Scolaris Content Display Scolaris Content Display

Beta‐blockers for suspected or diagnosed acute myocardial infarction

Esta versión no es la más reciente

Contraer todo Desplegar todo

Abstract

This is a protocol for a Cochrane Review (Intervention). The objectives are as follows:

To assess the benefits and harms of beta‐blockers for patients with suspected or diagnosed acute myocardial infarction compared with placebo or no intervention at one‐month follow‐up, and at maximum follow‐up.

Background

Description of the condition

Cardiovascular disease is the number one cause of death globally (Cooper 2000; Lloyd‐Jones 2010; Nichols 2014; Rosamond 2008; Schmidt 2012). Ischaemic heart disease accounts for almost 50% of the disease burden of the cardiovascular diseases (Nichols 2014). According to the World Health Organization (WHO), 7.4 million people died from ischaemic heart disease in 2012 (WHO 2015).

Ischaemic heart disease is caused by different underlying mechanisms: (1) atherosclerotic plaque‐related obstruction of the coronary arteries; (2) focal or diffuse spasms of normal or plaque‐diseased arteries; (3) microvascular dysfunction; and (4) left ventricular dysfunction caused by acute myocardial necrosis or ischaemic cardiomyopathy (Montalescot 2013). Ischaemic heart disease increases the risk of stable angina pectoris and acute coronary syndrome (see below).

Acute coronary syndrome is a collective term for: (1) unstable angina pectoris (chest pain during rest related to ischaemia or hypoxia of the heart muscle (Roffi 2016)); (2) acute non‐ST‐elevation myocardial infarction (NSTEMI); or (3) acute ST‐elevation myocardial infarction (STEMI) (O'Gara 2013; Steg 2012). Myocardial infarction is caused by death of cardiac myocytes (myocardial necrosis) due to ischaemia (Roffi 2016; Steg 2012). The clinical definition of myocardial infarction is elevated serum levels of cardiac biomarkers (cardiac specific troponins and CK‐MB among others) and changes of the ST‐segment on an electrocardiogram (ECG) (STEMI and NSTEMI) or symptoms of cardiac ischaemia (Roffi 2016; Steg 2012).

The diagnosis of myocardial infarction is dependent on an elevation of the serum levels of cardiac‐specific troponin I, troponin T, or the myocardial band (MB) isoenzyme of creatine kinase (CK‐MB), among others (Roffi 2016; Steg 2012). However, it will often take 8 to 24 hours after the first symptoms of the myocardial infarction occur before these enzymes are detectable in serum. Beta‐blockers may accordingly be commenced as an intervention in patients suspected of myocardial infarction or may be commenced as an intervention in patients diagnosed with myocardial infarction.

The cause of myocardial infarction is generally divided in to five main classes (Thygesen 2012).

  • Type 1: spontaneous myocardial infarction related to atherosclerotic plaque rupture, ulceration, fissuring, erosion, or dissection with resulting intraluminal thrombus in one or more of the coronary arteries often caused by coronary artery disease.

  • Type 2: myocardial infarction secondary to an ischaemic imbalance such as coronary artery spasm, coronary embolism, anaemia, arrhythmias, hypertension, or hypotension.

  • Type 3: myocardial infarction with symptoms suggestive of myocardial ischaemia and resulting in sudden unexpected cardiac death when biomarker values are unavailable or could not be obtained before death.

  • Type 4a: myocardial infarction associated with percutaneous coronary intervention (PCI).

  • Type 4b: myocardial infarction associated with stent thrombosis as documented by angiography or at autopsy.

  • Type 5: myocardial infarction associated with coronary artery bypass graft (CABG).

Major complications associated with myocardial infarction

  • Life‐threatening ventricular arrhythmias caused by changes in the electrophysiologic characteristics of the myocyte, electrolyte imbalance, continuous ischaemia, and variations in heart rate all due to obstruction and hence reduced flow to the myocardium and myocardial necrosis (Brieger 2009; Stevenson 1989).

  • Mechanical complications caused by necrosis of the myocardium such as ventricular wall rupture, septum rupture, and papillary muscle rupture (Brieger 2009; Stevenson 1989; Pohjola‐Sintonen 1989).

  • Cardiogenic shock caused by failure of the ventricle to pump adequate amount of blood leading to a systemic hypotension (Brieger 2009; Stevenson 1989).

