Références

     

[1] Carpenter DP. Reputation and power: Organizational image and pharmaceutical regulation at the FDA. Princeton (N.J.), Oxford: Princeton University Press 2010 ISBN:9780691141794;

[2] Hwang TJ, Carpenter D, Lauffenburger JC, et al. Failure of Investigational Drugs in Late-Stage Clinical Development and Publication of Trial Results. JAMA Intern Med 2016;176:1826–33 10.1001/jamainternmed.2016.6008 [27723879]

[3] Prasad V, Cifu A, Ioannidis JPA. Reversals of established medical practices: evidence to abandon ship. JAMA 2012;307:37–38 10.1001/jama.2011.1960 [22215160]

[4] Prasad V, Gall V, Cifu A. The frequency of medical reversal. Arch Intern Med 2011;171:1675–76 10.1001/archinternmed.2011.295 [21747003]

[5] Sutton D, Qureshi R, Martin J. Evidence reversal-when new evidence contradicts current claims: a systematic overview review of definitions and terms. J Clin Epidemiol 2018;94:76–84 10.1016/j.jclinepi.2017.10.004

[6] Tajika A, Ogawa Y, Takeshima N, et al. Replication and contradiction of highly cited research papers in psychiatry: 10-year follow-up. Br J Psychiatry 2015;207:357–62 10.1192/bjp.bp.113.143701 [26159600]

[7] Tatsioni A, Bonitsis NG, Ioannidis JPA. Persistence of contradicted claims in the literature. JAMA 2007;298:2517–26 10.1001/jama.298.21.2517 [18056905]

[8] Zarin DA, Goodman SN, Kimmelman J. Harms From Uninformative Clinical Trials. JAMA 2019;322:813–14 10.1001/jama.2019.9892 [31343666]

[9] The Centre for Evidence-Based Medicine. The ethics of COVID-19 treatment studies: too many are open, too few are double-masked - The Centre for Evidence-Based Medicine 2020. Available at: https://www.cebm.net/covid-19/the-ethics-of-covid-19-treatment-studies-too-many-are-open-too-few-are-double-masked/ Accessed August 23, 2021.

[10] Halpern SD, Karlawish JHT, Berlin JA. The continuing unethical conduct of underpowered clinical trials. JAMA 2002;288:358–62 10.1001/jama.288.3.358 [12117401]

[11] Williamson PR, Altman DG, Bagley H, et al. The COMET Handbook: version 1.0. Trials 2017;18:280 10.1186/s13063-017-1978-4 [28681707]

[12] Kozauer N, Katz R. Regulatory innovation and drug development for early-stage Alzheimer's disease. N Engl J Med 2013;368:1169–71 10.1056/NEJMp1302513 [23484795]

[13] CardioBrief: FDA's Gottlieb Preparing To Lower The Bar To Approval 2017. Available at: https://www.medpagetoday.com/cardiology/cardiobrief/68224 Accessed November 15, 2021.

[14] Nikolaidis GF, Woods B, Palmer S, et al. Classifying information-sharing methods. BMC Med Res Methodol 2021;21:107 10.1186/s12874-021-01292-z [34022810]

[15] Higgins JP, Whitehead A. Borrowing strength from external trials in a meta-analysis. Stat Med 1996;15:2733–49 10.1002/(SICI)1097-0258(19961230)15:24<2733:AID-SIM562>3.0.CO 2-0; [8981683]

[16] Woodcock J, LaVange LM. Master Protocols to Study Multiple Therapies, Multiple Diseases, or Both. N Engl J Med 2017;377:62–70 10.1056/NEJMra1510062 [28679092]

[17] Gyawali B, Rome BN, Kesselheim AS. Regulatory and clinical consequences of negative confirmatory trials of accelerated approval cancer drugs: retrospective observational study. BMJ 2021;374:n1959 10.1136/bmj.n1959 [34497044]

[18] Hilal T, Gonzalez-Velez M, Prasad V. Limitations in Clinical Trials Leading to Anticancer Drug Approvals by the US Food and Drug Administration. JAMA Internal Medicine 2020;180:1108–15 10.1001/jamainternmed.2020.2250 [32539071]

[19] Hilal T, Sonbol MB, Prasad V. Analysis of Control Arm Quality in Randomized Clinical Trials Leading to Anticancer Drug Approval by the US Food and Drug Administration. JAMA Oncol 2019;5:887–92 10.1001/jamaoncol.2019.0167 [31046071]

[20] Naci H, Davis C, Savović J, et al. Design characteristics, risk of bias, and reporting of randomised controlled trials supporting approvals of cancer drugs by European Medicines Agency, 2014-16: cross sectional analysis. BMJ 2019;366:l5221 10.1136/bmj.l5221 [31533922]

[21] Ladanie A, Schmitt AM, Speich B, et al. Clinical Trial Evidence Supporting US Food and Drug Administration Approval of Novel Cancer Therapies Between 2000 and 2016. JAMA Netw Open 2020;3:e2024406 10.1001/jamanetworkopen.2020.24406 [33170262]

[22] Kim C, Prasad V. Cancer Drugs Approved on the Basis of a Surrogate End Point and Subsequent Overall Survival: An Analysis of 5 Years of US Food and Drug Administration Approvals. JAMA Internal Medicine 2015;175:1992–94 10.1001/jamainternmed.2015.5868 [26502403]

[23] Gyawali B, Hey SP, Kesselheim AS. Assessment of the Clinical Benefit of Cancer Drugs Receiving Accelerated Approval. JAMA Internal Medicine 2019;179:906–13 10.1001/jamainternmed.2019.0462 [31135808]

[24] BEECHER HK. Surgery as placebo. A quantitative study of bias. JAMA 1961;176:1102–07 10.1001/jama.1961.63040260007008 [13688614]

[25] Cohen PJ. Failure to conduct a placebo-controlled trial may be unethical. Am J Bioeth 2002;2:24 10.1162/152651602317533604 [12189067]

[26] Heckerling PS. The Ethics of Single Blind Trials. IRB: Ethics and Human Research 2005;27:12 10.2307/3563956

[27] Zarin DA, Goodman SN, Kimmelman J. Harms From Uninformative Clinical Trials. JAMA 2019;322:813–14 10.1001/jama.2019.9892 [31343666]

[28] Hwang TJ, Ross JS, Vokinger KN, et al. Association between FDA and EMA expedited approval programs and therapeutic value of new medicines: retrospective cohort study. BMJ 2020;371:m3434 10.1136/bmj.m3434 [33028575]

[29] Schnog J-JB, Samson MJ, Gans ROB, et al. An urgent call to raise the bar in oncology. Br J Cancer 2021 10.1038/s41416-021-01495-7 [34400802]

[30] Tannock IF, Amir E, Booth CM, et al. Relevance of randomised controlled trials in oncology. The Lancet Oncology 2016;17:e560-e567 10.1016/S1470-2045(16)30572-1 [27924754]

[31] Tannock IF, Templeton AJ. Flawed trials for cancer. Annals of Oncology 2020;31:331–33 10.1016/j.annonc.2019.11.017 [32067676]

