Skip to main content
Top
Published in: Critical Care 1/2020

01-12-2020 | Research

Identification, collection, and reporting of harms among non-industry-sponsored randomized clinical trials of pharmacologic interventions in the critically ill population: a systematic review

Authors: Ari Moskowitz, Lars W. Andersen, Mathias J. Holmberg, Anne V. Grossestreuer, Katherine M. Berg, Asger Granfeldt

Published in: Critical Care | Issue 1/2020

Login to get access

Abstract

Background

Prescribing pharmacologic therapies for critically ill patients requires a careful balancing of risks and benefits. Defining, monitoring, and reporting harms that occur in clinical trials conducted in critically ill populations, however, is challenging given that the natural history of most critical illnesses includes progressive multiple organ failure and death. In this study, we assessed harms reporting in clinical trials performed in critically ill populations.

Methods

Randomized, non-industry-sponsored, human clinical trials of pharmacologic interventions in adult critically ill populations published between 2015 and 2018 in high-impact journals were included in this systematic review. Harms data, adherence to Consolidated Standards of Reporting Trials (CONSORT) harms reporting guidelines, and restrictions on harms reporting were recorded.

Results

A total of 707 abstracts were screened with 40 trials ultimately being included in the analysis. Included trials represent 28,636 randomized patients with a median of 292 (IQR 100–546) patients per trial. The most common disease states were general critical care (33%) and sepsis (28%). Of 18 included CONSORT items, the median number met was 12 (IQR 9, 14). The most commonly missed items were adverse event (AE) severity grading definitions and AE attribution (relationship of AE to study drug), which were only reported in 35 and 38% of manuscripts, respectively. Half of the manuscripts (48%) provided definitions for recorded AEs. There were 5 studies investigating the effects of corticosteroids in sepsis, with the number of AEs reported per analyzed patient ranging from 0.01 to 1.89. AE definitions in studies of similar/equivalent interventions often varied substantially. Study protocols were available for 30/40 (75%) of studies, with 13 (43%) of those not providing any guidance regarding AE attribution.

