Objectives
This is a protocol for a Cochrane Review (intervention). The objectives are as follows:
To evaluate the benefits and harms of oral bile acid‐based therapies versus no intervention or placebo, or versus a different bile acid‐based therapy, at any dose or regimen, in adults diagnosed with metabolic dysfunction‐associated steatotic liver disease (MASLD).
Background
Description of the condition
Metabolic dysfunction‐associated steatotic liver disease (MASLD), previously known as non‐alcoholic fatty liver disease (NAFLD), is the most common chronic liver disorder in the world, with a global prevalence of up to 30% of the world population [1, 2, 3]. MASLD has been steadily increasing in prevalence, along with obesity and type 2 diabetes mellitus [4]. Amongst people with type 2 diabetes mellitus, the global prevalence of MASLD was 55.5% (95% confidence interval (CI) 47.3% to 63.7%) [5]. Obesity, physical inactivity, age, sex, ethnicity, or dyslipidaemia are all factors that influence the prevalence of the MASLD [6, 7, 8, 9, 10, 11]. MASLD is associated with a wide spectrum of liver disorders, from simple fatty liver (i.e. steatosis, with accumulation of fat droplets in more than 5% of the liver cells, without cell injury) to metabolic dysfunction‐associated steatohepatitis, formerly known as non‐alcoholic steatohepatitis, with inflammatory hepatocyte injury (ballooning) with or without fibrosis, and increasing fibrosis leading to cirrhosis (end‐stage liver disease) [3, 12, 13]. Hepatocellular carcinoma can develop in genetically predisposed people with MASLD [14]. MASLD is the second cause of liver transplantation in the USA [15, 16]. It can develop 'de novo' even after liver transplantation [17]. People with MASLD also frequently have cardiovascular diseases, primarily affecting the heart or the brain [18]. Cardiovascular diseases are the leading cause of mortality in the MASLD population [19].
The definition of MASLD is based on the evidence of hepatic steatosis in the presence of at least one of the following five cardiometabolic criteria for adults [20, 21].
Overweight/obesity with body mass index (BMI) of 25 kg/m2 or greater (23 kg/m2 or greater in Asian people) or waist circumference of 94 cm or greater in Caucasian men and 80 cm or greater in Caucasian women (or ethnicity‐adjusted equivalent)
Fasting glucose levels of 5.6 mmol/L or greater (100 mg/dL or greater) or two‐hour postload glucose levels of 7.8 mmol/L or greater (140 mg/dL or greater) or glycated haemoglobin (HbA1c) of 5.7% or greater (39 mmol/L or greater) or type 2 diabetes or treatment for type 2 diabetes
Blood pressure of 130 mmHg/85 mmHg or greater (systolic/diastolic) or specific antihypertensive drug treatment
Plasma triglycerides of 150 mg/dL or greater (1.70 mmol/L or greater) or specific lipid‐lowering treatment
Plasma high‐density lipoprotein‐cholesterol less than 40 mg/dL (less than 1.0 mmol/L) for men and less than 50 mg/dL (less than 1.3 mmol/L) for women or specific lipid‐lowering treatment.
The pathogenesis of MASLD is unclear. The last 'multiple hit' hypothesis explains the mechanistic stages of liver injury [22]. It considers any interruption in the balance of the gut microbiome, insulin resistance, obesity, oxidative stress, and the disharmony of inflammatory cytokines, as well as epigenetic factors acting together, to predict progression and the development of MASLD in genetically predisposed individuals [22, 23]. There is no pathogenetic explanation for the clinical and morphological heterogeneity in MASLD [24].
The diagnosis of MASLD is often made on abnormal imaging of the liver or elevated serum liver enzymes, and it involves the exclusion of other causes of liver injury, such as excessive alcohol consumption, viruses, autoimmune liver disease, genetic disorders, starvation, and the use of hepatotoxic drugs [25, 26, 27]. Covert or hidden alcohol consumption continues to be a commonly missed problem in the diagnostic workup of people with suspected MASLD, and repeated interviews assisted by questionnaires to reveal covert or hidden alcohol problems are often needed. Imaging techniques, such as ultrasound screening or transient elastography, proton magnetic resonance spectroscopy, and quantitative fat or water‐selective magnetic resonance imaging, are used to establish the presence of hepatic steatosis in MASLD, assess liver fibrosis, or quantify liver fat [28, 29], in addition to liver biopsy [13, 30].
Description of the intervention and how it might work
Bile acids are naturally occurring molecules synthesised from cholesterol and conjugated to amino acids in the liver. These are called primary bile acids. Intestinal bacteria alter the primary bile acids into secondary bile acids. Then they return to the liver via the portal circulation.
Bile acids are stored in the gallbladder. They are essential for the digestion and absorption of dietary fats and fat‐soluble vitamins, and play a role in cholesterol homeostasis and gut microbiota composition. In addition to these physiological functions, bile acids act as signalling molecules that influence metabolic pathways, including lipid and glucose metabolism, through activation of nuclear and membrane receptors [31].
The spectrum of bile acids includes cholic acid, chenodeoxycholic acid, deoxycholic acid, lithocholic acid, ursodeoxycholic acid, norursodeoxycholic, obeticholic, taurocholic, and lithocholic acids.
Amongst the bile acids, oral administration of ursodeoxycholic acid and obeticholic acid is used as therapeutic agents in MASLD. Both acids are under investigation for their anti‐inflammatory and antifibrotic properties to reduce inflammation and fibrosis in liver diseases [32, 33]. Bile acids may reduce hepatic lipogenesis, steatosis, and insulin resistance [34, 35]. Oral administration of bile acid derivatives can modulate both the composition of the endogenous bile acid pool and the gut microbiota, potentially offering metabolic benefits in MASLD [36, 37, 38].
Beyond their digestive roles, bile acids interact with several receptors — such as the farnesoid X receptor, pregnane X receptor, vitamin D receptor, and G‐protein‐coupled bile acid receptor — to regulate energy homeostasis, detoxification, and immune responses [31, 34, 39]. Farnesoid X receptor activation, for example, induces fibroblast growth factor‐19, which downregulates bile acid synthesis by suppressing CYP7A1 and modulates cholesterol and glucose metabolism [40].
People with MASLD exhibit dysregulation in bile acid metabolism, including increased faecal bile acid excretion, elevated serum levels of primary bile acids, and impaired feedback regulation. This dysregulation may contribute to hepatic cholesterol accumulation and disease progression [40].
Lifestyle modification, a healthy diet, and controlling comorbidities such as type 2 diabetes mellitus are the usual interventions for MASLD [41, 42, 43]. A supplementary intervention for people with MASLD includes the oral bile acids, ursodeoxycholic acid and obeticholic acid [44]. The European Association for the Study of the Liver – European Association for the Study of Diabetes – European Association for the Study of Obesity (EASL–EASD–EASO) Clinical Practice Guidelines provide dose recommendations for two of the bile acids (i.e. ursodeoxycholic acid and obeticholic acid). The recommendations of the best doses and regimens for treating people with MASLD were selected using consensus, as evidence from systematic reviews with meta‐analyses of randomised clinical trials is lacking [45]. There are no recommendations for the best doses and regimens of the remaining bile acid derivatives in clinical settings.
Our knowledge of adverse events is also sparse. In one 2023 market report overview on ursodeoxycholic acid, under "Side effects of using ursodeoxycholic acid to bring down market growth," it is written that "using this acid continually for a very long time can still cause some side effects [meaning adverse effects] to the human body …" The report also states that "Ursodeoxycholic acid can cause certain problems which involve bloody or cloudy urine, faster heartbeat, severe nausea, stomach pain, skin rashes, body itchiness, frequent urge to urinate, severe bladder pain, and difficulty in urination" [46]. Obeticholic acid, also known as Ocaliva, is associated with well‐recognised adverse effects. The most common adverse events are pruritus, fatigue, abdominal pain and discomfort, arthralgia, sore throat, dizziness, and constipation [47]. In addition, although less frequent, there have been reports of hepatotoxicity, including people experiencing serious liver‐related adverse events, particularly in those with advanced cirrhosis [48].
Why it is important to do this review
Leoni and colleagues have critically reviewed and assessed five international guidelines for similarities and discrepancies in the diagnosis and treatment of MASLD [30]. They found that the five clinical guidelines, except for lifestyle modification treatments, are discordant in their recommendations on the management of people with MASLD. Evidence for possibly helpful medicines is either insufficient or lacking [30, 42, 49, 50]. A consensus report from 2022 also concluded that there is no high‐quality evidence for liver‐specific management in NAFLD (now known as MASLD) [51].