[32] Cohen D. Cancer drugs: high price, uncertain value. BMJ 2017;359:j4543 10.1136/bmj.j4543 [28978508]

[33] Sachs RE, Gavulic KA, Donohue JM, et al. Recent Trends in Medicaid Spending and Use of Drugs With US Food and Drug Administration Accelerated Approval. JAMA Health Forum 2021;2:e213177 10.1001/jamahealthforum.2021.3177

[34] Hernán MA. Methods of Public Health Research - Strengthening Causal Inference from Observational Data. The New England journal of medicine 2021 10.1056/NEJMp2113319 [34596980]

[35] Park K. The use of real-world data in drug repurposing. Transl Clin Pharmacol 2021;29:117–24 10.12793/tcp.2021.29.e18 [34621704]

[36] Hatswell AJ, Baio G, Berlin JA, et al. Regulatory approval of pharmaceuticals without a randomised controlled study: analysis of EMA and FDA approvals 1999-2014. BMJ open 2016;6:e011666 10.1136/bmjopen-2016-011666 [27363818]

[37] Mahase E. FDA allows drugs without proven clinical benefit to languish for years on accelerated pathway. BMJ 2021;374:n1898 10.1136/bmj.n1898 [34326042]

[38] Naci H, Smalley KR, Kesselheim AS. Characteristics of Preapproval and Postapproval Studies for Drugs Granted Accelerated Approval by the US Food and Drug Administration. JAMA 2017;318:626–36 10.1001/jama.2017.9415 [28810023]

[39] Boyle JM, Hegarty G, Frampton C, et al. Real-world outcomes associated with new cancer medicines approved by the Food and Drug Administration and European Medicines Agency: A retrospective cohort study. Eur J Cancer 2021;155:136–44 10.1016/j.ejca.2021.07.001 [34371443]

[40] Song F, Zang C, Ma X, et al. The use of real-world data/evidence in regulatory submissions. Contemporary Clinical Trials 2021;109:106521 10.1016/j.cct.2021.106521 [34339865]

[41] Soto-Becerra P, Culquichicón C, Hurtado-Roca Y, et al. Real-world effectiveness of hydroxychloroquine, azithromycin, and ivermectin among hospitalized COVID-19 patients: results of a target trial emulation using observational data from a nationwide healthcare system in Peru 2020.

[42] Greenland S, Pearl J, Robins JM. Causal diagrams for epidemiologic research. Epidemiology 1999;10:37–48 ; [9888278]

[43] Sterne JAC, Hernán MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016;355:i4919 10.1136/bmj.i4919

[44] Schünemann HJ, Cuello C, Akl EA, et al. GRADE Guidelines: 18. How ROBINS-I and other tools to assess risk of bias in non-randomized studies should be used to rate the certainty of a body of evidence. J Clin Epidemiol 2018 10.1016/j.jclinepi.2018.01.012

[45] Hernán MA, Alonso A, Logan R, et al. Observational studies analyzed like randomized experiments: an application to postmenopausal hormone therapy and coronary heart disease. Epidemiology 2008;19:766–79 10.1097/EDE.0b013e3181875e61 [18854702]

[46] Lash TL, VanderWeele TJ, Haneuse S, et al. Modern epidemiology. Philadelphia etc.: Wolters Kluwer 2021 ISBN:1451193289;

[47] Schuemie MJ, Ryan PB, Pratt N, et al. Principles of Large-scale Evidence Generation and Evaluation across a Network of Databases (LEGEND). J Am Med Inform Assoc 2020;27:1331–37 10.1093/jamia/ocaa103 [32909033]

[48] Bruns SB, Ioannidis JPA. p-Curve and p-Hacking in Observational Research. PLoS ONE 2016;11:e0149144 10.1371/journal.pone.0149144 [26886098]

[49] Patel CJ, Burford B, Ioannidis JPA. Assessment of vibration of effects due to model specification can demonstrate the instability of observational associations. Journal of Clinical Epidemiology 2015;68:1046–58 10.1016/j.jclinepi.2015.05.029 [26279400]

[50] Head ML, Holman L, Lanfear R, et al. The extent and consequences of p-hacking in science. PLoS Biology 2015;13:e1002106 10.1371/journal.pbio.1002106 [25768323]

[51] Silberzahn R, Uhlmann EL, Martin DP, et al. Many analysts, one dataset: Making transparent how variations in analytical choices affect results 2017.

[52] Chuard PJC, Vrtílek M, Head ML, et al. Evidence that nonsignificant results are sometimes preferred: Reverse P-hacking or selective reporting? PLoS Biol 2019;17:e3000127 10.1371/journal.pbio.3000127 [30682013]

[53] Michels KB, Rosner BA. Data trawling: to fish or not to fish. The Lancet 1996;348:1152–53 10.1016/S0140-6736(96)05418-9

[54] Data dredging - Wikipedia 2021. Available at: https://en.wikipedia.org/wiki/Data_dredging Accessed August 30, 2021.

[55] 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. Value Health 2017;20:1003–08 10.1016/j.jval.2017.08.3019 [28964430]

[56] Orsini LS, Monz B, Mullins CD, et al. Improving transparency to build trust in real-world secondary data studies for hypothesis testing-Why, what, and how: recommendations and a road map from the real-world evidence transparency initiative. Pharmacoepidemiol Drug Saf 2020;29:1504–13 10.1002/pds.5079 [32924243]

[57] Langan SM, Schmidt SA, Wing K, et al. The reporting of studies conducted using observational routinely collected health data statement for pharmacoepidemiology (RECORD-PE). BMJ 2018;363:k3532 10.1136/bmj.k3532 [30429167]

[58] Hernán MA, Sauer BC, Hernández-Díaz S, et al. Specifying a target trial prevents immortal time bias and other self-inflicted injuries in observational analyses. Journal of Clinical Epidemiology 2016;79:70–75 10.1016/j.jclinepi.2016.04.014 [27237061]

[59] Rothman KJ, Greenland S. Causation and causal inference in epidemiology. Am J Public Health 2005;95 Suppl 1:S144-50 10.2105/AJPH.2004.059204 [16030331]

[60] Pearl J. An introduction to causal inference. The International Journal of Biostatistics 2010;6:Article 7 10.2202/1557-4679.1203 [20305706]

[61] Hernán MA, Robins JM. Estimating causal effects from epidemiological data. J Epidemiol Community Health 2006;60:578–86 10.1136/jech.2004.029496 [16790829]

[62] Hernán MA. A definition of causal effect for epidemiological research. J Epidemiol Community Health 2004;58:265–71 10.1136/jech.2002.006361 [15026432]

[63] Belas N. P-hacking in Clinical Trials: A Meta-Analytical Approach ;

[64] Hripcsak G, Suchard MA, Shea S, et al. Comparison of Cardiovascular and Safety Outcomes of Chlorthalidone vs Hydrochlorothiazide to Treat Hypertension. JAMA Internal Medicine 2020 10.1001/jamainternmed.2019.7454 [32065600]

