Scolaris Content Display Scolaris Content Display

Industry sponsorship and research outcome

Collapse all Expand all

Background

Clinical research affecting how doctors practice medicine is increasingly sponsored by companies that make drugs and medical devices. Previous systematic reviews have found that pharmaceutical‐industry sponsored studies are more often favorable to the sponsor’s product compared with studies with other sources of sponsorship. A similar association between sponsorship and outcomes have been found for device studies, but the body of evidence is not as strong as for sponsorship of drug studies. This review is an update of a previous Cochrane review and includes empirical studies on the association between sponsorship and research outcome.

Objectives

To investigate whether industry sponsored drug and device studies have more favorable outcomes and differ in risk of bias, compared with studies having other sources of sponsorship.

Search methods

In this update we searched MEDLINE (2010 to February 2015), Embase (2010 to February 2015), the Cochrane Methodology Register (2015, Issue 2) and Web of Science (June 2015). In addition, we searched reference lists of included papers, previous systematic reviews and author files.

Selection criteria

Cross‐sectional studies, cohort studies, systematic reviews and meta‐analyses that quantitatively compared primary research studies of drugs or medical devices sponsored by industry with studies with other sources of sponsorship. We had no language restrictions.

Data collection and analysis

Two assessors screened abstracts and identified and included relevant papers. Two assessors extracted data, and we contacted authors of included papers for additional unpublished data. Outcomes included favorable results, favorable conclusions, effect size, risk of bias and whether the conclusions agreed with the study results. Two assessors assessed risk of bias of included papers. We calculated pooled risk ratios (RR) for dichotomous data (with 95% confidence intervals (CIs)).

Main results

Twenty‐seven new papers were included in this update and in total the review contains 75 included papers. Industry sponsored studies more often had favorable efficacy results, RR: 1.27 (95% CI: 1.17 to 1.37) (25 papers) (moderate quality evidence), similar harms results RR: 1.37 (95% CI: 0.64 to 2.93) (four papers) (very low quality evidence) and more often favorable conclusions RR: 1.34 (95% CI: 1.19 to 1.51) (29 papers) (low quality evidence) compared with non‐industry sponsored studies. Nineteen papers reported on sponsorship and efficacy effect size, but could not be pooled due to differences in their reporting of data and the results were heterogeneous. We did not find a difference between drug and device studies in the association between sponsorship and conclusions (test for interaction, P = 0.98) (four papers). Comparing industry and non‐industry sponsored studies, we did not find a difference in risk of bias from sequence generation, allocation concealment, follow‐up and selective outcome reporting. However, industry sponsored studies more often had low risk of bias from blinding, RR: 1.25 (95% CI: 1.05 to 1.50) (13 papers), compared with non‐industry sponsored studies. In industry sponsored studies, there was less agreement between the results and the conclusions than in non‐industry sponsored studies, RR: 0.83 (95% CI: 0.70 to 0.98) (six papers).

Authors' conclusions

Sponsorship of drug and device studies by the manufacturing company leads to more favorable efficacy results and conclusions than sponsorship by other sources. Our analyses suggest the existence of an industry bias that cannot be explained by standard 'Risk of bias' assessments.

Industry sponsorship and research outcome

Results from clinical studies on drugs and medical devices affect how doctors practice medicine and thereby the treatments offered to patients. However, clinical research is increasingly sponsored by companies that make these products, either because the companies directly perform the studies, or fully or partially fund them. Previous research has found that pharmaceutical industry sponsored studies tend to favor the sponsors’ drugs more than studies with any other sources of sponsorship. This suggests that industry sponsored studies are biased in favor of the sponsor’s products.

This review is an update of a previous review that looked at sponsorship of drug and device studies. The primary aim of the review was to find out whether the published results and overall conclusions of industry sponsored drug and device studies were more likely to favor the sponsors’ products, compared with studies with other sources of sponsorship. The secondary aim was to find out whether such industry sponsored studies used methods that increase the risk of bias, again compared with studies with other sources of sponsorship. In this update, we carried out a comprehensive search of all relevant papers of empirical studies published from 2010 to February 2015 and included 27 new papers, yielding a total of 75 papers included in our review.

Industry sponsored drug and device studies more often had efficacy results that were favorable to the sponsors' products, (risk ratio (RR): 1.27, 95% confidence interval (CI): 1.17 to 1.37), similar harms results (RR: 1.37, 95% CI: 0.64 to 2.93) and favorable overall conclusions (RR: 1.34, 95% CI: 1.19 to 1.51), compared with non‐industry sponsored drug and device studies. We did not find a difference between industry and non‐industry sponsored studies with respect to standard methodological factors that may increase the risk of bias, except for blinding: industry sponsored studies reported satisfactory blinding more often than non‐industry sponsored studies. In industry sponsored studies, there was less agreement between the results and the conclusions than in non‐industry sponsored studies, RR: 0.83 (95% CI: 0.70 to 0.98).We did not find a difference between drug and device studies in the association between sponsorship and conclusions. Our analysis suggests that industry sponsored drug and device studies are more often favorable to the sponsor’s products than non‐industry sponsored drug and device studies due to biases that cannot be explained by standard 'Risk of bias' assessment tools.

Authors' conclusions

Implication for methodological research

Currently, the Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 acknowledges problems in relation to sponsorship, but does not recommend assessing industry sponsorship as a separate domain in the 'Risk of bias' assessment (Higgins 2011b). The assumption is that the influence of the sponsor will be mediated through the mechanisms of bias that are currently assessed, such as selective reporting of favorable outcomes. A Cochrane review that examined the association of sponsorship and selective outcome reporting bias (Dwan 2011), found uncertain evidence for the association; however, assessment of selective outcome reporting is complex and bias may be difficult to detect (Kirkham 2010). Some studies that have documented the extensive selective reporting of favorable outcomes have examined only industry sponsored studies (Rising 2008; Vedula 2009), thus making comparison with non‐industry sponsored studies impossible.

Our data suggest that the more favorable outcomes in industry sponsored studies are mediated by factors other than those documented in the 'Risk of bias' assessment tool in Cochrane reviews. It has been suggested that industry bias should be regarded as a meta‐bias, as industry sponsorship in itself is not a bias‐producing process –  as for example lack of concealment of allocation is – but a risk factor for bias (Goodman 2011). However, the characteristics currently assessed in the standard risk of bias approach in Cochrane reviews likely do not capture the additional risk of bias in industry sponsored studies. For example, the Handbook states that design issues, such as dosage of comparators are not issues of bias, but of generalizability. Yet, pharmacological interventions have dose‐response curves, and testing drugs that are not in comparable places on their dose‐response curves sets up a systematic, unfair and biased comparison (Safer 2002).

Consequently, our data suggest that industry sponsorship should be treated as bias‐inducing and industry bias should be treated as a separate domain. There are many subtle mechanisms through which sponsorship may influence outcomes, and an assessment of sponsorship should therefore be used as a proxy for these mechanisms. Interestingly, the AMSTAR tool for methodological quality assessment of systematic reviews includes funding and conflicts of interest as a domain (Shea 2007). Adaptations of Cochrane tools for assessing risk of bias in studies assessing environmental risks have also included funding source and conflicts of interest as a domain (Johnson 2016). Methods for reporting, assessing and handling industry bias and other biases in future systematic reviews must be developed. Specifically, further methodological research should focus on how industry bias is handled in Cochrane reviews.

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Industry sponsored compared to non‐industry sponsored studies for research outcome

Results and conclusions: Industry sponsored compared to non‐industry sponsored studies

Patient or population: industry sponsorship and study results
Intervention: industry sponsored studies
Comparator: non‐industry sponsored studies

Outcomes

Illustrative comparative risks* (95% CI)

Risk ratio
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Non‐industry sponsored studies

Industry sponsored studies

Number of studies with favorable efficacy results

502 per 1000

638 per 1000
(588 to 688)

1.27
(1.17 to 1.37)

25 papers including 2923 studies

⊕ ⊕ ⊕ ○
MODERATE

Upgraded as control for confounders and analysis of low risk of bias papers gave similar results.

Number of studies with favorable harms results

474 per 1000

649 per 1000
(303 to 1388)

1.37
(0.64 to 2.93)

4 papers including 826 studies

⊕ ○ ○ ○
VERY LOW

Downgraded due to substantial heterogeneity (inconsistency) and wide confidence intervals (imprecision).

Number of studies with favorable conclusions

644 per 1000

863 per 1000
(766 to 972)

1.34
(1.19 to 1.51)

29 papers including 4583 studies

⊕ ⊕ ○ ○
LOW

Downgraded due to substantial heterogeneity (inconsistency).

Upgraded as control for confounders and analysis of low risk of bias papers gave similar results.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

The assumed risk of the control group (i.e. non‐industry group) was calculated as the mean risk (i.e. number of studies with favorable results divided by total number of studies).

Background

Description of the problem or issue

Clinical research sponsored by the pharmaceutical industry affects how doctors practice medicine (PhRMA 2008; Wyatt 1991). An increasing number of clinical trials at all stages in a product's life cycle are funded by the pharmaceutical industry, and the industry now spends more on medical research than do the National Institutes of Health in the USA (Moses 2015). Results and conclusions that are unfavorable to the sponsor (i.e. studies that find an expensive drug similarly or less effective or more harmful than drugs used to treat the same condition) can pose considerable financial risks to companies.

Several systematic reviews have documented that pharmaceutical industry sponsorship of drug studies is associated with findings that are favorable to the sponsor’s product (Bekelman 2003; Lexchin 2003; Schott 2010a; Sismondo 2008a). There are several ways that industry can sponsor a study, including single‐source sponsorship, shared sponsorship, and provision of free drugs or devices only. There are also several potential ways that industry sponsors can influence the outcome of a study, including the framing of the question, the design of the study, the conduct of the study, how data are analyzed, selective reporting of favorable results, and spin in reporting conclusions (Bero 1996; Lexchin 2012; Sismondo 2008b). Although some journals now require that the role of the sponsor in the design, conduct and publication of the study be described, this practice is not widespread (Tuech 2005). In addition, some have argued that because industry sponsored studies are often conducted for regulatory purposes, their methods must meet high standards (Rosefsky 2003). Therefore, it is important to examine differences not only in the outcomes of industry versus non‐industry sponsored studies, but also differences in the methods or risks of bias.

