Skip to main content
Top
Published in: BMC Medical Research Methodology 1/2020

Open Access 01-12-2020 | Research article

Statistical methods for the analysis of adverse event data in randomised controlled trials: a scoping review and taxonomy

Authors: Rachel Phillips, Odile Sauzet, Victoria Cornelius

Published in: BMC Medical Research Methodology | Issue 1/2020

Login to get access

Abstract

Background

Statistical methods for the analysis of harm outcomes in randomised controlled trials (RCTs) are rarely used, and there is a reliance on simple approaches to display information such as in frequency tables. We aimed to identify whether any statistical methods had been specifically developed to analyse prespecified secondary harm outcomes and non-specific emerging adverse events (AEs).

Methods

A scoping review was undertaken to identify articles that proposed original methods or the original application of existing methods for the analysis of AEs that aimed to detect potential adverse drug reactions (ADRs) in phase II-IV parallel controlled group trials. Methods where harm outcomes were the (co)-primary outcome were excluded.
Information was extracted on methodological characteristics such as: whether the method required the event to be prespecified or could be used to screen emerging events; and whether it was applied to individual events or the overall AE profile. Each statistical method was appraised and a taxonomy was developed for classification.

Results

Forty-four eligible articles proposing 73 individual methods were included. A taxonomy was developed and articles were categorised as: visual summary methods (8 articles proposing 20 methods); hypothesis testing methods (11 articles proposing 16 methods); estimation methods (15 articles proposing 24 methods); or methods that provide decision-making probabilities (10 articles proposing 13 methods). Methods were further classified according to whether they required a prespecified event (9 articles proposing 12 methods), or could be applied to emerging events (35 articles proposing 61 methods); and if they were (group) sequential methods (10 articles proposing 12 methods) or methods to perform final/one analyses (34 articles proposing 61 methods).

Conclusions

This review highlighted that a broad range of methods exist for AE analysis. Immediate implementation of some of these could lead to improved inference for AE data in RCTs. For example, a well-designed graphic can be an effective means to communicate complex AE data and methods appropriate for counts, time-to-event data and that avoid dichotomising continuous outcomes can improve efficiencies in analysis. Previous research has shown that adoption of such methods in the scientific press is limited and that strategies to support change are needed.
Appendix
Available only for authorised users
Footnotes
1
An adverse event is defined as ‘any untoward medical occurrence that may present during treatment with a pharmaceutical product but which does not necessarily have a causal relationship with this treatment’. An adverse drug reaction (ADR) is defined as ‘a response to a drug which is noxious and unintended …’ where a causal relationship is ‘at least a reasonable possibility’.
 