[65] Nyström T, Bodegard J, Nathanson D, et al. Second line initiation of insulin compared with DPP-4 inhibitors after metformin monotherapy is associated with increased risk of all-cause mortality, cardiovascular events, and severe hypoglycemia. Diabetes Research and Clinical Practice 2017;123:199–208 10.1016/j.diabres.2016.12.004 [28056431]

[66] Gerstein HC, Bosch J, Dagenais GR, et al. Basal insulin and cardiovascular and other outcomes in dysglycemia. N Engl J Med 2012;367:319–28 10.1056/NEJMoa1203858 [22686416]

[67] Concato J, Shah N, Horwitz RI. Randomized, controlled trials, observational studies, and the hierarchy of research designs. The New England journal of medicine 2000;342:1887–92 10.1056/nejm200006223422507

[68] Ioannidis JP, Haidich AB, Pappa M, et al. Comparison of evidence of treatment effects in randomized and nonrandomized studies. JAMA 2001;286:821–30 ;

[69] Benson K, Hartz AJ. A comparison of observational studies and randomized, controlled trials. New Engl J Med 2000;342:1878–86 10.1056/NEJM200006223422506 [10861324]

[70] Banerjee R, Prasad V. Are Observational, Real-World Studies Suitable to Make Cancer Treatment Recommendations? JAMA Netw Open 2020;3:e2012119 10.1001/jamanetworkopen.2020.12119 [32729916]

[71] Concato J. Observational versus experimental studies: what's the evidence for a hierarchy? NeuroRx the journal of the American Society for Experimental NeuroTherapeutics 2004;1:341–47 10.1602/neurorx.1.3.341

[72] Dahabreh IJ, Kent DM. Can the Learning Health Care System Be Educated With Observational Data? JAMA 2014;312:129–30 10.1001/jama.2014.4364

[73] Gerstein HC, McMurray J, Holman RR. Real-world studies no substitute for RCTs in establishing efficacy. The Lancet 2019;393:210–11 10.1016/s0140-6736(18)32840-x

[74] Kumar A, Guss ZD, Courtney PT, et al. Evaluation of the Use of Cancer Registry Data for Comparative Effectiveness Research. JAMA Netw Open 2020;3:e2011985 10.1001/jamanetworkopen.2020.11985 [32729921]

[75] Naudet F, Maria AS, Falissard B. Antidepressant response in major depressive disorder: a meta-regression comparison of randomized controlled trials and observational studies. PLoS ONE 2011;6:e20811 10.1371/journal.pone.0020811 [21687681]

[76] Oliver S, Bagnall AM, Thomas J, et al. Randomised controlled trials for policy interventions: a review of reviews and meta-regression. Health Technol Assess 2010;14:1-165, iii 10.3310/hta14160 [20338119]

[77] Papanikolaou PN, Christidi GD, Ioannidis JPA. Comparison of evidence on harms of medical interventions in randomized and nonrandomized studies. CMAJ 2006;174:635–41 10.1503/cmaj.050873 [16505459]

[78] Shikata S, Nakayama T, Noguchi Y, et al. Comparison of effects in randomized controlled trials with observational studies in digestive surgery. Ann Surg 2006;244:668–76 10.1097/01.sla.0000225356.04304.bc [17060757]

[79] Golder S, Loke YK, Bland M. Meta-analyses of adverse effects data derived from randomised controlled trials as compared to observational studies: methodological overview. PLOS Medicine 2011;8:e1001026 10.1371/journal.pmed.1001026 [21559325]

[80] Kuss O, Legler T, Börgermann J. Treatments effects from randomized trials and propensity score analyses were similar in similar populations in an example from cardiac surgery. Journal of Clinical Epidemiology 2011;64:1076–84 10.1016/j.jclinepi.2011.01.005 [21482068]

[81] Bhandari M, Tornetta P, Ellis T, et al. Hierarchy of evidence: differences in results between non-randomized studies and randomized trials in patients with femoral neck fractures. Arch Orthop Trauma Surg 2004;124:10–16 10.1007/s00402-003-0559-z [14576955]

[82] Edwards JP, Kelly EJ, Lin Y, et al. Meta-analytic comparison of randomized and nonrandomized studies of breast cancer surgery. Can J Surg 2012;55:155–62 10.1503/cjs.023410 [22449722]

[83] Furlan AD, Tomlinson G, Jadad AAR, et al. Examining heterogeneity in meta-analysis: comparing results of randomized trials and nonrandomized studies of interventions for low back pain. Spine (Phila Pa 1976) 2008;33:339–48 10.1097/BRS.0b013e31816233b5 [18303468]

[84] Müeller D, Sauerland S, Neugebauer EAM, et al. Reported effects in randomized controlled trials were compared with those of nonrandomized trials in cholecystectomy. Journal of Clinical Epidemiology 2010;63:1082–90 10.1016/j.jclinepi.2009.12.009 [20346627]

[85] Tannen RL, Weiner MG, Xie D. Use of primary care electronic medical record database in drug efficacy research on cardiovascular outcomes: comparison of database and randomised controlled trial findings. BMJ 2009;338:b81 10.1136/bmj.b81 [19174434]

[86] Dahabreh IJ, Sheldrick RC, Paulus JK, et al. Do observational studies using propensity score methods agree with randomized trials? A systematic comparison of studies on acute coronary syndromes. Eur. Heart J. 2012;33:1893–901 10.1093/eurheartj/ehs114

[87] Lonjon G, Boutron I, Trinquart L, et al. Comparison of treatment effect estimates from prospective nonrandomized studies with propensity score analysis and randomized controlled trials of surgical procedures. Ann Surg 2014;259:18–25 10.1097/SLA.0000000000000256

[88] Anglemyer A, Horvath HT, Bero L. Healthcare outcomes assessed with observational study designs compared with those assessed in randomized trials. Cochrane Database Syst Rev 2014:MR000034 10.1002/14651858.MR000034.pub2 [24782322]

[89] Califf RM, Hernandez AF, Landray M. Weighing the Benefits and Risks of Proliferating Observational Treatment Assessments: Observational Cacophony, Randomized Harmony. JAMA 2020;324:625–26 10.1001/jama.2020.13319 [32735313]

[90] Rush CJ, Campbell RT, Jhund PS, et al. Association is not causation: treatment effects cannot be estimated from observational data in heart failure. Eur Heart J 2018;39:3417–38 10.1093/eurheartj/ehy407 [30085087]

[91] Soni PD, Hartman HE, Dess RT, et al. Comparison of Population-Based Observational Studies With Randomized Trials in Oncology. JCO 2019;37:1209–16 10.1200/JCO.18.01074 [30897037]

[92] Klassen SA, Senefeld J, Johnson PW, et al. The Effect of Convalescent Plasma Therapy on COVID-19 Patient Mortality: Systematic Review and Meta-analysis. medRxiv 2021 10.1101/2020.07.29.20162917 [33140056]

[93] Janiaud P, Axfors C, Schmitt AM, et al. Association of Convalescent Plasma Treatment With Clinical Outcomes in Patients With COVID-19: A Systematic Review and Meta-analysis. JAMA 2021;325:1185–95 10.1001/jama.2021.2747 [33635310]

[94] Danaei G, Rodríguez LAG, Cantero OF, et al. Observational data for comparative effectiveness research: an emulation of randomised trials of statins and primary prevention of coronary heart disease 2013. Available at: http://journals.sagepub.com.docelec.univ-lyon1.fr/doi/pdf/10.1177/0962280211403603.