  • Acute decompensated heart failure caused by impairment in systolic and diastolic function due to myocardial ischaemia (Brieger 2009).

  • Depression (Thombs 2006).

Description of the intervention

The discovery of the difference between adrenergic receptors by Raymond Ahlquist in 1948 led Sir James Black to develop the first clinically useful beta‐receptor blocker (propranolol) in 1964 (Ahlquist 1948; Black 1964). This discovery was awarded the Nobel Prize in 1988 (Quirke 2006). Beta‐blockers are classified as non‐selective beta‐blockers or selective beta‐blockers according to their selectivity for one of the three subtypes of beta‐receptors.

  • The beta1‐receptor is mainly located in: (1) the heart, where it induces positive chronotropic effects (increases heart rate) and positive inotropic effects (increases contractility of the myocardium); and (2) in the kidneys where activation of the beta1‐receptor results in an increased release of renin which in turn increases blood pressure, among other effects (Golan 2011; Marlin 1975; Singh 1975).

  • The beta2‐receptor is mainly located in smooth muscle cells where it promotes relaxation, in skeletal muscle cells where it promotes tremor and increased glycogenolysis, and in the liver, where it increases glycogenolysis (Golan 2011).

  • The beta3‐receptor is mainly located in adipose tissue where it primarily induces lipolysis (Golan 2011).

Beta‐blockers may be administered both intravenously and orally. Three different classes of beta‐blockers exist: (1) the first generation non‐selective beta‐blockers (e.g. propranolol, oxprenolol, sotalol, timolol) affecting all beta‐receptors; (2) the second generation selective beta1‐blockers (e.g. metoprolol, bisoprolol, acebutolol, atenolol, esmolol) mainly affecting the heart; and (3) the third generation beta‐blockers which have combined non‐selective beta‐blocking effects and alpha‐blocking effects (e.g. carvedilol) affecting all beta‐receptors plus alpha‐receptors in the vessels lowering the blood pressure.

Several beta‐blockers have been used in the management of myocardial infarction. The first beta‐blockers used were the non‐selective beta‐blockers (e.g. propranolol) (Clausen 1966; Friedman 1986). Today the most frequently used beta‐blockers for managing myocardial infarction are the cardiac‐specific beta1‐blockers (Chen 2005; Roffi 2016; Steg 2012).

How the intervention might work

The beta‐receptor is an adrenergic Gs heterotrimeric G‐protein coupled receptor, located throughout the body. Beta‐receptors are stimulated by the sympathetic nervous system with catecholamines epinephrine (adrenaline) and norepinephrine (noradrenaline) as their primary endogenous agonists. The role of acute treatment or subacute treatment with beta‐blockers in patients suspected of or diagnosed with myocardial infarction, rests on their inhibition of the chronotropic and inotropic effects of the beta‐receptor. This may result in a reduction in heart rate, heart contractility, and blood pressure thereby decreasing the oxygen demand of the heart (Lopez‐Sendon 2004). Hence, the inhibition of the beta‐receptor is thought to decrease ischaemia and might decrease the risk of life‐threatening ventricular arrhythmias and other complications associated with myocardial infarction (Roffi 2016; Steg 2012).

Why it is important to do this review

The prevalence of ischaemic heart disease is considerable. According to the WHO, 7.4 million people died from ischaemic heart disease in 2012 (Lloyd‐Jones 2010; Nichols 2014; Rosamond 2008; WHO 2015). A considerable reduction in disease burden and healthcare cost may therefore be alleviated by effective treatment. However, as demonstrated below, previous meta‐analyses and guidelines show contrasting findings and recommendations.

The role of beta‐blockers in other settings is still debatable.

Beta‐blockers used to be contraindicated in patients with congestive heart failure. Non‐selective combined alpha‐ and beta‐blockers are now a part of standard treatment of congestive heart failure (Chatterjee 2013a; Yancy 2013).

Beta‐blockers are also considered an option in the treatment of hypertension, but are rarely used as first‐line treatment (Mancia 2013). A recent Cochrane review found that beta‐blockers were inferior when compared with other antihypertensive drugs (Wiysonge 2012). Non‐selective beta‐blockers are used in the treatment of anxiety due to their effect on decreasing tremor and tachycardia (Turner 1994).