Literature
2.
go back to reference The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH): ICH Harmonised Tripartite Guideline. E2A Clinical Safety Data Management: Definitions and Standards for Expedited Reporting. 1994. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH): ICH Harmonised Tripartite Guideline. E2A Clinical Safety Data Management: Definitions and Standards for Expedited Reporting. 1994.
4.
go back to reference Phillips R, Hazell L, Sauzet O, Cornelius V. Analysis and reporting of adverse events in randomised controlled trials: a review. BMJ Open. 2019;9(2):e024537.PubMedPubMedCentralCrossRef Phillips R, Hazell L, Sauzet O, Cornelius V. Analysis and reporting of adverse events in randomised controlled trials: a review. BMJ Open. 2019;9(2):e024537.PubMedPubMedCentralCrossRef
5.
go back to reference Seltzer JH, Li J, Wang W. Interdisciplinary safety evaluation and quantitative safety monitoring: introduction to a series of papers. Ther Innov Regul Sci. 2019;0(0):2168479018793130. Seltzer JH, Li J, Wang W. Interdisciplinary safety evaluation and quantitative safety monitoring: introduction to a series of papers. Ther Innov Regul Sci. 2019;0(0):2168479018793130.
6.
go back to reference Ioannidis JA, Evans SW, Gøtzsche PC, et al. Better reporting of harms in randomized trials: an extension of the consort statement. Ann Intern Med. 2004;141(10):781–8.PubMedCrossRef Ioannidis JA, Evans SW, Gøtzsche PC, et al. Better reporting of harms in randomized trials: an extension of the consort statement. Ann Intern Med. 2004;141(10):781–8.PubMedCrossRef
7.
go back to reference Zorzela L, Loke YK, Ioannidis JP, Golder S, Santaguida P, Altman DG, Moher D, Vohra S. PRISMA harms checklist: improving harms reporting in systematic reviews. BMJ. 2016;352. Zorzela L, Loke YK, Ioannidis JP, Golder S, Santaguida P, Altman DG, Moher D, Vohra S. PRISMA harms checklist: improving harms reporting in systematic reviews. BMJ. 2016;352.
8.
go back to reference Lineberry N, Berlin JA, Mansi B, Glasser S, Berkwits M, Klem C, Bhattacharya A, Citrome L, Enck R, Fletcher J, et al. Recommendations to improve adverse event reporting in clinical trial publications: A joint pharmaceutical industry/journal editor perspective. BMJ. 2016;355(linn033422):i5078.CrossRefPubMed Lineberry N, Berlin JA, Mansi B, Glasser S, Berkwits M, Klem C, Bhattacharya A, Citrome L, Enck R, Fletcher J, et al. Recommendations to improve adverse event reporting in clinical trial publications: A joint pharmaceutical industry/journal editor perspective. BMJ. 2016;355(linn033422):i5078.CrossRefPubMed
9.
go back to reference Crowe BJ, Xia HA, Berlin JA, Watson DJ, Shi H, Lin SL, Kuebler J, Schriver RC, Santanello NC, Rochester G, et al. Recommendations for safety planning, data collection, evaluation and reporting during drug, biologic and vaccine development: a report of the safety planning, evaluation, and reporting team. Clin Trials. 2009;6(5):430–40.PubMedCrossRef Crowe BJ, Xia HA, Berlin JA, Watson DJ, Shi H, Lin SL, Kuebler J, Schriver RC, Santanello NC, Rochester G, et al. Recommendations for safety planning, data collection, evaluation and reporting during drug, biologic and vaccine development: a report of the safety planning, evaluation, and reporting team. Clin Trials. 2009;6(5):430–40.PubMedCrossRef
10.
go back to reference Communication from the Commission. Detailed guidance on the collection, verification and presentation of adverse event/reaction reports arising from clinical trials on medicinal products for human use (‘CT-3’). Off J Eur Union. 2011; C 172/1. Communication from the Commission. Detailed guidance on the collection, verification and presentation of adverse event/reaction reports arising from clinical trials on medicinal products for human use (‘CT-3’). Off J Eur Union. 2011; C 172/1.