Why it is important to do this review

This systematic review is the update of an original systematic review by three of the authors (Lundh 2012), which investigated whether sponsorship by industry is associated with the publication of outcomes favorable to the sponsor. That review is now out of date. Developments, such as the adoption of trial registration could lessen the bias associated with industry sponsorship, as publication bias can be more readily detected (DeAngelis 2004). Furthermore, companies now publish results in trial registries suggesting a move toward increased transparency (Potthast 2014; Schwartz 2016). However, this may not be the case as a recent study found that reporting bias is also prevalent in registered trials, particular in industry sponsored trials (Jones 2013). In addition, the release of internal industry documents as a result of settlement agreements resulting from litigation against drug companies has revealed examples of industry manipulation of the conduct and publication of studies (Fugh‐Berman 2010; Ross 2008; Steinman 2006; Vedula 2009).

Objectives

The objectives were to investigate whether:

  • sponsorship of drug and device studies by the pharmaceutical and device industries is associated with outcomes, including conclusions, that are favorable to the sponsor;

  • drug and device studies sponsored by the pharmaceutical and device industries differ in their risk of bias compared with studies with other sources of sponsorship.

Methods

Criteria for considering studies for this review

Types of studies

This review includes reports of empirical studies that investigate samples of primary research studies. To avoid confusion we will use the terms 'studies' for the primary research studies and 'papers' for the reports of empirical studies of primary research studies. We will use the term trials to describe studies of a randomized clinical trial design.

We included papers of cross‐sectional studies, cohort studies, systematic reviews or meta‐analyses that quantitatively compared primary research of human drug or medical device studies sponsored by the pharmaceutical or device industries with studies that had other sources of sponsorship. These papers could report the results of methodological studies or systematic reviews that had a pre‐specified subgroup or sensitivity analysis by sponsorship source. We also included papers investigating sources of heterogeneity (e.g. using meta‐regression) if sponsorship was investigated. Drugs were defined as medications that require approval by a regulatory authority as a prescription drug, recognizing that these approval standards vary worldwide. Devices were defined based on the Food and Drug Administration (FDA) definition as instruments intended for use in the diagnosis, treatment or prevention of disease.

We excluded papers without quantitative data related to our primary or secondary outcomes. We excluded papers of the effects of sponsorship by non‐pharmaceutical or non‐device (e.g. tobacco, food or chemical) industries, and papers that evaluated the effectiveness of herbal supplements or medical procedures. Papers examining mixed interventions (e.g. pharmaceuticals and educational interventions) were included if drug or device data were reported separately or could be obtained from the authors.

We excluded papers that quantitatively compared the association of sponsorship and results of syntheses of research studies (i.e. systematic reviews or meta‐analyses) or pharmacoeconomic studies of drugs or devices. We also excluded analyses of pharmacokinetic studies and studies restricted to non humans (e.g. animal or cell cultures).

Only papers published in full, including structured research letters, were included. We excluded unstructured letters to the editor and conference abstracts. This decision was based on the poor reporting quality of data in letters and conference abstracts encountered in a previous version of our review (Lexchin 2003). A comment to the previous version of our review (Lundh 2012) suggested that the exclusion of conference abstracts and letters could have introduced publication bias. Therefore, we included conference abstracts and all types of letters in a sensitivity analysis (see below). We had no language restrictions.

Types of data

Drug and device papers including human research studies comparing drug to placebo, device to sham, drug to drug, drug to device, device to device, or mixed comparisons where the effectiveness, efficacy or harms of the drug or device were evaluated. A few papers included data from both unpublished and published studies. If data were reported separately for the published studies or were available from the authors we used data from published studies only. The reason for this decision was that published studies represent what is available to users of the medical literature and our focus was on assessing biases in published studies.

Types of methods

We defined sponsorship as funding or provision of free drug or devices. Drug or device studies with pharmaceutical or device industry funding versus those with other or undisclosed funding were included. We extracted the definition of industry funding verbatim from the included papers (see Data extraction and management) and reported this in the 'Characteristics of included studies' table. For analysis, we grouped the definitions into a variety of categories, including 100% pharmaceutical or device company funding, 100% non‐industry funding, mixed funding (e.g. non‐industry and industry collaboration), free provision of drug or device only, and undisclosed funding.

We included papers that compared industry sponsored studies with non‐industry sponsored studies and also papers that compared studies of products by competing manufacturers (i.e. studies sponsored by the manufacturer of the test treatment with studies sponsored by the manufacturer of the control treatment); we analyzed the two types of papers separately.

Types of outcome measures

Primary outcomes

We included two primary outcomes.

  • Whether the results were favorable to the sponsor.

  • Whether the conclusions were favorable to the sponsor. 

We used the definition of favorable results as described in the methods of the included papers. For efficacy results, most papers considered favorable results to be those that were statistically significant (e.g. P < 0.05 or a 95% confidence interval excluding the possibility of no difference) in favor of the sponsor's product. Based on the previous review (Lundh 2012), which found very few studies that reported results unfavorable to the sponsor, unfavorable results were combined with studies that reported results that were neutral or not statistically significant. For harms results, most papers regarded favorable results to be those where harms results were not statistically significant (e.g. P > 0.05 or a 95% confidence interval including the possibility of no difference) or results that had a statistically significant higher number of harms in the comparator group.

Conclusions in which the sponsor’s product was preferred over the control treatment were considered favorable to the sponsor. For conclusions we did not distinguish between efficacy and harms, as conclusions are often overall qualitative judgements based on a benefit to harm balance.

Secondary outcomes

We included three secondary outcomes.

  • The size of the effect estimate in industry sponsored studies versus those with other sources of sponsorship.

  • The risk of bias in industry sponsored studies versus those with other sources of sponsorship.

  • The concordance between study results and conclusions, i.e. whether the conclusions agreed with the study results, in industry sponsored studies versus those with other sources of sponsorship. 

We included papers that reported at least one of these secondary outcomes, even if it reported neither of the primary outcomes.

Search methods for identification of studies

Electronic searches

In this update, we searched Ovid MEDLINE (R) In‐Process and other non‐indexed citations and Ovid MEDLINE (R) (2010 to February 2015), Ovid Embase (2010 to February 2015) and the Cochrane Methodology Register (2015, Issue 2) (Wiley InterScience Online). We searched the Web of Science (June 2015) for papers that cited any of the papers included in our review.

Search strategy

We used the strategy shown in Appendix 1 for Ovid MEDLINE and adapted it for the other databases.

Searching other resources

Other sources of data included author files, searches of reference lists of included papers and previous systematic reviews.

Data collection and analysis

Selection of studies

Two pairs of assessors  (LB and JS or BM and JL) screened the titles and abstracts, when available, of all retrieved records for obvious exclusions, and assessed the remaining papers based on full text. Any disagreements were resolved by consensus and reasons for exclusions of potentially eligible papers are described in the 'Characteristics of excluded studies' table. There was no need for translation of non‐English papers.

Data extraction and management

Two pairs of assessors (AL and JS or BM and JL) independently extracted data from included papers; differences in data extraction were resolved by consensus. 

We extracted data on the following.

  • Year published.

  • Country of corresponding author.

  • Study objective.

  • Study design used in the paper (cohort, cross‐sectional, systematic review or meta‐analysis, other).

  • Study domain ‐ descriptive (e.g. oncology drug trials).

  • Study domain ‐ category (drug/device class, specific disease, medical specialty/type of diseases, mixed).

  • Type of studies (drug, device, drug and device, mixed).

  • Type of comparisons (drug versus drug, drug versus placebo, device versus device, device versus sham, device versus drug, mixed, other).

  • Sample strategy used to locate research studies (electronic search only, electronic plus other, sampling of journals, sampling by venue (e.g. conference abstracts)).

  • Whether there were language restrictions on the search.

  • Number of studies included in the sample.

  • Time period covered by studies in the paper.

  • Sponsorship categories coded in the paper. Categories were:

    • 100% pharmaceutical/device company funded;

    • 100% non‐profit funded;

    • mixed funding ‐ e.g. non‐industry and industry collaboration;

    • provision of drug or device only; and

    • undisclosed funding.

  • Sponsorship categories used in analysis in the paper (e.g. 100% industry funded grouped with mixed funding for industry category).

  • Description of role of the sponsor (if any). For example, definition of the sponsor’s role in the design, implementation or reporting in the sample of studies.

  • Criteria used to assess risk of bias of the studies included in the paper.

  • Primary purpose of the study.

  • Whether the paper commented on appropriateness of comparators.

  • Data on sponsorship and results.

  • Data on sponsorship and conclusions.

  • Data on sponsorship and effect size.

  • Data on sponsorship and risk of bias.

  • Data on sponsorship and concordance between study results and conclusions.

  • Additional relevant data.

  • Funding source of included paper. As this item was not included in the previous version of the review we also extracted data from papers included in the previous version.

Assessment of risk of bias in included studies

Since there are no validated criteria for assessing risk of bias in these types of papers, we developed our own criteria. We reviewed papers for high, low or unclear risk of bias for each of four criteria. If a criterion was met, it was regarded as having low risk of bias, and high risk of bias otherwise. If we could not determine whether a criterion was met, we coded it as unclear. We used the following criteria:

  • whether explicit and well‐defined criteria that could be replicated by others were used to select studies for inclusion/exclusion;

  • whether there was an adequate study inclusion method, with two or more assessors selecting studies;

  • whether the search for studies was comprehensive; and

  • whether methodological differences and other characteristics that could introduce bias were controlled for or explored.

Measures of the effect of the methods

We performed a meta‐analysis of the papers that reported the association of sponsorship with favorable study outcomes in cases where a pooled risk ratio (RR) and its 95% confidence interval could be computed.

The definition of a favorable outcome varied among papers. In some papers favorable outcomes were defined as those that were favorable to the sponsor's product and in others favorable to the test treatment. This difference in terminology did not matter when the comparison was between active treatment and placebo, since the sponsor's product was the active treatment and not placebo. For head‐to‐head comparisons, however, the sponsor could be either the manufacturer of the test treatment or the control treatment. In these cases, when data were available, we recoded outcomes as to whether they were favorable to the sponsor's product.

We separately analyzed papers of industry sponsored head‐to‐head studies, comparing studies sponsored by the manufacturer of the test treatment with studies sponsored by the manufacturer of the comparator treatment. This was done by assigning the newest treatment (most recent FDA approval date) as the 'test' treatment and the older treatment as the 'comparator' treatment using similar methods as described by Bero and colleagues (Bero 2007) and comparing the number of studies favorable to the test treatment in the two groups (i.e. sponsor produces test treatment or sponsor produces comparator treatment).

