A Review of Causal Inference for External Comparator Arm Studies.

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • Additional Information
    • Source:
      Publisher: Adis, Springer International Country of Publication: New Zealand NLM ID: 9002928 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1179-1942 (Electronic) Linking ISSN: 01145916 NLM ISO Abbreviation: Drug Saf Subsets: MEDLINE
    • Publication Information:
      Publication: Auckland : Adis, Springer International
      Original Publication: [Mairangi Bay, Auckland, N.Z. : ADIS Press Limited, c1990-
    • Subject Terms:
    • Abstract:
      Randomized controlled trials (RCTs) are the gold standard design to establish the efficacy of new drugs and to support regulatory decision making. However, a marked increase in the submission of single-arm trials (SATs) has been observed in recent years, especially in the field of oncology due to the trend towards precision medicine contributing to the rise of new therapeutic interventions for rare diseases. SATs lack results for control patients, and information from external sources can be compiled to provide context for better interpretability of study results. External comparator arm (ECA) studies are defined as a clinical trial (most commonly a SAT) and an ECA of a comparable cohort of patients-commonly derived from real-world settings including registries, natural history studies, or medical records of routine care. This publication aims to provide a methodological overview, to sketch emergent best practice recommendations and to identify future methodological research topics. Specifically, existing scientific and regulatory guidance for ECA studies is reviewed and appropriate causal inference methods are discussed. Further topics include sample size considerations, use of estimands, handling of different data sources regarding differential baseline covariate definitions, differential endpoint measurements and timings. In addition, unique features of ECA studies are highlighted, specifically the opportunity to address bias caused by unmeasured ECA covariates, which are available in the SAT.
      (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
    • References:
      Simon R, Blumenthal GM, Rothenberg ML, et al. The role of nonrandomized trials in the evaluation of oncology drugs. Clin Pharmacol Ther. 2015;97:502–7. https://doi.org/10.1002/cpt.86 . (PMID: 10.1002/cpt.8625676488)
      Arone B. The argument for external comparator adoption. Pharm Exerc. 2019;39(6):30–1.
      Love-Koh J, Peel A, Rejon-Parrilla JC, et al. The future of precision medicine: potential impacts for health technology assessment. Pharmacoeconomics. 2018;36:1439–51. https://doi.org/10.1007/s40273-018-0686-6 . (PMID: 10.1007/s40273-018-0686-6300034356244622)
      Gray CM, Grimson F, Layton D et al. A framework for methodological choice and evidence assessment for studies using external comparators from real-world data. Drug Saf. 2020;43:623–33. https://doi.org/10.1007/s40264-020-00944-1 . (PMID: 10.1007/s40264-020-00944-1324408477305259)
      Dreyer NA. Advancing a framework for regulatory use of real-world evidence: when real is reliable. Ther Innov Regul Sci. 2018;52(3):362–8. https://doi.org/10.1177/2168479018763591 . (PMID: 10.1177/2168479018763591297145755944086)
      Mack C, Pavesio A, Kelly K et al. Making the most of external comparators. A study of fracture healing in patients at risk of nonunion. Poster presented at: International Society of Pharmacoeconomics & Outcomes Research, 2017.
      Burcu M, Dreyer NA, Franklin JM, et al. Real-world evidence to support regulatory decision-making for medicines: considerations for external control arms. Pharmacoepidemiol Drug Saf. 2020;29:1228–35. https://doi.org/10.1002/pds.4975 . (PMID: 10.1002/pds.4975321623817687199)
      Berry DA, Elashoff M, Blotner S, et al. Creating a synthetic control arm from previous clinical trials: application to establishing early end points as indicators of overall survival in acute myeloid leukemia (AML). J Clin Oncol. 2017;25(suppl 15):7021–7021. https://doi.org/10.1200/JCO.2017.35.15_suppl.7021 . (PMID: 10.1200/JCO.2017.35.15_suppl.7021)
      Dron L, Golchi S, Hsu G, et al. Minimizing control group allocation in randomized trials using dynamic borrowing of external control data—an application to second line therapy for non-small cell lung cancer. Contemp Clin Trials Comm. 2019;16:1–6. https://doi.org/10.1016/j.conctc.2019.100446 . (PMID: 10.1016/j.conctc.2019.100446)
      YODA data portal. http://yoda.yale.edu/ . Accessed 25 Mar 2022.
      CSDR data portal. https://www.clinicalstudydatarequest.com/ . Accessed 25 Mar 2022.
      PDS data portal. https://projectdatasphere.org/projectdatasphere/html/home . Accessed 25 Mar 2022.
      Vivli data portal. https://vivli.org . Accessed 25 Mar 2022.
      SOAR data portal. https://dcri.org/about/who-we-are/our-approach/data-sharing/soar-data/ . Accessed 25 Mar 2022.
      TransCelerate data portal. https://www.transceleratebiopharmainc.com/initiatives/historical-trial-data-sharing/ . Accessed 25 Mar 2022.
      HOVON data portal. https://hovon.nl/en . Accessed 25 Mar 2022.
      Ventz S, Lai A, Chloughesy TF, et al. Design and evaluation of an external control arm using prior clinical trials and real-world data. Clin Cancer Res. 2019;25(16):4993–5001. https://doi.org/10.1158/1078-0432.CCR-19-0820 . (PMID: 10.1158/1078-0432.CCR-19-0820311750986697596)
      Largent JA and Velentgas P. External comparators supporting regulatory submissions 2017–2019. Pharmacoepidemiol Drug Saf. 2020;29(suppl 3):Abstract 4848.
      Mishra-Kalyani PS, Kordestani LA, Rivera DR, Singh H, Ibrahim A, DeClaro RA, Shen Y, Tang S, Sridhara R, Kluetz PG, Concato J, Pazdur R, Beaver JA. External control arms in oncology: current use and future directions. Ann Oncol. 2022;33(4):376–83. https://doi.org/10.1016/j.annonc.2021.12.015 . (PMID: 10.1016/j.annonc.2021.12.01535026413)
      Goring S, Taylor A, Müller K, et al. Characteristics of non-randomised studies using comparisons with external controls submitted for regulatory approval in the USA and Europe: a systematic review. BMJ Open. 2019;9: e024895. https://doi.org/10.1136/bmjopen-2018-024895 . (PMID: 10.1136/bmjopen-2018-024895308197086398650)
      FDA. FDA Briefing Document, NDA 212306, Selinexor, 2019. https://www.fda.gov/media/121667/download . Accessed 25 Mar 2022.