The adverse effects of beta‐blockers are both cardiac adverse effects and non‐cardiac adverse effects. Among the most serious cardiac adverse effects is exacerbation of heart failure in patients with acute decompensated heart failure, due to the need of sympathetic activity to maintain the cardiac output (Taylor 1982). In addition, beta‐blocker withdrawal has also been shown to cause exacerbation of ischaemic symptoms and precipitate acute myocardial infarction in patients with ischaemic heart disease (Houston 1981).

Perioperative beta‐blockade for major non‐cardiac surgery in patients with risk factors for ischaemic heart disease has been tested in several trials (Bangalore 2008; Devereaux 2008; Juul 2006) and seems to increase 30‐day all‐cause mortality, increase the risk of stroke, although the risk of non‐fatal myocardial infarction seems to be reduced (Bangalore 2008).

Case studies have suggested that depression, fatigue, and sexual dysfunction are among the beta‐blocker induced non‐cardiac adverse effects (Greenblatt 1974; Waal 1967; Warren 1977). However, a meta‐analysis comparing beta‐blockers versus placebo showed no difference on depressive symptoms and only a minor increase in sexual dysfunction and fatigue in patients randomised to beta‐blockers compared with placebo (Ko 2002).

Evidence on the effects of beta‐blockers for suspected or diagnosed acute myocardial infarction

Outcomes assessed at hospital discharge

Five meta‐analyses have compared the effects of beta‐blockers versus placebo, standard medical therapy, or late administration of beta‐blockers in participants with suspected or diagnosed myocardial infarction on outcomes reported at hospital discharge (Al‐Reesi 2008; Brandler 2010; Chatterjee 2013; Freemantle 1999; Yusuf 1985). While Chatterjee 2013 only assessed intravenous beta‐blockers and showed a beneficial effect of early beta‐blockers on mortality, Al‐Reesi 2008, Brandler 2010, and Freemantle 1999 assessed any type of beta‐blockers and could not demonstrate a beneficial effect of beta‐blockers on mortality. Yusuf 1985 assessed the effects of beta‐blockers on size of myocardial infarction and showed a beneficial effect when compared with no beta‐blockers. One of the meta‐analyses showed a beneficial effect of beta‐blockers on risk of myocardial reinfarction and ventricular arrhythmia, while no beneficial or harmful effects were found on cardiogenic shock (Chatterjee 2013). Al‐Reesi 2008, Brandler 2010, and Freemantle 1999 did not assess the effects of beta‐blockers on risk of myocardial reinfarction, ventricular arrhythmias, or cardiogenic shock.

Long‐term outcomes

Three meta‐analyses compared the effects of beta‐blockers versus no beta‐blockers in participants with suspected or diagnosed myocardial infarction on long‐term outcomes (Bangalore 2014; Freemantle 1999; Yusuf 1985). While Freemantle 1999 and Yusuf 1985 showed a beneficial effect of beta‐blockers on mortality, Bangalore 2014 only found a beneficial effect on mortality in trials where the participants did not receive reperfusion in the form of revascularisation (percutaneous coronary intervention or coronary artery bypass graft), or thrombolytics (e.g. streptokinase). Bangalore 2014 found a beneficial effect of beta‐blockers on symptoms of angina and risk of recurrent myocardial infarction regardless of whether the participants received intervention for reperfusion (revascularisation or thrombolytics) or not. However, Bangalore 2014 also showed that beta‐blockers seem to increase the severity of heart failure in participants receiving intervention for reperfusion (revascularisation or thrombolytics). It must be noted that Bangalore 2014 included a larger number of trials than Freemantle 1999 and Yusuf 1985, and only Bangalore 2014 included trials after the introduction of reperfusion strategies.

Current guidelines for using beta‐blockers in patients with suspected or diagnosed myocardial infarction

The American College of Cardiology Foundation/American Heart Association (ACCF/AHA) guideline recommends acute intravenous beta‐blockers in patients suspected of STEMI who are hypertensive or have ongoing ischaemia, unless there are contraindications to beta‐blockers (allergy towards beta‐blockers, signs of acute decompensated heart failure, increased risk of cardiogenic shock, atrio‐ventricular block, asthma, or chronic obstructive lung disease) (O'Gara 2013). The guideline recommends oral beta‐blockers within the first 24 hours in patients with a STEMI and no contraindications (see above) (O'Gara 2013).