Conclusions

Randomized trials of pharmacologic interventions conducted in critically ill populations and published in high impact journals often fail to adequately describe AE definitions, severity, attribution, and collection procedures. Among trials of similar interventions in comparable populations, variation in AE collection and reporting procedures is substantial. These factors may limit a clinician’s ability to accurately balance the potential benefits and harms of an intervention.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ioannidis JPA. Adverse events in randomized trials. Arch Intern Med. 2009;169(19):1737.CrossRef Ioannidis JPA. Adverse events in randomized trials. Arch Intern Med. 2009;169(19):1737.CrossRef
2.
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.CrossRef 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.CrossRef
3.
go back to reference Cook D, Lauzier F, Rocha MG, Sayles MJ, Finfer S. Serious adverse events in academic critical care research. CMAJ. 2008;178(9):1181–4.CrossRef Cook D, Lauzier F, Rocha MG, Sayles MJ, Finfer S. Serious adverse events in academic critical care research. CMAJ. 2008;178(9):1181–4.CrossRef
4.
go back to reference Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. 2009;339:b2535.CrossRef Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. 2009;339:b2535.CrossRef
5.
go back to reference Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer. 2019;145(3):639–648. Arnaud-Coffin P, Maillet D, Gan HK, et al. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int J Cancer. 2019;145(3):639–648.
6.
go back to reference Shukralla AA, Tudur-Smith C, Powell GA, Williamson PR, Marson AG. Reporting of adverse events in randomised controlled trials of antiepileptic drugs using the CONSORT criteria for reporting harms. Epilepsy Res. 2011;97(1–2):20–9.CrossRef Shukralla AA, Tudur-Smith C, Powell GA, Williamson PR, Marson AG. Reporting of adverse events in randomised controlled trials of antiepileptic drugs using the CONSORT criteria for reporting harms. Epilepsy Res. 2011;97(1–2):20–9.CrossRef
7.
go back to reference Smith SM, Chang RD, Pereira A, et al. Adherence to CONSORT harms-reporting recommendations in publications of recent analgesic clinical trials: an ACTTION systematic review. Pain. 2012;153(12):2415–21.CrossRef Smith SM, Chang RD, Pereira A, et al. Adherence to CONSORT harms-reporting recommendations in publications of recent analgesic clinical trials: an ACTTION systematic review. Pain. 2012;153(12):2415–21.CrossRef
8.
go back to reference Ioannidis JPA, Evans SJW, 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.CrossRef Ioannidis JPA, Evans SJW, 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.CrossRef
9.
go back to reference Recommendations to improve adverse event reporting in clinical trial publications: a joint pharmaceutical industry/journal editor perspective. BMJ. 2017;356:j1228. Recommendations to improve adverse event reporting in clinical trial publications: a joint pharmaceutical industry/journal editor perspective. BMJ. 2017;356:j1228.
10.
go back to reference Bagul NB, Kirkham JJ. The reporting of harms in randomized controlled trials of hypertension using the CONSORT criteria for harm reporting. Clin Exp Hypertens. 2012;34(8):548–54.CrossRef Bagul NB, Kirkham JJ. The reporting of harms in randomized controlled trials of hypertension using the CONSORT criteria for harm reporting. Clin Exp Hypertens. 2012;34(8):548–54.CrossRef
11.
go back to reference Breau RH, Gaboury I, Scales CD Jr, Fesperman SF, Watterson JD, Dahm P. Reporting of harm in randomized controlled trials published in the urological literature. J Urol. 2010;183(5):1693–7.CrossRef Breau RH, Gaboury I, Scales CD Jr, Fesperman SF, Watterson JD, Dahm P. Reporting of harm in randomized controlled trials published in the urological literature. J Urol. 2010;183(5):1693–7.CrossRef
12.
go back to reference Capili B, Anastasi JK, Geiger JN. Adverse event reporting in acupuncture clinical trials focusing on pain. Clin J Pain. 2010;26(1):43–8.CrossRef Capili B, Anastasi JK, Geiger JN. Adverse event reporting in acupuncture clinical trials focusing on pain. Clin J Pain. 2010;26(1):43–8.CrossRef
13.
go back to reference Pitrou I, Boutron I, Ahmad N, Ravaud P. Reporting of safety results in published reports of randomized controlled trials. Arch Intern Med. 2009;169(19):1756–61.CrossRef Pitrou I, Boutron I, Ahmad N, Ravaud P. Reporting of safety results in published reports of randomized controlled trials. Arch Intern Med. 2009;169(19):1756–61.CrossRef
14.
go back to reference Haidich AB, Birtsou C, Dardavessis T, Tirodimos I, Arvanitidou M. The quality of safety reporting in trials is still suboptimal: survey of major general medical journals. J Clin Epidemiol. 2011;64(2):124–35.CrossRef Haidich AB, Birtsou C, Dardavessis T, Tirodimos I, Arvanitidou M. The quality of safety reporting in trials is still suboptimal: survey of major general medical journals. J Clin Epidemiol. 2011;64(2):124–35.CrossRef
15.
go back to reference Hodkinson A, Kirkham JJ, Tudur-Smith C, Gamble C. Reporting of harms data in RCTs: a systematic review of empirical assessments against the CONSORT harms extension. BMJ Open. 2013;3(9):e003436.CrossRef Hodkinson A, Kirkham JJ, Tudur-Smith C, Gamble C. Reporting of harms data in RCTs: a systematic review of empirical assessments against the CONSORT harms extension. BMJ Open. 2013;3(9):e003436.CrossRef
17.
go back to reference Venkatesh B, Finfer S, Cohen J, et al. Adjunctive glucocorticoid therapy in patients with septic shock. N Engl J Med. 2018;378(9):797–808.CrossRef Venkatesh B, Finfer S, Cohen J, et al. Adjunctive glucocorticoid therapy in patients with septic shock. N Engl J Med. 2018;378(9):797–808.CrossRef
18.
go back to reference Annane D, Renault A, Brun-Buisson C, et al. Hydrocortisone plus fludrocortisone for adults with septic shock. N Engl J Med. 2018;378(9):809–18.CrossRef Annane D, Renault A, Brun-Buisson C, et al. Hydrocortisone plus fludrocortisone for adults with septic shock. N Engl J Med. 2018;378(9):809–18.CrossRef
19.
go back to reference Sivendran S, Galsky MD. Adverse event reporting in oncology clinical trials - lost in translation? Expert Opin Drug Saf. 2016;15(7):893–6.CrossRef Sivendran S, Galsky MD. Adverse event reporting in oncology clinical trials - lost in translation? Expert Opin Drug Saf. 2016;15(7):893–6.CrossRef
20.
go back to reference Belknap SM, Georgopoulos CH, Lagman J, et al. Reporting of serious adverse events during cancer clinical trials to the institutional review board: an evaluation by the research on adverse drug events and reports (RADAR) project. J Clin Pharmacol. 2013;53(12):1334–40.CrossRef Belknap SM, Georgopoulos CH, Lagman J, et al. Reporting of serious adverse events during cancer clinical trials to the institutional review board: an evaluation by the research on adverse drug events and reports (RADAR) project. J Clin Pharmacol. 2013;53(12):1334–40.CrossRef
21.
go back to reference Naranjo CA, Busto U, Sellers EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther. 1981;30(2):239–45.CrossRef Naranjo CA, Busto U, Sellers EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther. 1981;30(2):239–45.CrossRef
Metadata
Title
Identification, collection, and reporting of harms among non-industry-sponsored randomized clinical trials of pharmacologic interventions in the critically ill population: a systematic review
Authors
Ari Moskowitz
Lars W. Andersen
Mathias J. Holmberg
Anne V. Grossestreuer
Katherine M. Berg
Asger Granfeldt
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Critical Care / Issue 1/2020
Electronic ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03113-z

Other articles of this Issue 1/2020

Critical Care 1/2020 Go to the issue