      FDA. FDA Briefing Document, NDA 211723, Tazemetostat, 2019. https://www.fda.gov/media/133573/download . Accessed 25 Mar 2022.
      Center for Drug Evaluation and Research. Application number: 212725Orig1s000, 212726Orig1s000, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/212725Orig1s000,%20212726Orig1s000OtherR.pdf . Accessed 25 Mar 2022.
      Center for Drug Evaluation and Research. Application number: 212018Orig1s000, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/212018Orig1s000OtherR.pdf . Accessed 25 Mar 2022.
      European Medicines Agency. Assessment report Yescarta, 2018. https://www.ema.europa.eu/en/documents/assessment-report/yescarta-epar-public-assessment-report_en.pdf . Accessed 25 Mar 2022.
      Amgen. 07 March 2018 ODAC Meeting briefing document blinatumomab (BLINCYTO®), 2018. https://www.fda.gov/media/111622/download . Accessed 25 Mar 2022.
      Jahanshahi M, Gregg K, Davis G, Ndu A, Miller V, Vockley J, Ollivier C, Franolic T, Sakai S. The use of external controls in FDA regulatory decision making. Ther Innov Regul Sci. 2021;55:1019–35. https://doi.org/10.1007/s43441-021-00302-y . (PMID: 10.1007/s43441-021-00302-y340144398332598)
      Hatswell AJ, Thompson GJ, Maroudas PA, et al. Estimating outcomes and cost effectiveness using a single-arm clinical trial: ofatumumab for double-refractory chronic lymphocytic leukemia. Cost Eff Resour Alloc. 2017;15:8. https://doi.org/10.1186/s12962-017-0071-x . (PMID: 10.1186/s12962-017-0071-x285597465446681)
      Hatswell AJ, Sullivan WG. Creating historical controls using data from a previous line of treatment—two non-standard approaches. Stat Methods Med Res. 2020;29(6):1563–72. https://doi.org/10.1177/0962280219826609 . (PMID: 10.1177/096228021982660930698076)
      Strayhorn JM. Virtual controls as an alternative to randomized controlled trials for assessing efficacy of interventions. Med Res Methodol. 2021;21(3):1–14. https://doi.org/10.1186/s12874-020-01191-9 . (PMID: 10.1186/s12874-020-01191-9)
      Friends of Cancer Research, Beckers F, Capra W, Cassidy A et al. Characterizing the use of external controls for augmenting randomized control arms and confirming benefit. White Paper, 2019. https://www.focr.org/sites/default/files/Panel-1_External_Control_Arms2019AM.pdf . Accessed 25 Mar 2022.
      Schmidli H, Häring DA, Thomas M, et al. Beyond randomized clinical trials: use of external controls. Clin Pharmacol Ther. 2019;107(4):806–16. https://doi.org/10.1002/cpt.1723 . (PMID: 10.1002/cpt.172331725899)
      Mack C, Christian JC, Brinkley E, et al. When context is hard to come by: external comparators and how to use them. Ther Innov Regul Sci. 2020;54(4):932–8. https://doi.org/10.1007/s43441-019-00108-z . (PMID: 10.1007/s43441-019-00108-z32557316)
      Thorlund K, Dron L, Park JJH, et al. Synthetic and external controls in clinical trials—a primer for researchers. Clin Epidemiol. 2020;12:457–67. https://doi.org/10.2147/CLEP.S242097 . (PMID: 10.2147/CLEP.S242097324402247218288)
      Lodi S, Phillips A, Lundgren J, et al. Effect estimates in randomized trials and observational studies: comparing apples with apples. Am J Epidemiol. 2019;188:1569–77. https://doi.org/10.1093/aje/kwz100 . (PMID: 10.1093/aje/kwz100310631926670045)
      Ghadessi M, Tang R, Zhou J, et al. A roadmap to using historical controls in clinical trials—by Drug Information Association Adaptive Design Scientific Working Group (DIA-ADSWG). Orphanet J Rare Dis. 2020;15(1):1–19. https://doi.org/10.1186/s13023-020-1332-x . (PMID: 10.1186/s13023-020-1332-x)
      Burger HU, Gerlinger C, Harbron C, et al. The use of external controls: to what extent can it currently be recommended? Pharm Stat. 2021:1–15. https://doi.org/10.1002/pst.2120 .
      Seeger JD, Davis KJ, Innacone MR, et al. Methods for external control groups for single arm trials or long-term uncontrolled extensions to randomized clinical trials. Pharmacoepidemiol Drug Saf. 2020;29:1382–92. https://doi.org/10.1002/pds.5141 . (PMID: 10.1002/pds.5141329645147756307)
      European Medicines Agency, Committee for Medicinal Products for Human Use. Guideline on clinical trials in small populations. CHMP/EWP/83561/2005, 2006. https://www.ema.europa.eu/en/clinical-trials-small-populations . Accessed 25 Mar 2022.
      European Medicines Agency, International Council on Harmonization (ICH). ICH Topic E10: choice of control groups in clinical trials. CPMP/ICH/364/96, 2001. https://www.ema.europa.eu/en/documents/scientific-guideline/ich-e-10-choice-control-group-clinical-trials-step-5_en.pdf . Accessed 25 Mar 2022.
      European Network of Centres for Pharmacoepidemiology and Pharmacovigilance (ENCePP): ENCePP checklist for study protocols, 2010. https://www.encepp.eu/standards_and_guidances/checkListProtocols.shtml . Accessed 25 Mar 2022.
      European Network of Centres for Pharmacoepidemiology and Pharmacovigilance (ENCePP): ENCePP guide on methodological standards in pharmacoepidemiology, 2020. http://www.encepp.eu/standards_and_guidances/methodologicalGuide.shtml . Accessed 25 Mar 2022.
      U.S. Food and Drug Administration. Framework for FDA's real-world evidence, 2018. https://www.fda.gov/media/120060/download . Accessed 25 Mar 2022.
      Gottlieb S. Statement from FDA Commissioner Scott Gottlieb, M.D., on FDA’s new strategic framework to advance use of real-world evidence to support development of drugs and biologics. FDA press release, 2018. https://www.fda.gov/news-events/press-announcements/statement-fda-commissioner-scott-gottlieb-md-fdas-new-strategic-framework-advance-use-real-world . Accessed 25 Mar 2022.