[95] Dickerman BA, García-Albéniz X, Logan RW, et al. Avoidable flaws in observational analyses: an application to statins and cancer. Nat Med 2019;25:1601–06 10.1038/s41591-019-0597-x [31591592]

[96] Webster-Clark M, Lund JL, Stürmer T, et al. Reweighting Oranges to Apples: Transported RE-LY Trial Versus Nonexperimental Effect Estimates of Anticoagulation in Atrial Fibrillation. Epidemiology 2020;31:605–13 10.1097/EDE.0000000000001230 [32740469]

[97] Cain LE, Logan R, Robins JM, et al. When to initiate combined antiretroviral therapy to reduce mortality and AIDS-defining illness in HIV-infected persons in developed countries: an observational study. Ann. Intern. Med. 2011;154:509–15 10.7326/0003-4819-154-8-201104190-00001 [21502648]

[98] Franklin JM, Patorno E, Desai RJ, et al. Emulating Randomized Clinical Trials With Nonrandomized Real-World Evidence Studies: First Results From the RCT DUPLICATE Initiative. Circulation 2021;143:1002–13 10.1161/CIRCULATIONAHA.120.051718 [33327727]

[99] Dahabreh IJ, Robins JM, Hernán MA. Benchmarking Observational Methods by Comparing Randomized Trials and Their Emulations. Epidemiology 2020;31:614–19 10.1097/EDE.0000000000001231 [32740470]

[100] European Medicines Agency. Guideline on registry-based studies ;

[101] Mathes T, Pieper D. Study design classification of registry-based studies in systematic reviews. Journal of Clinical Epidemiology 2018;93:84–87 10.1016/j.jclinepi.2017.09.016 [28951107]

[102] Karanatsios B, Prang K-H, Verbunt E, et al. Defining key design elements of registry-based randomised controlled trials: a scoping review. Trials 2020;21:552 10.1186/s13063-020-04459-z [32571382]

[103] Li G, Sajobi TT, Menon BK, et al. Registry-based randomized controlled trials- what are the advantages, challenges, and areas for future research? Journal of Clinical Epidemiology 2016;80:16–24 10.1016/j.jclinepi.2016.08.003 [27555082]

[104] Lauer MS, D'Agostino RB. The randomized registry trial--the next disruptive technology in clinical research? N Engl J Med 2013;369:1579–81 10.1056/NEJMp1310102 [23991657]

[105] Nicholls SG, Carroll K, Hey SP, et al. A review of pragmatic trials found a high degree of diversity in design and scope, deficiencies in reporting and trial registry data, and poor indexing. Journal of Clinical Epidemiology 2021;137:45–57 10.1016/j.jclinepi.2021.03.021 [33789151]

[106] Loudon K, Treweek S, Sullivan F, et al. The PRECIS-2 tool: designing trials that are fit for purpose. BMJ 2015;350:h2147 10.1136/bmj.h2147 [25956159]

[107] Zuidgeest MGP, Goetz I, Groenwold RHH, et al. Series: Pragmatic trials and real world evidence: Paper 1. Introduction. Journal of Clinical Epidemiology 2017;88:7–13 10.1016/j.jclinepi.2016.12.023 [28549929]

[108] Ford I, Norrie J. Pragmatic Trials. N Engl J Med 2016;375:454–63 10.1056/NEJMra1510059 [27518663]

[109] Woodcock J, LaVange LM. Master Protocols to Study Multiple Therapies, Multiple Diseases, or Both. N Engl J Med 2017;377:62–70 10.1056/NEJMra1510062 [28679092]

[110] Berry SM, Connor JT, Lewis RJ. The platform trial: an efficient strategy for evaluating multiple treatments. JAMA 2015;313:1619–20 10.1001/jama.2015.2316 [25799162]

[111] Park JJH, Harari O, Dron L, et al. An overview of platform trials with a checklist for clinical readers. Journal of Clinical Epidemiology 2020;125:1–8 10.1016/j.jclinepi.2020.04.025 [32416336]

[112] Park JJH, Harari O, Dron L, et al. An overview of platform trials with a checklist for clinical readers. J Clin Epidemiol 2020;125:1–8 10.1016/j.jclinepi.2020.04.025

[113] Barker A, Sigman C, Kelloff G, et al. I-SPY 2: An Adaptive Breast Cancer Trial Design in the Setting of Neoadjuvant Chemotherapy. Clin Pharmacol Ther 2009;86:97–100 10.1038/clpt.2009.68

[114] Park JJH, Siden E, Zoratti MJ, et al. Systematic review of basket trials, umbrella trials, and platform trials: a landscape analysis of master protocols. Trials 2019;20:572 10.1186/s13063-019-3664-1 [31533793]

[115] James ND, Sydes MR, Clarke NW, et al. STAMPEDE: Systemic Therapy for Advancing or Metastatic Prostate Cancer — A Multi-Arm Multi-Stage Randomised Controlled Trial. Clinical Oncology 2008;20:577–81 10.1016/j.clon.2008.07.002

[116] LaVange L, Adam SJ, Currier JS, et al. Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV): Designing Master Protocols for Evaluation of Candidate COVID-19 Therapeutics. Ann Intern Med 2021;174:1293–300 10.7326/M21-1269

[117] Angus DC, Berry S, Lewis RJ, et al. The REMAP-CAP (Randomized Embedded Multifactorial Adaptive Platform for Community-acquired Pneumonia) Study. Rationale and Design. Ann Am Thorac Soc 2020;17:879–91 10.1513/AnnalsATS.202003-192SD [32267771]

[118] Pessoa-Amorim G, Campbell M, Fletcher L, et al. Making trials part of good clinical care: lessons from the RECOVERY trial. Future Healthc J 2021;8:e243-e250 10.7861/fhj.2021-0083 [34286192]

[119] Dodd LE, Freidlin B, Korn EL. Platform Trials - Beware the Noncomparable Control Group. N Engl J Med 2021;384:1572–73 10.1056/NEJMc2102446 [33882210]

[120] Park JJH, Harari O, Dron L, et al. An overview of platform trials with a checklist for clinical readers. J Clin Epidemiol 2020;125:1–8 10.1016/j.jclinepi.2020.04.025

[121] Collignon O, Gartner C, Haidich A-B, et al. Current Statistical Considerations and Regulatory Perspectives on the Planning of Confirmatory Basket, Umbrella, and Platform Trials. Clin Pharmacol Ther 2020;107:1059–67 10.1002/cpt.1804

[122] Choodari-Oskooei B, Bratton DJ, Gannon MR, et al. Adding new experimental arms to randomised clinical trials: Impact on error rates. Clin Trials 2020;17:273–84 10.1177/1740774520904346

[123] James ND, Bono JS de, Spears MR, et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N Engl J Med 2017;377:338–51 10.1056/NEJMoa1702900 [28578639]