The ACCF/AHA guideline does not recommend acute intravenous beta‐blockers in patients suspected of acute NSTEMI and advise against intravenous beta‐blockers in acute patients with risk factors for cardiogenic shock (Amsterdam 2014). However, the guideline recommends oral beta‐blockers commenced within the first 24 hours in patients with a NSTEMI or unstable angina pectoris and no contraindications (see above) (Amsterdam 2014).

Former meta‐analyses have shown conflicting results and no former reviews have used Cochrane methodology to assess the effects of beta‐blockers as an acute intervention in patients suspected or diagnosed with myocardial infarction. The present systematic review will be the first to take fully account of the risk of systematic errors ('bias'), design errors, and risks of random errors ('play of chance') (Higgins 2011; Jakobsen 2014; Keus 2010; Thorlund 2011), and include trials irrespective of outcome, follow‐up duration, number of participants, language, and publication status.

Objectives

To assess the benefits and harms of beta‐blockers for patients with suspected or diagnosed acute myocardial infarction compared with placebo or no intervention at one‐month follow‐up, and at maximum follow‐up.

Methods

Criteria for considering studies for this review

Types of studies

Randomised controlled trials (RCTs) irrespective of publication type, reported outcomes, publication status, publication date, and language. We will not specifically search for non‐randomised studies. However, if we, during the literature search, identify non‐randomised studies (quasi‐randomised studies or observational studies) with adequate reports of harmful effects, then we will narratively report these results.

Types of participants

Participants irrespective of age where the trialists have described the participants as either with suspected myocardial infarction or diagnosed myocardial infarction.

Types of interventions

Experimental group

We will include any type of beta‐blockers, both administered as intravenous therapy and as oral administration, as experimental intervention (non‐selective beta‐blockers (propranolol, oxprenolol, sotalol, timolol); selective beta1‐blockers (metoprolol, bisoprolol, acebutolol, atenolol, esmolol); and beta‐blockers which are combined alpha‐ and non‐selective beta‐blockers (carvedilol)) commenced in the acute or subacute phase of myocardial infarction.

Control group

We will accept placebo or no intervention as control interventions.

Co‐interventions

We will accept any co‐intervention (medical therapy as well as revascularisation strategy) provided they are intended to be delivered similarly to the experimental and the control groups.

Types of outcome measures

Primary outcomes

  • All‐cause mortality.

  • Major cardiovascular event defined as a composite outcome consisting of either cardiovascular mortality (defined by trialists) or myocardial infarction (defined by trialists). Additionally, we will assess cardiovascular mortality and myocardial infarction separately as secondary outcomes (see below).

  • Serious adverse event defined as any untoward medical occurrence that: resulted in death, was life‐threatening, was persistent, led to significant disability, prolonged hospitalisation, or jeopardised the participant (ICH‐GCP 1997).

Secondary outcomes

  • Quality of life measured on any valid scale, such as the Short‐Form (36) Health Survey (SF‐36) (Ware 1992).

  • Angina measured on any valid scale, such as the Canadian Cardiovascular Angina Score (CCS) (Campeau 1976).

  • Cardiovascular mortality.

  • Myocardial infarction.

We will narratively report adverse events, presenting them in a table.

We will estimate all outcomes at two different follow‐ups.

  • Outcomes assessed at the follow‐up time point closest to one month after randomisation (this will be the follow‐up time point of primary interest).

  • Outcomes assessed at maximum follow‐up.

We chose one‐month follow‐up as our primary follow‐up time point because the possible effects of beta‐blockers needs some time to show (one month), and the follow‐up period is not too long so other factors unrelated to the given trial affecting the outcomes might decrease the statistical power, i.e. the results are 'diluted' by events (e.g. traffic accidents) unrelated to the trial.

Search methods for identification of studies

Electronic searches

We will search the Cochrane Central Register of Controlled Trials (CENTRAL) in the Cochrane Library, MEDLINE Ovid, Embase Ovid, LILACS Bireme (Latin American and Caribbean Health Science Information database), Science Citation Index Expanded and BIOSIS Citation Index on Thomson Reuters Web of Science in order to identify relevant trials (Royle 2003). Additionally we will search the World Health Organization International Clinical Trials Registry Platform (WHO ICTRP) (apps.who.int/trialsearch), US National Institutes of Health Ongoing Trials Register ClinicalTrials.gov (www.clinicaltrials.gov), Turning Research Into Practice (TRIP), Google Scholar and Scisearch for finished trials as well as ongoing trials.