      Khozin S, Blumenthal GM, Pazdur R. Real-world data for clinical evidence generation in oncology. JNCI J Natl Cancer Inst. 2017;109(11):1–5. https://doi.org/10.1093/jnci/djx187 . (PMID: 10.1093/jnci/djx187)
      FDA. Real-World Data: Assessing electronic health records and medical claims data to support regulatory decision-making for drug and biological products. Draft Guidance for Industry, 2021. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/real-world-data-assessing-electronic-health-records-and-medical-claims-data-support-regulatory . Accessed 25 Mar 2022.
      Skovlund E, Leufkens HGM, Smyth JF. The use of real-world data in cancer drug development. Eur J Cancer. 2018;101:69–76. https://doi.org/10.1016/j.ejca.2018.06.036 . (PMID: 10.1016/j.ejca.2018.06.03630031168)
      Cave A, Kurz X, Arlett P. Real-World data for regulatory decision making: challenges and possible solutions for Europe. Clin Pharmacol Ther. 2019;106(1):36–8. https://doi.org/10.1002/cpt.1426 . (PMID: 10.1002/cpt.1426309701616617710)
      Eichler HG, Koenig F, Arlett P, et al. Are novel, nonrandomized analytic methods fit for decision making? The need for prospective, controlled, and transparent validation. Clin Pharmacol Ther. 2020;107(4):773–9. https://doi.org/10.1002/cpt.1638 . (PMID: 10.1002/cpt.163831574163)
      Baumfeld Andre E, Reynolds R, Caubel P, et al. Trial designs using real-world data: the changing landscape of the regulatory approval process. Pharmacoepidemiol Drug Saf. 2020;29(10):1201–12. https://doi.org/10.1002/pds.4932 . (PMID: 10.1002/pds.493231823482)
      Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG). Extract of rapid report A19-43: Concepts for the generation of routine practice data and their analysis for the benefit assessment of drugs according to §35a social code book v (sgb v), 2020. https://www.iqwig.de/download/a19-43_routine-practice-data-for-the-benefit-assessment-of-drugs_extract-of-rapid-report_v1-0.pdf?rev=184599 . accessed 25 Mar 2022.
      Faria R, Hernandez Alava M, Manca A et al. NICE DSU technical support document 17: the use of observational data to inform estimates of treatment effectiveness in technology appraisal: methods for comparative individual patient data, 2015. http://www.nicedsu.org.uk . Accessed 25 Mar 2022.
      Bell Gorrod H, Wailoo AJ, Hernandez M et al. The use of real world data for the estimation of treatment effects in NICE decision-making, 2016. Report by the Decision Support Unit. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2016. https://www.sheffield.ac.uk/sites/default/files/2022-02/RWD-DSU-REPORT-Updated-DECEMBER-2016.pdf . Accessed 25 Mar 2022.
      Makady A, Ham RT, de Boer A, et al. Policies for use of real-world data in Health Technology Assessment (HTA): A comparative study of six HTA agencies. Value in Health. 2017;20(4):520–32. https://doi.org/10.1016/j.jval.2016.12.003 . (PMID: 10.1016/j.jval.2016.12.00328407993)
      EUnetHTA. Comparators and comparisons: direct and indirect comparisons, 2015. https://eunethta.eu/wp-content/uploads/2018/01/Comparators-Comparisons-Direct-and-indirect-comparisons_Amended-JA1-Guideline_Final-Nov-2015.pdf . Accessed 25 Mar 2022.
      Gatto NM, Reynolds RF, Campbell UB. A structured preapproval and postapproval comparative study design framework to generate valid and transparent real-world evidence for regulatory decisions. Clin Pharmacol Ther. 2019;106:103–15. https://doi.org/10.1002/cpt.1480 . (PMID: 10.1002/cpt.1480310253116771466)
      Girman CJ, Ritchey ME, Zhou W, et al. Considerations in characterizing real-world data relevance and quality for regulatory purposes: a commentary. Pharmacoepidemiol Drug Saf. 2019;28:439–42. https://doi.org/10.1002/pds.4697 . (PMID: 10.1002/pds.469730515910)
      Miksad RA, Abernethy AP. Harnessing the power of real-world evidence (RWE): a checklist to ensure regulatory-grade data quality. Clin Pharmacol Ther. 2018;103:202–5. https://doi.org/10.1002/cpt.946 . (PMID: 10.1002/cpt.94629214638)
      von Elm E, Altman DG, Egger M, et al. The STrengthening the Reporting of OBservational studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Ann Intern Med. 2007;147(8):594–6. https://doi.org/10.7326/0003-4819-147-8-200710160-00010 . (PMID: 10.7326/0003-4819-147-8-200710160-00010)
      Dreyer NA, Bryant A, Velentgas P. The GRACE Checklist: A validated assessment tool for high quality observational studies of comparative effectiveness. J Manag Care Spec Pharm. 2016;22(10):1107–13. https://doi.org/10.18553/jmcp.2016.22.10.1107 .
      International Epidemiological Association (IEA). Good Epidemiological Practice (GEP) guidelines issued by the International Epidemiological Association (IEA), 2010. https://www.ieaweb.org/IEAWeb/Content/IEA_Publications.aspx . Accessed 25 Mar 2022.
      International Society for Pharmacoepidemiology (ISPE). Guidelines for Good Pharmacoepidemiology Practices (GPP) issued by the International Society for Pharmacoepidemiology (ISPE), 2019. https://www.pharmacoepi.org/resources/policies/guidelines-08027/ . Accessed 25 Mar 2022.
      Motheral B, Brooks J, Clark MA, et al. A checklist for retrospective database studies—report of the ISPOR Task Force on Retrospective Databases. Value Health. 2003;6(2):90–7. https://doi.org/10.1046/j.1524-4733.2003.00242.x . (PMID: 10.1046/j.1524-4733.2003.00242.x12641858)
      Berger ML, Sox H, Willke RJ, et al. 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. Pharmacoepidemiol Drug Saf. 2017;26:1033–9. https://doi.org/10.1002/pds.4297 . (PMID: 10.1002/pds.4297289139665639372)
      Faries DE, Zhang X, Zbigniew K, et al. Real world health care data analysis SAS. Cary: SAS Institute; 2020.
      Hernán MA, Robins JM. Causal inference: what if. Boca Raton: Chapman & Hall/CRC; 2020.