14.
go back to reference Zink RC, Marchenko O, Sanchez-Kam M, Ma H, Jiang Q. Sources of safety data and statistical strategies for design and analysis:clinical trials. Ther Innov Regul Sci. 2018;52(2):141–58.PubMedCrossRef Zink RC, Marchenko O, Sanchez-Kam M, Ma H, Jiang Q. Sources of safety data and statistical strategies for design and analysis:clinical trials. Ther Innov Regul Sci. 2018;52(2):141–58.PubMedCrossRef
15.
go back to reference Siddiqui O. Statistical methods to analyze adverse events data of randomized clinical trials. J Biopharm Stat. 2009;19(5):889–99.PubMedCrossRef Siddiqui O. Statistical methods to analyze adverse events data of randomized clinical trials. J Biopharm Stat. 2009;19(5):889–99.PubMedCrossRef
16.
go back to reference Ma H, Ke C, Jiang Q, Snapinn S. Statistical considerations on the evaluation of imbalances of adverse events in randomized clinical trials. Ther Innov Regul Sci. 2015;49(6):957–65.PubMedCrossRef Ma H, Ke C, Jiang Q, Snapinn S. Statistical considerations on the evaluation of imbalances of adverse events in randomized clinical trials. Ther Innov Regul Sci. 2015;49(6):957–65.PubMedCrossRef
17.
go back to reference Food and Drug Administration. Attachment B: Clinical Safety Review of an NDA or BLA of the Good Review Practice. Clinical Review Template (MAPP 6010.3 Rev. 1). Silver Spring: Food and Drug Administration; 2010. Food and Drug Administration. Attachment B: Clinical Safety Review of an NDA or BLA of the Good Review Practice. Clinical Review Template (MAPP 6010.3 Rev. 1). Silver Spring: Food and Drug Administration; 2010.
18.
go back to reference Unkel S, Amiri M, Benda N, Beyersmann J, Knoerzer D, Kupas K, Langer F, Leverkus F, Loos A, Ose C, et al. On estimands and the analysis of adverse events in the presence of varying follow-up times within the benefit assessment of therapies. Pharm Stat. 2019;18(2):166–83.PubMedCrossRef Unkel S, Amiri M, Benda N, Beyersmann J, Knoerzer D, Kupas K, Langer F, Leverkus F, Loos A, Ose C, et al. On estimands and the analysis of adverse events in the presence of varying follow-up times within the benefit assessment of therapies. Pharm Stat. 2019;18(2):166–83.PubMedCrossRef
19.
go back to reference Favier R, Crépin S. The reporting of harms in publications on randomized controlled trials funded by the “Programme Hospitalier de Recherche Clinique,” a French academic funding scheme. Clin Trials. 2018;0(0):1740774518760565. Favier R, Crépin S. The reporting of harms in publications on randomized controlled trials funded by the “Programme Hospitalier de Recherche Clinique,” a French academic funding scheme. Clin Trials. 2018;0(0):1740774518760565.
20.
go back to reference Arksey H, O'Malley L. Scoping studies: towards a methodological framework. Int J Soc Res Methodol. 2005;8(1):19–32.CrossRef Arksey H, O'Malley L. Scoping studies: towards a methodological framework. Int J Soc Res Methodol. 2005;8(1):19–32.CrossRef
21.
go back to reference O'Brien PC, Fleming TR. A multiple testing procedure for clinical trials. Biometrics. 1979;35(3):549–56.PubMedCrossRef O'Brien PC, Fleming TR. A multiple testing procedure for clinical trials. Biometrics. 1979;35(3):549–56.PubMedCrossRef
22.
go back to reference DeMets DL, Lan G. The alpha spending function approach to interim data analyses. In: Thall PF, editor. Recent Advances in Clinical Trial Design and Analysis. Boston: Springer US; 1995. p. 1–27. DeMets DL, Lan G. The alpha spending function approach to interim data analyses. In: Thall PF, editor. Recent Advances in Clinical Trial Design and Analysis. Boston: Springer US; 1995. p. 1–27.
23.
go back to reference Tricco AC, Lillie E, Zarin W, et al. Prisma extension for scoping reviews (prisma-scr): checklist and explanation. Ann Intern Med. 2018. Tricco AC, Lillie E, Zarin W, et al. Prisma extension for scoping reviews (prisma-scr): checklist and explanation. Ann Intern Med. 2018.
