2046-4053-2-79 2046-4053 Commentary <p>The evolution of assessing bias in Cochrane systematic reviews of interventions: celebrating methodological contributions of the Cochrane Collaboration</p> TurnerLucylturner@ohri.ca BoutronIsabelleisabelle.boutron@bch.aphp.fr HróbjartssonAsbjørnah@cochrane.dk AltmanGDouglasDoug.Altman@csm.ox.ac.uk MoherDaviddmoher@ohri.ca

Ottawa Hospital Research Institute, Ottawa, Canada

INSERM, U738, Paris, France

Hôpital Hôtel Dieu, Centre, d’Epidémiologie Clinique, Paris, France

Faculté de Médecine, University of Paris Descartes, Sorbonne, Paris Cité, Paris, France

French Cochrane Centre, Paris, France

Nordic Cochrane Centre, Rigshospitalet, Copenhagen, Denmark

Centre for Statistics in Medicine, University of Oxford, Oxford, UK

Department of Epidemiology and Community Medicine, University of Ottawa, Ottawa, Canada

Systematic Reviews 2046-4053 2013 2 1 79 http://www.systematicreviewsjournal.com/content/2/1/79 “…to manage large quantities of data objectively and effectively, standardized methods of appraising information should be included in review processes. … By using these systematic methods of exploration, evaluation, and synthesis, the good reviewer can accomplish the task of advancing scientific knowledge”. Cindy Mulrow, 1986, BMJ. 10.1186/2046-4053-2-7924059942
15201311920132392013 2013Turner et al.; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Cochrane Methodology <em>Anniversary Series</em>Mike Clarke

Background

The global evidence base for health care is extensive, and expanding; with nearly 2 million articles published annually. One estimate suggests 75 trials and 11 systematic reviews are published daily 1 . Research syntheses, in a variety of established and emerging forms, are well recognised as essential tools for summarising evidence with accuracy and reliability 2 . Systematic reviews provide health care practitioners, patients and policy makers with information to help make informed decisions. It is essential that those conducting systematic reviews are cognisant of the potential biases within primary studies and of how such biases could impact review results and subsequent conclusions.

Rigorous and systematic methodological approaches to conducting research synthesis emerged throughout the twentieth century with methods to identify and reduce biases evolving more recently 3 4 . The Cochrane Collaboration has made substantial contributions to the development of how biases are considered in systematic reviews and primary studies. Our objective within this paper is to review some of the landmark methodological contributions by members of the Cochrane Bias Methods Group (BMG) to the body of evidence which guides current bias assessment practices, and to outline the immediate and horizon objectives for future research initiatives.

Empirical works published prior to the establishment of the Cochrane Collaboration

In 1948, the British Medical Research Council published results of what many consider the first 'modern’ randomised trial 5 6 . Subsequently, the last 65 years has seen continual development of the methods used when conducting primary medical research aiming to reduce inaccuracy in estimates of treatment effects due to potential biases. A large body of literature has accumulated which supports how study characteristics, study reports and publication processes can potentially bias primary study and systematic review results. Much of the methodological research during the first 20 years of The Cochrane Collaboration has built upon that published before the Collaboration was founded. Reporting biases, or more specifically, publication bias and the influence of funding source(s) are not new concepts. Publication bias initially described as the file drawer problem as a bias concept in primary studies was early to emerge and has long been suspected in the social sciences 7 . In 1979 Rosenthal, a psychologist, described the issue in more detail 8 and throughout the 1980s and early 1990s an empirical evidence base began to appear in the medical literature 9 10 11 . Concurrent with the accumulation of early evidence, methods to detect and mitigate the presence of publication bias also emerged 12 13 14 15 . The 1980s also saw initial evidence of the presence of what is now referred to as selective outcome reporting 16 and research investigating the influence of source of funding on study results 10 11 17 18 .

The importance of rigorous aspects of trial design (e.g. randomisation, blinding, attrition, treatment compliance) were known in the early 1980s 19 and informed the development by Thomas Chalmers and colleagues of a quality assessment scale to evaluate the design, implementation, and analysis of randomized control trials 20 . The pre-Cochrane era saw the early stages of assessing quality of included studies, with consideration of the most appropriate ways to assess bias. Yet, no standardised means for assessing risk of bias, or “quality” as it was referred to at the time, were implemented when The Cochrane Collaboration was established. The use of scales for assessing quality or risk of bias is currently explicitly discouraged in Cochrane reviews based on more recent evidence 21 22 .

Methodological contributions of the Cochrane Collaboration: 1993 – 2013

In 1996, Moher and colleagues suggested that bias assessment was a new, emerging and important concept and that more evidence was required to identify trial characteristics directly related to bias 23 . Methodological literature pertaining to bias in primary studies published in the last 20 years has contributed to the evolution of bias assessment in Cochrane reviews. How bias is currently assessed has been founded on published studies that provide empirical evidence of the influence of certain study design characteristics on estimates of effect, predominately considering randomised controlled trials.

The publication of Ken Schulz’s work on allocation concealment, sequence generation, and blinding 24 25 the mid-1990s saw a change in the way the Collaboration assessed bias of included studies, and it was recommended that included studies were assessed in relation to how well the generated random sequence was concealed during the trial.

In 2001, the Cochrane Reporting Bias Methods Group now known as the Cochrane Bias Methods Group, was established to investigate how reporting and other biases influence the results of primary studies. The most substantial development in bias assessment practice within the Collaboration was the introduction of the Cochrane Risk of Bias (RoB) Tool in 2008. The tool was developed based on the methodological contributions of meta-epidemiological studies 26 27 and has since been evaluated and updated 28 , and integrated into Grading of Recommendations Assessment, Development and Evaluation (GRADE) 29 .