      Zhu H, Zhang S, Ahn C. Sample size considerations for historical control studies with survival outcomes. Biopharm Stat. 2016;26(4):657–71. https://doi.org/10.1080/10543406.2015.1052495 . (PMID: 10.1080/10543406.2015.1052495)
      Zhang S, Cao J, Ahn C. Sample size calculation for studies comparing binary outcomes using historical controls. Biom J. 2013;55:190–202. https://doi.org/10.1002/bimj.201200100 . (PMID: 10.1002/bimj.20120010023335222)
      Zhang S, Cao J, Ahn C. Calculating sample size in trials using historical controls. Clin Trials. 2010;7(4):343–53. https://doi.org/10.1177/1740774510373629 . (PMID: 10.1177/1740774510373629205736383085081)
      Han G. Designing historical control studies with survival endpoints using exact statistical inference. Pharm Stat. 2021;20(1):4–14. https://doi.org/10.1002/pst.2050 . (PMID: 10.1002/pst.205032743949)
      Chow S-C, Jun S, Wang H, Lokhnygina Y. Sample size calculations in clinical research. 3rd ed. Boca-Raton: CRC Press; 2017. (PMID: 10.1201/9781315183084)
      McCaffrey DF, Griffin BA, Almirall D, et al. A tutorial on propensity score estimation for multiple treatments using generalized boosted models. Stat Med. 2013;32(19):3388–414. https://doi.org/10.1002/sim.5753 . (PMID: 10.1002/sim.5753235086733710547)
      Austin PC. Statistical criteria for selecting the optimal number of untreated subjects matched to each treated subject when using many-to-one matching on the propensity score. Am J Epidemiol. 2010;172(9):1092–7. https://doi.org/10.1093/aje/kwq224 . (PMID: 10.1093/aje/kwq224208022412962254)
      ICH E9(R1) Expert Working Group. ICH E9(R1) addendum on estimands and sensitivity analysis in clinical trials to the guideline on statistical principles for clinical trials. EMA/CHMP/ICH/436221/2017, 2020. https://www.ema.europa.eu/en/documents/scientific-guideline/ich-e9-r1-addendum-estimands-sensitivity-analysis-clinical-trials-guideline-statistical-principles_en.pdf . Accessed 25 Mar 2022.
      Rufibach K. Treatment effect quantification for time-to-event endpoints—estimands, analysis strategies, and beyond. Pharm Stat. 2019;18(2):145–65. https://doi.org/10.1002/pst.1917 . (PMID: 10.1002/pst.191730478869)
      Sun S, Weber HJ, Butler E, et al. Estimands in hematologic oncology trials. Pharm Stat. 2021;20(4):793–805. https://doi.org/10.1002/pst.2108 . (PMID: 10.1002/pst.210833686762)
      Guo S, Fraser MW. Propensity score analysis. 2nd ed. Thousand Oaks: Sage Publications; 2015.
      Li F, Morgan KL, Zaslavsky AM. Balancing covariates via propensity score weighting. J Am Stat Assoc. 2018;113(521):390–400. https://doi.org/10.1080/01621459.2016.1260466 . (PMID: 10.1080/01621459.2016.1260466)
      Li F, Thomas LE, Li F. Addressing extreme propensity scores via the overlap weights. Am J Epidemiol. 2019;188(1):250–7. https://doi.org/10.1093/aje/kwy201 . (PMID: 10.1093/aje/kwy20130189042)
      Li F. Introducing the overlap weights in causal inference, 2018. https://stat.duke.edu/~fl35/OW/JASA_talk.pdf . Accessed 25 Mar 2022.
      Thomas LE, Li F, Pencina M. Overlap weighting: a propensity score method that mimics attributes of a randomized clinical trial. J Am Med Assoc. 2020;323(23):2417–8. https://doi.org/10.1001/jama.2020.7819 . (PMID: 10.1001/jama.2020.7819)
      Li H, Wang C, Chen WC, Lu N, Song C, Tiwari R, Xu Y, Yue LQ. Estimands in observational studies: Some considerations beyond ICH E9 (R1). Pharm Stat. 2022;1–10. https://doi.org/10.1002/pst.2196 .
      Aalen OO, Cook RJ, Roysland K. Does Cox analysis of a randomized survival study yield a causal treatment effect? Lifetime Data Anal. 2015;21(4):579–93. https://doi.org/10.1007/s10985-015-9335-y . (PMID: 10.1007/s10985-015-9335-y26100005)
      Mao H, Li L, Yang W, Shen Y. On the propensity score weighting analysis with survival outcome: estimands, estimation, and inference. Stat Med. 2018;37(26):3745–63. https://doi.org/10.1002/sim.7839 . (PMID: 10.1002/sim.783929855060)
      Royston P, Parmar MKB. Restricted mean survival time: an alternative to the hazard ratio for the design and analysis of randomized trials with a time-to-event outcome. BMC Med Res Methodol. 2013;13(152):1–15. https://doi.org/10.1186/1471-2288-13-152 . (PMID: 10.1186/1471-2288-13-152)
      Chen P, Tsiatis A. Causal inference on the difference of the restricted mean lifetime between two groups. Biometrics. 2001;57(4):1030–8. https://doi.org/10.1111/j.0006-341X.2001.01030.x .
      Andersen PK, Hansen MG, Klein JP. Regression analysis of restricted mean survival time based on pseudo-observations. Lifetime Data Anal. 2004;10:335–50. https://doi.org/10.1007/s10985-004-4771-0 . (PMID: 10.1007/s10985-004-4771-015690989)
      Tian L, Zhao L, Wei LJ. Predicting the restricted mean event time with the subject’s baseline covariates in survival analysis. Biostatistics. 2014;15(2):222–33. https://doi.org/10.1093/biostatistics/kxt050 .
      Rubin DB. Discussion of “Randomization analysis of experimental data in the Fisher randomization test” by D. Basu. J Am Stat Assoc. 1980;75:591–3.
      Holland PW. Statistics and causal inference. J Am Stat Assoc. 1986;81(396):945–60. https://doi.org/10.1080/01621459.1986.10478354 . (PMID: 10.1080/01621459.1986.10478354)
      Xu R, O’Quigley J. Estimating average regression effect under non-proportional hazards. Biostatistics. 2000;1:423–39. https://doi.org/10.1093/biostatistics/1.4.423 . (PMID: 10.1093/biostatistics/1.4.42312933565)
      Boyd AP, Kittelson JM, Gillen DL. Estimation of treatment effect under non-proportional hazards and conditionally independent censoring. Stat Med. 2012;31:3504–15. https://doi.org/10.1002/sim.5440 . (PMID: 10.1002/sim.544022763957)
      European Medicines Agency. Questions and answer on adjustment for cross-over in estimating effects in oncology trials, 2018. https://www.ema.europa.eu/en/adjustment-cross-over-estimating-effects-oncology-trials . Accessed 25 Mar 2022.