24.
go back to reference Moher D, Liberati A, Tetzlaff J, Altman DG, The PG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.PubMedPubMedCentralCrossRef Moher D, Liberati A, Tetzlaff J, Altman DG, The PG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097.PubMedPubMedCentralCrossRef
25.
go back to reference Amit O, Heiberger RM, Lane PW. Graphical approaches to the analysis of safety data from clinical trials. Pharm Stat. 2008;7(1):20–35.PubMedCrossRef Amit O, Heiberger RM, Lane PW. Graphical approaches to the analysis of safety data from clinical trials. Pharm Stat. 2008;7(1):20–35.PubMedCrossRef
26.
go back to reference Chuang-Stein C, Le V, Chen W. Recent advancements in the analysis and presentation of safety data. Drug Information Journal. 2001;35(2):377–97.CrossRef Chuang-Stein C, Le V, Chen W. Recent advancements in the analysis and presentation of safety data. Drug Information Journal. 2001;35(2):377–97.CrossRef
27.
go back to reference Chuang-Stein C, Xia HA. The practice of pre-marketing safety assessment in drug development. J Biopharm Stat. 2013;23(1):3–25.PubMedCrossRef Chuang-Stein C, Xia HA. The practice of pre-marketing safety assessment in drug development. J Biopharm Stat. 2013;23(1):3–25.PubMedCrossRef
28.
go back to reference Karpefors M, Weatherall J. The tendril plot—a novel visual summary of the incidence, significance and temporal aspects of adverse events in clinical trials. J Am Med Inform Assoc. 2018;25(8):1069–73.PubMedCrossRefPubMedCentral Karpefors M, Weatherall J. The tendril plot—a novel visual summary of the incidence, significance and temporal aspects of adverse events in clinical trials. J Am Med Inform Assoc. 2018;25(8):1069–73.PubMedCrossRefPubMedCentral
29.
go back to reference Southworth H. Detecting outliers in multivariate laboratory data. J Biopharm Stat. 2008;18(6):1178–83.PubMedCrossRef Southworth H. Detecting outliers in multivariate laboratory data. J Biopharm Stat. 2008;18(6):1178–83.PubMedCrossRef
30.
go back to reference Trost DC, Freston JW. Vector analysis to detect hepatotoxicity signals in drug development. Ther Innov Regul Science. 2008;42(1):27–34. Trost DC, Freston JW. Vector analysis to detect hepatotoxicity signals in drug development. Ther Innov Regul Science. 2008;42(1):27–34.
31.
go back to reference Zink RC, Wolfinger RD, Mann G. Summarizing the incidence of adverse events using volcano plots and time intervals. Clin Trials. 2013;10(3):398–406.PubMedCrossRef Zink RC, Wolfinger RD, Mann G. Summarizing the incidence of adverse events using volcano plots and time intervals. Clin Trials. 2013;10(3):398–406.PubMedCrossRef
32.
go back to reference Bolland K, Whitehead J. Formal approaches to safety monitoring of clinical trials in life-threatening conditions. Stat Med. 2000;19(21):2899–917.PubMedCrossRef Bolland K, Whitehead J. Formal approaches to safety monitoring of clinical trials in life-threatening conditions. Stat Med. 2000;19(21):2899–917.PubMedCrossRef
33.
go back to reference Fleishman AN, Parker RA. Stopping guidelines for harm in a study designed to establish the safety of a marketed drug. J Biopharm Stat. 2012;22(2):338–50.PubMedCrossRef Fleishman AN, Parker RA. Stopping guidelines for harm in a study designed to establish the safety of a marketed drug. J Biopharm Stat. 2012;22(2):338–50.PubMedCrossRef
34.
go back to reference Lieu TA, Kulldorff M, Davis RL, Lewis EM, Weintraub E, Yih K, Yin R, Brown JS, Platt R. Real-time vaccine safety surveillance for the early detection of adverse events. Med Care. 2007;45(10 SUPPL. 2):S89–95.PubMedCrossRef Lieu TA, Kulldorff M, Davis RL, Lewis EM, Weintraub E, Yih K, Yin R, Brown JS, Platt R. Real-time vaccine safety surveillance for the early detection of adverse events. Med Care. 2007;45(10 SUPPL. 2):S89–95.PubMedCrossRef