Throughout this paper we define bias as a systematic error or deviation in results or inferences from the truth 30 and should not be confused with “quality”, or how well a trial was conducted. The distinction between internal and external validity is important to review. When we describe bias we are referring to internal validity as opposed to the external validity or generalizability which is subject to demographic or other characteristics 31 . Here, we highlight landmark methodological publications which contribute to understanding how bias influences estimates of effects in Cochrane reviews (Figure  1).

<p>Figure 1</p>

Timeline of landmark methods research 8131617202226313233343536373839404142434445

Timeline of landmark methods research 8 13 16 17 20 22 26 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 .

Sequence generation and allocation concealment

Early meta-epidemiological studies assessed the impact of inadequate allocation concealment and sequence generation on estimates of effect 24 25 . Evidence suggests that adequate or inadequate allocation concealment modifies estimates of effect in trials 31 . More recently, several other methodological studies have examined whether concealment of allocation is associated with magnitude of effect estimates in controlled clinical trials while avoiding confounding by disease or intervention 42 46 .

More recent methodological studies have assessed the importance of proper generation of a random sequence in randomised clinical trials. It is now mandatory, in accordance with the Methodological Expectations for Cochrane Interventions Reviews (MECIR) conduct standards, for all Cochrane systematic reviews to assess potential selection bias (sequence generation and allocation concealment) within included primary studies.

Blinding of participants, personnel and outcome assessment

The concept of the placebo effect has been considered since the mid-1950s 47 and the importance of blinding trial interventions to participants has been well known, with the first empirical evidence published in the early 1980s 48 . The body of empirical evidence on the influence on blinding has grown since the mid-1990s, especially in the last decade, with some evidence highlighting that blinding is important for several reasons 49 . Currently, the Cochrane risk of bias tool suggests blinding of participants and personnel, and blinding of outcome assessment be assessed septely. Moreover consideration should be given to the type of outcome (i.e. objective or subjective outcome) when assessing bias, as evidence suggests that subjective outcomes are more prone to bias due to lack of blinding 42 44 As yet there is no empirical evidence of bias due to lack of blinding of participants and study personnel. However, there is evidence for studies described as 'blind’ or 'double-blind’, which usually includes blinding of one or both of these groups of people. In empirical studies, lack of blinding in randomized trials has been shown to be associated with more exaggerated estimated intervention effects 42 46 50 .

Different people can be blinded in a clinical trial 51 52 . Study reports often describe blinding in broad terms, such as 'double blind’. This term makes it impossible to know who was blinded 53 . Such terms are also used very inconsistently 52 54 55 and the frequency of explicit reporting of the blinding status of study participants and personnel remains low even in trials published in top journals 56 , despite explicit recommendations. Blinding of the outcome assessor is particularly important, both because the mechanism of bias is simple and foreseeable, and because evidence for bias is unusually clear 57 . A review of methods used for blinding highlights the variety of methods used in practice 58 . More research is ongoing within the Collaboration to consider the best way to consider the influence of lack of blinding within primary studies. Similar to selection bias, performance and detection bias are both mandatory components of risk of bias assessment in accordance with the MECIR standards.

Reporting biases

Reporting biases have long been identified as potentially influencing the results of systematic reviews. Bias arises when the dissemination of research findings is influenced by the nature and direction of results, there is still debate over explicit criteria for what constitutes a 'reporting bias’. More recently, biases arising from non-process related issues (i.e. source of funding, publication bias) have been referred to as meta-biases 59 . Here we discuss the literature which has emerged in the last twenty years with regards to two well established reporting biases, non-publication of whole studies (often simply called publication bias) and selective outcome reporting.

Publication bias

The last two decades have seen a large body of evidence of the presence of publication bias 60 61 62 63 and why authors fail to publish 64 65 . Given that it has long been recognized that investigators frequently fail to report their research findings 66 , many more recent papers have been geared towards methods of detecting and estimating the effect of publication bias. An array of methods to test for publication bias and additional recommendations are now available 38 43 67 68 69 70 71 72 73 74 75 76 , many of which have been evaluated 77 78 79 80 . Automatic generation of funnel plots have been incorporated when producing a Cochrane review and software (RevMan) and are encouraged for outcomes with more than ten studies 43 .A thorough overview of methods is included in Chapter 10 of the Cochrane Handbook for Systematic Reviews of Interventions 81 .

Selective outcome reporting

While the concept of publication bias has been well established, studies reporting evidence of the existence of selective reporting of outcomes in trial reports have appeared more recently 39 41 82 83 84 85 86 87 . In addition, some studies have investigated why some outcomes are omitted from published reports 41 88 89 90 as well as the impact of omission of outcomes on the findings of meta-analyses 91 . More recently, methods for evaluating selective reporting, namely, the ORBIT (Outcome Reporting Bias in Trials) classification system have been developed. One attempt to mitigate selective reporting is to develop field specific core outcome measures 92 the work of COMET (Core Outcome Measures in Effectiveness Trials) initiative 93 is supported by many members within the Cochrane Collaboration. More research is being conducted with regards to selective reporting of outcomes and selective reporting of trial analyses, within this concept there is much overlap with the movement to improve primary study reports, protocol development and trial registration.

