data to Generate Real-World Evidence in Regulatory Decision-Making (U01) Clinical Trials Optional,” available at https://grants.nih.gov/grants/guide/rfa-files/RFA-FD-20-030.html.
  • FDA (2020b), “FDA Announces 4 Grant Awards for Projects Exploring the Use of Real-World Data to Generate Real-World Evidence in Regulatory Decision-Making,” available at https://www.fda.gov/drugs/science-and-research-drugs/fda-announces-4-grant-awards-projects-exploring-use-real-world-data-generate-real-world-evidence.
  • Feinberg, B. A., Gajra, A., Zettler, M. E., Phillips, T. D., Phillips Jr, E. G., and Kish, J. K. (2020), “Use of Real-World Evidence to Support FDA Approval of Oncology Drugs,” Value in Health, 23, 1358–1365. DOI: 10.1016/j.jval.2020.06.006.
  • Franklin, J. M., Patorno, E., Desai, R. J., Glynn, R. J., Martin, D., Quinto, K., Pawar, A., Bessette, L. G., Lee, H., and Garry, E. M. (2020a), “Emulating Randomized Clinical Trials with Nonrandomized Real-World Evidence Studies: First Results from the RCT DUPLICATE Initiative,” Circulation, 143, 1002–1013. DOI: 10.1161/CIRCULATIONAHA.120.051718.
  • Franklin, J. M., Pawar, A., Martin, D., Glynn, R. J., Levenson, M., Temple, R., and Schneeweiss, S. (2020b), “Nonrandomized Real-World Evidence to Support Regulatory Decision Making: Process for a Randomized Trial Replication Project,” Clinical Pharmacology & Therapeutics, 107, 817–826. DOI: 10.1002/cpt.1633.
  • Franklin, J. M., and Schneeweiss, S. (2017), “When and How Can Real World Data Analyses Substitute for Randomized Controlled Trials?,” Clinical Pharmacology and Therapeutics, 102, 924–933. DOI: 10.1002/cpt.857.
  • Goldstein, B. A., Navar, A. M., Pencina, M. J., and Ioannidis, J. (2017), “Opportunities and Challenges in Developing Risk Prediction Models with Electronic Health Records Data: A Systematic Review,” Journal of the American Medical Informatics Association, 24, 198–208. DOI: 10.1093/jamia/ocw042.
  • Hastie, T., Tibshirani, R., and Friedman, J. (2009), The Elements of Statistical Learning: Data Mining, Inference, and Prediction (2nd ed.). Springer Science & Business Media.
  • Haynes, R. B. (2006), “Forming Research Questions,” Journal of Clinical Epidemiology, 59, 881–886.
  • He, W., and Cao, X. (2014), “Approaches for Patient Recruitment Modeling and Simulation,” in Practical Considerations for Adaptive Trial Design and Implementation, eds. W. He, J. Pinheiro, O. Kuznetsova, Springer, pp. 299–318.
  • Hernan, M. A., and Robins, J. M. (2020), Causal Inference: What If, Boca Raton: Chapman & Hall/CRC.
  • Ho, M., van der Laan, M., Lee, H., Chen, J., Kee, K., Fang, Y., He, W., Irony, T., Jiang, Q., Lin, X., Meng, Z., Mishra-Kalyani, P., Rockhold, F., Song, Y., Wang, H., and White, R. (2021), “The Current Landscape in Causal Inference Frameworks for Design and Analysis of Studies Using Real-world Data and Evidence,” Statistics in Biopharmaceutical Research.
  • Hulley, S. B. (2007), Designing Clinical Research, Lippincott Williams & Wilkins.
  • ICH (1998), “ICH E9 Statisitial Principles for Clinical Trials,” available at https://www.ich.org/page/efficacy-guidelines.
  • ICH (2016), “ICH E6(R2) Integrated addendum to E6(R1): Guideline for Good Clinical Practice,” available at https://www.ich.org/page/efficacy-guidelines.
  • ICH (2019), “ICH E9(R1) Addendum to Statistical Principles for Clinical Trials on Choosing Appropriate Estimands and Defining Sensitivity Analyses in Clinical Trials,” available at https://www.ich.org/page/efficacy-guidelines.