36.
37.
go back to reference Agresti AaK B. Multivariate tests comparing binomial probabilities, with application to safety studies for drugs. Appl Stat. 2005;54(4):691–706. Agresti AaK B. Multivariate tests comparing binomial probabilities, with application to safety studies for drugs. Appl Stat. 2005;54(4):691–706.
38.
go back to reference Bristol DR, Patel HI. A Markovian model for comparing incidences of side effects. Stat Med. 1990;9(7):803–9.PubMedCrossRef Bristol DR, Patel HI. A Markovian model for comparing incidences of side effects. Stat Med. 1990;9(7):803–9.PubMedCrossRef
39.
go back to reference Chuang-Stein C, Mohberg NR, Musselman DM. Organization and analysis of safety data using a multivariate approach. Stat Med. 1992;11(8):1075–89.PubMedCrossRef Chuang-Stein C, Mohberg NR, Musselman DM. Organization and analysis of safety data using a multivariate approach. Stat Med. 1992;11(8):1075–89.PubMedCrossRef
40.
go back to reference Huang L, Zalkikar J, Tiwari R. Likelihood ratio based tests for longitudinal drug safety data. Stat Med. 2014;33(14):2408–24.PubMedCrossRef Huang L, Zalkikar J, Tiwari R. Likelihood ratio based tests for longitudinal drug safety data. Stat Med. 2014;33(14):2408–24.PubMedCrossRef
41.
go back to reference Mehrotra DV, Adewale AJ. Flagging clinical adverse experiences: reducing false discoveries without materially compromising power for detecting true signals. Stat Med. 2012;31(18):1918–30.PubMedCrossRef Mehrotra DV, Adewale AJ. Flagging clinical adverse experiences: reducing false discoveries without materially compromising power for detecting true signals. Stat Med. 2012;31(18):1918–30.PubMedCrossRef
42.
go back to reference Mehrotra DV, Heyse JF. Use of the false discovery rate for evaluating clinical safety data. Stat Methods Med Res. 2004;13(3):227–38.PubMedCrossRef Mehrotra DV, Heyse JF. Use of the false discovery rate for evaluating clinical safety data. Stat Methods Med Res. 2004;13(3):227–38.PubMedCrossRef
43.
go back to reference Allignol A, Beyersmann J, Schmoor C. Statistical issues in the analysis of adverse events in time-to-event data. Pharm Stat. 2016;15(4):297–305.PubMedCrossRef Allignol A, Beyersmann J, Schmoor C. Statistical issues in the analysis of adverse events in time-to-event data. Pharm Stat. 2016;15(4):297–305.PubMedCrossRef
44.
go back to reference Borkowf CB. Constructing binomial confidence intervals with near nominal coverage by adding a single imaginary failure or success. Stat Med. 2006;25(21):3679–95.PubMedCrossRef Borkowf CB. Constructing binomial confidence intervals with near nominal coverage by adding a single imaginary failure or success. Stat Med. 2006;25(21):3679–95.PubMedCrossRef
45.
go back to reference Evans SJW, Nitsch D. Statistics: Analysis and Presentation of Safety Data. In: Talbot J, Aronson JK, editors. Stephens' Detection and Evaluation of Adverse Drug Reactions: Principles and Practice. 6th ed: Wiley; 2012. p. 349–88.. Evans SJW, Nitsch D. Statistics: Analysis and Presentation of Safety Data. In: Talbot J, Aronson JK, editors. Stephens' Detection and Evaluation of Adverse Drug Reactions: Principles and Practice. 6th ed: Wiley; 2012. p. 349–88..
46.
go back to reference Gong Q, Tong B, Strasak A, Fang L. Analysis of safety data in clinical trials using a recurrent event approach. Pharm Stat. 2014;13(2):136–44.PubMedCrossRef Gong Q, Tong B, Strasak A, Fang L. Analysis of safety data in clinical trials using a recurrent event approach. Pharm Stat. 2014;13(2):136–44.PubMedCrossRef
47.
go back to reference Hengelbrock J, Gillhaus J, Kloss S, Leverkus F. Safety data from randomized controlled trials: applying models for recurrent events. Pharm Stat. 2016;15(4):315–23.PubMedCrossRef Hengelbrock J, Gillhaus J, Kloss S, Leverkus F. Safety data from randomized controlled trials: applying models for recurrent events. Pharm Stat. 2016;15(4):315–23.PubMedCrossRef