At the time many of the papers were published, the approach was to assess the methodological quality of studies as opposed to an assessment of the risk of bias of studies. We therefore recoded the data on methodological quality into 'Risk of bias' categories. So, for example, a trial with adequate concealment of allocation was coded as low risk of bias and a trial with inadequate concealment of allocation as high risk of bias. Some papers assessed risk of bias by summarizing results for individual domains into an overall methodological quality score (i.e. a scale approach). There are substantial methodological problems related to quality scales (Jüni 1999), and their use is not recommended. We therefore did not combine the results obtained with these scales, but report the results descriptively. The included papers assessed blinding using different approaches. Some papers rated blinding for the study overall, for example whether a study used matching placebo tablets (which could be considered blinded) and some papers assessed who was blinded, similar to the Cochrane 'Risk of bias' tool. The Cochrane 'Risk of bias' tool assesses blinding to protect against performance bias (e.g. clinicians or patients are blinded) and to protect against detection bias (e.g. outcome assessors are blinded). We therefore categorized each 'Risk of bias' assessment into the items blinding‐overall, blinding‐performance bias and blinding‐detection bias.

Dealing with missing data

We contacted authors of the original papers in an attempt to obtain missing data. If papers included studies reporting conflicts of interest, but not the source of funding, we contacted the authors in order to obtain separate data for funding. In total, we contacted authors of eight papers included in this update and received additional data for five of these papers.

Assessment of heterogeneity

We assessed heterogeneity using I2. We defined substantial statistical heterogeneity as an I2 > 50% (Higgins 2011a).

Data synthesis

We used Review Manager (RevMan 2014) to analyze data. For dichotomous data we used the Mantel‐Haenszel random‐effects model to create a pooled RR. In the previous version of this review (Lundh 2012), we used a fixed‐effect model as default and a random‐effects model when substantial heterogeneity was observed. However, due to the large clinical heterogeneity between papers (e.g. study domains, study designs and definition of outcomes), we decided that a random‐effects model was more appropriate.

Subgroup analysis and investigation of heterogeneity

We considered the following factors as potential explanations for heterogeneity and investigated them in separate subgroup analyses for our primary outcomes.    

  • We hypothesized that the association of industry sponsorship and favorable outcomes may be larger in high risk of bias papers. We assessed overall risk of bias of the included papers using the criteria described in 'Assessment of risk of bias in included studies'. We regarded papers with adequate study inclusion, a comprehensive search and controlling for bias as having a low risk of bias; others as having a high risk. We compared low risk of bias papers with high risk of bias papers in a subgroup analysis.

  • We compared papers of drug studies with device studies, as the mechanisms of influencing study outcomes may differ between the industries. For example, drug trials are more regulated than device trials, which could have an influence on biases in the design, conduct and reporting of the trials. We compared this in a subgroup analysis.   

  • As the study domain might contribute to heterogeneity, we compared papers on specific treatments or diseases with papers of mixed domains in another subgroup analysis.

Sensitivity analysis

We undertook the following sensitivity analyses to test the robustness of our findings for our primary outcomes.

  • The primary analyses compared the number of favorable results and conclusions in papers with industry sponsorship to those with other sources of sponsorship; 'industry sponsorship' included 100% pharmaceutical/device company funding, mixed funding and provision of drug or device only. 'Non‐industry sponsorship' included 100% government funding, 100% non‐industry funding and undisclosed funding. In a sensitivity analysis, we excluded those studies with mixed funding sources and those with funding consisting solely of free product from the 'industry sponsorship' category, and excluded studies with undisclosed funding from the category of 'non‐industry sponsorship', to determine if these had an impact on the initial analysis. As noted under 'Data extraction and management', we were reliant on how the studies in our review defined 'funding'.

  • A sensitivity analysis restricted to papers that adjusted for confounders (e.g. adjusted for sample size and concealment of allocation using logistic regression) using adjusted estimates. We used the generic inverse variance method to pool adjusted odds ratios in a random‐effects model.

  • A sensitivity analysis where all analyses were based on a fixed‐effect model.

  • One paper (Finucane 2004), included unpublished abstracts from conference proceedings. One paper (Lynch 2007), included manuscripts submitted to a medical journal of which the majority were never published. However, based on the reported data it was not possible to extract data from the published studies separately. As these two papers included unpublished data and we planned to analyze only published data, we conducted a sensitivity analysis that excluded these two papers.

  • Many of the included papers investigated similar domains (e.g. antidepressants or oncology drugs) or included studies from similar journals in overlapping time‐periods. This is likely to lead to double counting if data from the same studies are included more than once and to an overestimate of the precision of effect estimates. Due to the way data were reported in the papers, it was not possible ensure that data was not double counted. Instead we performed a sensitivity analysis restricted to papers on specific treatments or diseases where none of the other included papers were related to the same domain.

  • We included letters and conference abstracts reporting quantitative data in a sensitivity analysis.

Quality of Evidence

We created a 'Summary of findings' table using the GRADE approach (Higgins 2011a) and using the MAGICapp software (MAGICapp; Vandvik 2013).

Results

Description of studies

See:Characteristics of included studies; Characteristics of excluded studies.

Results of the search

See: Figure 1.


Study flow diagram.

Study flow diagram.

After removal of duplicates, 3052 references were identified. From reading titles and abstracts, 2925 were eliminated as being not relevant to the review. Full‐text papers were obtained for 127 references. From these 127 papers, 104 papers were excluded (see Characteristics of excluded studies) and 23 included (see Characteristics of included studies). Four additional papers were included from searching additional sources and 48 were included from the previous version of the review (see Characteristics of included studies). In total, 75 papers were included.

Included studies

See: Characteristics of included studies.

The 75 papers were published between 1986 and 2015. Seventy‐two papers included mainly published studies, one included studies presented at a conference, one included studies submitted to a medical journal, and one included studies submitted to eight medical journals. Fifty‐seven papers included only drug studies, three only device studies, two drug and device studies and 13 included different types of interventions (e.g. drugs, devices, behavioral interventions). Thirty‐four papers included studies related to specific drug classes, 16 related to specific medical specialties or types of diseases (e.g. endocrinology), 10 related to a specific disease, three related to a specific type of device, 11 included all types of research studies, and one did not state the domain. Various aspects of medicine were covered, but 16 (21%) papers were restricted to psychiatric diseases or drugs and eight (11%) to cancer treatment. Fifty‐eight papers included only clinical trials, two only observational studies, and 15 both clinical trials and observational studies. Thirteen papers included only drug versus drug comparisons, eight only drug versus placebo, 49 mixed comparisons (e.g. drug versus drug, drug versus placebo) and five did not describe the kind of comparisons. The median number of included studies per paper was 105 (range: nine to 930). Of the 75 papers, 27 reported data on both favorable outcomes and risk of bias, 44 on favorable outcomes only, and four on risk of bias only. Twenty‐five papers were non‐industry funded, two were industry funded (Freemantle 2000; van Lent 2014), one was funded by both industry and non‐industry sources (Lynch 2007), 17 reported that they did not receive funding and 30 did not reported on funding.

Risk of bias in included studies

See: Figure 2; Figure 3.


'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included papers.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included papers.


'Risk of bias' summary: review authors' judgements about each risk of bias item for each included paper.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included paper.

Sixty‐eight papers had low risk of bias for the selection criteria for inclusion of studies, three were unclear and four had high risk. Twenty‐one papers had low risk of bias for the study inclusion process, 40 were unclear and 14 had high risk. Sixty‐six papers had low risk of bias from the search, one was unclear and eight had high risk. Thirty‐one papers had low risk of bias due to lack of control for bias in the studies, five were unclear and 39 had high risk. Fourteen papers were regarded as having an overall low risk of bias and 61 as a high risk of bias according to our criteria.

Effect of methods

See: Summary of findings for the main comparison Industry sponsored compared to non‐industry sponsored studies for research outcome

Favorable results: industry sponsored versus non‐industry sponsored studies

See: summary of findings Table for the main comparison

Twenty‐six papers, including 3081 studies (3062 drug studies and 19 device studies), reported on sponsorship and efficacy results; 25 could be combined in a pooled analysis. An analysis based on these 25 papers, including 2923 studies, found that industry sponsored studies more often had favorable efficacy results (e.g. those with significant P values) compared with non‐industry sponsored studies, risk ratio (RR): 1.27 (95% confidence interval (CI): 1.17 to 1.37), I2: 28% (Analysis 1.1). The paper that could not be included in the pooled analysis (Bhandari 2004), which had included 158 drug studies in general medicine, found similar results, odds ratio (OR): 1.6 (95% CI: 1.1 to 2.8).

Four papers, including 826 studies, did not find a difference in favorable harms results in industry sponsored studies compared with non‐industry sponsored studies, RR: 1.37 (95% CI: 0.64 to 2.93), I2: 96%. (Analysis 1.2). The results of one paper (Als‐Nielsen 2003), were opposite in direction to the other papers and resulted in the substantial heterogeneity. .

Favorable results: industry sponsorship by test treatment company versus industry sponsorship by comparator treatment company

Three papers, including 151 studies (all drug trials), compared efficacy results of trials sponsored by the manufacturer of the test treatment with trials sponsored by the manufacturer of the comparator treatment; two could be combined in a pooled analysis. An analysis based on these two papers (Bero 2007; Rattinger 2009), which included 131 industry sponsored trials of statins and thiazolidinediones, found that trials were much more likely to favor the test treatment when they were sponsored by the manufacturer of the test treatment than when they were sponsored by the manufacturer of the comparator treatment, RR: 3.88 (95% CI: 1.26 to 11.94), I2: 50% (Analysis 2.1). The paper that could not be included in the pooled analysis, which had included 20 selective serotonin reuptake inhibitor head‐to‐head trials, found that two trials favored the sponsor's drug, 18 had similar efficacy and none favored the comparator drug (Gartlehner 2010).