  • IMI (2013), “Innovative Medicines Initiative GetReal,” available at https://www.imi-getreal.eu/.
  • James, G., Witten, D., Hastie, T., and Tibshirani, R. (2013), An Introduction to Statistical Learning, Springer Texts in Statistics, New York, NY: Springer.
  • Johnston, A., Jones, W. S., and Hernandez, A. F. (2016), “The ADAPTABLE Trial and Aspirin Dosing in Secondary Prevention for Patients with Coronary Artery Disease,” Current Cardiology Reports, 18, 1–9. DOI: 10.1007/s11886-016-0749-2.
  • Kalincik, T., Kuhle, J., Pucci, E., Rojas, J. I., Tsolaki, M., Sirbu, C.-A., Slee, M., Butzkueven, H., MSBase Scientific Leadership Group, and MSBase Study Group (2017), “Data Quality Evaluation for Observational Multiple Sclerosis Registries,” Multiple Sclerosis Journal, 23, 647–655. DOI: 10.1177/1352458516662728.
  • Levenson, M., He, W., Chen, J., Fang, Y., Faries, D., Goldstein, B. A., Ho, M., Lee, K., Mishra-Kalyani, P., Rockhold, F., Wang, H., and Zink, R. (2021), “Biostatistical Considerations When Using RWD and RWE in Clinical Studies for Regulatory Purposes: A Landscape Assessment,” Statistics in Biopharmaceutical Research, 1–11.
  • Longhurst, C. A., Harrington, R. A., and Shah, N. H. (2014), “A ‘Green Button’ for Using Aggregate Patient Data at the Point of Care,” Health Affairs, 33, 1229–1235.
  • Lou, J., Sarin, K. C., Toh, K. Y., Dabak, S., Adler, A., Ahn, J., Bayani, D. B. S., Chan, K., Choiphel, D., and Chua, B. (2020), “Real-World Data for Health Technology Assessment for Reimbursement Decisions in Asia: Current Landscape and A Way Forward,” International Journal of Technology Assessment in Health Care, 36, 474–480. DOI: 10.1017/S0266462320000628.
  • Loudon, K., Treweek, S., Sullivan, F., Donnan, P., Thorpe, K. E., and Zwarenstein, M. (2015), “The PRECIS-2 Tool: Designing Trials That Are Fit for Purpose,” BMJ, 350, h2147. DOI: 10.1136/bmj.h2147.
  • Makady, A., Ten Ham, R., de Boer, A., Hillege, H., Klungel, O., and Goettsch, W. (2017), “Policies for Use of Real-World Data in Health Technology Assessment (HTA): A Comparative Study of Six HTA Agencies,” Value in Health, 20, 520–532. DOI: 10.1016/j.jval.2016.12.003.
  • Makady, A., van Veelen, A., Jonsson, P., Moseley, O., D’Andon, A., de Boer, A., Hillege, H., Klungel, O., and Goettsch, W. (2018), “Using Real-World Data in Health Technology Assessment (HTA) Practice: A Comparative Study of Five HTA Agencies,” Pharmacoeconomics, 36, 359–368. DOI: 10.1007/s40273-017-0596-z.
  • MHRA (2020), “MHRA Draft Guidance on Randomised Controlled Trials Generating Real-World Evidence to Support Regulatory Decisions,” GOV.UK, available at https://www.gov.uk/government/consultations/mhra-draft-guidance-on-randomised-controlled-trials-generating-real-world-evidence-to-support-regulatory-decisions.
  • NASEM (2019), Examining the Impact of Real-World Evidence on Medical Product Development: Proceedings of a Workshop Series, The National Academies Press. DOI: 10.17226/25352.
  • National Research Council (2013), The Prevention and Treatment of Missing Data in Clinical Trials, National Academies Press.
  • NICE (2020), “Changes We’re Making to Health Technology Evaluation,” available at https://www.nice.org.uk/about/what-we-do/our-programmes/nice-guidance/nice-technology-appraisal-guidance/changes-to-health-technology-evaluation.