One Cochrane review by Orlando and colleagues published in 2007 included four clinical trials with 279 participants randomised to ursodeoxycholic acid versus placebo (three trials) with or without a low‐calorie diet, or versus vitamin C and E. The review found insufficient data to support or refute the use of bile acids in people with MASLD [52]. Though we identified two meta‐analyses on ursodeoxycholic acid and obeticholic acid in people with MASLD, the included trials used non‐invasive markers to assess progression of fibrosis, and the majority of the assessed outcomes were unvalidated surrogate outcomes [53, 54]. Some randomised clinical trials of norursodeoxycholic acid and obeticholic acid in people with MASLD showed beneficial results [55, 56, 57].
Given the increasing prevalence of MASLD, the growing administration of bile acids and their derivatives, with no proven effects, the long‐term risks associated with comorbidities, and the current guideline recommendations on bile acids in people with MASLD, there is a need to conduct a systematic review with meta‐analyses of randomised clinical trials, aiming to evaluate the available evidence on the use of bile acids versus no intervention, placebo, or another bile acid in people with MASLD.
Objectives
To evaluate the benefits and harms of oral bile acid‐based therapies versus no intervention or placebo, or versus a different bile acid‐based therapy, at any dose or regimen, in adults diagnosed with metabolic dysfunction‐associated steatotic liver disease (MASLD).
Methods
Criteria for considering studies for this review
Types of studies
We will include randomised clinical trials with a parallel‐group design, irrespective of language, format, year, status of publication, and the outcomes measured or reported.
Though unlikely, if we identify cluster‐randomised trials or trials with a cross‐over design, we will include them. In cluster‐randomised trials, groups of individuals are randomised to different interventions [58]. In trials with a cross‐over design, each participant is allocated to a sequence of interventions [58].
We will exclude quasi‐randomised studies as the method of allocation of participants to the study groups is not truly random (e.g. based on date of birth, data entry) and any other observational studies.
Types of participants
We will include adults aged 18 years or older, any sex, gender, race, ethnicity, and setting, diagnosed with MASLD at any stage (i.e. MASLD in people with or without liver fibrosis, or cirrhosis with compensated stage). We will accept any definitions used by the trial authors when describing the diagnosed trial participants.
If we identify a trial that includes a subset of participants with diseases other than MASLD (i.e. the arbitrary greater than 10%), and we cannot separate their data from the data of interest for our review, we will contact trial authors to request the data. If we cannot obtain the data or if the trial authors do not respond, then we will exclude the trial. If the subset is 10% or less, we will include the trial, irrespective of response, and we will perform a sensitivity analysis.
We will exclude trials with participants having chronic liver disease other than MASLD, such as alcoholic liver disease, hepatitis B, or hepatitis C. We will record if and how people with alcohol problems were excluded in the individual trials. We will also register whether alcohol consumption was monitored during the trial.
Types of interventions
Experimental intervention
Any bile acid drug administered at any dose, treatment duration, or regimen
Control interventions
No drug intervention or placebo
A different experimental bile acid drug administered at any dose or regimen
We will allow co‐interventions if administered equally to the trial intervention groups.
Outcome measures
If data are provided at more than one time point (i.e. by the end of treatment, during post‐treatment, and at the longest follow‐up), we will record and analyse the data of the three time points. Our primary analyses for conclusions will consist of outcome data at the longest follow‐up. We will use the outcome data at the remaining two time points for secondary analyses.
We will also include trials that do not report results of outcomes they might have measured or do not provide usable data.
We have selected our critical and important outcomes to align with clinically meaningful outcomes commonly used in regulatory trials of treatments for liver disease, including all‐cause mortality, liver‐related mortality, serious adverse events, liver‐related morbidity, and health‐related quality of life measured by validated tools.
Critical outcomes
All‐cause mortality.
Liver‐related mortality.
Proportion of people with one or more serious adverse events. We will use the definitions employed by the trial authors for serious adverse events as well as the definition in the International Conference on Harmonisation (ICH) Guidelines for Good Clinical Practice [59] (i.e. any untoward medical occurrence that results in death, is life‐threatening, requires hospitalisation or prolongation of existing hospitalisation, results in persistent or significant disability or incapacity, or is a congenital anomaly or birth defect). We will consider any other adverse event as non‐serious (see Important outcomes). As the 'serious adverse events' outcome is a composite outcome, we will also perform secondary analyses of the individual components of the composite outcome.
Important outcomes
Liver‐related morbidity (a composite of gastrointestinal bleeding, ascites, hepatic encephalopathy, hepatorenal syndrome, jaundice, hepatocellular carcinoma, or need for liver transplantation).
Health‐related quality of life. We will consider trial data only if trialists have used validated tools to measure health‐related quality of life, such as World Health Organization Quality of Life (WHOQOL), EuroQoL Group Quality of Life Questionnaire based on 5 dimensions (EQ‐5D), 36‐item Short Form Health Survey (SF‐36). We will use these tools in the order provided.
Proportion of people with one or more adverse events considered non‐serious (see Critical outcomes).
Proportion of people with no change in fibrosis or worsening of fibrosis.
Proportion of people with no resolution of metabolic dysfunction‐associated steatohepatitis.
Search methods for identification of studies
To minimise bias in our search results, we will follow the guidance in Chapter 4 of the Cochrane Handbook for Systematic Reviews of Interventions [60] and in PRISMA‐S [61] on planning and describing the search process for the review. The Cochrane Hepato‐Biliary Group Information Specialist will develop the final search strategies and perform the electronic searches. We will not impose any restrictions on the language of publication, date, or status.
Electronic searches
We will search the Cochrane Hepato‐Biliary Group Controlled Trials Register via the Cochrane Register of Studies Web, Cochrane Central Register of Controlled Trials in the Cochrane Library, MEDLINE ALL Ovid, Embase Ovid, LILACS (Latin American and Caribbean Health Science Information database; VHL Regional Portal), Science Citation Index Expanded, and Conference Proceedings Citation Index – Science. We will search the latter two simultaneously through the Web of Science.
We will use the Cochrane Highly Sensitive Search Strategies for identifying randomised trials in MEDLINE and Embase, modified to include relevant search terms for identifying retraction statements and errata, as recommended in Chapter 4 of the Cochrane Handbook for Systematic Reviews of Interventions [60, 62].
Supplementary material 1 shows the preliminary search strategies with the expected date range of the searches. We will provide the actual date of the electronic searches at the review stage.
Searching other resources
We will search online trial registries such as the US National Institutes of Health Ongoing Trials Register ClinicalTrials.gov (https://clinicaltrials.gov), European Medicines Agency (https://euclinicaltrials.eu/search-for-clinical-trials/), World Health Organization International Clinical Trials Registry Platform (https://trialsearch.who.int), the US Food and Drug Administration (https://www.fda.gov), eLibrary (https://www.elibrary.ru), VINITI (https://www.viniti.ru/database), Russian Medicine (https://medjrf.com/0869-2106), and pharmaceutical company sources for ongoing or unpublished trials. We will attempt to search Chinese databases through the Chinese Cochrane Centre.
We will search for grey literature such as meeting abstracts and internal reports in Europe OpenGrey (https://www.opengrey.eu), and Google Scholar (https://scholar.google.com).
We will use the PubMed/MEDLINE 'similar articles search' tool on all included trials. We will manually check citations and reference lists of the included trials, and of any relevant systematic reviews identified.
We will search for and examine any relevant retraction statements through the Retraction Watch Database (https://retractionwatch.com/retraction-watch-database-user-guide/), and errata for information, as errata can reveal important limitations or even fatal flaws in studies [62].
We will screen reference lists of included trials, and may contact the trial authors, to identify further published or unpublished trials of potential interest for our review.
We will provide the search terms used for the above‐listed resources and the dates of search at the review stage.
We will use the relevant sections of the PRISMA‐S checklist for our review to ensure that we have reported and documented our searches as advised (PRISMA‐S Checklist [61]).
Data collection and analysis
We will follow the guidelines provided in the Cochrane Handbook for Systematic Reviews of Interventions [63]. We will perform our main analyses using Review Manager [64]. We will assess imprecision with Trial Sequential Analysis and compare the assessment with our assessment with GRADE in a sensitivity analysis [65, 66, 67, 68].
Selection of studies
We will use Covidence software for screening of search results and trial selection [69]. One review author (REA) will upload the list of all candidate studies in Covidence and will remove duplicates of the same study. Three review authors (REA, DN, and CSP) will independently screen titles and abstracts to identify the studies of possible interest. We will retrieve the full‐text publications of potentially eligible studies, and the same three review authors will assess the trials based on the inclusion and exclusion criteria described in this protocol. The same three review authors will screen reference lists of trial reports to identify further trials of potential interest for our review.