48.
go back to reference Lancar R, Kramar A, Haie-Meder C. Non-parametric methods for analysing recurrent complications of varying severity. Stat Med. 1995;14(24):2701–12.PubMedCrossRef Lancar R, Kramar A, Haie-Meder C. Non-parametric methods for analysing recurrent complications of varying severity. Stat Med. 1995;14(24):2701–12.PubMedCrossRef
49.
go back to reference Leon-Novelo LG, Zhou X, Bekele BN, Muller P. Assessing toxicities in a clinical trial: Bayesian inference for ordinal data nested within categories. Biometrics. 2010;66(3):966–74.PubMedPubMedCentralCrossRef Leon-Novelo LG, Zhou X, Bekele BN, Muller P. Assessing toxicities in a clinical trial: Bayesian inference for ordinal data nested within categories. Biometrics. 2010;66(3):966–74.PubMedPubMedCentralCrossRef
50.
go back to reference Liu GF, Wang J, Liu K, Snavely DB. Confidence intervals for an exposure adjusted incidence rate difference with applications to clinical trials. Stat Med. 2006;25(8):1275–86.PubMedCrossRef Liu GF, Wang J, Liu K, Snavely DB. Confidence intervals for an exposure adjusted incidence rate difference with applications to clinical trials. Stat Med. 2006;25(8):1275–86.PubMedCrossRef
51.
go back to reference Nishikawa M, Tango T, Ogawa M. Non-parametric inference of adverse events under informative censoring. Stat Med. 2006;25(23):3981–4003.PubMedCrossRef Nishikawa M, Tango T, Ogawa M. Non-parametric inference of adverse events under informative censoring. Stat Med. 2006;25(23):3981–4003.PubMedCrossRef
52.
go back to reference O'Gorman TW, Woolson RF, Jones MP. A comparison of two methods of estimating a common risk difference in a stratified analysis of a multicenter clinical trial. Control Clin Trials. 1994;15(2):135–53.PubMedCrossRef O'Gorman TW, Woolson RF, Jones MP. A comparison of two methods of estimating a common risk difference in a stratified analysis of a multicenter clinical trial. Control Clin Trials. 1994;15(2):135–53.PubMedCrossRef
53.
go back to reference Rosenkranz G. Analysis of adverse events in the presence of discontinuations. Drug Inform J. 2006;40(1):79–87.CrossRef Rosenkranz G. Analysis of adverse events in the presence of discontinuations. Drug Inform J. 2006;40(1):79–87.CrossRef
54.
go back to reference Sogliero-Gilbert G, Ting N, Zubkoff L. A statistical comparison of drug safety in controlled clinical trials: The Genie score as an objective measure of lab abnormalities. Ther Innov Regul Sci. 1991;25(1):81–96. Sogliero-Gilbert G, Ting N, Zubkoff L. A statistical comparison of drug safety in controlled clinical trials: The Genie score as an objective measure of lab abnormalities. Ther Innov Regul Sci. 1991;25(1):81–96.
55.
go back to reference Wang J, Quartey G. Nonparametric estimation for cumulative duration of adverse events. Biom J. 2012;54(1):61–74.PubMedCrossRef Wang J, Quartey G. Nonparametric estimation for cumulative duration of adverse events. Biom J. 2012;54(1):61–74.PubMedCrossRef
56.
go back to reference Wang J, Quartey G. A semi-parametric approach to analysis of event duration and prevalence. Comput Stat Data Anal. 2013;67:248–57.CrossRef Wang J, Quartey G. A semi-parametric approach to analysis of event duration and prevalence. Comput Stat Data Anal. 2013;67:248–57.CrossRef
57.
58.
go back to reference French JL, Thomas N, Wang C. Using historical data with Bayesian methods in early clinical trial monitoring. Stat Biopharm Res. 2012;4(4):384–94.CrossRef French JL, Thomas N, Wang C. Using historical data with Bayesian methods in early clinical trial monitoring. Stat Biopharm Res. 2012;4(4):384–94.CrossRef
60.