Evidence on how to conduct risk of bias assessments

Often overlooked are the processes behind how systematic evaluations or assessments are conducted. In addition to empirical evidence of specific sources of bias, other methodological studies have led to changes in the processes used to assess risk of bias. One influential study published in 1999 highlighted the hazards of scoring 'quality’ of clinical trials when conducting meta-analysis and is one of reasons why each bias is assessed septely as 'high’, 'low’ or 'unclear’ risk rather than using a combined score 22 94 . Prior work investigated blinding of readers, data analysts and manuscript writers 51 95 . More recently, work has been completed to assess blinding of authorship and institutions in primary studies when conducting risk of bias assessments, suggesting that there is discordance in results between blind and unblinded RoB assessments. However uncertainty over best practice remains due to time and resources needed to implement blinding 96 .

Complementary contributions

Quality of reporting and reporting guidelines

Assessing primary studies for potential biases is a challenge 97 . During the early 1990s, poor reporting in randomized trials and consequent impediments to systematic review conduct, especially when conducting what is now referred to as 'risk of bias assessment’, were observed. In 1996, an international group of epidemiologists, statisticians, clinical trialists, and medical editors, some of whom were involved with establishing the Cochrane Collaboration, published the CONSORT Statement 32 , a checklist of items to be addressed in a report of the findings of an RCT. CONSORT has twice been revised and updated 35 36 and over time, the impact of CONSORT has been noted, for example, CONSORT was considered one of the major milestones in health research methods over the last century by the Patient-Centered Outcomes Research Institute (PCORI) 98 .

Issues of poor reporting extend far beyond randomized trials, and many groups have developed guidance to aid reporting of other study types. The EQUATOR Network’s library for health research reporting includes more than 200 reporting guidelines 99 . Despite evidence that the quality of reporting has improved over time, systemic issues with the clarity and transparency of reporting remain 100 101 . Such inadequacies in primary study reporting result in systematic review authors’ inability to assess the presence and extent of bias in primary studies and the possible impact on review results, continued improvements in trial reporting are needed to lead to more informed risk of bias assessments in systematic reviews.

Trial registration

During the 1980s and 1990s there were several calls to mitigate publication bias and selective reporting via trial registration 102 103 104 . After some resistance, in 2004, the BMJ and The Lancet reported that they would only publish registered clinical trials 105 with the International Committee of Medical Journal Editors making a statement to the same effect 40 . Despite the substantial impact of trial registration 106 uptake is still not optimal and it is not mandatory for all trials. A recent report indicated that only 22% of trials mandated by the FDA were reporting trial results on clinicaltrials.gov 107 . One study suggested that despite trial registration being strongly encouraged and even mandated in some jurisdictions only 45.5% of a sample of 323 trials were adequately registered 108 .

Looking forward

Currently, there are three major ongoing initiatives which will contribute to how The Collaboration assesses bias. First, there has been some criticism of the Cochrane risk of bias tool 109 concerning its ease of use and reliability 110 111 and the tool is currently being revised. As a result, a working group is established to improve the format of the tool, with version 2.0 due to be released in 2014. Second, issues of study design arise when assessing risk of bias when including non-randomised studies in systematic reviews 112 113 114 . Even 10 years ago there were 114 published tools for assessing risk of bias in non-randomised studies 115 . An ongoing Cochrane Methods Innovation Fund project will lead to the release a tool for assessing non-randomized studies as well as tools for cluster and cross-over trials 116 . Third, selective reporting in primary studies is systemic 117 yet further investigation and adoption of sophisticated means of assessment remain somewhat unexplored by the Collaboration. A current initiative is ongoing to explore optimal ways to assess selective reporting within trials. Findings of this initiative will be considered in conjunction with the release of revised RoB tool and its extension for non-randomized studies.

More immediate issues

Given the increase in meta-epidemiological research, an explicit definition of evidence needed to identify study characteristics which may lead to bias (es) needs to be defined. One long debated issue is the influence of funders as a potential source of bias. In one empirical study, more than half of the protocols for industry-initiated trials stated that the sponsor either owns the data or needs to approve the manuscript, or both; none of these constraints were stated in any of the trial publications 118 . It is important that information about vested interests is collected and presented when relevant 119 .

There is an on-going debate related to the risk of bias of trials stopping early because of benefit. A systematic review and a meta-epidemiologic study showed that such truncated RCTs were associated with greater effect sizes than RCTs not stopped early, particularly for trials with small sample size 120 121 . These results were widely debated and discussed 122 and recommendations related to this item are being considered.

In addition, recent meta-epidemiological studies of binary and continuous outcomes showed that treatment effect estimates in single-centre RCTs were significantly larger than in multicenter RCTs even after controlling for sample size 123 124 . The Bias in Randomized and Observational Studies (BRANDO) project combining data from all available meta-epidemiologic studies 44 found consistent results for subjective outcomes when comparing results from single centre and multi-centre trials. Several reasons may explain these differences between study results: small study effect, reporting bias, higher risk of bias in single centre studies, or factors related to the selection of the participants, treatment administration and care providers’ expertise. Further studies are needed to explore the role and effect of these different mechanisms.

Longer term issues

The scope of methodological research and subsequent contributions and evolution in bias assessment over the last 20 years has been substantial. However, there remains much work to be done, particularly in line with innovations in systematic review methodology itself. There is no standardised methodological approach to the conduct of systematic reviews. Subject to a given clinical question, it may be most appropriate to conduct a network meta-analysis, scoping review, a rapid review, or update any of these reviews. Along with the development of these differing types of review, there is the need for bias assessment methods to develop concurrently.

The way in which research synthesis is conducted may change further with technological advances 125 . Globally, there are numerous initiatives to establish integrated administrative databases which may open up new research avenues and methodological questions about assessing bias when primary study results are housed within such databases.