We will include conference abstracts, unpublished trials, and grey literature reports if we can use the data for analysis. We will also include trials that do not assess or report data on outcomes of interest for our review, even if trial investigators, after personal communication, do not send us the requested data. We will resolve any disagreements by discussion or by involving any of the three remaining review authors (OB, PA, or CG), for arbitration. We will list the studies excluded during the full‐test assessment in 'Excluded studies'. We will record the selection process in sufficient detail to complete a PRISMA 2020 flow diagram (for new systematic reviews, which includes searches of databases, registers, and other sources) [70, 71].
To screen non‐English‐language publications, and if we do not have command of a certain language, we will use Google Translate to assist with eligibility assessment (https://translate.google.com). If needed, we will look for translators with the help of the Cochrane Hepato‐Biliary Group Editorial Team office to assist with eligibility assessment and data extraction.
Data extraction and management
We will use Covidence software for data extraction [69]. Three review authors (REA, DN, and CSP) will independently extract data from the trials fulfilling the protocol inclusion criteria, using a data extraction form, piloted on a few trials. If a trial is reported in more than one publication, we will extract data from each report separately and then combine the information across the multiple data collection forms ensuring there is no duplication of data. We will resolve disagreements by discussion or with any of the remaining three review authors (OB, AC, or CG).
For our review, we will record the information below.
Publication data (i.e. title, format of publication, year (range of years) of conductance of the trial and language of publication, place and country of the recruited participants, setting, authors; contact author email for correspondence).
Study design.
Funding.
Registered trial protocol.
Setting: inclusion and exclusion criteria; methods of randomisation, including allocation sequence and concealment, to identify quasi‐randomised studies.
Sample size calculation performed and reached, or not.
Population data (i.e. age, sex, gender, race, ethnicity, history of the disease, ethnicity, and MASLD stage).
Intervention data (type of intervention in the experimental and control group(s), dose, frequency, duration of intervention, and concurrent medications used).
Outcomes.
Dropouts and reasons for them.
Length of follow‐up; other time points with follow‐up data.
Types of data analyses (i.e. intention‐to‐treat, modified intention‐to‐treat, per‐protocol).
Number of participants randomised.
Number of participants included in the analysis.
Number of participants with events for binary outcomes, mean and standard deviation (SD) for continuous outcomes, number of events and the mean follow‐up period for count outcomes, and number of participants with events and the mean follow‐up period for time‐to‐event outcomes.
Any valid information that can be extracted from ongoing trials.
Any information on withdrawn or retracted trials if such is found.
We will ensure that we have retrieved and collected all available and relevant data for our review from the trial publications and through correspondence with study publication authors. We will enter details of our correspondence with the trial publication authors in the 'Characteristics of included studies' table.
Risk of bias assessment in included studies
Two review authors (REA and CSP) will independently assess the risk of bias for each outcome at the longest follow‐up, using the Cochrane RoB 2 tool (https://www.riskofbias.info/; 72, 73; Critical outcomes; Important outcomes). For the outcome 'health‐related quality of life', we will follow the guidance in Section 8.6 of the Cochrane Handbook for Systematic Reviews of Interventions [72]. We will resolve disagreements by discussion with a third review author (PA, DN, or CG).
We will assess the effect of the assignment on the intervention using the intention‐to‐treat (ITT) principle, which includes all randomised participants, irrespective of the interventions that participants received.
We will assess the following five RoB 2 domains for each outcome in the randomised trials.
Bias arising from the randomisation process
Bias due to deviations from intended interventions
Bias due to missing outcome data
Bias in the measurement of outcome
Bias in the selection of the reported result
Each domain contains a series of signalling questions, with the answers (yes, probably yes, no information, probably no, no). Elaborations on these signalling questions can be found in Chapter 8 of the Cochrane Handbook for Systematic Reviews of Interventions [72]. We will determine the risk of bias in each domain (low risk, some concerns, and high risk). We will include text alongside the judgements to provide supporting information for our decisions, including when our risk of bias assessment is due to additional correspondence with trial authors.
Each domain will obtain one of the three levels below.
Low risk of bias: the trial is at low risk of bias for all domains for this result.
Some concerns: the trial is judged to raise some concerns in at least one domain for this result, but is not at high risk of bias for any of the remaining domains.
High risk of bias: the trial is at high risk of bias in at least one domain for this result, or the trial is judged to have some concerns for multiple domains in a way that substantially lowers confidence in the result.
The overall risk‐of‐bias judgement is the same as for an individual domain, such as low risk of bias, some concerns, or high risk of bias. Judging a result to be at a particular level of risk of bias for an individual domain implies that the result has an overall risk of bias at least this severe.
We will summarise the risk of bias in the forest plots and in the text based on ITT.
We will assess the risk of bias of all our Critical outcomes and Important outcomes at the longest follow‐up time point. As risk of bias is one of the five GRADE domains, we will use our risk of bias assessments to assess the certainty of the body of evidence [74]. In our 'Summary of findings' tables, we will present the outcomes below as these outcomes are considered most clinically important.
All‐cause mortality
Liver‐related mortality
Proportion of people with one or more serious adverse events
Liver‐related morbidity
Health‐related quality of life
Proportion of people with one or more adverse events considered non‐serious
In case of cross‐over trials of interest to our review, we will use the standard version of RoB 2 as we will use the data from the first period only [58, 72, 73]. If we identify cluster‐randomised clinical trials, we will consider an additional domain that specifically applies to that design, RoB 2 Domain 1b, 'Bias arising from the timing of identification and recruitment of individual participants within clusters in relation to the timing of randomisation' (https://www.riskofbias.info/). We will follow the suggested algorithm for reaching the risk of bias judgements, for bias arising from the timing of the identification and recruitment of participants in a cluster‐randomised trial.
We will use the RoB 2 Microsoft Excel tool to store the data (it will be available upon request, or an address will be provided in the review).
Measures of treatment effect
If data in a trial are not reported in a format that we can enter directly into a meta‐analysis, we will convert the data to the required format using guidance in Chapter 6 of the Cochrane Handbook for Systematic Reviews of Interventions [75].
Dichotomous outcomes
For dichotomous outcomes, we will calculate the risk ratio (RR) with a 95% confidence interval (CI). For count outcomes (e.g. number of adverse events per individual), we will calculate the rate ratio (i.e. the ratio of the rate in the experimental group to the rate in the control group) with 95% CI.
We will dichotomise time‐to‐event data, if found.
Continuous outcomes
For continuous outcomes, such as health‐related quality of life, we will calculate the mean difference (MD) between the experimental and control groups with a 95% CI if the trials used the same validated tool. If more than one trial measures health‐related quality of life using different validated tools, we will calculate the standardised mean difference (SMD) and 95% CI. The SMD expresses the size of the intervention effect when the MD between groups is divided by the SD amongst participants [75]. As the SMD method does not correct for differences in the direction of the scale, if necessary, we will multiply the mean values from one set of studies by −1 to ensure that all the scale points are in the same direction [75]. We will interpret SMD as follows: SMD less than 0.40 for small intervention effects; SMD between 0.40 and 0.70 for moderate intervention effects; and SMD greater than 0.70 for large intervention effects [76]. We plan to present median and interquartile ranges for continuous data that are not normally distributed (skewed data), in a narrative format. We will present a forest plot to display effect estimates and CIs for individual trials [77]. We will conduct meta‐analyses only when the study groups are sufficiently homogeneous [77].
We will analyse participants in the intervention groups to which they were randomised, regardless of the intervention they received, and we will include all randomised participants in the outcome analyses (i.e. ITT).
Unit of analysis issues
The unit of analysis will be participants with MASLD as originally randomised to the trial groups. We do not expect to find parallel‐group design trials with more than two intervention groups. However, if we identify parallel‐group trials with more than two groups having a common control group, we will divide the control group if it is to be used within the same comparison. We do not expect to find cluster‐randomised or cross‐over trials. However, if we find cluster‐randomised trials, then we will analyse cluster‐randomised trials separately from the randomised parallel‐group clinical trials included in the review as such trials may be at an increased risk of bias [58, 78]. If we find cross‐over trials, then, as already described, we will use only the first trial period for analysis, to avoid the cross‐over effect of the intervention [58].
To avoid repeated observations of study participants, our main analyses will include trial data for the trial participants at the longest follow‐up [75].
Dealing with missing data
If data are missing in a published report, one review author (RE) will contact trial investigators to request the missing data. If trialists have used ITT analysis to deal with missing data, we will use the ITT data. If the required data for ITT analysis are missing in the trial publications, we will use the data as available (i.e. this could be ITT, modified ITT, per‐protocol data, or all of these) in our primary analysis.
As data may be biased (e.g. treatment was withdrawn due to adverse events or participants were excluded from analysis), we will conduct sensitivity analyses as described below. We will include missing data by considering participants as treatment failures or treatment successes, by imputing them according to the following two scenarios.