Favorable conclusions: industry sponsored versus non‐industry sponsored studies

See: summary of findings Table for the main comparison

Thirty‐two papers, including 5258 studies (4761 drug studies and 497 device studies), reported on sponsorship and conclusions, 29 of which could be combined in a pooled analysis. An analysis based on these 29 papers, including 4583 studies (4179 drug studies and 404 device studies), found that industry sponsored studies more often had favorable conclusions than non‐industry sponsored studies, RR: 1.34 (95% CI: 1.19 to 1.51), I2: 92% (Analysis 3.1). Three papers could not be included in the pooled analysis due to the reporting of data. Of these, one paper reporting on 301 psychiatric drug studies (Kelly 2006) found that industry sponsored studies more often had favorable conclusions than non‐industry sponsored studies (P < 0.001) and similar findings were reported in a paper of 59 trials of antipsychotics (P = 0.02) (Montgomery 2004). A paper on 315 gastroenterology trials (222 drug trials and 93 device trials) did not find a difference in conclusions between industry sponsored trials and non‐industry sponsored trials (industry: 86% favorable, non‐industry: 83% favorable; P = 0.57) (Brown 2006).

Favorable conclusions: industry sponsorship by test treatment company versus sponsorship by comparator treatment company

Five papers, including 348 drug trials, compared conclusions of studies sponsored by the manufacturer of the test treatment with studies sponsored by the manufacturer of the comparator treatment, and three could be combined in a pooled analysis. These three papers (Bero 2007; Heres 2006; Rattinger 2009) including 154 industry sponsored trials of statins, antipsychotics and thiazolidinediones, found that trials were much more likely to favor the test treatment when they were sponsored by the manufacturer of the test treatment than when they were sponsored by the manufacturer of the control treatment, RR: 5.92 (95% CI: 2.80 to 12.54). No heterogeneity was observed (Analysis 4.1). A paper including 138 psychiatric drug studies (Kelly 2006) had similar findings, RR 2.80 (95% CI: 2.02 to 3.88), and a paper on 56 non‐steroidal anti‐inflammatory drug (NSAID) trials (Rochon 1994), found that 16 trials favored the sponsor's drug, 40 concluded that the drugs had similar effect and none favored the comparator drug.

Effect size: industry sponsored versus non‐industry sponsored studies

Twenty‐four papers, including 1517 studies (1476 drug studies and 41 device studies), reported on sponsorship and effect size, but could not be pooled due to differences in reporting of data. The results were heterogeneous and are described in Table 1 below.

Table 1. Effect size in industry and non‐industry sponsored studies.

Paper ID

Study domain

Effect size of industry versus non‐industry studies

Efficacy

Als‐Nielsen 2003

370 drug RCTs in Cochrane reviews

Primary outcome. Mean z‐scores: industry: ‐1.48 (95% CI: ‐1.77 to ‐1.19); mixed: ‐1.77 (95% CI: ‐2.28 to ‐1.26); non‐industry: ‐1.20 (95% CI: ‐1.81 to ‐0.59); not stated: ‐1.20 (95% CI: ‐1,49 to ‐0.91) (P > 0.05).

Avni 2014

36 RCTs of antibiotics for pneumonia

No difference in mortality and clinical failure.

Barden 2006

176 acute pain and migraine drug RCTs

No difference in pain relief.

Clark 2002

19 RCTs of erythropoietin for cancer‐related anemia

Number of transfusions: industry: OR: 0.43 (95% CI: 0.35 to 0.54); non‐industry OR: 0.22 (95% CI: 0.11 to 0.45).

Corona 2014a

25 RCTs of testosterone for male sexual dysfunction

Erectile dysfunction. Standardized mean difference (SMD): industry: SMD: 1.36 (95% CI: 0.55 to 1.16); non‐industry: SMD: 0.33 (95% CI: 0.13 to 0.54) (P = 0.02).

Davis 2008

124 RCTs of 2nd generation versus 1st generation antipsychotics

Effect on psychotic symptoms (P = 0.57).

Djulbegovic 2013

126 oncology drug RCTs

Primary outcome: industry: OR/hazard ratio (HR): 0.61 (99% CI: 0.47 to 0.78); non‐industry OR/HR: 0.86 (99% CI: 0.74 to 1.00) (P = 0.003). Overall survival (P = 1.00).

Etter 2007

34 RCTs of nicotine replacement therapy

Effect: industry: OR: 1.90 (95% CI: 1.67 to 2.16); non‐industry: OR: 1.61 (95% CI: 1.43 to 1.80) (P = 0.06).

Freemantle 2000

105 RCTs of selective serotonin reuptake inhibitors versus alternative antidepressants

No difference in effect.

Jinapriya 2011

31 of prostaglandin analogues for open‐angle glaucoma

Effect on intraocular pressure (P = 0.83).

Killin 2014

14 RCTs of donepezil for Alzheimer's disease

Effect on cognitive scales: industry: SMD: 0.46 (95% CI: 0.38 to 0.54); non‐industry: SMD: 0.33 (95% CI: 0.18 to 0.48) (P = 0.13).

Lubowitz 2007

23 studies of chondrocyte implantation

No difference in effect on various outcomes.

Moncrieff 2003

9 RCTs of clozapine versus conventional antipsychotics

Psychotic symptoms: industry: SMD: ‐0.83 (95% CI: ‐1.06 to ‐0.61); non‐industry: SMD: ‐0.21 (95% CI: ‐0.34 to ‐0.07) (P < 0.001).

Naci 2014

183 statin RCTs

No difference in effect on mean change in LDL levels, after controlling for statin dose.

Popelut 2010

41 clinical studies of dental implants

Failure rates. Industry versus non‐industry: OR: 0.21 (95% CI: 0.12 to 0.38).

Rösner 2010

24 RCTs of acamprosate for alcohol dependence

Return to any drinking: industry: RR: 0.88 (95% CI: 0.80 to 0.97); mixed: RR: 0.84 (95% CI: 0.78 to 0.89); non‐industry: RR: 0.86 (95%: CI 0.81 to 0.91).

Rösner 2010a

26 RCTs of opioid antagonists for alcohol dependence

Return to any drinking: industry: RR: 0.90 (95% CI: 0.78 to 1.05); non‐industry: RR: 0.84 (95% CI: 0.77 to 0.91).

Vlad 2007

15 RCTs of glucosamine for osteoarthritis

Primary outcome: industry: SMD: 0.47 (95% CI: 0.24 to 0.70); non‐industry: SMD: 0.05 (95% CI: ‐0.32 to 0.41) (P = 0.05).

Zhang 2013

12 RCTs of 2nd generation versus 1st generation antipsychotics

Short‐term symptom reduction and response (P = 0.007 and P = 0.046).

Harms

Corona 2014

26 RCTs of testosterone therapy for men

Cardiovascular events: industry: OR: 1.07 (95% CI: 0.54 to 2.24); non‐industry: OR: 0.94 (95% CI: 0.39 to 2.24).

Kemmeren 2001

9 observational studies of 3rd generation versus 2nd generation oral contraceptives

Thrombosis: industry: OR: 1.3 (95% CI: 1.0 to 1.7); non‐industry: OR 2.3 (95% CI: 1.7 to 3.2).

Ma 2014

4 RCTs of fluoxetine for major depressive disorder

Harms: industry: OR: 2.34 (95% CI: 1.62 to 3.36); non‐industry: OR: 2.78 (95% CI:1.76 to 4.38).

Xu 2013

27 RCTs of testosterone therapy for men

Cardiovascular events: industry: OR 0.89 (95%: CI 0.50 to 1.60); non‐industry: OR 2.06 (95% CI: 1.34 to 3.17) (P = 0.03).

Efficacy and dosage

Sinyor 2012

58 industry head‐to‐head RCTs of antidepressants

Remission: sponsor's drug at higher dose OR: 1.28 (95% CI: 1.11 to 1.47) versus sponsor's drug at comparable or lower dose OR: 1.06 (95% CI: 0.96 to 1.17) (P = 0.04).

Most papers (19 out of 24) compared effect sizes for efficacy results. Twelve of these papers, including 1131 drug studies, did not find a difference in effect sizes for efficacy estimates between industry sponsored studies and non‐industry sponsored studies. In contrast, seven papers, including 262 studies (221 drug studies and 41 device studies) found higher effect sizes of efficacy estimates in industry sponsored studies. Two papers, including 30 drug studies, did not find a difference in effect size of harms between industry sponsored studies and non‐industry sponsored studies. In contrast, two papers, including 36 drug studies, found lower effect size of harms in industry sponsored studies. Lastly, a paper on 58 industry‐sponsored head‐to‐head trials of antidepressants (Sinyor 2012), found that the sponsor's antidepressants were often given at a higher dose than the comparator. Effects on remission were higher when the sponsor's drug was given at a higher dose, as compared to trials in which the sponsor's drug was given in comparable or lower dose OR: 1.28 (95% CI: 1.11 to 1.47) versus OR: 1.06 (95% CI: 0.96 to 1.17) (P = 0.04).

Risk of bias: industry sponsored versus non‐industry sponsored studies

Twelve papers, including 1660 studies (1482 drug studies and 178 device studies), compared risk of bias in industry versus non‐industry studies using six different composite quality scales (Brown, Cho, Cochrane, Jadad, PEDro or Sackett) and the results were heterogeneous. Seven papers did not find a difference in risk of bias between industry sponsored and non‐industry sponsored studies (Cho 1996; Clark 2002; Corona 2014; Jefferson 2009; Lynch 2007; Sung 2013; Vlad 2007), whereas five papers found lower risk of bias (i.e. higher methodological quality scores) in industry sponsored studies (Brown 2006; Djulbegovic 2000; Montgomery 2004; Pengel 2009; Perlis 2005a).   

Nine papers, including 913 drug trials, did not find a difference in risk of bias from sequence generation in industry sponsored trials compared with non‐industry sponsored trials, RR: 0.99 (95% CI: 0.78 to 1.27), I2: 73% (Analysis 5.1). Sixteen papers, including 1886 trials (1867 drug trials and 19 device trials), did not find a difference in risk of bias from concealment of allocation in industry sponsored trials compared with non‐industry sponsored trials, RR: 1.06 (95% CI: 0.85 to 1.31), I2: 71% (Analysis 5.2). Thirteen papers, including 1578 trials (1559 drug trials and 19 device trials), found that industry sponsored trials more often had low risk of bias from overall blinding compared with non‐industry sponsored trials, RR: 1.25 (95% CI: 1.05 to 1.50), I2: 72% (Analysis 5.3). Three papers, including 128 drug trials, did not find a difference in risk of performance bias (e.g. blinding of clinicians or patients) in industry sponsored trials compared with non‐industry sponsored trials, RR: 1.26 (95% CI: 0.60 to 2.62), I2: 66% (Analysis 5.4). Four papers, including 307 drug trials, found that industry sponsored trials more often had low risk of detection bias (e.g. blinding of outcome assessors) compared with non‐industry sponsored trials, RR: 1.47 (95% CI: 1.02 to 2.12). No heterogeneity was observed (Analysis 5.5). Six papers, including 416 drug trials, did not find a difference in risk of bias from loss to follow‐up in industry sponsored trials compared with non‐industry sponsored trials, RR: 1.05 (95% CI: 0.92 to 1.18), I2: 2% (Analysis 5.6). Two papers, including 193 drug trials, did not find a difference in risk of reporting bias in industry sponsored trials compared with non‐industry sponsored trials, RR: 1.49 (95% CI: 0.61 to 3.60), I2: 79% (Analysis 5.7).