      Parra CO, Daniel RM, Bartlett JW. Hypothetical estimands in clinical trials: a unification of causal inference and missing data methods. https://arXiv.org/abs/2107.04392 . Accessed 25 Mar 2022.
      Bornkamp B, Rufibach K, Lin J, et al. Principal stratum strategy: potential role in drug development. Pharm Stat. 2021;20(4):737–51. https://doi.org/10.1002/pst.2104 . (PMID: 10.1002/pst.210433624407)
      Degtyarev E, Zhang Y, Sen K, et al. Estimands and the patient journey: addressing the right question in oncology clinical trials. JCO Precis Oncol. 2019;3:1–10. https://doi.org/10.1200/PO.18.00381 . (PMID: 10.1200/PO.18.0038135100723)
      Pocock S. The combination of randomized and historical controls in clinical trials. J Chronic Dis. 1976;29:175–88. https://doi.org/10.1016/0021-9681(76)90044-8 . (PMID: 10.1016/0021-9681(76)90044-8770493)
      Hatswell AJ, Freemantle N, Baio G, et al. Summarising salient information on historical controls: a structured assessment of validity and comparability across studies. Clin Trials. 2020;17(6):607–16. https://doi.org/10.1177/1740774520944855 . (PMID: 10.1177/1740774520944855329578047649932)
      Abrahami D, Pradhan R, Yin H, et al. Use of real-world data to emulate a clinical trial and support regulatory decision making: assessing the impact of temporality, comparator choice, and method of adjustment. Clin Pharmacol Ther. 2021;109(2):452–61. https://doi.org/10.1002/cpt.2012 . (PMID: 10.1002/cpt.201232767673)
      Suissa S. Single-arm trials with historical controls. Study designs to avoid time-related biases. Epidemiol. 2021;32(1):94–100. https://doi.org/10.1097/EDE.0000000000001267 .
      Zhang S, Liang F, Li W, et al. Comparison of eligibility criteria between protocols, registries, and publications of cancer clinical trials. JNCI J Natl Cancer Inst. 2016;108(11):1–3. https://doi.org/10.1093/jnci/djw129 . (PMID: 10.1093/jnci/djw129)
      Schwartz LH, Litière S, de Vries E, et al. RECIST 1.1—update and clarification: from the RECIST committee. Eur J Cancer 2016;62:132–37. https://doi.org/10.1016/j.ejca.2016.03.081 .
      Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32:3059. https://doi.org/10.1200/JCO.2013.54.8800 . (PMID: 10.1200/JCO.2013.54.8800251137534979083)
      Griffith SD, Tucker M, Bowser B, et al. Generate real-world tumor burden endpoints from electronic health record data: comparison of RECIST, radiology-anchored approaches for abstracting real-world progression in non-small cell lung cancer. Adv Ther. 2019;39:2122–36. https://doi.org/10.1007/s12325-019-00970-1 . (PMID: 10.1007/s12325-019-00970-1)
      Stewart M, Norden AD, Dreyer N, et al. An exploratory analysis of real-world endpoints for assessing outcomes among immunotherapy-treated patients with advanced non-small-cell lung cancer. JCO Clin Cancer Informa. 2019;3:1–15. https://doi.org/10.1200/CCI.18.00155 . (PMID: 10.1200/CCI.18.00155)
      Davies J, Martinec M, Delmar P, et al. Comparative effectiveness from a single-arm trial and real-world data: alectinib versus ceritinib. J Comp Eff Res. 2018;7(9):855–65. https://doi.org/10.2217/cer-2018-0032 . (PMID: 10.2217/cer-2018-003229944008)
      Carrigan G, Whipple S, Capra WB, 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. 2019;107(2):369–77. https://doi.org/10.1002/cpt.1586 . (PMID: 10.1002/cpt.1586313508537006884)
      Yin X, Mishra-Kalyan PS, Sridharab R, Stewart MD, Stuart EA, Davi RC. Exploring the potential of external control arms created from patient level data: a case study in non-small cell lung cancer. J Biopharm Stat. 2022. https://doi.org/10.1080/10543406.2021.2011901 . (PMID: 10.1080/10543406.2021.201190134986069)
      Franklin JM, Glynn RJ, Martin D, et al. Evaluating the use of nonrandomized real-world data analyses for regulatory decision making. Clin Pharmacol Ther. 2019;105(4):867–77. https://doi.org/10.1002/cpt.1351 . (PMID: 10.1002/cpt.135130636285)
      Franklin JM, Pawar A, Martin D, et al. Nonrandomized real-world evidence to support regulatory decision making: process for a randomized trial replication project. Clin Pharmacol Ther. 2020;107(4):817–26. https://doi.org/10.1002/cpt.1633 . (PMID: 10.1002/cpt.163331541454)
      Carrigan G, Whipple S, Taylor MD, et al. An evaluation of the impact of missing deaths on overall survival analyses of advanced non-small cell lung cancer patients conducted in an electronic health records database. Pharmacoepidemiol Drug Saf. 2019;28:572–81. https://doi.org/10.1002/pds.4758 . (PMID: 10.1002/pds.4758308737296594237)
      Rubin DB. For objective causal inference, design trumps analysis. Ann Appl Stat. 2008;2(3):808–40. https://doi.org/10.1214/08-AOAS187 . (PMID: 10.1214/08-AOAS187)
      Imbens GW, Rubin DW. Causal inference for statistics, social and biomedical sciences: an introduction. Cambridge: Cambridge University Press; 2015. (PMID: 10.1017/CBO9781139025751)
      Andrillon A, Pirrachio R, Chevret S. Performance of propensity score matching to estimate causal effects in small samples. Stat Methods Med Res. 2020;29(3):644–58. https://doi.org/10.1177/0962280219887196 . (PMID: 10.1177/096228021988719632186264)
      Stuart EA. Matching methods for causal inference: a review and a look forward. Stat Sci. 2010;25(1):1–21. https://doi.org/10.1214/09-STS313 . (PMID: 10.1214/09-STS313208718022943670)
      Austin PC, Stuart EA. Optimal full matching for survival outcomes: a method that merits more widespread use. Stat Med. 2015;34(30):3949–67. https://doi.org/10.1002/sim.6602 . (PMID: 10.1002/sim.6602262506114715723)
      Austin PC. The use of propensity score methods with survival or time-to-event outcomes: reporting measures of effect similar to those used in randomized experiments. Stat Med. 2014;33:1242–58. https://doi.org/10.1002/sim.5984 . (PMID: 10.1002/sim.598424122911)