  • NMPA (2020), “NMPA (CFDA) English Regulations,” China Med Device, available at https://chinameddevice.com/resources2/nmpa-cfda-regulations/.
  • Rockhold, F., Bromley, C., Wagner, E. K., and Buyse, M. (2019), “Open Science: The Open Clinical Trials Data Journey,” Clinical Trials, 16, 539–546. DOI: 10.1177/1740774519865512.
  • Rockhold, F. W., Tenenbaum, J. D., Richesson, R., Marsolo, K. A., and O’Brien, E. C. (2020), “Design and Analytic Considerations for Using Patient-Reported Health Data in Pragmatic Clinical Trials: Report from an NIH Collaboratory Roundtable,” Journal of the American Medical Informatics Association, 27, 634–638. DOI: 10.1093/jamia/ocz226.
  • Rubin, D. B. (2007), “The Design versus the Analysis of Observational Studies for Causal Effects: Parallels with the Design of Randomized Trials,” Statistics in Medicine, 26, 20–36. DOI: 10.1002/sim.2739.
  • Rubin, D. B. (2008), “For Objective Causal Inference, Design Trumps Analysis,” The Annals of Applied Statistics, 2, 808–840. DOI: 10.1214/08-AOAS187.
  • Seifu, Y., Gamalo-Siebers, M., Barthel, F. M.-S., Lin, J., Qiu, J., Cooner, F., Ruan, S., and Walley, R. (2020), “Real-World Evidence Utilization in Clinical Development Reflected by US Product Labeling: Statistical Review,” Therapeutic Innovation & Regulatory Science, 54, 1436–1443.
  • Stewart, M., Norden, A. D., Dreyer, N., Henk, H. J., Abernethy, A. P., Chrischilles, E., Kushi, L., Mansfield, A. S., Khozin, S., Sharon, E., Arunajadai, S., Carnahan, R., Christian, J. B., Miksad, R. A., Sakoda, L. C., Torres, A. Z., Valice, E., and Allen, J. (2019), “An Exploratory Analysis of Real-World End Points for Assessing Outcomes Among Immunotherapy-Treated Patients With Advanced Non–Small-Cell Lung Cancer,” JCO Clinical Cancer Informatics, 1–15. DOI: 10.1200/CCI.18.00155.
  • US Congress (2016), “21st Century Cures Act. H.R. 34, 114th Congress,” available at https://www.gpo.gov/fdsys/pkg/BILLS-114hr34enr/pdf/BILLS-114hr34enr.pdf.
  • Wang, S. V., Schneeweiss, S., Berger, M. L., Brown, J., de Vries, F., Douglas, I., Gagne, J. J., Gini, R., Klungel, O., and Mullins, C. D. (2017), “Reporting to Improve Reproducibility and Facilitate Validity Assessment for Healthcare Database Studies V1. 0,” Value in Health, 20, 1009–1022.
  • Westreich, D. (2012), “Berkson’s Bias, Selection Bias, and Missing Data,” Epidemiology, 23, 159. DOI: 10.1097/EDE.0b013e31823b6296.
  • Yue, L. Q., Lu, N., and Xu, Y. (2014), “Designing Premarket Observational Comparative Studies Using Existing Data as Controls: Challenges and Opportunities,” Journal of Biopharmaceutical Statistics, 24, 994– 1010. DOI: 10.1080/10543406.2014.926367.
  • Zamakhshary, M. F., Hassanain, M., Farhat, G. N., Higashi, M., Kruzikas, D., Basulaiman, M., Saeedi, M. Y., and Tahan, F. (2014), “Using Innovative Modeling Analytics with Real World Data to Develop a National Breast Cancer Screening Program in the Kingdom of Saudi Arabia,” Value in Health, 17, A656. DOI: 10.1016/j.jval.2014.08.2395.
  • Zhang, X., Faries, D. E., Li, H., Stamey, J. D., and Imbens, G. W. (2018), “Addressing Unmeasured Confounding in Comparative Observational Research,” Pharmacoepidemiology and Drug Safety, 27, 373–382. DOI: 10.1002/pds.4394.

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.