Concordance between study results and conclusions: industry sponsored versus non‐industry sponsored studies

Six papers, including 751 drug studies, reported on concordance between study efficacy results (e.g. as judged by their P values) and conclusions. Industry sponsored studies were less concordant than non‐industry sponsored studies, RR: 0.83 (95% CI: 0.70 to 0.98), I2: 63% (Analysis 6.1). One paper (Alasbali 2009), including 39 drug studies, found markedly higher lack of concordance in industry studies than the other four papers, and this was the reason for the high heterogeneity between papers.

One paper, of 211 corticosteroid studies with statistically significant harms results, found that industry sponsored studies more often concluded that the drug was safe than non‐industry sponsored studies, RR: 3.68 (95% CI: 2.14 to 6.33) (Nieto 2007).

Subgroup analysis and investigation of heterogeneity

For efficacy results, the association between industry sponsorship and favorable results was stronger in papers with a low risk of bias than in those with a high risk of bias, RR: 1.46 (95% CI: 1.25 to 1.71) versus RR: 1.20 (95% CI: 1.11 to 1.30) (test for subgroup differences P = 0.03) (Analysis 7.1). For harms results, the association between industry sponsorship and favorable harms results differed in papers with a low risk of bias compared with those with a high risk of bias, RR: 0.82 (95%: 0.72 to 0.93) versus RR: 1.87 (95% CI: 1.54 to 2.27) (test for subgroup differences P < 0.0001) (Analysis 7.2). For conclusions, the differences between the groups went in the same direction as for efficacy results, RR: 1.42 (95% CI: 1.12 to 1.79) versus RR: 1.32 (95% CI: 1.15 to 1.50) (test for subgroup differences P = 0.60) (Analysis 7.3).

For efficacy results, the association between industry sponsorship and favorable results were opposite in direction in drug studies compared with device studies, RR: 1.27 (95% CI: 1.17 to 1.38) versus RR: 0.50 (95% CI: 0.26 to 0.97) (test for subgroup differences P = 0.006) (Analysis 7.4). However, the analysis only included 19 device studies, of which only three were non‐industry. We did not find a difference in the association between sponsorship and conclusions in drug studies compared with device studies (test for subgroup differences P = 0.98) (Analysis 7.5) or between sponsorship and efficacy results or conclusion in studies limited to specific treatments or diseases compared with studies of mixed domains (test for subgroup differences P = 0.67 and P = 0.49 ) (Analysis 7.6; Analysis 7.8). However, for harms results the association between industry sponsorship and favorable harms results differed in papers with mixed study domain compared with those of specific treatments or diseases, RR: 0.82 (95%: 0.72 to 0.93) versus RR: 1.87 (95% CI: 1.54 to 2.27) (test for subgroup differences P < 0.0001) (Analysis 7.7).

Sensitivity analysis

Our re‐analyses of the outcomes using variations in definition of sponsorship categories gave similar results as our main analyses for efficacy results, harms results and conclusions (Analysis 8.1; Analysis 8.2; Analysis 8.3). Our analyses, taking into account papers that adjusted for confounding, based on pooling adjusted odds ratios, confirmed our findings that industry sponsored trials compared with non‐industry sponsored trials more often had favorable results, OR: 3.15 (95% CI: 2.07 to 4.80), I2: 0% and favorable conclusions, OR: 3.13 (95% CI: 1.66 to 5.93), I2: 38% (Analysis 8.4; Analysis 8.5). Similarly, sensitivity analyses using a fixed‐effect model rather than a random‐effects model did not affect our results (Analysis 8.6; Analysis 8.8; Analysis 8.9; Analysis 8.10), except for harms results where they changed from RR: 1.37 (95% CI: 0.64 to 2.93) to RR: 1.29 (95% CI: 1.15 to 1.46) (Analysis 8.7). When we excluded papers sampling unpublished studies, it did not affect our analysis on favorable conclusions (Analysis 8.11). The same was found when we limited our analyses to papers from specific domains (i.e. papers on specific treatments or diseases where none of the other included papers were related to the same domain) (Analysis 8.12; ; Analysis 8.14; Analysis 8.15; Analysis 8.16), except for harms results where they changed from RR: 1.37 (95% CI: 0.64 to 2.93) to RR: 1.87 (95% CI: 1.54 to 2.27) (Analysis 8.13). Lastly, if we included the two additional papers that were only published as letters (Mandelkern 1999; Thomas 2002) our analysis on the association between sponsorship and favorable conclusions gave similar results RR: 1.35 (95% CI: 1.20 to 1.52), I2: 91%.

Discussion

Summary of main results

We found that drug and device studies sponsored by the manufacturing company more often had favorable efficacy results (e.g. those with statistically significant results, usually defined using P values) and conclusions than those that were sponsored by other sources. The findings were consistent across a wide range of diseases and treatments. We did not find any differences in harms results and risk of bias of drug and device trials sponsored by industry compared with non‐industry sponsored trials, except in relation to blinding, where industry sponsored trials seemed to have lower risk of bias. Industry sponsored studies also had less concordance between results and conclusions than non‐industry sponsored studies. The evidence from device studies was limited due to fewer data, but the association between sponsorship and favorable conclusions was similar to drug studies.

Reasons for observed heterogeneity

For the association between sponsorship and favorable efficacy results of drug and device studies the data had acceptable heterogeneity, but heterogeneity for the association between sponsorship and harms results and study conclusions was substantial with an I2 of 96% and 92%, respectively.

The reason for the large heterogeneity related to harms results is attributed to the results of the Als‐Nielsen paper (Als‐Nielsen 2003), that was opposite in direction to the other papers (i.e. industry had less favorable harms results as opposed to more favorable harms results) (Halpern 2005; Kemmeren 2001; Nieto 2007), and the interaction tests in the subgroup analyses were statistically significant. The Als‐Nielsen paper differs in some aspects from the three other included papers. First, it samples trials from various therapeutic areas, whereas the other papers each deal with harms results of single‐drug classes (HIV drugs, oral contraceptives and inhaled corticosteroids). Second, the three papers related to single‐drug classes had harms results as their primary outcome and only included studies with quantitative data, but Als‐Nielsen had harms as a secondary outcome and also included trials without quantitative harms data. The number of trials without harms data was high, particularly in the non‐industry group, 28% and 52%, respectively. Third, Als‐Nielsen only included trials, whereas Halpern 2005 included both trials and observational studies and Kemmeren 2001 and Nieto 2007 only included observational studies. Finally, we assessed Als‐Nielsen 2003 as having an overall low risk of bias, compared to high risk of bias in the other three papers.

In relation to the substantial heterogeneity for study conclusions, one reason was likely that the coding of favorable results was similar across the different papers, using statistical significance as the cut‐off, but coding varied for favorable conclusions. Some papers did not describe what they considered a favorable conclusion and this would involve some judgement. Others used scales, but for similar scales the cut‐off varied between papers. For example, on the same six‐point scale one paper used four as the cut‐off (Djulbegovic 2000) and another used six as the cut‐off (Als‐Nielsen 2003).

Also, the proportion of studies with favorable conclusions in the non‐industry sponsored group might have contributed to the size of the association and thereby the heterogeneity. For example, while the Chard and Liss papers (Chard 2000; Liss 2006) had a similar proportion of favorable industry sponsored studies (both 98%), they reported very different proportions of favorable non‐industry sponsored studies (32% and 97%) and this explains why the risk ratios reported in the two studies were not the same: RR: 3.03 in Liss and RR: 1.01 in Chard. Variations in the definition of favorable conclusions might explain why the risk ratios reported in the two papers were not similar. For example, in the Chard paper, a conclusion was coded as favorable if the study authors supported the use of the treatment, even in the absence of a statistically significant result.

Our subgroup analyses stratifying papers in relation to risk of bias (low versus high), type of intervention (drug versus device) or study domain (mixed versus specific treatments or diseases) did not explain the observed heterogeneity, though this was a simplistic comparison and other factors might also contribute to heterogeneity.

We found mixed results on the relationship between sponsorship and effect size, with most papers not finding a difference. All but one of these papers were restricted to specific treatments, which may explain the different findings. A recent study of systematic reviews of nine different drugs found that the influence of reporting biases on effect sizes varied considerably between drugs (Hart 2012). Furthermore, one paper found that even when adjusting for effect size, industry sponsored studies more often had favorable conclusions, compared with non‐industry sponsored studies (Als‐Nielsen 2003). Therefore, while the direction of the relationship between sponsorship and favorable outcomes was consistent, the size of the effect likely varies depending on the type of treatment or treated condition.

Reasons for favorable outcomes in industry sponsored studies

The pharmaceutical and medical device industries have strong interests in scientific publications that present their products positively, as publications are the basis of regulatory, purchasing, and medical decisions. These interests can influence the design, conduct and publication of studies in ways that make the sponsor’s product appear better than the comparator product (Bero 1996).

Several possible factors can explain the relationship between industry sponsorship and favorable outcomes. It has been argued that since many industry sponsored studies are undertaken to fulfill regulatory requirements, industry sponsored studies could have a lower risk of bias than non‐industry sponsored studies (Rosefsky 2003). Even if this were true, it would not explain the association of industry sponsorship and favorable efficacy results and conclusions. In addition, we did not find evidence for differences in risk of bias except in relation to blinding, where industry sponsored trials tended to have a lower risk of bias, even when restricted to head‐to‐head trials (Bero 2007). The papers comparing blinding between trials with different sponsorship often used a description of double blinding as an indicator for low risk of bias. Double blinding is an inconsistent term and does not ensure that, for example, outcome assessors are blinded (Devereaux 2001). The more frequent use of double blinding may therefore be a reporting issue, with industry trials being better reported. This is further substantiated by the fact that nearly all the papers finding a higher methodological quality score in industry studies used the Jadad scale, a scale which has been criticized for having more focus on the quality of reporting than on methodological quality (Lundh 2008).   