go back to reference Zhu L, Yao B, Xia HA, Jiang Q. Statistical monitoring of safety in clinical trials. Stat Biopharm Res. 2016;8(1):88–105.CrossRef Zhu L, Yao B, Xia HA, Jiang Q. Statistical monitoring of safety in clinical trials. Stat Biopharm Res. 2016;8(1):88–105.CrossRef
61.
go back to reference Berry SM, Berry DA. Accounting for multiplicities in assessing drug safety: a three-level hierarchical mixture model. Biometrics. 2004;60(2):418–26.PubMedCrossRef Berry SM, Berry DA. Accounting for multiplicities in assessing drug safety: a three-level hierarchical mixture model. Biometrics. 2004;60(2):418–26.PubMedCrossRef
62.
go back to reference Chen W, Zhao N, Qin G, Chen J. A bayesian group sequential approach to safety signal detection. J Biopharm Stat. 2013;23(1):213–30.PubMedCrossRef Chen W, Zhao N, Qin G, Chen J. A bayesian group sequential approach to safety signal detection. J Biopharm Stat. 2013;23(1):213–30.PubMedCrossRef
63.
go back to reference Gould AL. Detecting potential safety issues in clinical trials by Bayesian screening. Biom J. 2008;50(5):837–51.PubMedCrossRef Gould AL. Detecting potential safety issues in clinical trials by Bayesian screening. Biom J. 2008;50(5):837–51.PubMedCrossRef
64.
go back to reference Gould AL. Detecting potential safety issues in large clinical or observational trials by bayesian screening when event counts arise from poisson distributions. J Biopharm Stat. 2013;23(4):829–47.PubMedCrossRef Gould AL. Detecting potential safety issues in large clinical or observational trials by bayesian screening when event counts arise from poisson distributions. J Biopharm Stat. 2013;23(4):829–47.PubMedCrossRef
65.
go back to reference McEvoy BW, Nandy RR, Tiwari RC. Bayesian approach for clinical trial safety data using an Ising prior. Biometrics. 2013;69(3):661–72.PubMedCrossRef McEvoy BW, Nandy RR, Tiwari RC. Bayesian approach for clinical trial safety data using an Ising prior. Biometrics. 2013;69(3):661–72.PubMedCrossRef
66.
go back to reference Xia HA, Ma H, Carlin BP. Bayesian hierarchical modeling for detecting safety signals in clinical trials. J Biopharm Stat. 2011;21(5):1006–29.PubMedCrossRef Xia HA, Ma H, Carlin BP. Bayesian hierarchical modeling for detecting safety signals in clinical trials. J Biopharm Stat. 2011;21(5):1006–29.PubMedCrossRef
67.
go back to reference Whone A, Luz M, Boca M, Woolley M, Mooney L, Dharia S, Broadfoot J, Cronin D, Schroers C, Barua NU, et al. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain. 2019;142(3):512–25.PubMedPubMedCentralCrossRef Whone A, Luz M, Boca M, Woolley M, Mooney L, Dharia S, Broadfoot J, Cronin D, Schroers C, Barua NU, et al. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain. 2019;142(3):512–25.PubMedPubMedCentralCrossRef
68.
go back to reference Wang W, Whalen E, Munsaka M, Li J, Fries M, Kracht K, Sanchez-Kam M, Singh K, Zhou K. On quantitative methods for clinical safety monitoring in drug development. Stat Biopharm Res. 2018;10(2):85–97.CrossRef Wang W, Whalen E, Munsaka M, Li J, Fries M, Kracht K, Sanchez-Kam M, Singh K, Zhou K. On quantitative methods for clinical safety monitoring in drug development. Stat Biopharm Res. 2018;10(2):85–97.CrossRef
69.
go back to reference Phillips R, Cornelius V. Understanding current practice, identifying barriers and exploring priorities for adverse event analysis in randomised controlled trials: an online, cross-sectional survey of statisticians from academia and industry. BMJ Open. 2020;10(6):e036875.PubMedPubMedCentralCrossRef Phillips R, Cornelius V. Understanding current practice, identifying barriers and exploring priorities for adverse event analysis in randomised controlled trials: an online, cross-sectional survey of statisticians from academia and industry. BMJ Open. 2020;10(6):e036875.PubMedPubMedCentralCrossRef
70.