      Elze MC, Gregson J, Baber U, et al. Comparison of propensity score methods and covariate adjustment. J Am Coll Cardiol. 2017;69(3):345–57. https://doi.org/10.1016/j.jacc.2016.10.060 . (PMID: 10.1016/j.jacc.2016.10.06028104076)
      Austin PC, Grootendorst P, Normand SL, et al. Conditioning on the propensity score can result in biased estimation of common measures of treatment effect: a Monte Carlo study. Stat Med. 2007;26(4):754–68. https://doi.org/10.1002/sim.2618 . (PMID: 10.1002/sim.261816783757)
      Austin PC, Schuster T. The performance of different propensity score methods for estimating absolute effects of treatments on survival outcomes: a simulation study. Stat Methods Med Res. 2016;25(5):2214–37. https://doi.org/10.1177/0962280213519716 . (PMID: 10.1177/096228021351971624463885)
      Austin PC. Variance estimation when using inverse probability of treatment weighting (IPTW) with survival analysis. Stat Med. 2016;35:5642–55. https://doi.org/10.1002/sim.7084 . (PMID: 10.1002/sim.7084275490165157758)
      Hajage D, Chauvet G, Belin L, et al. Closed-form variance estimator for weighted propensity score estimators with survival outcome. Biom J. 2018;60(6):1151–63. https://doi.org/10.1002/bimj.201700330 . (PMID: 10.1002/bimj.20170033030257058)
      Austin PC, Cafri G. Variance estimation when using propensity-score matching with replacement with survival or time-to-event outcomes. Stat Med. 2020;39:1623–40. https://doi.org/10.1002/sim.8502 . (PMID: 10.1002/sim.8502321093197217182)
      Shu D, Young JG, Toh S, et al. Variance estimation in inverse probability weighted Cox models. Biometrics. 2021;77(3):1101–17. https://doi.org/10.1111/biom.13332 . (PMID: 10.1111/biom.1333232662087)
      Austin PC, Stuart EA. Moving towards best practice when using inverse probability of treatment weighting (IPTW) using the propensity score to estimate causal treatment effects in observational studies. Stat Med. 2015;34(28):3661–79. https://doi.org/10.1002/sim.6607 . (PMID: 10.1002/sim.6607262389584626409)
      Brookhart MA, Schneeweiss S, Rothman KJ. Variable selection for propensity score models. Am J Epidemiol. 2006;163(12):1149–56. https://doi.org/10.1093/aje/kwj149 . (PMID: 10.1093/aje/kwj14916624967)
      Austin PC. Editorial: advances in propensity score analysis. Stat Methods Med Res. 2020;29(3):641–64. https://doi.org/10.1177/0962280219899248 . (PMID: 10.1177/096228021989924832186265)
      Leite W. Practical propensity score methods using R. Thousand Oaks: Sage Publications; 2017. (PMID: 10.4135/9781071802854)
      Leyrat C, Seaman SR, White IR, et al. Propensity score analysis with partially observed covariates: How should multiple imputation be used? Stat Methods Med Res. 2017;28(1):3–19. https://doi.org/10.1177/0962280217713032 . (PMID: 10.1177/0962280217713032285739196313366)
      Mitra R, Reiter JP. A comparison of two methods of estimating propensity scores after multiple imputation. Stat Methods Med Res. 2012;25(1):188–204. https://doi.org/10.1177/0962280212445945 . (PMID: 10.1177/096228021244594522687877)
      Rassen JA, Glynn RJ, Rothman KJ, et al. Applying propensity scores estimated in a full cohort to adjust for confounding in subgroup analyses. Pharmacoepidemiol Drug Saf. 2012;21(7):697–709. https://doi.org/10.1002/pds.2256 . (PMID: 10.1002/pds.225622162077)
      Marcus SM, Gibbons RD. Caution should be used in applying propensity scores estimated in a full cohort to adjust for confounding in subgroup analyses: commentary on “Applying propensity scores estimated in a full cohort to adjust for confounding in subgroup analyses.” Pharmacoepidemiol Drug Saf. 2012;21:710–2. https://doi.org/10.1002/pds.3202 . (PMID: 10.1002/pds.320222645065)
      Girman CJ, Gokhale M, Kou TG, et al. Assessing the impact of propensity score estimation and implementation on covariate balance and confounding control within and across important subgroups in comparative effectiveness research. Med Care. 2014;52(3):280–7. https://doi.org/10.1097/MLR.0000000000000064 . (PMID: 10.1097/MLR.0000000000000064243744224042911)
      Wang SV, Jin Y, Fireman B, et al. Relative performance of propensity score matching strategies for subgroup analyses. Am J Epidemiol. 2018;187:1799–807. https://doi.org/10.1093/aje/kwy049 . (PMID: 10.1093/aje/kwy04929554199)
      Liu S, Liu C, Nehus E, et al. Propensity score analysis for correlated subgroup effects. Stat Methods Med Res. 2019;29(4):1067–80. https://doi.org/10.1177/0962280219850595 . (PMID: 10.1177/096228021985059531144601)
      Dong J, Zhang JL, Li F. Subgroup balancing propensity score. Stat Methods Med Res. 2020;29(3):659–76. https://doi.org/10.1177/0962280219870836 . (PMID: 10.1177/096228021987083631456486)
      Imai K, Ratkovic M. Covariate balancing propensity score. J R Stat Soc B. 2014;76(1):243–63. https://doi.org/10.1111/rssb.12027 . (PMID: 10.1111/rssb.12027)
      Lunceford J, Davidian M. Stratification and weighting via the propensity score in estimation of causal treatment effect: a comparative study. Stat Med. 2004;23:2937–60. https://doi.org/10.1002/sim.1903 . (PMID: 10.1002/sim.190315351954)
      Wooldridge JM. Econometric analysis of cross section and panel data. 2nd ed. Cambridge: MT Press; 2010.
      Barlev A, Brookhart MA, Xun P, Thirumalai D, Sadetsky N, Suissa S. Comparison of estimation methods for single-arm trials in rare diseases with historical control groups. ISPOR conference, 2021. https://www.ispor.org/docs/default-source/euro2021/ispor-20eu-20podium-20presentationmethods-20comparative-20analysis2021.pdf?sfvrsn=20df850d_0 . Accessed 25 Mar 2022.