A few papers assessing a more specific definition of blinding related to performance bias and detection bias also found that industry sponsored studies had lower risk of bias. Evidence also suggests that for non‐industry trials, companies may prevent proper blinding by restricting access to placebo drugs (Christensen 2012), and therefore differences in adequate blinding may be real. In addition, double blinding can be used as a proxy for low risk of bias and trials without double blinding are on average more likely to have favorable results (Pildal 2007). The effect of this bias is in the opposite direction of our findings, as it would lead to industry sponsored studies having less favorable results and conclusions, and our findings can therefore, not be explained by differences in risk of bias related to blinding between industry and non‐industry sponsored studies.

Another possible explanation for our findings could be that industry studies have larger sample sizes, and would have a higher chance of achieving statistically significant results. Although industry trials seem in general to be of larger size (Als‐Nielsen 2003; Booth 2008; Bourgeois 2010; Djulbegovic 2013; Etter 2007; Flacco 2015; Perlis 2005a), when we restricted our analysis to studies controlling for sample size and other confounders, the relationship between industry sponsorship and favorable results or conclusions was still present.

Industry representatives argues that the trials they sponsor are more likely to have favorable outcomes because they fund research that has a high chance of achieving success (Palmer 2003). However, when independent investigators conduct non‐industry sponsored trials, they in most cases test treatments that have been approved based on favorable industry trial results. Non‐industry sponsored trials would therefore also be expected to achieve successful results, unless they are designed to answer different questions than industry sponsored trials. For example, non‐industry sponsored studies may test a new treatment against a well‐established treatment, while industry sponsored studies might test the new treatment against placebo or against an outdated, inferior treatment.

Accordingly, it seems most plausible that industry achieves overly positive results through a variety of biasing choices in the design, conduct and reporting of their studies. For example, industry protocols might include inferior comparators that will increase the chance of their product’s success. Djulbegovic and colleagues (Djulbegovic 2003) have argued that industry sponsored studies violate equipoise by choosing inferior competing treatment alternatives. Previous studies have found that industry sponsored trials more often use placebo control (Als‐Nielsen 2003Djulbegovic 2000; Dunn 2013; Estellat 2012Katz 2006Lathyris 2010), active comparators in inferior doses (Rochon 1994Safer 2002; Sinyor 2012), or inappropriate administration of the drugs (Johansen 1999). Industry may also selectively choose less clinical relevant outcomes as their primary outcome in order to get a higher chance of achieving an effect. For example, in the paper by Djulbegovic (Djulbegovic 2013) industry sponsored trials had higher effect size than non‐industry sponsored trials on primary outcomes, but not overall survival. This could also be one of the possible explanations as to why industry sponsored trials more often have favorable results and conclusions while the effect sizes are often similar when comparing similar outcomes.

Industry sponsored studies may also be biased in the coding of events and their data analysis (Furukawa 2004Psaty 2008Psaty 2010). Industry and its sponsored investigators also may selectively report favorable outcomes, fail to publish whole studies with unfavorable results, or publish studies with favorable results multiple times (Chan 2004Dwan 2008Gøtzsche 2011McGauran 2010Melander 2003Rising 2008Vedula 2009). While such biases in analyses and reporting have been documented in a number of cases, the papers included in this review focused on comparisons of published studies. Only two papers (Killin 2014; Naci 2014) included in our review compared risk of selective reporting between industry sponsored trials and non‐industry sponsored trials and found no difference. However, confidence intervals were wide and the analysis was limited to two types of drugs (donepezil for Alzheimer's disease and statins). Therefore, we are unable to determine the extent to which selective analysis or reporting contribute to our findings. Similarly, we found no difference in harms results between industry sponsored and non‐industry sponsored studies. Under‐reporting of harms seems to be a major problem in both industry and non‐industry sponsored studies with 28% of trials included in Als‐Nielsen 2003 not reporting any harms data, which is in line with a recent systematic review that found that a median of 54% studies do not report harms data (Golder 2016).

Favorable conclusions in industry‐sponsored trials may also be reached by over‐interpreting results and use of spin in conclusions (Boutron 2010). We found that industry sponsored studies had less concordance between results and conclusions compared with non‐industry sponsored studies, suggesting that conclusions of industry sponsored studies are less reliable.

It should also be noted that some studies in the non‐industry group likely had authors with conflicts of interest related to the pharmaceutical or device industry, which may have influenced their interpretation of study results (Stelfox 1998; Wang 2010), thereby diluting the measured effect of industry bias on study conclusions. Also, we coded studies as non‐industry sponsored if they did not state who sponsored the study. As some of these studies were likely industry sponsored, this misclassification will have led to similar bias towards the null. However, in our sensitivity analyses, we excluded studies without sponsorship statements and did not see a change in results.

Further evidence for industry bias stems from our comparison of studies sponsored by the manufacturer of the test treatment with those sponsored by the manufacturer of the control treatment. These studies had the advantage of comparing like with like, as they are restricted to specific drug classes or types of devices and have similar methodologies. Though limited to only three papers on drug trials, the findings show associations that are stronger than the comparison between industry and non‐industry sponsored studies. These comparisons are restricted to drugs competing for the same market, which may put pressure on companies to influence outcomes to a greater degree than what is needed in placebo‐controlled trials to present the drug in a good light.

In sum, the industry bias associated with favorable efficacy results and conclusions may be mediated by factors other than traditional measures of the risk of bias (e.g. lack of concealment of allocation, blinding and dropout) and sample size. This industry bias may be partially mediated by such factors as the choice of comparators, dosing and timing of comparisons, choice of outcomes, selective analysis, and selective reporting.

Quality of the evidence

The majority of included papers were regarded as having a high risk of bias. Many lacked information on study conduct and did not control for confounders that could influence the relationship. Nevertheless, we did identify 14 papers with low risk of bias and analyses restricted to these papers actually strengthened the relationship between industry sponsorship and conclusions. In general, there is convincing and consistent evidence for the existence of an industry bias in studies; however, the body of evidence for device studies is not as strong as for drug studies. While many papers, including studies of devices and other interventions, have been published in the surgical field (Amiri 2014; Cunningham 2007; Khan 2008; Leopold 2003; Roach 2008; Shah 2005; Sun 2013; Yao 2007), the papers do not report separate data for device studies.

Potential biases in the review process

We did a comprehensive search, our methods were based on pre‐specified criteria in a protocol as outlined in Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (Higgins 2011a), and this updated review has substantially increased the number of included papers from our previous review (Lundh 2012). Nevertheless, there are some limitations. First, we decided only to include published papers. In our first version of the review (Lexchin 2003), we found problems with the completeness and quality of the data in conference abstracts and unstructured letters and therefore decided not to include them in this review. A comment to the previous version of this review (Lundh 2012), suggested that the exclusion of conference abstracts and letters could have introduced publication bias. In this update, we decided to include these papers in a sensitivity analysis, which gave similar results. However, we could only include quantitative data from two papers and we might have missed relevant papers. We expect that due to the high number of included papers such unidentified papers would not have major impact on our results. Due to the heterogeneity of included papers we decided not to assess publication bias using a funnel plot as it would be difficult to interpret.

Second, our assessment of risk of bias in the included papers was not based on validated criteria similar to 'Risk of bias' assessment for clinical trials (Higgins 2011b). As no validated assessment tools exist for these type of papers, we developed our own criteria and included items similar to assessment tools for systematic reviews (Oxman 1991; Shea 2007).

Third, one item not included in our assessment of risk of bias in the papers was whether coders of outcomes were blinded to the sponsorship status of the studies. If these types of papers were undertaken by authors with a particular view on the drug industry, knowledge of sponsorship status could introduce bias in the assessment of whether outcomes were favorable, particularly for conclusions, as this is an outcome that is qualitative in nature. Some of the included papers were written by authors who had published multiple times in the area, and as such could be at increased risk of bias. These papers used coders who were both blinded and unblinded to the sponsorship status of the studies. The agreement in coding was high, suggesting a lack of bias (Als‐Nielsen 2003; Bero 2007; Kjaergard 2002). Likewise, all review authors (AL, BM, JL, JS, LB) have published several times in the field and one review author (LB) is the author of four of the included papers (Bero 2007; Cho 1996; Rasmussen 2009; Rattinger 2009), which could have introduced bias. Because of the way data were presented in the papers, it was not possible to blind our data extraction process. None of the data extractors were co‐authors of the included papers. Furthermore, our data extraction of outcomes did not involve any qualitative interpretation as we extracted actual numbers.

Fourth, if the papers included in this review included some of the same studies, their findings would not be independent. Furthermore, some papers included some of the same studies (Corona 2014; Xu 2013), but had different results, which could be explained by differences in inclusion criteria and data extraction. In the majority of cases, it was not possible to assess the potential overlap of studies as most papers did not provide a reference list of included studies and we rarely had access to raw data. Instead we undertook sensitivity analyses restricted to papers on specific treatments or diseases where none of the other included papers were related to that domain. The analyses gave similar results, however with wider confidence intervals.

Agreements and disagreements with other studies or reviews

Our results are in agreement with previous systematic reviews (Bekelman 2003; Lexchin 2003; Schott 2010a; Sismondo 2008a), though the risk ratios for the associations are less than previous quantitative estimates, but similar to our previous estimates (Lundh 2012). Previous reviews did not distinguish between favorable efficacy results or conclusions, but looked at the association between sponsorship and outcomes. Bekelman 2003 found OR 3.60 (95% CI: 2.63 to 4.91) and Lexchin 2003 OR 4.05 (95% CI: 2.98 to 5.51). Translated to odds ratios, we found 2.05 (95% CI: 1.66 to 2.52) for results and 2.69 (95% CI 2.04 to 3.54) for conclusions in our review. This difference could be due to chance or it could be because the earlier reviews also included pharmacoeconomic analyses, non‐drug studies, unstructured letters and conference abstracts. It is also possible that the degree of industry bias has diminished over time, for example with a decrease in reporting bias due to trial registration. One paper on oncology drug trials (Djulbegovic 2013), suggested that the treatment effect size between industry sponsored and non‐industry trials became more similar over time. However, the analysis included both published and unpublished trials and did not investigate the association between sponsorship and results or conclusions over time. In contrast, a recent paper found that reporting bias is also prevalent in registered trials, particular in industry sponsored trials (Jones 2013). Second, one of the most recent papers (Flacco 2015) sampled drug trials published in 2011 and found OR: 2.8 (95% CI: 1.6 to 4.7) for results, suggesting that industry bias has not changed over time. 

