Randomized Controlled Trials Versus Real World Evidence: Neither Magic Nor Myth

Hans-Georg Eichler, Francesco Pignatti, Brigitte Schwarzer-Daum, Ana Hidalgo-Simon, Irmgard Eichler, Peter Arlett, Anthony Humphreys, Spiros Vamvakas, Nikolai Brun, Guido Rasi, Hans-Georg Eichler, Francesco Pignatti, Brigitte Schwarzer-Daum, Ana Hidalgo-Simon, Irmgard Eichler, Peter Arlett, Anthony Humphreys, Spiros Vamvakas, Nikolai Brun, Guido Rasi

Abstract

Compared with drugs from the blockbuster era, recently authorized drugs and those expected in the future present a heterogenous mix of chemicals, biologicals, and cell and gene therapies, a sizable fraction being for rare diseases, and even individualized treatments or individualized combinations. The shift in the nature of products entails secular trends for the definitions of "drugs" and "target population" and for clinical use and evidence generation. We discuss that the lessons learned from evidence generation for 20th century medicines may have limited relevance for 21st century medicines. We explain why the future is not about randomized controlled trials (RCTs) vs. real-world evidence (RWE) but RCTs and RWE-not just for the assessment of safety but also of effectiveness. Finally, we highlight that, in the era of precision medicine, we may not be able to reliably describe some small treatment effects-either by way of RCTs or RWE.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2020 The Authors. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
The complex matrix of research questions and methods. The graphic conceptualizes the complexities of research questions associated with a hypothetical drug treatment intended for a disease condition caused by different mutations in individual patients. Complexity is defined along three axes: the x‐axis depicts breadth of information (i.e., different patient subgroups, based on mutation, phenotype, or disease stage), the z‐axis depicts depth of information (i.e., different types of efficacy or safety end points of interest), and the y‐axis depicts context of information (i.e., different comparators or treatment combinations). Each cell in the three‐dimensional matrix represents an item of information that may be relevant for a particular decision maker and/or patient subgroup. Different study types (symbolized by different colors) will be required to generate the information, given the appropriateness of methods for different research questions as well as practical constraints on evidence generation. Note that for some research questions, there will be no data and information available at all, at least at the time of market launch. See main text for real‐life examples that fit the schematic. ECA, external control arm; RCT, randomized controlled trial.

References

    1. Collins, R. , Bowman, L. , Landray, M. & Peta, R. The magic of randomization versus the myth of real‐world evidence. N. Engl. J. Med. 382, 674–678 (2020).
    1. Trusheim, M.R. , Berndt, E.R. & Douglas, F.L. Stratified medicine: strategic and economic implications of combining drugs and clinical biomarkers. Nat. Rev. Drug Discov. 6, 287–293 (2007).
    1. Alliance for Regenerative Medicines website: ARM Annual Report & Sector Year in Review <> (2019).
    1. Statement from FDA Commissioner Scott Gottlieb, M.D. and Peter Marks, M.D., Ph.D., Director of the Center for Biologics Evaluation and Research on new policies to advance development of safe and effective cell and gene therapies <> (2019).
    1. Chasman, D.I. , Posada, D. , Subrahmanyan, L. , Cook, N.R. , Stanton, V.P. Jr & Ridker, P.M. Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA 291, 2821–2827 (2004).
    1. Kitzmiller, J.P. , Mikulik, E.B. , Dauki, A.M. , Murkherjee, C. & Luzum, J.A. Pharmacogenomics of statins: understanding susceptibility to adverse effects. Pharmgenomics Pers. Med. 9, 97–106 (2016).
    1. Tambuyzer, E. et al. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat. Rev. Drug Discov. 19, 93–111 (2020).
    1. EMA website: Kymriah EPAR – Public Assessment Report <>.
    1. EMA website: Yescarta EPAR – Public Assessment Report <>.
    1. Neelapu, S.S. CAR‐T efficacy: is conditioning the key? Blood 133, 1799–1800 (2019).
    1. Majzner, R.G. & Mackall, C.L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 25, 1341–1355 (2019).
    1. Kim, J. et al. Patient‐customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381, 1644–1652 (2019).
    1. Woodcock, J. & Marks, P. Drug regulation in the era of individualized therapies. N. Engl. J. Med. 381, 1678–1680 (2019).
    1. EMA webstite: Zynteglo EPAR – Public Assessment Report <>.
    1. Eichler, H.G. et al. Data rich, information poor: can we use electronic health records to create a learning healthcare system for pharmaceuticals? Clin. Pharmacol. Ther. 105, 912–922 (2019).
    1. EMA website: Qualification opinion on Cellular therapy module of the European Society for Blood & Marrow Transplantation (EBMT) Registry <>.
    1. European Cystic Fibrosis Society Patient Registry website <>.
    1. Cystic Fibrosis Foundation website: Annual report 2018. <>.
    1. Eichler, H.G. et al. "Threshold‐crossing": a useful way to establish the counterfactual in clinical trials? Clin. Pharmacol. Ther. 100, 699–712 (2016).
    1. Mulberg, A.E. et al. Regulatory strategies for rare diseases under current global regulatory statutes: a discussion with stakeholders. Orphanet. J. Rare Dis. 14, 36 (2019).
    1. Carrigan, G. et al. Using electronic health records to derive control arms for early phase single‐arm lung cancer trials: proof‐of‐concept in randomized controlled trials. Clin. Pharmacol. Ther. 107, 369–377 (2020).
    1. Wainwright, C.E. et al. Lumacaftor‐ivacaftor in patients with cystic fibrosis homozygous for Phe508del CFTR. N. Engl. J. Med. 373, 220–231 (2015).
    1. FDA website. Highlights of prescribing information <>.
    1. Carpten, J.D. et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 448, 439–444 (2007).
    1. Bleeker, F.E. et al. AKT1(E17K) in human solid tumours. Oncogene 27, 5648–5650 (2008).
    1. Hyman, D.M. , Taylor, B.S. & Baselga, J. Implementing genome‐driven oncology. Cell 168, 584–599 (2017).
    1. Flynn, R. et al. Ability of primary care health databases to assess medicinal products discussed by the European Union Pharmacovigilance Risk Assessment Committee. Clin. Pharmacol. Ther. 107, 957–965 (2020).
    1. Tervonen, T. , Angelis, A. , Hockley, K. , Pignatti, F. & Phillips, L.D. Quantifying preferences in drug benefit‐risk decisions. Clin. Pharmacol. Ther. 106, 955–959 (2019).

Source: PubMed

3
Subscribe