      King G, Nielsen R. Why propensity scores should not be used for matching. Polit Anal. 2019;27(4):1–20. https://doi.org/10.1017/pan.2019.11 . (PMID: 10.1017/pan.2019.11)
      Diamond A, Sekhon JS. Genetic matching for estimating causal effects: a general multivariate method for achieving balance in observational studies. Rev Econ Stat. 2013;95(3):932–45. https://doi.org/10.1162/REST_a_00318 . (PMID: 10.1162/REST_a_00318)
      Rubin DB. Bias reduction using Mahalanobis-metric matching. Biometrics. 1980;36:293–8. https://doi.org/10.2307/2529981 . (PMID: 10.2307/2529981)
      Iacus SM, King G, Porro G. Causal inference without balance checking: coarsened exact matching. Polit Anal. 2012;20:1–24. https://doi.org/10.1093/pan/mpr013 . (PMID: 10.1093/pan/mpr013)
      Visconti G, Zubizarreta JR. Handling limited overlap in observational studies with cardinality matching. Obs Stud. 2018;4:217–49. https://doi.org/10.1353/obs.2018.0012 . (PMID: 10.1353/obs.2018.0012)
      Hainmüller J. Entropy balancing for causal effects: a multivariate reweighting method to produce balanced samples in observational studies. Polit Anal. 2012;20(1):25–46. https://doi.org/10.2139/ssrn.1904869 . (PMID: 10.2139/ssrn.1904869)
      Zubizarreta JR. Stable weights that balance covariates for estimation with incomplete outcome data. J Am Stat Assess. 2015;110:910–22. https://doi.org/10.1080/01621459.2015.1023805 . (PMID: 10.1080/01621459.2015.1023805)
      Wang Y, Zubizarreta JR. Minimal dispersion approximately balancing weights: asymptotic properties and practical considerations. Biometrika. 2017;103(1):1–29. https://doi.org/10.1093/biomet/asz050 . (PMID: 10.1093/biomet/asz050)
      Yiu S, Su L. Covariate association eliminating weights: a unified weighting framework for causal effect estimation. Biometrika. 2018;105(3):709–22. https://doi.org/10.1093/biomet/asy015 . (PMID: 10.1093/biomet/asy01531031408)
      Ning Y, Peng S, Imai K. Robust estimation of causal effects via high-dimensional covariate balancing propensity score. Biometrika. 2020;107(3):533–54. https://doi.org/10.1093/biomet/asaa020 . (PMID: 10.1093/biomet/asaa020)
      Huling JD, Mak S. Energy balancing of covariate distributions. 2020. https://arxiv.org/abs/2004.13962 . Accessed 25 Mar 2022.
      Chan KCG, Yam SCP, Zhang Z. Globally efficient non-parametric inference of average treatment effects by empirical balancing calibration weighting. J R Stat Soc B Stat Methodol. 2016;78(3):673–700. https://doi.org/10.1111/rssb.12129 . (PMID: 10.1111/rssb.12129)
      Austin PC. An introduction to propensity score methods for reducing the effects of confounding in observational studies. Multivar Behav Res. 2011;46(3):399–424. https://doi.org/10.1080/00273171.2011.568786 . (PMID: 10.1080/00273171.2011.568786)
      Samuel M, Schuster T, Kaufman JS, et al. Differences between conditional and marginal propensity score estimates. J Am Coll Cardiol. 2017;70(1):117. https://doi.org/10.1016/j.jacc.2017.03.610 . (PMID: 10.1016/j.jacc.2017.03.61028662799)
      Andersen PK, Syriopoulou E, Parner ET. Causal inference in survival analysis using pseudo-observations. Stat Med. 2017;36(17):2669–81. https://doi.org/10.1002/sim.7297 . (PMID: 10.1002/sim.729728384840)
      Pearl J. The seven tools of causal inference, with reflections on machine learning. Commun ACM. 2019;62:54–60. https://doi.org/10.1145/3241036 . (PMID: 10.1145/3241036)
      Baiocchi M, Cheng J, Small D. Instrumental variable methods for causal inference. Stat Med. 2014;33(13):2297–340. https://doi.org/10.1002/sim.6128 . (PMID: 10.1002/sim.6128245998894201653)
      Lash TL, Fox MP, MacLehose RF, et al. Good practices for quantitative bias analysis. Int J Epidemiol. 2014;43(6):1969–85. https://doi.org/10.1093/ije/dyu149 . (PMID: 10.1093/ije/dyu14925080530)
      Lash TL, Fox MP, Cooney D, et al. Quantitative bias analysis in regulatory settings. Am J Public Health. 2016;106:1227–30. https://doi.org/10.2105/AJPH.2016.303199 . (PMID: 10.2105/AJPH.2016.303199271966524984770)
      Arah OA. Bias analysis for uncontrolled confounding in the health sciences. Annu Rev Public Health. 2017;38:23–38. https://doi.org/10.1146/annurev-publhealth-032315-021644 . (PMID: 10.1146/annurev-publhealth-032315-02164428125388)
      VanderWheele TJ, Ding P. Sensitivity analysis in observational research: introducing the E-value. Ann Intern Med. 2017;167(4):268–74. https://doi.org/10.7326/M16-2607 . (PMID: 10.7326/M16-2607)
      Molenbergh G, Kenward MG. Missing data in clinical studies. Hoboken: Wiley; 2007. (PMID: 10.1002/9780470510445)
      Carpenter JR, Kenward MG. Multiple imputation and its application. Hoboken: Wiley; 2013. (PMID: 10.1002/9781119942283)
      Enders CK. Applied missing data analysis. New York: Guilford Press; 2010.
      O’Kelly M, Ratitch B. Clinical trials with missing data. Hoboken: Wiley; 2014. (PMID: 10.1002/9781118762516)
      Lee CH, Wang H. Multiple imputation confidence intervals for the mean of the discrete distributions for incomplete data. Stat Med. 2022;14(7):1172–90. https://doi.org/10.1002/sim.9254 . (PMID: 10.1002/sim.9254)
      Moscovici JL, Ratitch B. Combining survival analysis results after multiple imputation of censored event times. Proceedings of PharmaSUG, 2017. https://www.lexjansen.com/pharmasug/2017/SP/PharmaSUG-2017-SP05.pdf . Accessed 25 Mar 2022.