For harms results one other systematic review (Golder 2008) has been published. Due to the anticipated heterogeneity of the data, the authors decided not to perform a meta‐analysis. The results of the review are in line with our findings of no differences in harms results between industry sponsored and non‐industry sponsored studies, but there are large variation in findings among individual papers.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included papers.
Figures and Tables -
Figure 2

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included papers.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included paper.
Figures and Tables -
Figure 3

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included paper.

Comparison 1 Results: Industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with favorable efficacy results.
Figures and Tables -
Analysis 1.1

Comparison 1 Results: Industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with favorable efficacy results.

Comparison 1 Results: Industry sponsored versus non‐industry sponsored studies, Outcome 2 Number of studies with favorable harms results.
Figures and Tables -
Analysis 1.2

Comparison 1 Results: Industry sponsored versus non‐industry sponsored studies, Outcome 2 Number of studies with favorable harms results.

Comparison 2 Results: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company, Outcome 1 Number of studies with favorable test treatment efficacy results.
Figures and Tables -
Analysis 2.1

Comparison 2 Results: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company, Outcome 1 Number of studies with favorable test treatment efficacy results.

Comparison 3 Conclusions: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with favorable conclusions.
Figures and Tables -
Analysis 3.1

Comparison 3 Conclusions: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with favorable conclusions.

Comparison 4 Conclusions: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company, Outcome 1 Number of studies with favorable test treatment conclusions.
Figures and Tables -
Analysis 4.1

Comparison 4 Conclusions: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company, Outcome 1 Number of studies with favorable test treatment conclusions.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with low risk of bias from sequence generation.
Figures and Tables -
Analysis 5.1

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with low risk of bias from sequence generation.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 2 Number of studies with low risk of bias from concealment of allocation.
Figures and Tables -
Analysis 5.2

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 2 Number of studies with low risk of bias from concealment of allocation.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 3 Number of studies with low risk of bias from blinding‐overall.
Figures and Tables -
Analysis 5.3

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 3 Number of studies with low risk of bias from blinding‐overall.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 4 Number of studies with low risk from blinding‐performance bias.
Figures and Tables -
Analysis 5.4

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 4 Number of studies with low risk from blinding‐performance bias.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 5 Number of studies with low risk from blinding‐detection bias.
Figures and Tables -
Analysis 5.5

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 5 Number of studies with low risk from blinding‐detection bias.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 6 Number of studies with low risk of bias from loss to follow‐up.
Figures and Tables -
Analysis 5.6

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 6 Number of studies with low risk of bias from loss to follow‐up.

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 7 Number of studies with low risk of bias from selective outcome reporting.
Figures and Tables -
Analysis 5.7

Comparison 5 Risk of bias: industry sponsored versus non‐industry sponsored studies, Outcome 7 Number of studies with low risk of bias from selective outcome reporting.

Comparison 6 Concordance between study results and conclusions: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with concordant study results and conclusions.
Figures and Tables -
Analysis 6.1

Comparison 6 Concordance between study results and conclusions: industry sponsored versus non‐industry sponsored studies, Outcome 1 Number of studies with concordant study results and conclusions.

Comparison 7 Subgroup analysis, Outcome 1 Number of studies with favorable efficacy results, stratified by risk of bias.
Figures and Tables -
Analysis 7.1

Comparison 7 Subgroup analysis, Outcome 1 Number of studies with favorable efficacy results, stratified by risk of bias.

Comparison 7 Subgroup analysis, Outcome 2 Number of studies with favorable harms results, stratified by risk of bias.
Figures and Tables -
Analysis 7.2

Comparison 7 Subgroup analysis, Outcome 2 Number of studies with favorable harms results, stratified by risk of bias.

Comparison 7 Subgroup analysis, Outcome 3 Number of studies with favorable conclusions, stratified by risk of bias.
Figures and Tables -
Analysis 7.3

Comparison 7 Subgroup analysis, Outcome 3 Number of studies with favorable conclusions, stratified by risk of bias.

Comparison 7 Subgroup analysis, Outcome 4 Number of studies with favorable efficacy results, stratified by type of intervention.
Figures and Tables -
Analysis 7.4

Comparison 7 Subgroup analysis, Outcome 4 Number of studies with favorable efficacy results, stratified by type of intervention.

Comparison 7 Subgroup analysis, Outcome 5 Number of studies with favorable conclusions, stratified by type of intervention.
Figures and Tables -
Analysis 7.5

Comparison 7 Subgroup analysis, Outcome 5 Number of studies with favorable conclusions, stratified by type of intervention.

Comparison 7 Subgroup analysis, Outcome 6 Number of studies with favorable efficacy results, stratified by type of domain.
Figures and Tables -
Analysis 7.6

Comparison 7 Subgroup analysis, Outcome 6 Number of studies with favorable efficacy results, stratified by type of domain.

Comparison 7 Subgroup analysis, Outcome 7 Number of studies with favorable harms results, stratified by type of domain.
Figures and Tables -
Analysis 7.7

Comparison 7 Subgroup analysis, Outcome 7 Number of studies with favorable harms results, stratified by type of domain.

Comparison 7 Subgroup analysis, Outcome 8 Number of studies with favorable conclusions, stratified by type of domain.
Figures and Tables -
Analysis 7.8

Comparison 7 Subgroup analysis, Outcome 8 Number of studies with favorable conclusions, stratified by type of domain.

Comparison 8 Sensitivity analysis, Outcome 1 Number of studies with favorable efficacy results, sponsorship recoded.
Figures and Tables -
Analysis 8.1

Comparison 8 Sensitivity analysis, Outcome 1 Number of studies with favorable efficacy results, sponsorship recoded.

Comparison 8 Sensitivity analysis, Outcome 2 Number of studies with favorable harms results, sponsorship recoded.
Figures and Tables -
Analysis 8.2

Comparison 8 Sensitivity analysis, Outcome 2 Number of studies with favorable harms results, sponsorship recoded.

Comparison 8 Sensitivity analysis, Outcome 3 Number of studies with favorable conclusions, sponsorship recoded.
Figures and Tables -
Analysis 8.3

Comparison 8 Sensitivity analysis, Outcome 3 Number of studies with favorable conclusions, sponsorship recoded.

Comparison 8 Sensitivity analysis, Outcome 4 Number of studies with favorable efficacy results, analysis adjusted for confounders.
Figures and Tables -
Analysis 8.4

Comparison 8 Sensitivity analysis, Outcome 4 Number of studies with favorable efficacy results, analysis adjusted for confounders.

Comparison 8 Sensitivity analysis, Outcome 5 Number of studies with favorable conclusions, analysis adjusted for confounders.
Figures and Tables -
Analysis 8.5

Comparison 8 Sensitivity analysis, Outcome 5 Number of studies with favorable conclusions, analysis adjusted for confounders.

Comparison 8 Sensitivity analysis, Outcome 6 Number of studies with favorable efficacy results, fixed‐effect model.
Figures and Tables -
Analysis 8.6

Comparison 8 Sensitivity analysis, Outcome 6 Number of studies with favorable efficacy results, fixed‐effect model.

Comparison 8 Sensitivity analysis, Outcome 7 Number of studies with favorable harms results, fixed‐effect model.
Figures and Tables -
Analysis 8.7

Comparison 8 Sensitivity analysis, Outcome 7 Number of studies with favorable harms results, fixed‐effect model.

Comparison 8 Sensitivity analysis, Outcome 8 Number of studies with favorable test treatment efficacy results, fixed‐effect model.
Figures and Tables -
Analysis 8.8

Comparison 8 Sensitivity analysis, Outcome 8 Number of studies with favorable test treatment efficacy results, fixed‐effect model.

Comparison 8 Sensitivity analysis, Outcome 9 Number of studies with favorable conclusions, fixed‐effect model.
Figures and Tables -
Analysis 8.9

Comparison 8 Sensitivity analysis, Outcome 9 Number of studies with favorable conclusions, fixed‐effect model.

Comparison 8 Sensitivity analysis, Outcome 10 Number of studies with favorable test treatment conclusions, fixed‐effect model.
Figures and Tables -
Analysis 8.10

Comparison 8 Sensitivity analysis, Outcome 10 Number of studies with favorable test treatment conclusions, fixed‐effect model.

Comparison 8 Sensitivity analysis, Outcome 11 Number of studies with favorable conclusions, papers with unpublished studies excluded.
Figures and Tables -
Analysis 8.11

Comparison 8 Sensitivity analysis, Outcome 11 Number of studies with favorable conclusions, papers with unpublished studies excluded.

Comparison 8 Sensitivity analysis, Outcome 12 Number of studies with favorable efficacy results, restricted to specific domains.
Figures and Tables -
Analysis 8.12

Comparison 8 Sensitivity analysis, Outcome 12 Number of studies with favorable efficacy results, restricted to specific domains.

Comparison 8 Sensitivity analysis, Outcome 13 Number of studies with favorable harms results, restricted to specific domains.
Figures and Tables -
Analysis 8.13

Comparison 8 Sensitivity analysis, Outcome 13 Number of studies with favorable harms results, restricted to specific domains.

Comparison 8 Sensitivity analysis, Outcome 14 Number of studies with favorable test treatment efficacy results, restricted to specific domains.
Figures and Tables -
Analysis 8.14

Comparison 8 Sensitivity analysis, Outcome 14 Number of studies with favorable test treatment efficacy results, restricted to specific domains.

Comparison 8 Sensitivity analysis, Outcome 15 Number of studies with favorable conclusions, restricted to specific domains.
Figures and Tables -
Analysis 8.15

Comparison 8 Sensitivity analysis, Outcome 15 Number of studies with favorable conclusions, restricted to specific domains.

Comparison 8 Sensitivity analysis, Outcome 16 Number of studies with favorable test treatment conclusions, restricted to specific domains.
Figures and Tables -
Analysis 8.16

Comparison 8 Sensitivity analysis, Outcome 16 Number of studies with favorable test treatment conclusions, restricted to specific domains.