Extreme‐case analysis favouring the experimental intervention ('best‐worse' case scenario); none of the dropouts/participants lost from the experimental group, but all the dropouts/participants lost from the control group, experienced the outcome, including all randomised participants in the denominator.
Extreme‐case analysis favouring the control intervention ('worst‐best' case scenario); all dropouts/participants lost from the experimental group, but none from the control group, experienced the outcome, including all randomised participants in the denominator.
We will perform the two sensitivity analyses only for the critical outcomes.
Reporting bias assessment
If we include 10 or more trials in a meta‐analysis, we will draw funnel plots to assess reporting biases from the individual trials by plotting the RR on a logarithmic scale against its standard error [79, 80]. For dichotomous outcomes, we will test asymmetry using the Harbord test in cases where Tau² is less than 0.1 [81]; we will use the Rücker test if Tau² is more than 0.1 [82]. For continuous outcomes, we will use the regression asymmetry test [79] and the adjusted rank correlation [83].
Synthesis methods
We will perform the meta‐analyses using Review Manager [64], and we will follow the recommendations provided in Chapter 10 of the Cochrane Handbook for Systematic Reviews of Interventions [77]. We will perform our meta‐analysis, including outcome data from the trials at any risk of bias.
We will present the results of dichotomous outcomes as RR with 95% CI, and the results of continuous outcomes, such as quality of life, as MD or SMD with 95% CI. In the presence of rare events, we will use the Peto method [77]. We will use random‐effects model meta‐analysis as our primary analyses and the fixed‐effect model meta‐analysis as sensitivity analyses [76, 77]. When it is not appropriate to perform a meta‐analysis, we will present data using forest plots without summary estimates, and we will present a summary of the results. We will follow Chapter 12 of the Cochrane Handbook for Systematic Reviews of Interventions to identify the most appropriate approach for our review to present findings using other methods [84].
We will address the presence of clinical, methodological, and statistical heterogeneity. We will describe clinical and methodological heterogeneity by exploring the design features of the trial, participants' characteristics, interventions, comparators, outcomes, dropouts and withdrawals, treatment duration, follow‐up, and funding. We will summarise this information, using subheadings, and we will provide a summary of our risk of bias assessments to identify similarities and discrepancies.
We will assess statistical heterogeneity of meta‐analysed trials by visual inspection of forest plots, the Chi2 test for heterogeneity, and the I2 statistic [85]. For the Chi2 test, we will consider a P ≤ 0.10 to indicate statistically significant heterogeneity (i.e. variation in effect estimates beyond that expected by chance). We will use the I2 statistic to classify heterogeneity. As thresholds for the interpretation of the I² statistic can be misleading, we will use the following approximate guide for the interpretation of heterogeneity provided in Chapter 10 of the Cochrane Handbook for Systematic Reviews of Interventions [77].
0% to 40%: might not be important
30% to 60%: may represent moderate heterogeneitya
50% to 90%: may represent substantial heterogeneitya
75% to 100%: considerable heterogeneity
aThe importance of the observed value of the I² statistic depends on the magnitude and direction of effects, and the strength of evidence for heterogeneity (e.g. P value from the Chi² test, or a CI for the I² statistic).
If the heterogeneity is considerable, then we may decide not to meta‐analyse the data.
Investigation of heterogeneity and subgroup analysis
We plan to perform the following subgroup analyses as we expect that they may be a cause for heterogeneity.
Trials with no for‐profit support compared to trials with for‐profit support, and trials without reported funding. The reason is that trials with vested interests may overestimate beneficial intervention effects or underestimate harmful intervention effects [86].
Follow‐up at the end of treatment compared to follow‐up at the longest time of follow‐up to determine if the effect of the drugs has been retained or waned.
Based on the steatosis‐only stage of MASLD compared to metabolic dysfunction‐associated steatohepatitis, and compared to liver cirrhosis, to compare the effect of the drugs in terms of severity of the disease.
We may consider additional subgroup analyses at the review stage.
We will use a formal statistical test in Review Manager to assess subgroup differences that could account for heterogeneity (e.g. Chi² test) [64]. It is recommended to have at least five trials providing data for an outcome in a subgroup to ascertain a valid subgroup difference in effect [77]. We plan to conduct subgroup analyses with two of our critical outcomes (i.e. all‐cause mortality and liver‐related mortality), at the longest follow‐up.
We will use the I² statistic to quantify inconsistencies amongst the trials in each analysis when P value is 0.10 or less. We will also interpret the P value of 0.10 or less from the Chi² test as evidence of statistical heterogeneity [77]. We will use the random‐effects model meta‐analysis to account for the presence of between‐study heterogeneity.
Equity‐related assessment
We do not plan to conduct a formal equity‐related assessment in this review. Our rationale is that the available trial data in this field are unlikely to provide sufficient information disaggregated across PROGRESS‐Plus factors (such as place of residence, race/ethnicity, socioeconomic status, sex, education, occupation, or gender identity). Most trials in this area do not report relevant subgroup analyses or stratified outcome data.
However, we will remain attentive to any equity‐relevant subgroup data that may be reported in the included trials and will extract and report such data where available.
Sensitivity analysis
We plan to perform the below‐listed sensitivity analyses on all our critical and important outcomes (i.e. all‐cause mortality; liver‐related mortality; proportion of people with one or more serious adverse events; liver‐related morbidity; health‐related quality of life; proportion of people with one or more adverse events considered non‐serious; proportion of people with no change in fibrosis or worsening of fibrosis; and proportion of people with no resolution of metabolic dysfunction‐associated steatohepatitis), whenever there are data.
Restricting the analysis to trials at low risk of bias (see Synthesis methods)
Restricting the analysis to trials published in an abstract form or unpublished trials, or both
Conducting the analyses with a fixed‐effect model (see Synthesis methods)
'Best‐worst' case scenario analysis (see Dealing with missing data)
'Worst‐best' case scenario analysis (see Dealing with missing data)
Trial Sequential Analysis and assessment of significance. We will compare our assessment of imprecision with GRADE [68] to that performed with the Trial Sequential Analysis [87, 88].
Trial Sequential Analysis
We will conduct Trial Sequential Analysis as cumulative meta‐analyses are at risk of producing random errors due to sparse data and repetitive testing of the accumulating data [65, 66, 67, 89]. To control random errors, we will calculate the diversity‐adjusted required information size (DARIS) (i.e. the number of participants needed in a meta‐analysis to detect or reject a certain intervention effect) [67, 89, 90, 91, 92]. In our dichotomous outcome meta‐analysis, we will base the DARIS on the event proportion in the control group of our meta‐analyses; assumption of a plausible relative risk reduction of 20% or the risk observed in the included trials at low risk of bias; a risk of type I error of 2.5% due to three critical and important secondary outcomes, a risk of type II error of 10% [87], and the diversity of the included trials in the meta‐analysis. For our continuous outcome meta‐analysis (health‐related quality of life), we will estimate DARIS using the SD observed in the control group and the minimal relevant difference of 50% of this SD; alpha of 2.5% due to three important outcomes [93]; beta of 10% [87]; and the diversity as estimated from the trials in the meta‐analysis [94]. We will also calculate and report the Trial Sequential Analysis‐adjusted 95% CI if the cumulative Z‐score does not cross any of the trial sequential monitoring boundaries for benefit, harm, or futility [65, 67]. The underlying assumption of Trial Sequential Analysis is that testing for statistical significance may be performed each time a new trial is added to the meta‐analysis. We will add the trials according to the year of publication, and, if more than one trial has been published in a year, we will add trials alphabetically according to the last name of the first author. Based on the DARIS, we will construct the trial sequential monitoring boundaries for benefit, harm, and futility [65, 67, 89]. These boundaries will determine the statistical inference one may draw regarding the cumulative meta‐analysis that has not reached the DARIS; if the trial sequential monitoring boundary for benefit or harm is crossed before the DARIS is reached, firm evidence for an intervention effect may be established, and further trials may be superfluous. However, if the boundaries are not crossed, it is most probably necessary to continue doing trials to detect or reject a certain intervention effect. If the cumulative z‐curve crosses the trial sequential monitoring boundaries for futility, no more trials may be needed. A more detailed description of Trial Sequential Analysis can be found at https://www.ctu.dk/tsa/ [65, 66, 67]. We will perform Trial Sequential Analysis with Trial Sequential Analysis software, version 0.9.5.10 beta [66]. We will use random‐effects meta‐analysis.