Despite the increase in meta-epidemiological research identifying study characteristics which could contribute to bias in studies, further investigation is needed. For example, as yet there has been little research on integration of risk of bias results into review findings. This is done infrequently and guidance on how to do it could be improved 126 . Concurrently, although some work has been done, little is known about how magnitude and direction in estimates of effect for a given bias and across biases for a particular trial and in turn, set of trials 127 .

Conclusion

To summarise, there has been much research conducted to develop understanding of bias in trials and how these biases could influence the results of systematic reviews. Much of this work has been conducted since the Cochrane Collaboration was established either as a direct initiative of the Collaboration or thanks to the work of many affiliated individuals. There has been clear advancement in mandatory processes for assessing bias in Cochrane reviews. These processes, based on a growing body of empirical evidence have aimed to improve the overall quality of the systematic review literature, however, many areas of bias remain unexplored and as the evidence evolves, the processes used to assess and interpret biases and review results will also need to adapt.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

This paper was invited for submission by the Cochrane Bias Methods Group. AH and IB conceived of the initial outline of the manuscript, LT collected information and references and drafted the manuscript, DM reviewed the manuscript and provided guidance on structure, DM, DGA, IB and AH all provided feedback on the manuscript and suggestions of additional literature. All authors read and approved the final manuscript.

Acknowledgements

We would like to thank the Canadian Institutes of Health Research for their long-term financial support (2005–2015)-CIHR Funding Reference Number—CON-105529 this financing has enabled much of the progress of the group in the last decade. We would also like to thank Jodi Peters for help preparing the manuscript, Jackie Chandler for coordinating this paper as one of the series. We would also like to thank the Bias Methods Group membership for their interest and involvement with the group. We would especially like to thank Matthias Egger and Jonathan Sterne, previous BMG convenors, and Julian Higgins, Jennifer Tetzlaff and Laura Weeks for their extensive contributions to the group.

Cochrane awards received by BMG members

The Cochrane BMG currently has a membership of over 200 statisticians, clinicians, epidemiologists and researchers interested in issues of bias in systematic reviews. For a full list of BMG members, please login to Archie or visit http://www.bmg.cochrane.org for contact information.

Bill Silverman prize recipients

2009 - Moher D, Tetzlaff J, Tricco AC, Sampson M, Altman DG. Epidemiology and reporting characteristics of systematic reviews. PLoS Medicine 2007 4(3): e78. doi: http://dx.doi.org/10.1371/journal.pmed.0040078 [full-text PDF].

Thomas C. Chalmers award recipients

2001 (tie) - Henry D, Moxey A, O’Connell D. Agreement between randomised and non-randomised studies - the effects of bias and confounding [abstract]. Proceedings of the Ninth Cochrane Colloquium, 2001.

2001 (runner-up) - Full publication: Sterne JAC, Jüni P, Schulz KF, Altman DG, Bartlett C, and Egger M. Statistical methods for assessing the influence of study characteristics on treatment effects in “meta-epidemiological” research. Stat Med 2002;21:1513–1524.

2010 - Kirkham JJ, Riley R, Williamson P. Is multivariate meta-analysis a solution for reducing the impact of outcome reporting bias in systematic reviews? [abstract] Proceedings of the Eighteenth Cochrane Colloquium, 2010.

2012 - Page MJ, McKenzie JE, Green SE, Forbes A. Types of selective inclusion and reporting bias in randomised trials and systematic reviews of randomised trials [presentation]. Proceedings of the Twentieth Cochrane Colloquium, 2012.