Summary of findings for the main comparison. Industry sponsored compared to non‐industry sponsored studies for research outcome

Results and conclusions: Industry sponsored compared to non‐industry sponsored studies

Patient or population: industry sponsorship and study results
Intervention: industry sponsored studies
Comparator: non‐industry sponsored studies

Outcomes

Illustrative comparative risks* (95% CI)

Risk ratio
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Non‐industry sponsored studies

Industry sponsored studies

Number of studies with favorable efficacy results

502 per 1000

638 per 1000
(588 to 688)

1.27
(1.17 to 1.37)

25 papers including 2923 studies

⊕ ⊕ ⊕ ○
MODERATE

Upgraded as control for confounders and analysis of low risk of bias papers gave similar results.

Number of studies with favorable harms results

474 per 1000

649 per 1000
(303 to 1388)

1.37
(0.64 to 2.93)

4 papers including 826 studies

⊕ ○ ○ ○
VERY LOW

Downgraded due to substantial heterogeneity (inconsistency) and wide confidence intervals (imprecision).

Number of studies with favorable conclusions

644 per 1000

863 per 1000
(766 to 972)

1.34
(1.19 to 1.51)

29 papers including 4583 studies

⊕ ⊕ ○ ○
LOW

Downgraded due to substantial heterogeneity (inconsistency).

Upgraded as control for confounders and analysis of low risk of bias papers gave similar results.

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

The assumed risk of the control group (i.e. non‐industry group) was calculated as the mean risk (i.e. number of studies with favorable results divided by total number of studies).

Figures and Tables -
Summary of findings for the main comparison. Industry sponsored compared to non‐industry sponsored studies for research outcome
Comparison 1. Results: Industry sponsored versus non‐industry sponsored studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable efficacy results Show forest plot

25

2923

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.17, 1.37]

2 Number of studies with favorable harms results Show forest plot

4

826

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.64, 2.93]

Figures and Tables -
Comparison 1. Results: Industry sponsored versus non‐industry sponsored studies
Comparison 2. Results: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable test treatment efficacy results Show forest plot

2

131

Risk Ratio (M‐H, Random, 95% CI)

3.88 [1.26, 11.94]

Figures and Tables -
Comparison 2. Results: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company
Comparison 3. Conclusions: industry sponsored versus non‐industry sponsored studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable conclusions Show forest plot

29

4583

Risk Ratio (M‐H, Random, 95% CI)

1.34 [1.19, 1.51]

Figures and Tables -
Comparison 3. Conclusions: industry sponsored versus non‐industry sponsored studies
Comparison 4. Conclusions: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable test treatment conclusions Show forest plot

3

154

Risk Ratio (M‐H, Random, 95% CI)

5.92 [2.80, 12.54]

Figures and Tables -
Comparison 4. Conclusions: Industry sponsorship by test treatment company versus sponsorship by comparator treatment company
Comparison 5. Risk of bias: industry sponsored versus non‐industry sponsored studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with low risk of bias from sequence generation Show forest plot

9

913

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.78, 1.27]

2 Number of studies with low risk of bias from concealment of allocation Show forest plot

16

1886

Risk Ratio (M‐H, Random, 95% CI)

1.06 [0.85, 1.31]

3 Number of studies with low risk of bias from blinding‐overall Show forest plot

13

1578

Risk Ratio (M‐H, Random, 95% CI)

1.25 [1.05, 1.50]

4 Number of studies with low risk from blinding‐performance bias Show forest plot

3

128

Risk Ratio (M‐H, Random, 95% CI)

1.26 [0.60, 2.62]

5 Number of studies with low risk from blinding‐detection bias Show forest plot

4

307

Risk Ratio (M‐H, Random, 95% CI)

1.47 [1.02, 2.12]

6 Number of studies with low risk of bias from loss to follow‐up Show forest plot

6

416

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.92, 1.18]

7 Number of studies with low risk of bias from selective outcome reporting Show forest plot

2

193

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.61, 3.60]

Figures and Tables -
Comparison 5. Risk of bias: industry sponsored versus non‐industry sponsored studies
Comparison 6. Concordance between study results and conclusions: industry sponsored versus non‐industry sponsored studies

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with concordant study results and conclusions Show forest plot

6

751

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.70, 0.98]

Figures and Tables -
Comparison 6. Concordance between study results and conclusions: industry sponsored versus non‐industry sponsored studies
Comparison 7. Subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable efficacy results, stratified by risk of bias Show forest plot

25

2923

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.17, 1.37]

1.1 High risk of bias

20

2107

Risk Ratio (M‐H, Random, 95% CI)

1.20 [1.11, 1.30]

1.2 Low risk of bias

5

816

Risk Ratio (M‐H, Random, 95% CI)

1.46 [1.25, 1.71]

2 Number of studies with favorable harms results, stratified by risk of bias Show forest plot

4

826

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.64, 2.93]

2.1 High risk of bias

3

561

Risk Ratio (M‐H, Random, 95% CI)

1.87 [1.54, 2.27]

2.2 Low risk of bias

1

265

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.72, 0.93]

3 Number of studies with favorable conclusions, stratified by risk of bias Show forest plot

29

4583

Risk Ratio (M‐H, Random, 95% CI)

1.34 [1.19, 1.51]

3.1 High risk of bias

23

3515

Risk Ratio (M‐H, Random, 95% CI)

1.32 [1.15, 1.50]

3.2 Low risk of bias

6

1068

Risk Ratio (M‐H, Random, 95% CI)

1.42 [1.12, 1.79]

4 Number of studies with favorable efficacy results, stratified by type of intervention Show forest plot

25

2923

Risk Ratio (M‐H, Random, 95% CI)

1.26 [1.15, 1.38]

4.1 Drug studies

25

2904

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.17, 1.38]

4.2 Device studies

1

19

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.26, 0.97]

5 Number of studies with favorable conclusions, stratified by type of intervention Show forest plot

29

4583

Risk Ratio (M‐H, Random, 95% CI)

1.33 [1.18, 1.49]

5.1 Drug studies

27

4179

Risk Ratio (M‐H, Random, 95% CI)

1.33 [1.17, 1.52]

5.2 Device studies

4

404

Risk Ratio (M‐H, Random, 95% CI)

1.33 [1.13, 1.57]

6 Number of studies with favorable efficacy results, stratified by type of domain Show forest plot

25

2923

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.17, 1.37]

6.1 Specific treatments or diseases

20

1845

Risk Ratio (M‐H, Random, 95% CI)

1.27 [1.13, 1.42]

6.2 Mixed domain

5

1078

Risk Ratio (M‐H, Random, 95% CI)

1.31 [1.18, 1.46]

7 Number of studies with favorable harms results, stratified by type of domain Show forest plot

4

826

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.64, 2.93]

7.1 Specific treatments or diseases

3

561

Risk Ratio (M‐H, Random, 95% CI)

1.87 [1.54, 2.27]

7.2 Mixed study domain

1

265

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.72, 0.93]

8 Number of studies with favorable conclusions, stratified by type of domain Show forest plot

29

4583

Risk Ratio (M‐H, Random, 95% CI)

1.34 [1.19, 1.51]

8.1 Specific treatments or diseases

24

3416

Risk Ratio (M‐H, Random, 95% CI)

1.37 [1.17, 1.61]

8.2 Mixed study domain

5

1167

Risk Ratio (M‐H, Random, 95% CI)

1.26 [1.07, 1.49]

Figures and Tables -
Comparison 7. Subgroup analysis
Comparison 8. Sensitivity analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of studies with favorable efficacy results, sponsorship recoded Show forest plot

8

726

Risk Ratio (M‐H, Random, 95% CI)

1.35 [1.18, 1.55]

2 Number of studies with favorable harms results, sponsorship recoded Show forest plot

3

501

Risk Ratio (M‐H, Random, 95% CI)

1.42 [0.31, 6.50]

3 Number of studies with favorable conclusions, sponsorship recoded Show forest plot

8

1029

Risk Ratio (M‐H, Random, 95% CI)

1.24 [1.04, 1.47]

4 Number of studies with favorable efficacy results, analysis adjusted for confounders Show forest plot

3

Odds Ratio (Random, 95% CI)

3.15 [2.07, 4.80]

5 Number of studies with favorable conclusions, analysis adjusted for confounders Show forest plot

4

Odds Ratio (Random, 95% CI)

3.13 [1.66, 5.93]

6 Number of studies with favorable efficacy results, fixed‐effect model Show forest plot

25

2923

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [1.23, 1.40]

7 Number of studies with favorable harms results, fixed‐effect model Show forest plot

4

826

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [1.15, 1.46]

8 Number of studies with favorable test treatment efficacy results, fixed‐effect model Show forest plot

2

131

Risk Ratio (M‐H, Fixed, 95% CI)

4.64 [2.08, 10.32]

9 Number of studies with favorable conclusions, fixed‐effect model Show forest plot

29

4583

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [1.24, 1.35]

10 Number of studies with favorable test treatment conclusions, fixed‐effect model Show forest plot

3

154

Risk Ratio (M‐H, Fixed, 95% CI)

5.90 [2.79, 12.49]

11 Number of studies with favorable conclusions, papers with unpublished studies excluded Show forest plot

27

4436

Risk Ratio (M‐H, Random, 95% CI)

1.35 [1.19, 1.54]

12 Number of studies with favorable efficacy results, restricted to specific domains Show forest plot

13

797

Risk Ratio (M‐H, Random, 95% CI)

1.28 [1.07, 1.51]

13 Number of studies with favorable harms results, restricted to specific domains Show forest plot

3

561

Risk Ratio (M‐H, Random, 95% CI)

1.87 [1.54, 2.27]

14 Number of studies with favorable test treatment efficacy results, restricted to specific domains Show forest plot

2

131

Risk Ratio (M‐H, Random, 95% CI)

3.88 [1.26, 11.94]

15 Number of studies with favorable conclusions, restricted to specific domains Show forest plot

15

1803

Risk Ratio (M‐H, Random, 95% CI)

1.42 [1.25, 1.61]

16 Number of studies with favorable test treatment conclusions, restricted to specific domains Show forest plot

3

154

Risk Ratio (M‐H, Random, 95% CI)

5.92 [2.80, 12.54]

Figures and Tables -
Comparison 8. Sensitivity analysis