Downgrading for imprecision with Trial Sequential Analysis
In Trial Sequential Analysis where the cumulative z‐value does not cross the monitoring boundaries for benefit, harm, or futility, we will downgrade our assessment of imprecision by three levels if the accrued number of participants is below 33% of the DARIS, by two levels if between 33% and 65%, and by one level if between 66% and 99% of DARIS. We will not downgrade for imprecision if the cumulative z‐value reaches or crosses benefit, harm, futility, or DARIS.
Certainty of the evidence assessment
We will create two summary of findings tables with the following comparisons.
Any bile acid administered at any dose or regimen versus no intervention or placebo
Any bile acid administered at any dose or regimen versus another bile acid administered at any dose or regimen
In both tables, we will present the outcomes: all‐cause mortality; liver‐related mortality; proportion of people with one or more serious adverse events; liver‐related morbidity; health‐related quality of life; and proportion of people with one or more adverse events considered non‐serious.
The GRADE approach appraises the certainty of a body of evidence based on the extent to which one can be confident that an estimate of effect or association reflects the item being assessed [95]. The certainty of a body of evidence considers within‐study risk of bias, indirectness of the evidence, heterogeneity of the data, imprecision of effect estimates, and risk of publication bias [96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111].
Regarding risk of bias, we will use the overall judgement for the result of an outcome. Low risk of bias indicates no limitation (the certainty is not downgraded); some concerns indicates either no limitation or serious limitation (the certainty is downgraded one level); and high risk of bias indicates either serious limitation or very serious limitation (the certainty is downgraded two levels) [95]. For the assessment of imprecision, we will use the latest guidance based on the use of a minimally contextualised approach which is based on thresholds and CI of absolute effects as the primary criterion for imprecision rating (i.e. the CI approach) [112]. The primary considerations for this include the following.
If the CIs are wide and considerably crossed prespecified thresholds of interest (we will consider 0.25 as trivial), we will downgrade two levels for imprecision, and if the CIs are very wide and both boundaries of the CIs suggest very different inferences, we will consider downgrading three levels.
If the CIs do not cross the threshold and the relative effect is large, we will consider implementing the optimal information size (OIS) approach, downgrading more than one level for imprecision if the sample size of the meta‐analysis is far less than the OIS.
We will interpret the GRADE Working Group grades of evidence as follows.
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.
Two review authors (RE and PA) will independently conduct GRADE assessments using GRADEpro GDT [68]. The two review authors will resolve any discrepancies through discussion with a third review author (CP, OB, or CG). We will justify all decisions to downgrade the certainty of the evidence for each outcome in footnotes, and we will make comments to aid readers' understanding of our judgements if necessary.
We will conduct the review according to this protocol. We will report any deviations from it in the 'Methods' section.
Consumer involvement
One of the co‐authors of this protocol (PA) has previously been listed as a consumer representative in the Editorial Manager system and contributed to this protocol from the perspective of someone with lived experience relevant to MASLD.
Supporting Information
Supplementary materials are available with the online version of this article: 10.1002/14651858.CD014850.
Supplementary materials are published alongside the article and contain additional data and information that support or enhance the article. Supplementary materials may not be subject to the same editorial scrutiny as the content of the article and Cochrane has not copyedited, typeset or proofread these materials. The material in these sections has been supplied by the author(s) for publication under a Licence for Publication and the author(s) are solely responsible for the material. Cochrane accordingly gives no representations or warranties of any kind in relation to, and accepts no liability for any reliance on or use of, such material.
Supplementary material 1 Search strategies
New
Additional information
Acknowledgements
Cochrane Review Group funding acknowledgement: the Danish State is the largest single funder of the Cochrane Hepato‐Biliary Group (CHBG) through its investment in the Copenhagen Trial Unit, Centre for Clinical Intervention Research, the Capital Region, Rigshospitalet, Copenhagen, Denmark. Disclaimer: the views and opinions expressed in this protocol are those of the authors and do not necessarily reflect those of the Danish State or the Copenhagen Trial Unit.
We thank the following people from the CHBG Editorial Team.
CHBG Contact Editor: Goran Poropat, Croatia, for providing expert comments on this review protocol.
Sarah Louise Klingenberg, CHBG Information Specialist, Denmark, for designing searches and checking the style of references.
As three of the authors have also published other Cochrane interventional reviews, text in the Methods section may overlap with their other published articles.
The authors, Dimitrinka Nikolova, Chavdar Pavlov, Paola Andrenacci, and Christian Gluud are Cochrane Editors but were not involved in the editorial process of this article.
Cochrane Central Editorial Service
Editorial and peer‐reviewer contributions
The following people conducted the editorial process for this article.
Sign‐off Editor (final editorial decision): Emmanuel Tsochatzis, Royal Free Hospital and the University College of London, Institute of Liver and Digestive Health
Managing Editor (selected peer reviewers, provided editorial guidance to authors, edited the article): Anne‐Marie Stephani, Cochrane Central Editorial Service
Editorial Assistant (conducted editorial policy checks, collated peer‐reviewer comments and supported editorial team): Addie‐Ann Smyth, Cochrane Central Editorial Service; Andrew Savage (Cochrane Central Editorial Service)
Copy Editor (copy editing and production): Anne Lawson, Cochrane Central Production Service
Peer‐reviewers (provided comments and recommended an editorial decision): Karla Duque Jacome MD, MPH, CET, Cochrane (consumer review); Jo‐Ana Chase, Cochrane Evidence Production and Methods Directorate (methods review); Jo Platt, Central Editorial Information Specialist (search review); Anna Fichera, Ospedale Santa Marta e Venera, Italy (clinical review); Rachel Richardson, Cochrane (methods review)
Contributions of authors
REA: conceptualisation, project administration, methodology, writing of original draft, investigation, and writing – review and editing.
OB: writing – review and editing.
DN: conceptualisation, methodology, investigation, supervision, and writing – review and editing.
CP: conceptualisation, methodology, supervision, writing and reviewing the protocol.
PA: writing – review and editing.
CG: conceptualisation, methodology, investigation, supervision, writing – review and editing.
All authors approved the current version for publication.
Declarations of interest
REA: none.
OB: none.
DN is the Managing Editor of the Cochrane Hepato‐Biliary Group (CHBG).
CP is a CHBG Editor.
CG is the Co‐ordinating Editor of the CHBG.
PA is a CHBG Editor and external CHBG consumer peer reviewer.
The authors, DN, CP, PA, and CG, are Cochrane Editors but were not involved in the editorial process of this article.
Sources of support
Internal sources
-
The Cochrane Hepato‐Biliary Group Editorial Team office, Denmark
DN: is paid by the Danish State in the form of a monthly salary through the Rigshospitalet, which hosts the Cochrane Hepato‐Biliary Group (CHBG) Editorial Team office.
CG: is paid by the Danish State in the form of a monthly salary through the Rigshospitalet, which hosts the CHBG Editorial Team office.
-
The Cochrane Hepato‐Biliary Group, Denmark
Design of search strategies
External sources
-
The Cochrane Hepato‐Biliary Group, Denmark
Design of search strategies
Registration and protocol
Not relevant
Data, code and other materials
Data sharing is not applicable to this article as it is a protocol, so no datasets were generated or analysed.