[124] Sydes MR, Parmar MKB, James ND, et al. Issues in applying multi-arm multi-stage methodology to a clinical trial in prostate cancer: the MRC STAMPEDE trial. Trials 2009;10:39 10.1186/1745-6215-10-39 [19519885]

[125] James ND, Sydes MR, Clarke NW, et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. The Lancet 2016;387:1163–77 10.1016/S0140-6736(15)01037-5

[126] Parmar MKB, Sydes MR, Cafferty FH, et al. Testing many treatments within a single protocol over 10 years at MRC CTU at UCL: Multi-arm, multi stage platform, umbrella and basket protocols. Clin Trials 2017;14:451–61 10.1177/1740774517725697

[127] Normand S-LT. The RECOVERY Platform. New Engl J Med 2021;384:757–58 10.1056/NEJMe2025674

[128] Horby PW, Mafham M, Bell JL, et al. Lopinavir–ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. The Lancet 2020;396:1345–52 10.1016/S0140-6736(20)32013-4

[129] Horby P, Lim WS, Emberson JR, et al. Dexamethasone in Hospitalized Patients with Covid-19 - Preliminary Report. N Engl J Med 2020 10.1056/NEJMoa2021436 [32678530]

[130] Siden EG, Park JJ, Zoratti MJ, et al. Reporting of master protocols towards a standardized approach: A systematic review. Contemp Clin Trials Commun 2019;15:100406 10.1016/j.conctc.2019.100406 [31334382]

[131] Kapur J, Elm J, Chamberlain JM, et al. Randomized Trial of Three Anticonvulsant Medications for Status Epilepticus. N Engl J Med 2019;381:2103–13 10.1056/NEJMoa1905795 [31774955]

[132] Azithromycin for community treatment of suspected COVID-19 in people at increased risk of an adverse clinical course in the UK (PRINCIPLE): a randomised, controlled, open-label, adaptive platform trial. Lancet 2021;397:1063–74 10.1016/S0140-6736(21)00461-X [33676597]

[133] Korley FK, Durkalski-Mauldin V, Yeatts SD, et al. Early Convalescent Plasma for High-Risk Outpatients with Covid-19. N Engl J Med 2021 10.1056/NEJMoa2103784 [34407339]

[134] Kaul S. Is the Mortality Benefit With Empagliflozin in Type 2 Diabetes Mellitus Too Good To Be True? Circulation 2016;134:94–96 10.1161/CIRCULATIONAHA.116.022537 [27400894]

[135] Laptook AR, Shankaran S, Tyson JE, et al. Effect of Therapeutic Hypothermia Initiated After 6 Hours of Age on Death or Disability Among Newborns With Hypoxic-Ischemic Encephalopathy: A Randomized Clinical Trial. JAMA 2017;318:1550–60 10.1001/jama.2017.14972 [29067428]

[136] Reardon MJ, van Mieghem NM, Popma JJ, et al. Surgical or Transcatheter Aortic-Valve Replacement in Intermediate-Risk Patients. N Engl J Med 2017;376:1321–31 10.1056/NEJMoa1700456 [28304219]

[137] Lawler PR, Goligher EC, Berger JS, et al. Therapeutic Anticoagulation with Heparin in Noncritically Ill Patients with Covid-19. N Engl J Med 2021 10.1056/NEJMoa2105911 [34351721]

[138] Effect of anakinra versus usual care in adults in hospital with COVID-19 and mild-to-moderate pneumonia (CORIMUNO-ANA-1): a randomised controlled trial. Lancet Respir Med 2021;9:295–304 10.1016/S2213-2600(20)30556-7 [33493450]

[139] Angus DC, Derde L, Al-Beidh F, et al. Effect of Hydrocortisone on Mortality and Organ Support in Patients With Severe COVID-19: The REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA 2020;324:1317–29 10.1001/jama.2020.17022 [32876697]

[140] Arabi YM, Gordon AC, Derde LPG, et al. Lopinavir-ritonavir and hydroxychloroquine for critically ill patients with COVID-19: REMAP-CAP randomized controlled trial. Intensive Care Med 2021;47:867–86 10.1007/s00134-021-06448-5 [34251506]

[141] Gordon AC, Mouncey PR, Al-Beidh F, et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19. N Engl J Med 2021;384:1491–502 10.1056/NEJMoa2100433 [33631065]

[142] Hermine O, Mariette X, Tharaux P-L, et al. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2021;181:32–40 10.1001/jamainternmed.2020.6820 [33080017]

[143] Houston BL, Lawler PR, Goligher EC, et al. Anti-Thrombotic Therapy to Ameliorate Complications of COVID-19 (ATTACC): Study design and methodology for an international, adaptive Bayesian randomized controlled trial. Clin Trials 2020:1740774520943846 10.1177/1740774520943846 [32815416]

[144] U.S. Department of Health, Human Services - Food, Drug Administration. Adaptive Designs for Clinical Trials of Drugs and Biologics - Guidance for Industry 2019 ;

[145] Bhatt DL, Mehta C, Drazen JM, et al. Adaptive Designs for Clinical Trials. New Engl J Med 2016;375:65–74 10.1056/NEJMra1510061

[146] Porcher R, Lecocq B, Vray M, et al. Les méthodes adaptatives quand et comment les utiliser dans les essais cliniques ? Thérapie 2011;66:309–17 10.2515/therapie/2011042

[147] Lachin JM. A review of methods for futility stopping based on conditional power. Stat Med 2005;24:2747–64 10.1002/sim.2151

[148] Thall P, Fox P, Wathen J. Statistical controversies in clinical research: scientific and ethical problems with adaptive randomization in comparative clinical trials. Annals of Oncology 2015;26:1621–28 10.1093/annonc/mdv238

[149] McMurray JJV, Packer M, Desai AS, et al. Angiotensin–Neprilysin Inhibition versus Enalapril in Heart Failure. New Engl J Med 2014 10.1056/NEJMoa1409077

[150] McMurray JJV, Ostergren J, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: the CHARM-Added trial. The Lancet 2003;362:767–71 10.1016/S0140-6736(03)14283-3 [13678869]

[151] Boulware DR, Pullen MF, Bangdiwala AS, et al. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for Covid-19. New Engl J Med 2020 10.1056/NEJMoa2016638

[152] van Eijk RPA, Nikolakopoulos S, Ferguson TA, et al. Increasing the efficiency of clinical trials in neurodegenerative disorders using group sequential trial designs. Journal of Clinical Epidemiology 2018;98:80–88 10.1016/j.jclinepi.2018.02.013

[153] Wilson N, Biggs K, Bowden S, et al. Costs and staffing resource requirements for adaptive clinical trials: quantitative and qualitative results from the Costing Adaptive Trials project. BMC Med 2021;19:1–17 10.1186/s12916-021-02124-z

[154] Prowell TM, Theoret MR, Pazdur R. Seamless Oncology-Drug Development. N Engl J Med 2016;374:2001–03 10.1056/NEJMp1603747

[155] Mulligan MJ, Lyke KE, Kitchin N, et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature 2020;586:589–93 10.1038/s41586-020-2639-4 [32785213]