<p>Seventy-five trials and eleven systematic reviews a day: how will we ever keep up?</p>BastianHGlasziouPChalmersIPLoS Med20107e100032610.1371/journal.pmed.1000326294343920877712<p>The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration</p>LiberatiAAltmanDGTetzlaffJMulrowCGotzschePCIoannidisJPJ Clin Epidemiol200962e1e3410.1016/j.jclinepi.2009.06.00619631507<p>Commentary</p>HedgesLVStatist Med1987638138510.1002/sim.4780060333<p>A brief history of research synthesis</p>ChalmersIHedgesLVCooperHEval Health Prof200225123710.1177/016327870202500100311868442<p>STREPTOMYCIN treatment of pulmonary tuberculosis</p>Medical Research CouncilBr Med J19482769782209187218890300<p>Suspended judgment. Memories of the British streptomycin trial in tuberculosis. The first randomized clinical trial</p>HillABControl Clin Trials199011777910.1016/0197-2456(90)90001-I2161313<p>Publication decisions and their possible effects on inferences drawn from tests of significance - or vice versa</p>SterlingTDJ Am Stat Assoc1959543034<p>The file drawer problem and tolerance for null results</p>RosenthalRPsycholog Bull197986638641<p>Publication bias and clinical trials</p>DickersinKChanSChalmersTCSacksHSSmithHJrControl Clin Trials1987834335310.1016/0197-2456(87)90155-33442991<p>Publication bias in clinical research</p>EasterbrookPJBerlinJAGopalanRMatthewsDRLancet199133786787210.1016/0140-6736(91)90201-Y1672966<p>Factors influencing publication of research results. Follow-up of applications submitted to two institutional review boards</p>DickersinKMinYIMeinertCLJAMA199226737437810.1001/jama.1992.034800300520361727960<p>Analyzing nonlinear scatchard plots</p>LightKEScience1984223767810.1126/science.65463236546323<p>Publication bias: a problem in interpreting medical data</p>BeggCBBerlinJAJ Royal Stat Soc Series A (Stat Soc)198815141946310.2307/2982993<p>An approach for assessing publication bias prior to performing a meta-analysis</p>DearKBGBeggCBStat Sci1992723724510.1214/ss/1177011363<p>Modeling publication selection effects in meta-analysis</p>HedgesLVStat Sci1992724625510.1214/ss/1177011364<p>Statistical problems in the reporting of clinical trials. A survey of three medical journals</p>PocockSJHughesMDLeeRJN Engl J Med198731742643210.1056/NEJM1987081331707063614286<p>Study of information submitted by drug companies to licensing authorities</p>HemminkiEBr Med J198028083383610.1136/bmj.280.6217.83316010117370687<p>Multiple publication of reports of drug trials</p>GotzschePCEur J Clin Pharmacol19893642943210.1007/BF005580642666138<p>Scientific challenges in the application of randomized trials</p>KramerMSShapiroSHJAMA19842522739274510.1001/jama.1984.033501900410176492351<p>A method for assessing the quality of a randomized control trial</p>ChalmersTCSmithHJrBlackburnBSilvermanBSchroederBReitmanDControl Clin Trials19812314910.1016/0197-2456(81)90056-87261638<p>An empirical study of the possible relation of treatment differences to quality scores in controlled randomized clinical trials</p>EmersonJDBurdickEHoaglinDCMostellerFChalmersTCControl Clin Trials19901133935210.1016/0197-2456(90)90175-21963128<p>The hazards of scoring the quality of clinical trials for meta-analysis</p>JuniPWitschiABlochREggerMJAMA19992821054106010.1001/jama.282.11.105410493204<p>Assessing the quality of randomized controlled trials. Current issues and future directions</p>MoherDJadadARTugwellPInt J Technol Assess Health Care19961219520810.1017/S02664623000095708707495<p>Subverting randomization in controlled trials</p>SchulzKFJAMA19952741456145810.1001/jama.1995.035301800500297474192<p>Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials</p>SchulzKFChalmersIHayesRJAltmanDGJAMA199527340841210.1001/jama.1995.035202900600307823387<p>Meta-analysis and the meta-epidemiology of clinical research</p>NaylorCDBMJ199731561761910.1136/bmj.315.7109.61721274359310553<p>Statistical methods for assessing the influence of study characteristics on treatment effects in 'meta-epidemiological’ research</p>SterneJAJuniPSchulzKFAltmanDGBartlettCEggerMStat Med2002211513152410.1002/sim.118412111917<p>The cochrane collaboration’s tool for assessing risk of bias in randomised trials</p>HigginsJPAltmanDGGotzschePCJuniPMoherDOxmanADBMJ2011343d592810.1136/bmj.d5928319624522008217<p>GRADE: an emerging consensus on rating quality of evidence and strength of recommendations</p>GuyattGHOxmanADVistGEKunzRFalck-YtterYAlonso-CoelloPBMJ200833692492610.1136/bmj.39489.470347.AD233526118436948GreenSHigginsJGlossary. Cochrane handbook for systematic reviews of interventions 4.2. 5 [Updated May 2005]2009<p>Systematic reviews in health care: assessing the quality of controlled clinical trials</p>JuniPAltmanDGEggerMBMJ2001323424610.1136/bmj.323.7303.42112067011440947<p>Improving the quality of reporting of randomized controlled trials. The CONSORT statement</p>BeggCChoMEastwoodSHortonRMoherDOlkinIJAMA199627663763910.1001/jama.1996.035400800590308773637<p>Effectiveness and efficiency: random reflections on health services</p>CochraneAL1973<p>A proposal for structured reporting of randomized controlled trials. The standards of reporting trials group</p>JAMA1994272241926193110.1001/jama.1994.035202400540417990245<p>The CONSORT statement: revised recommendations for improving the quality of reports of pllel-group randomised trials</p>MoherDSchulzKFAltmanDGLancet20013571191119410.1016/S0140-6736(00)04337-311323066<p>CONSORT 2010 statement: updated guidelines for reporting pllel group randomised trials</p>SchulzKFAltmanDGMoherDBMJ2010340c33210.1136/bmj.c332284494020332509<p>The methodologic quality of randomization as assessed from reports of trials in specialist and general medical journals</p>SchulzKFChalmersIAltmanDGGrimesDADoreCJOnline J Curr Clin Trials199519781<p>Bias in meta-analysis detected by a simple, graphical test</p>EggerMDaveySGSchneiderMMinderCBMJ199731562963410.1136/bmj.315.7109.62921274539310563<p>Empirical evidence for selective reporting of outcomes in randomized trials: comparison of protocols to published articles</p>ChanAWHrobjartssonAHaahrMTGotzschePCAltmanDGJAMA20042912457246510.1001/jama.291.20.245715161896<p>Clinical trial registration: a statement from the International Committee of Medical Journal Editors</p>DeACDrazenJMFrizelleFAHaugCHoeyJHortonRN Engl J Med20043511250125110.1056/NEJMe04822515356289<p>Systematic review of the empirical evidence of study publication bias and outcome reporting bias</p>DwanKAltmanDGArnaizJABloomJChanAWCroninEPLoS One20083e308110.1371/journal.pone.0003081251811118769481<p>Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta-epidemiological study</p>WoodLEggerMGluudLLSchulzKFJuniPAltmanDGBMJ200833660160510.1136/bmj.39465.451748.AD226799018316340<p>Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials</p>SterneJASuttonAJIoannidisJPTerrinNJonesDRLauJBMJ2011343d400210.1136/bmj.d400221784880<p>Influence of reported study design characteristics on intervention effect estimates from randomized, controlled trials</p>SavovicJJonesHEAltmanDGHarrisRJJuniPPildalJAnn Intern Med201215742943822945832<p>The impact of outcome reporting bias in randomised controlled trials on a cohort of systematic reviews</p>KirkhamJJDwanKMAltmanDGGambleCDoddSSmythRBMJ2010340c36510.1136/bmj.c36520156912<p>Impact of allocation concealment on conclusions drawn from meta-analyses of randomized trials</p>PildalJHrobjartssonAJorgensenKJHildenJAltmanDGGotzschePCInt J Epidemiol20073684785710.1093/ije/dym08717517809<p>The powerful placebo</p>BeecherHKJ Am Med Assoc19551591602160610.1001/jama.1955.0296034002200613271123<p>Bias in treatment assignment in controlled clinical trials</p>ChalmersTCCelanoPSacksHSSmithHJrN Engl J Med19833091358136110.1056/NEJM1983120130922046633598<p>Placebo interventions for all clinical conditions</p>HróbjartssonAGøtzschePCCochrane Database Syst Rev20101<p>Observer bias in randomised clinical trials with binary outcomes: systematic review of trials with both blinded and non-blinded outcome assessors</p>HrobjartssonAThomsenASEmanuelssonFTendalBHildenJBoutronIBMJ2012344e111910.1136/bmj.e111922371859<p>Blinding during data analysis and writing of manuscripts</p>GotzschePCControl Clin Trials19961728529010.1016/0197-2456(95)00263-48889343<p>Who is blinded in randomized clinical trials? A study of 200 trials and a survey of authors</p>HaahrMTHrobjartssonAClin Trials2006336036517060210<p>The landscape and lexicon of blinding in randomized trials</p>SchulzKFChalmersIAltmanDGAnn Intern Med200213625425910.7326/0003-4819-136-3-200202050-0002211827510<p>Physician interpretations and textbook definitions of blinding terminology in randomized controlled trials</p>DevereauxPJMannsBJGhaliWAQuanHLacchettiCMontoriVMJAMA20012852000200310.1001/jama.285.15.200011308438<p>A review of blinding in randomized controlled trials found results inconsistent and questionable</p>BoutronIEstellatCRavaudPJ Clin Epidemiol2005581220122610.1016/j.jclinepi.2005.04.00616291465<p>In the dark: the reporting of blinding status in randomized controlled trials</p>MontoriVMBhandariMDevereauxPJMannsBJGhaliWAGuyattGHJ Clin Epidemiol20025578779010.1016/S0895-4356(02)00446-812384193<p>Observer bias in randomized clinical trials with measurement scale outcomes: a systematic review of trials with both blinded and nonblinded assessors</p>HróbjartssonAThomsenASSEmanuelssonFTendalBHildenJBoutronICan Med Assoc J2013185E201E21110.1503/cmaj.120744<p>Methods of blinding in reports of randomized controlled trials assessing pharmacologic treatments: a systematic review</p>BoutronIEstellatCGuittetLDechartresASackettDLHrobjartssonAPLoS Med20063e42510.1371/journal.pmed.0030425162655317076559<p>Metabias: a challenge for comptive effectiveness research</p>GoodmanSDickersinKAnn Intern Med2011155616210.7326/0003-4819-155-1-201107050-0001021727295<p>Publication decisions revisited: the effect of the outcome of statistical tests on the decision to publish and vice versa</p>SterlingTDRosenbaumWLWeinkamJJAm Stat199549108112<p>Positive-outcome bias: comparison of emergency medicine and general medicine literatures</p>MoscatiRJehleDEllisDFiorelloALandiMAcad Emerg Med199412672717621207<p>Evidence of publication bias in reporting acute stroke clinical trials</p>LiebeskindDSKidwellCSSayreJWSaverJLNeurology20066797397910.1212/01.wnl.0000237331.16541.ac17000963<p>A survey identified publication bias in the secondary literature</p>CarterAOGriffinGHCarterTPJ Clin Epidemiol20065924124510.1016/j.jclinepi.2005.08.01116488354<p>Unpublished research from a medical specialty meeting: why investigators fail to publish</p>WeberEJCallahamMLWearsRLBartonCYoungGJAMA199828025725910.1001/jama.280.3.2579676674<p>NIH clinical trials and publication bias</p>DickersinKMinYIOnline J Curr Clin Trials1993504967<p>How important is publication bias? A synthesis of available data</p>DickersinKAIDS Educ Prev1997915219083596<p>A