References
- 1.Córdova-Gallardo J, Keaveny AP, Qi X, Méndez-Sánchez N. Metabolic associated fatty liver disease and acute-on-chronic liver failure: common themes for common problems. European Journal of Gastroenterology & Hepatology 2021;33(1S Suppl 1):e84-e93. [DOI: 10.1097/MEG.0000000000002335] [DOI] [PubMed] [Google Scholar]
- 2.Younossi ZM, Golabi P, Paik JM, Henry A, Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology (Baltimore, Md.) 2023;77(4):1335-47. [DOI: 10.1097/HEP.0000000000000004] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sanyal AJ. Past, present and future perspectives in nonalcoholic fatty liver disease. Nature Reviews. Gastroenterology & Hepatology 2019;16:377-86. [DOI: 10.1038/s41575-019-0144-8] [DOI] [PubMed] [Google Scholar]
- 4.Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology (Baltimore, Md.) 2018;67(1):123-33. [DOI: 10.1002/hep.29466] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Younossi ZM, Golabi P, Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. Journal of Hepatology 2019;71(4):793-801. [DOI] [PubMed] [Google Scholar]
- 6.Frith J, Day CP, Henderson E, Burt AD, Newton JL. Non-alcoholic fatty liver disease in older people. Gerontology 2009;55:607-13. [DOI: 10.1159/000235677] [DOI] [PubMed] [Google Scholar]
- 7.Jimba S, Nakagami T, Takahashi M, Wakamatsu T, Hirota Y, Iwamoto Y, et al. Prevalence of non-alcoholic fatty liver disease and its association with impaired glucose metabolism in Japanese adults. Diabetic Medicine 2005;22:1141-5. [DOI: 10.1111/j.1464-5491.2005.01582.x] [DOI] [PubMed] [Google Scholar]
- 8.Lonardo A, Bellentani S, Argo CK, Ballestri S, Byrne CD, Caldwell SH, et al; Non-alcoholic Fatty Liver Disease Study Group. Epidemiological modifiers of non-alcoholic fatty liver disease: focus on high-risk groups. Digestive and Liver Disease 2015;47(12):997-1006. [DOI: 10.1016/j.dld.2015.08.004] [DOI] [PubMed] [Google Scholar]
- 9.Papatheodoridis GV, Goulis J, Christodoulou D, Manolakopoulos S, Raptopoulou M, Andrioti E, et al. High prevalence of elevated liver enzymes in blood donors: associations with male gender and central adiposity. European Journal of Gastroenterology & Hepatology 2007;19:281-7. [DOI: 10.1097/MEG.0b013e328011438b] [DOI] [PubMed] [Google Scholar]
- 10.Saki S, Saki N, Poustchi H, Malekzadeh R. Assessment of genetic aspects of non-alcoholic fatty liver and premature cardiovascular events. Middle East Journal of Digestive Diseases 2020;12(2):65-88. [DOI: 10.34172/mejdd.2020.166] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Singh SP, Kar SK, Panigrahi MK, Misra B, Pattnaik K, Bhuyan P, et al. Profile of patients with incidentally detected nonalcoholic fatty liver disease (IDNAFLD) in coastal eastern India. Tropical Gastroenterology 2013;34(3):144-52. [DOI: 10.7869/tg.118] [DOI] [PubMed] [Google Scholar]
- 12.Brunt EM. Non-alcoholic fatty liver disease: what's new under the microscope? Gut 2011;60:1152-8. [DOI] [PubMed] [Google Scholar]
- 13.Wong VW, Ekstedt M, Wong JL, Hagström H. Changing epidemiology, global trends and implications for outcomes of NAFLD. Journal of Hepatology 2023;79:842-52. [DOI: 10.1016/j.jhep.2023.04.036] [DOI] [PubMed] [Google Scholar]
- 14.Anstee QM, Harrison S, Sanyal AJ. Obeticholic acid (OCA) improves non-invasive markers of fibrosis in patients with non-alcoholic steatohepatitis (NASH): a secondary analysis of the phase 3 Regenerate study. Digestive and Liver Disease 2020;52:e41-2. [Google Scholar]
- 15.Adam R, Karam V, Cailliez V, O'Grady JG, Mirza D, Cherqui D, et al. Annual Report of the European Liver Transplant Registry (ELTR) – 50-year evolution of liver transplantation. Transplant International 2018;31:1293-317. [DOI: 10.1111/tri.13358] [DOI] [PubMed] [Google Scholar]
- 16.Younossi Z, Stepanova M, Ong JP, Jacobson IM, Bugianesi E, Duseja A, et al. Nonalcoholic steatohepatitis is the fastest growing cause of hepatocellular carcinoma in liver transplant candidates. Clinical Gastroenterology and Hepatology 2019;17(4):748-55. [DOI: 10.1016/j.cgh.2020.05.064] [DOI] [PubMed] [Google Scholar]
- 17.Galvin Z, Rajakumar R, Chen E, Adeyi O, Selzner M, Grant D, et al. Predictors of de novo nonalcoholic fatty liver disease after liver transplantation and associated fibrosis. Liver Transplantation 2019;25:56-67. [DOI: 10.1002/lt.25338] [DOI] [PubMed] [Google Scholar]
- 18.Zhou XD, Cai J, Targher G, Byrne CD, Shapiro MD, Sung KC, et al. Metabolic dysfunction-associated fatty liver disease and implications for cardiovascular risk and disease prevention. Cardiovascular Diabetology 2022;21(1):270. [DOI: 10.1186/s12933-022-01697-0] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chan WK, Chuah KH, Rajaram RB, Lim LL, Ratnasingam J, Vethakkan SR. Metabolic dysfunction-associated steatotic liver disease (MASLD): a state-of-the-art review. Journal of Obesity & Metabolic Syndrome 2023;32:197-213. [DOI: 10.7570/jomes23052] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al. NAFLD Nomenclature consensus group (2023). A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Journal of Hepatology 2023;79(6):1542-56. [DOI: 10.1016/j.jhep.2023.06.003] [DOI] [PubMed] [Google Scholar]
- 21.Kanwal F, Neuschwander-Tetri B, Loomba R, Rinella M. Metabolic dysfunction-associated steatotic liver disease: update and impact of new nomenclature on the American Association for the Study of Liver Diseases practice guidance on nonalcoholic fatty liver disease. Hepatology (Baltimore, Md.) 2024;79:1212-9. [DOI: 10.1097/HEP.0000000000000670] [DOI] [PubMed] [Google Scholar]
- 22.Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism: Clinical and Experimental 2016;65(8):1038-48. [DOI] [PubMed] [Google Scholar]
- 23.Collins SL, Stine JG, Bisanz JE, Okafor CD, Patterson AD. Bile acids and the gut microbiota: metabolic interactions and impacts on disease. Nature Reviews Microbiology 2023;21:236-47. [PMID: 10.1038/s41579-022-00805-x] [DOI] [PubMed] [Google Scholar]
- 24.Parthasarathy G, Revelo X, Malhi H. Pathogenesis of nonalcoholic steatohepatitis: an overview. Hepatology Communications 2020;4:478-92. [DOI: 10.1002/hep4.1479] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, et al. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology (Baltimore, Md.) 2023;77(5):1797-835. [DOI: 10.1097/HEP.0000000000000323] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology (Baltimore, Md.) 2018;67(1):328-57. [DOI] [PubMed] [Google Scholar]
- 27.European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. Journal of Hepatology 2016;64(6):1388-402. [DOI] [PubMed] [Google Scholar]
- 28.Stern C, Castera L. Non-invasive diagnosis of hepatic steatosis. Hepatology International 2017;11(1):70-8. [DOI: 10.1007/s12072-016-9772-z] [DOI] [PubMed] [Google Scholar]
- 29.Tapper E, Loomba R. Noninvasive imaging biomarker assessment of liver fibrosis by elastography in NAFLD. Nature Reviews Gastroenterology and Hepatology 2018;15:274-82. [DOI: 10.1038/nrgastro.2018.10] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Leoni S, Tovoli F, Napoli L, Serio I, Ferri S, Bolondi L. Current guidelines for the management of non-alcoholic fatty liver disease: a systematic review with comparative analysis. World Journal of Gastroenterology 2018;24(30):336173. [DOI: 10.3748/wjg.v24.i30.3361] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Copple BL, Li T. Pharmacology of bile acid receptors: evolution of bile acids from simple detergents to complex signalling molecules. Pharmacological Research 2016;104:9-21. [DOI: 10.1016/j.phrs.2015.12.007] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Abenavoli L, Falalyeyeva T, Boccuto L, Tsyryuk O, Kobyliak N. Obeticholic acid: a new era in the treatment of nonalcoholic fatty liver disease. Pharmaceuticals (Basel, Switzerland) 2018;11(4):104. [DOI: 10.3390/ph11040104] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang YD, Chen WD, Wang M, Yu D, Forman BM, Huang W, et al. Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology (Baltimore, Md.) 2008;48:1632-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Arab JP, Karpen SJ, Dawson PA, Arrese M, Trauner M. Bile acids and nonalcoholic fatty liver disease: molecular insights and therapeutic perspectives. Hepatology (Baltimore, Md.) 2017;65(1):350-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Altamirano-Barrera A, Barranco-Fragoso B, Méndez-Sánchez N. Management strategies for liver fibrosis. Annals of Hepatology 2017;16(1):48-56. [DOI: 10.5604/16652681.1226814] [DOI] [PubMed] [Google Scholar]
- 36.Ikegami T, Honda A. Reciprocal interactions between bile acids and gut microbiota in human liver diseases. Hepatology Research 2018;48(1):15-27. [DOI: 10.1111/hepr.13001] [DOI] [PubMed] [Google Scholar]
- 37.Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nature Reviews Gastroenterology & Hepatology 2018;15(2):111-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schaap FG, Trauner M, Jansen PL. Bile acid receptors as targets for drug development. Nature Reviews. Gastroenterology & Hepatology 2014;11:55-67. [DOI] [PubMed] [Google Scholar]
- 39.Perino A, Schoonjans K. TGR5 and immunometabolism: insights from physiology and pharmacology. Trends in Pharmacological Sciences 2015;36(12):847-57. [DOI] [PubMed] [Google Scholar]
- 40.Zhang X, Deng R. Dysregulation of bile acids in patients with NAFLD [Internet]. In: Nonalcoholic Fatty Liver Disease – An Update. London (UK): IntechOpen, 2019:1-144. [DOI: 10.5772/intechopen.81474] [DOI] [Google Scholar]
- 41.Centis E, Marzocchi R, Suppini A, Dalle Grave R, Villanova N, Hickman IJ, et al. The role of lifestyle change in the prevention and treatment of NAFLD. Current Pharmaceutical Design 2013;19(29):5270-9. [PubMed] [Google Scholar]
- 42.Houghton D, Thoma C, Hallsworth K, Cassidy S, Hardy T, Burt AD, et al. Exercise reduces liver lipids and visceral adiposity in patients with nonalcoholic steatohepatitis in a randomized controlled trial. Clinical Gastroenterology and Hepatology 2017;15(1):96-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Puri P, Sanyal AJ. Nonalcoholic fatty liver disease: definitions, risk factors, and workup. Clinics in Liver Disease 2012;1:99-103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kuang J, Wang J, Li Y, Li M, Zhao M, Ge K, et al. Hyodeoxycholic acid alleviates non-alcoholic fatty liver disease through modulating the gut-liver axis. Cell Metabolism 2023;35:10. [DOI: 10.1016/j.cmet.2023.07.011] [DOI] [PubMed] [Google Scholar]
- 45.European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO clinical practice guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). Journal of Hepatology 2024;81:492-542. [DOI: 10.1016/j.jhep.2024.04.031] [DOI] [PubMed] [Google Scholar]
- 46.Ursodeoxycholic acid market size, share, growth and industry analysis by type (synthetic UDCA, extraction UDCA) by application (drugs, health care products), regional insights, and forecast from 2024 to 2032. https://www.businessresearchinsights.com/market-reports/ursodeoxycholic-acid-market-103041 (accessed 10 October 2024).