[156] Walsh EE, Frenck RW, Falsey AR, et al. Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates. N Engl J Med 2020;383:2439–50 10.1056/NEJMoa2027906 [33053279]

[157] Polack FP, Thomas SJ, Kitchin N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med 2020;383:2603–15 10.1056/NEJMoa2034577 [33301246]

[158] Thomas SJ, Moreira ED, Kitchin N, et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine through 6 Months. N Engl J Med 2021 10.1056/NEJMoa2110345 [34525277]

[159] Hobbs BP, Barata PC, Kanjanapan Y, et al. Seamless Designs: Current Practice and Considerations for Early-Phase Drug Development in Oncology. JNCI Journal of the National Cancer Institute 2019;111:118–28 10.1093/jnci/djy196

[160] Chalmers I, Bracken MB, Djulbegovic B, et al. How to increase value and reduce waste when research priorities are set. The Lancet 2014;383:156–65 10.1016/S0140-6736(13)62229-1

[161] Chalmers I, Glasziou P. Avoidable waste in the production and reporting of research evidence. The Lancet 2009;374:86–89 10.1016/S0140-6736(09)60329-9

[162] Glasziou P, Altman DG, Bossuyt P, et al. Reducing waste from incomplete or unusable reports of biomedical research. The Lancet 2014;383:267–76 10.1016/S0140-6736(13)62228-X

[163] Glasziou P, Chalmers I. Research waste is still a scandal—an essay by Paul Glasziou and Iain Chalmers. BMJ 2018:k4645 10.1136/bmj.k4645

[164] Lu J, Xu B, Shen L, et al. Characteristics and Research Waste Among Randomized Clinical Trials in Gastric Cancer. JAMA Netw Open 2021;4:e2124760 10.1001/jamanetworkopen.2021.24760 [34533573]

[165] The BMJ. Paul Glasziou and Iain Chalmers: Can it really be true that 50% of research is unpublished? - The BMJ 2017. Available at: https://blogs.bmj.com/bmj/2017/06/05/paul-glasziou-and-iain-chalmers-can-it-really-be-true-that-50-of-research-is-unpublished/ Accessed August 22, 2021.

[166] Yordanov Y, Dechartres A, Porcher R, et al. Avoidable waste of research related to inadequate methods in clinical trials. BMJ 2015;350:h809 10.1136/bmj.h809 [25804210]

[167] Downing NS, Aminawung JA, Shah ND, et al. Clinical trial evidence supporting FDA approval of novel therapeutic agents, 2005-2012. JAMA 2014;311:368–77 10.1001/jama.2013.282034 [24449315]

[168] Bours MJL. A nontechnical explanation of the counterfactual definition of confounding. J Clin Epidemiol 2020;121:91–100 10.1016/j.jclinepi.2020.01.021 [32068101]

[169] International Conference on Harmonisation of technical requirements for registration of pharmaceuticals for human use. CHOICE OF CONTROL GROUP AND RELATED ISSUES IN CLINICAL TRIALS E10.

[170] INTERNATIONAL COUNCIL FOR HARMONISATION OF TECHNICAL REQUIREMENTS FOR PHARMACEUTICALS FOR HUMAN USE. ADDENDUM ON ESTIMANDS AND SENSITIVITY ANALYSIS IN CLINICAL TRIALS TO THE GUIDELINE ON STATISTICAL PRINCIPLES FOR CLINICAL TRIALS E9(R1).

[171] Grein J, Ohmagari N, Shin D, et al. Compassionate Use of Remdesivir for Patients with Severe Covid-19. N Engl J Med 2020;382:2327–36 10.1056/NEJMoa2007016 [32275812]

[172] Rittberg R, Czaykowski P, Niraula S. Feasibility of Randomized Controlled Trials for Cancer Drugs Approved by the Food and Drug Administration Based on Single-Arm Studies. JNCI Cancer Spectrum 2021;5:pkab061 10.1093/jncics/pkab061 [34409254]

[173] International Conference on Harmonisation of technical requirements for registration of pharmaceuticals for human use. Choice of control group and related issues in clinical trials E10 2000.

[174] Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. The Lancet 2016;387:1909–20 10.1016/S0140-6736(16)00561-4

[175] Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. The Lancet 2018;391:748–57 10.1016/S0140-6736(17)33297-X [29268948]

[176] Signorovitch JE, Sikirica V, Erder MH, et al. Matching-adjusted indirect comparisons: a new tool for timely comparative effectiveness research. Value Health 2012;15:940–47 10.1016/j.jval.2012.05.004 [22999145]

[177] Signorovitch JE, Wu EQ, Yu AP, et al. Comparative effectiveness without head-to-head trials: a method for matching-adjusted indirect comparisons applied to psoriasis treatment with adalimumab or etanercept. PharmacoEconomics 2010;28:935–45 10.2165/11538370-000000000-00000 [20831302]

[178] Fox MP, Lash TL. On the Need for Quantitative Bias Analysis in the Peer-Review Process. Am J Epidemiol 2017;185:865–68 10.1093/aje/kwx057 [28430833]

[179] Lash TL, Fox MP, Cooney D, et al. Quantitative Bias Analysis in Regulatory Settings. Am J Public Health 2016;106:1227–30 10.2105/AJPH.2016.303199 [27196652]

[180] Lash TL, Fox MP, MacLehose RF, et al. Good practices for quantitative bias analysis. Int J Epidemiol 2014;43:1969–85 10.1093/ije/dyu149 [25080530]

[181] David M. Phillippo, A. E. Ades, Sofia Dias, Stephen Palmer, Keith R. Abrams, Nicky J. Welton. NICE DSU technical support document 18: methods for population-adjusted indirect comparisons in submissions to NICE 2016.

[182] Sridhara R, Mandrekar SJ, Dodd LE. Missing data and measurement variability in assessing progression-free survival endpoint in randomized clinical trials. Clin Cancer Res 2013;19:2613–20 10.1158/1078-0432.CCR-12-2938 [23669421]

[183] Denne JS, Stone AM, Bailey-Iacona R, et al. Missing data and censoring in the analysis of progression-free survival in oncology clinical trials. Journal of biopharmaceutical statistics 2013;23:951–70 10.1080/10543406.2013.813515 [23957509]

[184] Marks HM. A rational therapeutics: Science and the reform of therapeutics in the United States, 1900-1990. Cambridge England, New York: Cambridge University Press 1997 ISBN:0521581427;

[185] Debray TPA, Damen JAAG, Snell KIE, et al. A guide to systematic review and meta-analysis of prediction model performance. BMJ 2017;356:i6460 10.1136/bmj.i6460 [28057641]

[186] 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 2020;107:369–77 10.1002/cpt.1586 [31350853]

[187] Larrouquere L, Giai J, Cracowski J-L, et al. Externally Controlled Trials: Are We There Yet? Clin Pharmacol Ther 2020;108:918–19 10.1002/cpt.1881 [32542679]

[188] Neuenschwander B, Capkun-Niggli G, Branson M, et al. Summarizing historical information on controls in clinical trials. Clin Trials 2010;7:5–18 10.1177/1740774509356002 [20156954]

[189] FDA/CBER. Interacting with the Food and Drug Administration on Complex Innovative Clinical Trial Designs for Drugs and Biological Products, Guidance for Industry ;

[190] FDA/CDER/mccrayk. Rare Diseases: Common Issues in Drug Development: Guidance for Industry ;

[191] EWP. Committee for Medicinal Product for Human Use (CHMP). In: D'Agostino RB, Sullivan L, Massaro J, eds. Wiley Encyclopedia of Clinical Trials. Hoboken, NJ, USA: John Wiley & Sons, Inc 2007.