general linear model for estimating effect size in the presence of publication bias</p>VeveaJLHedgesLVPsychometrika19956041943510.1007/BF02294384<p>Practical estimates of the effect of publication bias in meta-analysis</p>TaylorSJTweedieRLAust Epidemiol199851417<p>Publication bias in meta-analysis: a Bayesian data-augmentation approach to account for issues exemplified in the passive smoking debate</p>GivensGHSmithDDTweedieRLStat Sci1997221240<p>Trim and fill: a simple funnel-plot-based method of testing and adjusting for publication bias in meta-analysis</p>DuvalSTweedieRBiometrics20005645546310.1111/j.0006-341X.2000.00455.x10877304<p>Funnel plots for detecting bias in meta-analysis: guidelines on choice of axis</p>SterneJAEggerMJ Clin Epidemiol2001541046105510.1016/S0895-4356(01)00377-811576817<p>Adjusting for publication bias in the presence of heterogeneity</p>TerrinNSchmidCHLauJOlkinIStat Med2003222113212610.1002/sim.146112820277<p>A test for publication bias in meta-analysis with sparse binary data</p>SchwarzerGAntesGSchumacherMStat Med20072672173310.1002/sim.258816755545<p>Contour-enhanced meta-analysis funnel plots help distinguish publication bias from other causes of asymmetry</p>PetersJLSuttonAJJonesDRAbramsKRRushtonLJ Clin Epidemiol20086199199610.1016/j.jclinepi.2007.11.01018538991<p>Assessment of regression-based methods to adjust for publication bias through a comprehensive simulation study</p>MorenoSGSuttonAJAdesAEStanleyTDAbramsKRPetersJLBMC Med Res Methodol20099210.1186/1471-2288-9-2264915819138428<p>Novel methods to deal with publication biases: secondary analysis of antidepressant trials in the FDA trial registry database and related journal publications</p>MorenoSGSuttonAJTurnerEHAbramsKRCooperNJPalmerTMBMJ2009339b298110.1136/bmj.b2981272321719666685<p>In an empirical evaluation of the funnel plot, researchers could not visually identify publication bias</p>TerrinNSchmidCHLauJJ Clin Epidemiol20055889490110.1016/j.jclinepi.2005.01.00616085192<p>Comparison of two methods to detect publication bias in meta-analysis</p>PetersJLSuttonAJJonesDRAbramsKRRushtonLJAMA200629567668010.1001/jama.295.6.67616467236<p>Performance of the trim and fill method in the presence of publication bias and between-study heterogeneity</p>PetersJLSuttonAJJonesDRAbramsKRRushtonLStat Med2007264544456210.1002/sim.288917476644<p>The appropriateness of asymmetry tests for publication bias in meta-analyses: a large survey</p>IoannidisJPTrikalinosTACMAJ20071761091109610.1503/cmaj.060410183979917420491<p>Chapter 10: addressing reporting biases</p>SterneJACEggerMMoherDCochrane handbook for systematic reviews of intervention. Version 5.1.0 (Updated march 2011) editionThe Cochrane CollaborationHiggins JPT, Green S2011<p>Bias in metaGÇÉanalysis due to outcome variable selection within studies</p>HuttonJLWilliamsonPRJ Royal Stat Soc : Series C (Appl Stat)200249359370<p>Investigation of within-study selective reporting in clinical research: follow-up of applications submitted to a local research ethics committee</p>HahnSWilliamsonPRHuttonJLJ Eval Clin Pract2002835335910.1046/j.1365-2753.2002.00314.x12164983<p>Outcome reporting bias in randomized trials funded by the Canadian Institutes of Health Research</p>ChanAWKrleza-JericKSchmidIAltmanDGCMAJ200417173574010.1503/cmaj.104108651785815451835<p>Selective reporting of outcomes of drug trials. Comparison of study protocols and published articles</p>von ElmERollinABlumleASenessieCLowNEggerM2006<p>Association between unreported outcomes and effect size estimates in Cochrane meta-analyses</p>FurukawaTAWatanabeNOmoriIMMontoriVMGuyattGHJAMA200729746847017284696<p>Many scenarios exist for selective inclusion and reporting of results in randomized trials and systematic reviews</p>PageMJMcKenzieJEForbesAJ Clin Epidemiol2013<p>Identifying outcome reporting bias in randomised trials on PubMed: review of publications and survey of authors</p>ChanAWAltmanDGBMJ200533075310.1136/bmj.38356.424606.8F55587515681569<p>Frequency and reasons for outcome reporting bias in clinical trials: interviews with trialists</p>SmythRMKirkhamJJJacobyAAltmanDGGambleCWilliamsonPRBMJ2011342c715310.1136/bmj.c7153301681621212122<p>Systematic review of the empirical evidence of study publication bias and outcome reporting BiasGÇöAn updated review</p>DwanKGambleCWilliamsonPRKirkhamJJPloS one20138e6684410.1371/journal.pone.0066844370253823861749<p>Outcome selection bias in meta-analysis</p>WilliamsonPRGambleCAltmanDGHuttonJLStat Methods Med Res20051451552410.1191/0962280205sm415oa16248351<p>Driving up the quality and relevance of research through the use of agreed core outcomes</p>WilliamsonPAltmanDBlazebyJClarkeMGargonEJ Health Serv Res Policy2012171222572709The COMET Initiativehttp://www.comet-initiative.org/, Last accessed 19th September 2013<p>On the bias produced by quality scores in meta-analysis, and a hierarchical view of proposed solutions</p>GreenlandSO’RourkeKBiostat2001246347110.1093/biostatistics/2.4.463<p>Does blinding of readers affect the results of meta-analyses? University of Pennsylvania Meta-analysis Blinding Study Group</p>BerlinJALancet19973501851869250191<p>Blinded versus unblinded assessments of risk of bias in studies included in a systematic review</p>MorissetteKTriccoACHorsleyTChenMHMoherDCochrane Database Syst Rev2011MR000025<p>Does quality of reports of randomised trials affect estimates of intervention efficacy reported in meta-analyses?