      Eekhout I, van de Wiel MA, Heymans MW. Methods for significance testing of categorical covariates in logistic regression models after multiple imputation: power and applicability analysis. BMC Med Res Methodol. 2017;17:129. https://doi.org/10.1186/s12874-017-0404-7 . (PMID: 10.1186/s12874-017-0404-7288304665568368)
      Madley-Dowd P, Hughes R, Tilling K, Heron J. The proportion of missing data should not be used to guide decisions on multiple imputation. J Clin Epidemiol. 2019;110:63–73. https://doi.org/10.1016/j.jclinepi.2019.02.016 . (PMID: 10.1016/j.jclinepi.2019.02.016308786396547017)
      Hernán MA, Robins JM. Using big data to emulate a target trial when a randomized trial is not available. Pract Epidemiolog. 2016;183(8):758–64. https://doi.org/10.1093/aje/kwv254 . (PMID: 10.1093/aje/kwv254)
      Adams DC, Gurevitch J, Rosenberg MS. Resampling tests for meta-analysis of ecological data. Ecol. 1997;78(5):1277–83. https://doi.org/10.1890/0012-9658(1997)078%5B1277:RTFMAO%5D2.0.CO;2 . (PMID: 10.1890/0012-9658(1997)078%5B1277:RTFMAO%5D2.0.CO;2)
      Van den Noortgate W, Onghena P. Parametric and nonparametric bootstrap methods for meta-analysis. Behav Res Methods. 2005;37(1):11–22. https://doi.org/10.3758/BF03206394 . (PMID: 10.3758/BF03206394)
      Tanner-Smith EE, Tipton E, Polanin JR. Handling complex meta-analytic data structures using robust variance estimates: a tutorial in R. J Dev Life Course Criminol. 2016;2:85–112. https://doi.org/10.1007/s40865-016-0026-5 . (PMID: 10.1007/s40865-016-0026-5)
      Cheung MW. A guide to conducting a meta-analysis with non-independent effect sizes. Neuropsychol Rev. 2019;29:387–96. https://doi.org/10.1007/s11065-019-09415-6 . (PMID: 10.1007/s11065-019-09415-6314465476892772)
      Salles G, Schuster SJ, Dreyling M et al. Efficacy comparison of tisagenlecleucel versus usual care in patients with relapsed or refractory follicular lymphoma. Poster presented at the American Society of Hematology Annual Meeting & Exposition, 11-14 December 2021. https://www.medicalcongress.novartisoncology.com/ASHQR/CART/pdf/Salles_Poster_3528.pdf . Accessed 25 Mar 2022.
      Food and Drug Administration. Guidance for industry: enrichment strategies for clinical trials to support approval of human drugs and biological products, 2012. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enrichment-strategies-clinical-trials-support-approval-human-drugs-and-biological-products . Accessed 25 Mar 2022.
      European Medicines Agency. Guideline on the investigation of subgroups in confirmatory clinical trials, EMA/CHMP/539146/2013, 2019.  https://www.ema.europa.eu/en/investigation-subgroups-confirmatory-clinical-trials . Accessed 25 Mar 2022.
      Lipkovich I, Dmitrienko A, D’Agostino RB. Tutorial in biostatistics: data-driven subgroup identification and analysis in clinical trials. Stat Med. 2016;36(1):136–96. https://doi.org/10.1002/sim.7064 . (PMID: 10.1002/sim.706427488683)
      Ballarini NM, Rosenkranz GK, Jaki T et al. Subgroup identification in clinical trials via the predicted individual treatment effect. 2018. PLoS ONE. 13(10):e0205971. https://doi.org/10.1371/journal.pone.0205971 .
      Behrendt CE, Gehan EA. Treatment-subgroup interaction: an example from a published, phase II clinical trial. Contemp Clin Trials. 2009;30(3):279–81. https://doi.org/10.1016/j.cct.2009.02.002 . (PMID: 10.1016/j.cct.2009.02.002192325492732105)
      Peng Y, Yu B. Cure models. Boca Raton: Chapman & Hall/CRC; 2020.
      Lim J, Walley R, Yuan J, et al. Minimizing patient burden through the use of historical subject-level data in innovative confirmatory clinical trials: review of methods and opportunities. Ther Innov Regul Sci. 2018;52(5):546–59. https://doi.org/10.1177/2168479018778282 . (PMID: 10.1177/216847901877828229909645)
      Collignon O, Schritz A, Spezia R, et al. Implementing historical controls in oncology trials. The Oncol. 2021;26(5):859–62. https://doi.org/10.1002/onco.13696 . (PMID: 10.1002/onco.13696)
      van Rosmalen J, Dejardin D, van Norden Y, et al. Including historical data in the analysis of clinical trials: Is it worth the effort? Stat Methods Med Res. 2018;27(10):3167–82. https://doi.org/10.1177/0962280217694506 . (PMID: 10.1177/096228021769450628322129)
      Collins R, Bowman L, Landray M, et al. The magic of randomization versus the myth of real-world evidence. New Engl J Med. 2020;382(7):674–8. https://doi.org/10.1056/NEJMsb1901642 . (PMID: 10.1056/NEJMsb190164232053307)
      Eichler HG, Bloechl-Daum B, Bauer P, et al. “Threshold-crossing”: a useful way to establish the counterfactual in clinical trials? Clin Pharmacol & Ther. 2016;100(6):699–712. https://doi.org/10.1002/cpt.515 . (PMID: 10.1002/cpt.515)
      Jarow JP. Use of external controls in regulatory decision-making. Clin Pharmacol Ther. 2017;101(5):595–6. https://doi.org/10.1002/cpt.652 . (PMID: 10.1002/cpt.65228182270)
      Grayling MJ, Mander AP. Do single-arm trials have a role in drug development plans incorporating randomised trials? Pharm Stat. 2016;15:143–51. https://doi.org/10.1002/pst.1726 . (PMID: 10.1002/pst.172626609689)
      Lasch F, Weber K, Chao MM, et al. A plea to provide best evidence in trials under sample-size restrictions: the example of pioglitazone to resolve leukoplakia and erythroplakia in Fanconi anemia patients. Orphanet J Rare Dis. 2017;12(102):1–6. https://doi.org/10.1186/s13023-017-0655-8 . (PMID: 10.1186/s13023-017-0655-8)
      Jemielita T, Tse A, Chen C. Oncology phase II proof-of-concept studies with multiple targets: randomized controlled trial or single arm? Pharm Stat. 2020;19(2):117–25. https://doi.org/10.1002/pst.1972 . (PMID: 10.1002/pst.197231424631)
    • Publication Date:
      Date Created: 20220727 Date Completed: 20220810 Latest Revision: 20221012
    • Publication Date:
      20240104
    • Accession Number:
      10.1007/s40264-022-01206-y
    • Accession Number:
      35895225