[192] Pocock SJ. The combination of randomized and historical controls in clinical trials. J Chronic Dis 1976;29:175–88 10.1016/0021-9681(76)90044-8 [770493]

[193] Neuenschwander B, Branson M, Spiegelhalter DJ. A note on the power prior. Stat Med 2009;28:3562–66 10.1002/sim.3722 [19735071]

[194] Schmidli H, Gsteiger S, Roychoudhury S, et al. Robust meta-analytic-predictive priors in clinical trials with historical control information. Biometrics 2014;70:1023–32 10.1111/biom.12242 [25355546]

[195] Baeten D, Baraliakos X, Braun J, et al. Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial. The Lancet 2013;382:1705–13 10.1016/S0140-6736(13)61134-4

[196] Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001;69:89–95 10.1067/mcp.2001.113989 [11240971]

[197] Buyse M, Molenberghs G, Burzykowski T, et al. The validation of surrogate endpoints in meta-analyses of randomized experiments. Biostatistics 2000;1:49–67 10.1093/biostatistics/1.1.49 [12933525]

[198] Buyse M, Molenberghs G, Paoletti X, et al. Statistical evaluation of surrogate endpoints with examples from cancer clinical trials: Statistical evaluation of surrogate endpoints. Biometrical Journal 2016;58:104–32 10.1002/bimj.201400049

[199] IQWiG. Validity of surrogate endpoints in oncology. Cologne 2011.

[200] Burzykowski T, Buyse M. Surrogate threshold effect: an alternative measure for meta-analytic surrogate endpoint validation. Pharm Stat 2006;5:173–86 10.1002/pst.207 [17080751]

[201] Baker SG. Five criteria for using a surrogate endpoint to predict treatment effect based on data from multiple previous trials. Stat Med 2018;37:507–18 10.1002/sim.7561 [29164641]

[202] Papanikos T, Thompson JR, Abrams KR, et al. A novel approach to bivariate meta-analysis of binary outcomes and its application in the context of surrogate endpoints 2020.

[203] Bujkiewicz S, Jackson D, Thompson JR, et al. Bivariate network meta-analysis for surrogate endpoint evaluation. Stat Med 2019;38:3322–41 10.1002/sim.8187 [31131475]

[204] Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90 056 participants in 14 randomised trials of statins. The Lancet 2005;366:1267–78 10.1016/S0140-6736(05)67394-1 [16214597]

[205] Landray MJ, Haynes R, Hopewell JC, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014;371:203–12 10.1056/NEJMoa1300955 [25014686]

[206] Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N Engl J Med 2017;376:1713–22 10.1056/NEJMoa1615664 [28304224]

[207] Mok TSK, Wu Y-L, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. The Lancet 2019;393:1819–30 10.1016/S0140-6736(18)32409-7

[208] Xie W, Regan MM, Buyse M, et al. Metastasis-Free Survival Is a Strong Surrogate of Overall Survival in Localized Prostate Cancer. Journal of Clinical Oncology 2017:JCO.2017.73.9987 10.1200/JCO.2017.73.9987

[209] Naci H, Davis C. Inappropriate use of progression-free survival in cancer drug approvals. BMJ-BRITISH MEDICAL JOURNAL 2020;368:m770 10.1136/bmj.m770 [32156802]

[210] Kemp R, Prasad V. Surrogate endpoints in oncology: when are they acceptable for regulatory and clinical decisions, and are they currently overused? BMC Med 2017;15:134 10.1186/s12916-017-0902-9 [28728605]

[211] Chen EY, Joshi SK, Tran A, et al. Estimation of Study Time Reduction Using Surrogate End Points Rather Than Overall Survival in Oncology Clinical Trials. JAMA Internal Medicine 2019;179:642–47 10.1001/jamainternmed.2018.8351 [30933235]

[212] Downing NS, Aminawung JA, Shah ND, et al. Clinical trial evidence supporting FDA approval of novel therapeutic agents, 2005-2012. JAMA 2014;311:368–77 10.1001/jama.2013.282034 [24449315]

[213] Pease AM, Krumholz HM, Downing NS, et al. Postapproval studies of drugs initially approved by the FDA on the basis of limited evidence: systematic review. BMJ-BRITISH MEDICAL JOURNAL 2017;357:j1680 10.1136/bmj.j1680 [28468750]

[214] Prasad V, Kim C, Burotto M, et al. The Strength of Association Between Surrogate End Points and Survival in Oncology: A Systematic Review of Trial-Level Meta-analyses. JAMA Internal Medicine 2015;175:1389–98 10.1001/jamainternmed.2015.2829 [26098871]

[215] Buyse M, Burzykowski T, Carroll K, et al. Progression-free survival is a surrogate for survival in advanced colorectal cancer. J Clin Oncol 2007;25:5218–24 10.1200/JCO.2007.11.8836 [18024867]

[216] Ciani O, Buyse M, Garside R, et al. Meta-analyses of randomized controlled trials show suboptimal validity of surrogate outcomes for overall survival in advanced colorectal cancer. J Clin Epidemiol 2015;68:833–42 10.1016/j.jclinepi.2015.02.016 [25863582]

[217] Yu T, Hsu Y-J, Fain KM, et al. Use of surrogate outcomes in US FDA drug approvals, 2003-2012: a survey. BMJ open 2015;5:e007960 10.1136/bmjopen-2015-007960 [26614616]

[218] Ciani O, Buyse M, Garside R, et al. Comparison of treatment effect sizes associated with surrogate and final patient relevant outcomes in randomised controlled trials: meta-epidemiological study. BMJ 2013;346:f457 10.1136/bmj.f457 [23360719]

[219] Ciani O, Buyse M, Drummond M, et al. Time to Review the Role of Surrogate End Points in Health Policy: State of the Art and the Way Forward. Value in Health 2017;20:487–95 10.1016/j.jval.2016.10.011

[220] Ciani O, Buyse M, Drummond M, et al. Use of surrogate end points in healthcare policy: a proposal for adoption of a validation framework. Nature Reviews Drug Discovery 2016;15:516 10.1038/nrd.2016.81

[221] Ciani O, Davis S, Tappenden P, et al. VALIDATION OF SURROGATE ENDPOINTS IN ADVANCED SOLID TUMORS: SYSTEMATIC REVIEW OF STATISTICAL METHODS, RESULTS, AND IMPLICATIONS FOR POLICY MAKERS. International Journal of Technology Assessment in Health Care 2014;30:312–24 10.1017/S0266462314000300

[222] Validity of surrogate endpoints in oncology: IQWiG Reports – Commission No. A10-05.