</p>MoherDPhamBJonesACookDJJadadARMoherMLancet199835260961310.1016/S0140-6736(98)01085-X9746022<p>Getting the methods right–the foundation of patient-centered outcomes research</p>GabrielSENormandSLN Engl J Med201236778779010.1056/NEJMp120743722830434<p>A catalogue of reporting guidelines for health research</p>SimeraIMoherDHoeyJSchulzKFAltmanDGEur J Clin Invest201040355310.1111/j.1365-2362.2009.02234.x20055895<p>The quality of reports of randomised trials in 2000 and 2006: comptive study of articles indexed in PubMed</p>HopewellSDuttonSYuLMChanAWAltmanDGBMJ2010340c72310.1136/bmj.c723284494120332510<p>Consolidated standards of reporting trials (CONSORT) and the completeness of reporting of randomised controlled trials (RCTs) published in medical journals</p>TurnerLShamseerLAltmanDGWeeksLPetersJKoberTCochrane Database Syst Rev201211MR000030<p>Publication bias: the case for an international registry of clinical trials</p>SimesRJJ Clin Oncol19864152915413760920<p>Clinical-trial registration: a call for its implementation in Canada</p>MoherDCMAJ19931491657165814859748242504<p>Time to register randomised trials. The case is now unanswerable</p>HortonRSmithRBMJ199931986586610.1136/bmj.319.7214.865111670410506022<p>Compulsory registration of clinical trials</p>AbbasiKBMJ200432963763810.1136/bmj.329.7467.63751763015374895<p>Compliance of clinical trial registries with the World Health Organization minimum data set: a survey</p>MojaLPMoschettiINurbhaiMCompagnoniALiberatiAGrimshawJMTrials2009105610.1186/1745-6215-10-56273455219624821<p>Compliance with mandatory reporting of clinical trial results on ClinicalTrials.gov: cross sectional study</p>PrayleAPHurleyMNSmythARBMJ2012344d737310.1136/bmj.d737322214756<p>Comparison of registered and published primary outcomes in randomized controlled trials</p>MathieuSBoutronIMoherDAltmanDGRavaudPJAMA200930297798410.1001/jama.2009.124219724045<p>Chapter 8: assessing risk of bias in included studies</p>HigginsJPTAltmanDGSterneJACCochrane handbook for systematic reviews of interventions. Version 5.1.0 (Updated march 2011) editionThe Cochrane CollaborationHiggins JPT, Green S2011<p>Risk of bias versus quality assessment of randomised controlled trials: cross sectional study</p>HartlingLOspinaMLiangYDrydenDMHootonNKrebsSJBMJ2009339b401210.1136/bmj.b4012276403419841007<p>Testing the risk of bias tool showed low reliability between individual reviewers and across consensus assessments of reviewer pairs</p>HartlingLHammMPMilneAVandermeerBSantaguidaPLAnsariMJ Clin Epidemiol2012<p>Issues relating to study design and risk of bias when including non-randomized studies in systematic reviews on the effects of interventions</p>HigginsJPRamsayCReevesBDeeksJJSheaBValentineJRes Syn Methods201210.1002/jrsm.1056<p>Issues relating to selective reporting when including non-randomized studies in systematic reviews on the effects of healthcare interventions</p>NorrisSLMoherDReevesBSheaBLokeYGarnerSResearch Synthesis Methods201210.1002/jrsm.1062<p>Issues relating to confounding and meta-analysis when including non-randomized studies in systematic reviews on the effects of interventions</p>ValentineJThompsonSGRes Syn Methods201210.1002/jrsm.1064<p>Evaluating non-randomised intervention studies</p>DeeksJJDinnesJD’AmicoRSowdenAJSakarovitchCSongFHealth Technol Assess20037iii17314636486<p>Cochrane methods</p>ChandlerJClarkeMHigginsJCochrane Database Syst Rev2012Suppl 1156<p>Comparison of protocols and registry entries to published reports for randomised controlled trials</p>DwanKAltmanDGCresswellLBlundellMGambleCLWilliamsonPRCochrane Database Syst Rev2011MR000031<p>Constraints on publication rights in industry-initiated clinical trials</p>GotzschePCHrobjartssonAJohansenHKHaahrMTAltmanDGChanAWJAMA20062951645164616609085<p>Reporting of conflicts of interest from drug trials in Cochrane reviews: cross sectional study</p>RosemanMTurnerEHLexchinJCoyneJCBeroLAThombsBDBMJ2012345e515510.1136/bmj.e5155342363522906823<p>Stopping randomized trials early for benefit and estimation of treatment effects: systematic review and meta-regression analysis</p>BasslerDBrielMMontoriVMLaneMGlasziouPZhouQJAMA20103031180118710.1001/jama.2010.31020332404<p>Randomized trials stopped early for benefit: a systematic review</p>MontoriVMDevereauxPJAdhikariNKBurnsKEEggertCHBrielMJAMA20052942203220910.1001/jama.294.17.220316264162<p>Bias and trials stopped early for benefit</p>GoodmanSBerryDWittesJJAMA201030415715920628126<p>Single-center trials show larger treatment effects than multicenter trials: evidence from a meta-epidemiologic study</p>DechartresABoutronITrinquartLCharlesPRavaudPAnn Intern Med2011155395110.7326/0003-4819-155-1-201107050-0000621727292<p>Impact of single centre status on estimates of intervention effects in trials with continuous outcomes: meta-epidemiological study</p>BafetaADechartresATrinquartLYavchitzABoutronIRavaudPBMJ2012344e81310.1136/bmj.e813327932822334559<p>A Web-based archive of systematic review data</p>IpSHadarNKeefeSParkinCIovinRBalkEMSyst Rev201211510.1186/2046-4053-1-15335173722588052<p>Incorporation of assessments of risk of bias of primary studies in systematic reviews of randomized trials: a cross-sectional review</p>HopewellSBoutronIAltmanDGRavaudPJournal TBDin press<p>Bias modelling in evidence synthesis</p>TurnerRMSpiegelhalterDJSmithGCThompsonSGJ R Stat Soc Ser A Stat Soc2009172214710.1111/j.1467-985X.2008.00547.x266730319381328