- 47.US Food and Drug Administration (FDA). Drug trials snapshots: OCALIVA. U.S. Food and Drug Administration (FDA) 2020;1-1:1. [Google Scholar]
- 48.US Food and Drug Administration (FDA). Ocaliva (obeticholic acid) by intercept pharmaceuticals: drug safety communication – due to risk of serious liver injury, FDA restricts use of Ocaliva in primary biliary cholangitis patients with advanced cirrhosis. U.S. Food and Drug Administration (FDA) 2021;1-1:1. [Google Scholar]
- 49.Bertha M, Choi G, Mellinger J. Diagnosis and treatment of alcohol-associated liver disease: a patient-friendly summary of the 2019 AASLD guidelines. Clinical Liver Disease 2021;17:418-23. [DOI: 10.1002/cld.1129] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology (Baltimore, Md.) 2018;67(1):328-57. [DOI: 10.1002/hep.29367] [DOI] [PubMed] [Google Scholar]
- 51.McPherson S, Armstrong M, Cobbold J, Corless L, Anstee Q, Aspinall R, et al. Quality standards for the management of NAFLD: consensus recommendations from the British Association for the Study of the Liver (BASL) and British Society of Gastroenterology (BSG) NAFLD special interest group. Lancet. Gastroenterology & Hepatology 2022;7(8):755-69. [DOI: 10.1016/S2468-1253(22)00061-9] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Orlando R, Azzalini L, Orando S, Lirussi F. Bile acids for non-alcoholic fatty liver disease and/or steatohepatitis. Cochrane Database of Systematic Reviews 2007, Issue 1. Art. No: CD005160. [DOI: 10.1002/14651858.CD005160] [DOI] [PubMed] [Google Scholar]
- 53.Zhang W, Tang Y, Huang J, Ren H, Yang Y, Yang Q, et al. Efficacy of ursodeoxycholic acid in nonalcoholic fatty liver disease: an updated meta-analysis of randomized controlled trials. https://www.researchsquare.com/article/rs-6563/v1 (accessed 24 October 2024). [DOI: 10.21203/rs.2.15906/v1.] [DOI] [PubMed]
- 54.Rind DM, Hansen R, Guzauskas G, Beinfeld M, Chapman R, Bradt P, et al. Obeticholic acid for the treatment of nonalcoholic steatohepatitis with fibrosis: effectiveness and value. Institute for Clinical and Economic Review, July 21, 2020. https://icer.org/wp-content/uploads/2020/10/ICER_NASH_Evidence_Report_072120.pdf (accessed 8 August 2025).
- 55.Neuschwander-Tetri BA, Kowdley KV, Sanyal AJ, Abdelmalek MF, Terrault N, McCullough AJ, et al. Efficacy and safety of obeticholic acid in patients with non-alcoholic steatohepatitis and significant fibrosis using endpoint definitions and populations accepted for registrational studies. Hepatology (Baltimore, Md.) 2018;68:952A. [Google Scholar]
- 56.Traussnigg S, Schattenberg JM, Demir M, Wiegand J, Geier A, Teuber G, et al. Norursodeoxycholic acid versus placebo in the treatment of non-alcoholic fatty liver disease: a double-blind, randomised, placebo-controlled, phase 2 dose-finding trial. Lancet. Gastroenterology and Hepatology 2019;4(10):781-93. [DOI] [PubMed] [Google Scholar]
- 57.Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2019;394:2184-96. [DOI] [PubMed] [Google Scholar]
- 58.Higgins JP, Eldridge S, Li T. Chapter 23: Including variants on randomized trials (last updated October 2019). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 59.International Council for Harmonisation of technical requirements for pharmaceuticals for human use (ICH). ICH Harmonised Guideline. Integrated addendum to ICH E6(R1): guideline for good clinical practice E6(R2). https://database.ich.org/sites/default/files/E6_R2_Addendum.pdf (accessed 15 June 2025).
- 60.Lefebvre C, Glanville J, Briscoe S, Featherstone R, Littlewood A, Metzendorf M-I, et al. Chapter 4: Searching for and selecting studies (last updated September 2024). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 61.Rethlefsen ML, Kirtley S, Waffenschmidt S, Ayala AP, Moher D, Page MJ, et al; PRISMA-S Group. PRISMA-S: an extension to the PRISMA statement for reporting literature searches in systematic reviews. Systematic Reviews 2021;10(1):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lefebvre C, Glanville J, Briscoe S, Featherstone R, Littlewood A, Metzendorf M-I, et al. Technical Supplement to Chapter 4: Searching for and selecting studies (last updated September 2024). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 63.Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5 (updated August 2024). Cochrane, 2024. Available from https://www.cochrane.org/handbook. [DOI] [PMC free article] [PubMed]
- 64.Review Manager (RevMan). Version 9.4.0. The Cochrane Collaboration, 2024. Available at https://revman.cochrane.org.
- 65.Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User Manual for Trial Sequential Analysis (TSA); 2nd edition. Copenhagen Trial Unit, 2017. Available from https://ctu.dk/tsa/ (accessed 15 January 2024).
- 66.TSA – Trial Sequential Analysis. Version 0.9.5.10 Beta. Copenhagen: Copenhagen Trial Unit, 2021. https://ctu.dk/tsa/downloads/.
- 67.Wetterslev J, Jakobsen JC, Gluud C. Trial Sequential Analysis in systematic reviews with meta-analysis. BMC Medical Research Methodology 2017;17(1):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.GRADEpro GDT. Version (accessed 10 July 2020). Hamilton (ON): McMaster University (developed by Evidence Prime), 2020. Available at https://www.gradepro.org.
- 69.Covidence systematic review software. Version (accessed 13 December 2023). Melbourne, Australia: Veritas Health Innovation, 2023. Available at https://www.covidence.org.
- 70.Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ (Clinical Research Ed.) 2021;372:n71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ (Clinical Research Ed.) 2021;372:n160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Higgins JP, Savović J, Page MJ, Elbers RG, Sterne JA. Chapter 8: Assessing risk of bias in a randomized trial (last updated October 2019). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 73.Sterne JA, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ (Clinical Research Ed.) 2019;366:l4898. [DOI] [PubMed] [Google Scholar]
- 74.Schünemann HJ, Higgins JP, Vist GE, Glasziou P, Akl EA, Skoetz N, et al. Chapter 14: Completing 'Summary of findings' tables and grading the certainty of the evidence (last updated August 2023). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://cochrane.org/handbook.
- 75.Higgins JP, Li T, Deeks JJ. Chapter 6: Choosing effect measures and computing estimates of effect (last updated August 2023). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 76.Schünemann HJ, Vist GE, Higgins JP, Santesso N, Deeks JJ, Glasziou P, et al. Chapter 15: Interpreting results and drawing conclusions (last updated August 2023). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://cochrane.org/handbook.