go back to reference Cornelius VR, Sauzet O, Williams JE, Ayis S, Farquhar-Smith P, Ross JR, Branford RA, Peacock JL. Adverse event reporting in randomised controlled trials of neuropathic pain: considerations for future practice. PAIN. 2013;154(2):213–20.PubMedCrossRef Cornelius VR, Sauzet O, Williams JE, Ayis S, Farquhar-Smith P, Ross JR, Branford RA, Peacock JL. Adverse event reporting in randomised controlled trials of neuropathic pain: considerations for future practice. PAIN. 2013;154(2):213–20.PubMedCrossRef
71.
go back to reference Hum SW, Golder S, Shaikh N. Inadequate harms reporting in randomized control trials of antibiotics for pediatric acute otitis media: a systematic review. Drug Saf. 2018;41(10):933–8.PubMedCrossRef Hum SW, Golder S, Shaikh N. Inadequate harms reporting in randomized control trials of antibiotics for pediatric acute otitis media: a systematic review. Drug Saf. 2018;41(10):933–8.PubMedCrossRef
72.
go back to reference Patson N, Mukaka M, Otwombe KN, Kazembe L, Mathanga DP, Mwapasa V, Kabaghe AN, Eijkemans MJC, Laufer MK, Chirwa T. Systematic review of statistical methods for safety data in malaria chemoprevention in pregnancy trials. Malar J. 2020;19(1):119.PubMedPubMedCentralCrossRef Patson N, Mukaka M, Otwombe KN, Kazembe L, Mathanga DP, Mwapasa V, Kabaghe AN, Eijkemans MJC, Laufer MK, Chirwa T. Systematic review of statistical methods for safety data in malaria chemoprevention in pregnancy trials. Malar J. 2020;19(1):119.PubMedPubMedCentralCrossRef
75.
go back to reference Spiegelhalter DJ, Abrams KR, Myles JP. An Overview of the Bayesian Approach. In: Bayesian Approaches to Clinical Trials and Health-Care Evaluation: John Wiley & Sons, Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK: Wiley; 2004. Spiegelhalter DJ, Abrams KR, Myles JP. An Overview of the Bayesian Approach. In: Bayesian Approaches to Clinical Trials and Health-Care Evaluation: John Wiley & Sons, Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK: Wiley; 2004.
76.
go back to reference Gelman A, Hill J, Yajima M. Why We (Usually) Don't Have to Worry About Multiple Comparisons. J Res Educ Effect. 2012;5(2):189–211. Gelman A, Hill J, Yajima M. Why We (Usually) Don't Have to Worry About Multiple Comparisons. J Res Educ Effect. 2012;5(2):189–211.
77.
go back to reference Spiegelhalter DJ. Incorporating Bayesian ideas into health-care evaluation. Stat Sci. 2004;19(1):156–74.CrossRef Spiegelhalter DJ. Incorporating Bayesian ideas into health-care evaluation. Stat Sci. 2004;19(1):156–74.CrossRef
78.
go back to reference Ball G, Piller LB. Continuous safety monitoring for randomized controlled clinical trials with blinded treatment information part 2: statistical considerations. Contemp Clin Trials. 2011;32:S5–7.PubMedCrossRef Ball G, Piller LB. Continuous safety monitoring for randomized controlled clinical trials with blinded treatment information part 2: statistical considerations. Contemp Clin Trials. 2011;32:S5–7.PubMedCrossRef
79.
go back to reference Gould AL, Wang WB. Monitoring potential adverse event rate differences using data from blinded trials: the canary in the coal mine. Stat Med. 2017;36(1):92–104.PubMedCrossRef Gould AL, Wang WB. Monitoring potential adverse event rate differences using data from blinded trials: the canary in the coal mine. Stat Med. 2017;36(1):92–104.PubMedCrossRef
Metadata
Title
Statistical methods for the analysis of adverse event data in randomised controlled trials: a scoping review and taxonomy
Authors
Rachel Phillips
Odile Sauzet
Victoria Cornelius
Publication date
01-12-2020
Publisher
BioMed Central
Published in
BMC Medical Research Methodology / Issue 1/2020
Electronic ISSN: 1471-2288
DOI
https://doi.org/10.1186/s12874-020-01167-9

Other articles of this Issue 1/2020

BMC Medical Research Methodology 1/2020 Go to the issue