[223] A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). The Lancet 1996;348:1329–39 10.1016/s0140-6736(96)09457-3 [8918275]

[224] Collaborative overview of randomised trials of antiplatelet therapy--I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various categories of patients. Antiplatelet Trialists'Collaboration. BMJ 1994;308:81–106 ; [8298418]

[225] Collaborative overview of randomised trials of antiplatelet therapy--II: Maintenance of vascular graft or arterial patency by antiplatelet therapy. Antiplatelet Trialists' Collaboration. BMJ 1994;308:159–68 ; [8312766]

[226] Egger M, Davey Smith G, Schneider M, et al. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997;315:629–34 10.1136/bmj.315.7109.629 [9310563]

[227] Villar J, Carroli G, Belizán JM. Predictive ability of meta-analyses of randomised controlled trials. The Lancet 1995;345:772–76 10.1016/s0140-6736(95)90646-0 [7891492]

[228] LeLorier J, Grégoire G, Benhaddad A, et al. Discrepancies between meta-analyses and subsequent large randomized, controlled trials. New Engl J Med 1997;337:536–42 10.1056/NEJM199708213370806 [9262498]

[229] Cappelleri JC, Ioannidis JP, Schmid CH, et al. Large trials vs meta-analysis of smaller trials: how do their results compare? JAMA 1996;276:1332–38 ; [8861993]

[230] Meta-analysis under scrutiny. The Lancet 1997;350:675 ; [9291895]

[231] Cranney A, Welch V, Adachi JD, et al. Etidronate for treating and preventing postmenopausal osteoporosis. Cochrane Database Syst Rev 2001:CD003376 10.1002/14651858.CD003376 [11687195]

[232] Granger CB, McMurray JJV, Yusuf S, et al. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function intolerant to angiotensin-converting-enzyme inhibitors: the CHARM-Alternative trial. The Lancet 2003;362:772–76 10.1016/S0140-6736(03)14284-5 [13678870]

[233] Yusuf S, Pfeffer MA, Swedberg K, et al. Effects of candesartan in patients with chronic heart failure and preserved left-ventricular ejection fraction: the CHARM-Preserved Trial. The Lancet 2003;362:777–81 10.1016/S0140-6736(03)14285-7 [13678871]

[234] Pfeffer MA, Swedberg K, Granger CB, et al. Effects of candesartan on mortality and morbidity in patients with chronic heart failure: the CHARM-Overall programme. The Lancet 2003;362:759–66 10.1016/s0140-6736(03)14282-1 [13678868]

[235] Hauser SL, Bar-Or A, Cohen JA, et al. Ofatumumab versus Teriflunomide in Multiple Sclerosis. N Engl J Med 2020;383:546–57 10.1056/NEJMoa1917246 [32757523]

[236] Salanti G, Del Giovane C, Chaimani A, et al. Evaluating the quality of evidence from a network meta-analysis. PLoS ONE 2014;9:e99682 10.1371/journal.pone.0099682 [24992266]

[237] Salanti G, Higgins J, Ades AE, et al. Evaluation of networks of randomized trials. Statistical methods in medical research 2007 ;

[238] Song F, Xiong T, Parekh-Bhurke S, et al. Inconsistency between direct and indirect comparisons of competing interventions: meta-epidemiological study. BMJ (Clinical research ed.) 2011;343:d4909-d4909 10.1136/bmj.d4909

[239] Song F, Altman DG, Glenny AM, et al. Validity of indirect comparison for estimating efficacy of competing interventions: empirical evidence from published meta-analyses. BMJ 2003;326:472 ;

[240] Song F, Glenny AM, Altman DG. Indirect comparison in evaluating relative efficacy illustrated by antimicrobial prophylaxis in colorectal surgery. Control Clin Trials 2000;21:488–97 ;

[241] Veroniki AA, Tsokani S, White IR, et al. Prevalence of evidence of inconsistency and its association with network structural characteristics in 201 published networks of interventions. BMC Med Res Methodol 2021;21:224 10.1186/s12874-021-01401-y [34689743]

[242] Salanti G, Nikolakopoulou A, Efthimou O, et al. Introducing the treatment hierarchy question in network meta-analysis 2020.

[243] Pontes C, Fontanet JM, Vives R, et al. Evidence supporting regulatory-decision making on orphan medicinal products authorisation in Europe: methodological uncertainties. Orphanet J Rare Dis 2018;13:206 10.1186/s13023-018-0926-z [30442155]

[244] Meekings KN, Williams CSM, Arrowsmith JE. Orphan drug development: an economically viable strategy for biopharma R&D. Drug Discov Today 2012;17:660–64 10.1016/j.drudis.2012.02.005 [22366309]

[245] Gaddipati H, Liu K, Pariser A, et al. Rare cancer trial design: lessons from FDA approvals. Clin Cancer Res 2012;18:5172–78 10.1158/1078-0432.CCR-12-1135 [22718862]

[246] Day S, Jonker AH, Lau LPL, et al. Recommendations for the design of small population clinical trials. Orphanet J Rare Dis 2018;13:195 10.1186/s13023-018-0931-2 [30400970]

[247] Cucherat M, Laporte S. Les résultats faux positifs ou quelle est la probabilité que le traitement soit efficace quand p < 0,05 ? Thérapie 2017;72:421–26 10.1016/j.therap.2016.09.021 [28577824]

[248] Prasad V, Oseran A. Do we need randomised trials for rare cancers? Eur J Cancer 2015;51:1355–57 10.1016/j.ejca.2015.04.015 [25963018]

[249] Mitsumoto J, Dorsey ER, Beck CA, et al. Pivotal studies of orphan drugs approved for neurological diseases. Ann Neurol 2009;66:184–90 10.1002/ana.21676 [19743448]

[250] Hilgers R-D. Design and analysis of clinical trials for small rare disease populations. J Rare Dis Res Treat 2016;1:53–60 10.29245/2572-9411/2016/3.1054

[251] Kesselheim AS, Myers JA, Avorn J. Characteristics of clinical trials to support approval of orphan vs nonorphan drugs for cancer. JAMA 2011;305:2320–26 10.1001/jama.2011.769 [21642684]

[252] Jiao F, Tu W, Jimenez S, et al. Utilizing shared internal control arms and historical information in small-sized platform clinical trials. Journal of biopharmaceutical statistics 2019;29:845–59 10.1080/10543406.2019.1657132 [31462131]

[253] Ursino M, Stallard N. Bayesian Approaches for Confirmatory Trials in Rare Diseases: Opportunities and Challenges. Int J Environ Res Public Health 2021;18 10.3390/ijerph18031022 [33498915]

[254] Wandel S, Neuenschwander B, Röver C, et al. Using phase II data for the analysis of phase III studies: An application in rare diseases. Clin Trials 2017;14:277–85 10.1177/1740774517699409 [28387537]

[255] Finkel RS, Mercuri E, Darras BT, et al. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med 2017;377:1723–32 10.1056/NEJMoa1702752 [29091570]