- 77.Deeks JJ, Higgins JP, Altman DG, McKenzie JE, Veroniki AA. Chapter 10: Analysing data and undertaking meta-analyses (last updated November 2024). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 78.Bjelakovic G, Nikolova D, Bjelakovic M, Pavlov CS, Sethi NJ, Korang SK, et al. Effects of primary or secondary prevention with vitamin A supplementation on clinically important outcomes: a systematic review of randomised clinical trials with meta-analysis and trial sequential analysis. BMJ Open 2024;14(5):e078053. [DOI: 10.1136/bmjopen-2023-078053] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Egger M, Davey SG, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ (Clinical Research Ed.) 1997;315(7109):629-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Page MJ, Higgins JP, Sterne JA. Chapter 13: Assessing risk of bias due to missing evidence in a meta-analysis (last updated August 2024). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 81.Harbord RM, Egger M, Sterne JA. A modified test for small-study effects in meta-analyses of controlled trials with binary endpoints. Statistics in Medicine 2006;25(20):3443-57. [DOI] [PubMed] [Google Scholar]
- 82.Rücker G, Schwarzer G, Carpenter J. Arcsine test for publication bias in meta-analyses with binary outcomes. Statistics in Medicine 2008;27:746-63. [DOI] [PubMed] [Google Scholar]
- 83.Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics 1994;50(4):1088-101. [PubMed] [Google Scholar]
- 84.McKenzie JE, Brennan SE. Chapter 12: Synthesizing and presenting findings using other methods (last updated October 2019). In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.5. Cochrane, 2024. Available from https://www.cochrane.org/handbook.
- 85.Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Statistics in Medicine 2002;21(11):1539-58. [DOI] [PubMed] [Google Scholar]
- 86.Lundh A, Lexchin J, Mintzes B, Schroll JB, Bero L. Industry sponsorship and research outcome: systematic review with meta-analysis. Intensive Care Medicine 2018;44(10):1603-12. [DOI] [PubMed]
- 87.Castellini G, Bruschettini M, Gianola S, Gluud C, Moja L. Assessing imprecision in Cochrane systematic reviews: a comparison of GRADE and Trial Sequential Analysis. Systematic Reviews 2018;7(110):1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gartlehner G, Nussbaumer-Streit B, Wagner G, Patel S, Swinson-Evans T, Dobrescu A, et al. Increased risks for random errors are common in outcomes graded as high certainty of evidence. Journal of Clinical Epidemiology 2019;106:50-9. [DOI: 10.1016/j.jclinepi.2018.10.009] [DOI] [PubMed] [Google Scholar]
- 89.Wetterslev J, Thorlund K, Brok J, Gluud C. Trial sequential analysis may establish when firm evidence is reached in cumulative meta-analysis. Journal of Clinical Epidemiology 2008;61(1):64-75. [DOI] [PubMed] [Google Scholar]
- 90.Brok J, Thorlund K, Gluud C, Wetterslev J. Trial sequential analysis reveals insufficient information size and potentially false positive results in many meta-analyses. Journal of Clinical Epidemiology 2008;61:763-9. [DOI] [PubMed] [Google Scholar]
- 91.Brok J, Thorlund K, Wetterslev J, Gluud C. Apparently conclusive meta-analyses may be inconclusive – Trial Sequential Analysis adjustment of random error risk due to repetitive testing of accumulating data in apparently conclusive neonatal meta-analyses. International Journal of Epidemiology 2009;38(1):287-98. [DOI] [PubMed] [Google Scholar]
- 92.Thorlund K, Anema A, Mills E. Interpreting meta-analysis according to the adequacy of sample size. An example using isoniazid chemoprophylaxis for tuberculosis in purified protein derivative negative HIV-infected individuals. Clinical Epidemiology 2010;2:57-66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Jakobsen J, Wetterslev J, Winkel P, Lange T, Gluud C. Thresholds for statistical and clinical significance in systematic reviews with meta-analytic methods. BMC Medical Research Methodology 2014;14:120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wetterslev J, Thorlund K, Brok J, Gluud C. Estimating required information size by quantifying diversity in a random-effects meta-analysis. BMC Medical Research Methodology 2009;9:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Schünemann H, Brożek J, Guyatt G, Oxman A, editor(s). Handbook for grading the quality of evidence and the strength of recommendations using the GRADE approach (updated October 2013). GRADE Working Group, 2013. Available from https://gdt.guidelinedevelopment.org/app/handbook/handbook.html.
- 96.Balshem H, Helfand M, Schünemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401-6. [DOI] [PubMed] [Google Scholar]
- 97.Guyatt G, Oxman AD, Vist GE, Kunz R, Falck Y, Alonso CP, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ (Clinical Research Ed.) 2008;336(7650):924-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction – GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383-94. [DOI] [PubMed] [Google Scholar]
- 99.Guyatt GH, Oxman AD, Kunz R, Atkins D, Brozek J, Vist G, et al. GRADE guidelines: 2. Framing the question and deciding on important outcomes. Journal of Clinical Epidemiology 2011;64(4):395-400. [DOI] [PubMed] [Google Scholar]
- 100.Guyatt GH, Oxman AD, Vist G, Kunz R, Brozek J, Alonso-Coello P, et al. GRADE guidelines: 4. Rating the quality of evidence – study limitations (risk of bias). Journal of Clinical Epidemiology 2011;64(4):407-15. [DOI] [PubMed] [Google Scholar]
- 101.Guyatt GH, Oxman AD, Montori V, Vist G, Kunz R, Brozek J, et al. GRADE guidelines: 5. Rating the quality of evidence – publication bias. Journal of Clinical Epidemiology 2011;64(12):1277-82. [DOI] [PubMed] [Google Scholar]
- 102.Guyatt GH, Oxman AD, Kunz R, Brozek J, Alonso-Coello P, Rind D, et al. GRADE guidelines 6. Rating the quality of evidence – imprecision. Journal of Clinical Epidemiology 2011;64(12):1283-93. [DOI] [PubMed] [Google Scholar]
- 103.Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 7. Rating the quality of evidence – inconsistency. Journal of Clinical Epidemiology 2011;64(12):1294-302. [DOI] [PubMed] [Google Scholar]
- 104.Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 8. Rating the quality of evidence – indirectness. Journal of Clinical Epidemiology 2011;64(12):1303-10. [DOI] [PubMed] [Google Scholar]
- 105.Guyatt GH, Oxman AD, Sultan S, Glasziou P, Akl EA, Alonso-Coello P, et al. GRADE guidelines: 9. Rating up the quality of evidence. Journal of Clinical Epidemiology 2011;64(12):1311-6. [DOI] [PubMed] [Google Scholar]
- 106.Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso-Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66(2):151-7. [DOI] [PubMed] [Google Scholar]
- 107.Guyatt GH, Oxman AD, Santesso N, Helfand M, Vist G, Kunz R, et al. GRADE guidelines: 12. Preparing summary of findings tables – binary outcomes. Journal of Clinical Epidemiology 2013;66(2):158-72. [DOI] [PubMed] [Google Scholar]
- 108.Guyatt GH, Thorlund K, Oxman AD, Walter SD, Patrick D, Furukawa TA, et al. GRADE guidelines: 13. Preparing summary of findings tables and evidence profiles – continuous outcomes. Journal of Clinical Epidemiology 2013;66(2):173-83. [DOI] [PubMed] [Google Scholar]
- 109.Guyatt G, Andrews J, Oxman AD, Alderson P, Dahm P, Falck-Ytter Y, et al. GRADE guidelines: 15. Going from evidence to recommendations: the significance and presentation of recommendations. Journal of Clinical Epidemiology 2013;66(7):719-25. [DOI] [PubMed] [Google Scholar]
- 110.Guyatt GH, Ebrahim S, Alonso-Coello P, Johnston BC, Mathioudakis AG, Briel M, et al. GRADE guidelines 17: assessing the risk of bias associated with missing participant outcome data in a body of evidence. Journal of Clinical Epidemiology 2017;87:14-22. [DOI] [PubMed] [Google Scholar]
- 111.Mustafa RA, Santesso N, Brozek J, Akl EA, Walter SD, Norman G, et al. The GRADE approach is reproducible in assessing the quality of evidence of quantitative evidence syntheses. Journal of Clinical Epidemiology 2013;66(7):736-42. [DOI] [PubMed] [Google Scholar]
- 112.Zeng L, Brignardello-Petersen R, Hultcrantz M, Mustafa RA, Murad MH, Iorio A, et al. GRADE guidance 34: update on rating imprecision using a minimally contextualized approach. Journal of Clinical Epidemiology 2022;150:216-24. [DOI: 10.1016/j.jclinepi.2022.07.014] [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplementary material 1 Search strategies
Data Availability Statement
Data sharing is not applicable to this article as it is a protocol, so no datasets were generated or analysed.