The preliminary search strategy for MEDLINE Ovid (Appendix 1) will be adapted for use in the other databases. We will apply the Cochrane sensitivity‐maximising RCT filter (Lefebvre 2011) to MEDLINE Ovid and adaptations of it to the other databases, except CENTRAL.

We will search all databases from their inception to the present and we will impose no restriction on language of publication. If we identify any papers in a language not known by the author group, we will seek help. This will be acknowledged in the Acknowledgements section.

Searching other resources

Additional trials will be identified and included where relevant by searching the bibliographies of review articles and identified trials.

Data collection and analysis

The analyses will be performed using Review Manager 5 (RevMan 2014), Stata 14 (Stata 2015), and Trial Sequential Analysis (TSA) (CTU 2011; Thorlund 2011) software.

Selection of studies

Two review authors (Emil Eik Nielsen (EEN) and Joshua Feinberg (JF)) will assess each identified trial independently. If a trial is identified as relevant by one author, but not by another, the reasoning behind each decision will be discussed. If no agreement can be reached, a third review author (Janus C Jakobsen (JCJ)) will resolve the discussion.

Data extraction and management

We will use a data collection form for trial characteristics and outcome data which has been piloted on at least one trial in the review. Two review authors (JF and EEN) will extract trial characteristics from included trials. We will extract the following trial characteristics.

  1. Methods:duration of the trial, details of any 'run in' period, and date of publication.

  2. Participants: number randomised, number analysed, mean age, sex, inclusion and exclusion criteria.

  3. Interventions: intervention, comparison, concomitant medications, and excluded medications.

  4. Outcomes: primary and secondary outcomes specified and collected, and time points reported.

  5. Notes: funding for trial, and notable conflicts of interest of trial authors.

Four review authors (EEN, JF, Sanam Safi (SF) and Naqash J Sethi (NJS)) will independently extract outcome data in pairs from included studies. We will resolve disagreements by consensus or by involving a third person (JCJ). One review author will transfer data into the Review Manager file (RevMan 2014). We will double‐check that data are entered correctly by comparing the data presented in the systematic review with the study reports. A second review author will spot‐check study characteristics for accuracy against the trial report.

Assessment of risk of bias in included studies

We will use the instructions given in the Cochrane Handbook for Systematic Reviews of Interventions in our evaluation of the methodology and the risk of bias of the included trials (Higgins 2011). Two review authors will assess the included trials independently. We will evaluate the risks of bias in random sequence generation, allocation concealment, blinding of participants and treatment providers, blinding of outcome assessment, incomplete outcome data, selective outcome reporting, and other bias sources. This is done because these domains enable classification of randomised controlled trials with low risk of bias and of randomised controlled trials with unclear or high risk of bias. The latter trials overestimate benefits and underestimate harms (Gluud 2006; Kjaergard 2001; Lundh 2012; Moher 1998; Savovic 2012; Savovic 2012a; Schulz 1995; Wood 2008). For additional details on how the risk of bias will be assessed see Appendix 2.

Overall risk of bias

  • Low risk of bias: the outcome result will be classified as at overall 'low risk of bias' only if all of the bias domains described in the above paragraphs are classified as at low risk of bias.

  • High risk of bias: the outcome result will be classified as at 'high risk of bias' if any of the bias risk domains described above are classified as at 'unclear' or 'high risk of bias'.

We will assess the domains blinding of outcome assessment, incomplete outcome data, and selective outcome reporting for each outcome. Thus, we will be able to assess the bias risk for each result in addition to each trial. We will base our primary conclusions as well as our presentation in the 'Summary of findings' table on the results of our primary outcomes with low risk of bias.

Measures of treatment effect

Dichotomous outcomes

We will calculate risk ratios (RR) with 95% confidence intervals (CI) and TSA‐adjusted CIs (Thorlund 2011) for dichotomous outcomes.

Continous outcomes

We will calculate the mean differences (MD) with 95% CIs and TSA‐adjusted CIs (Thorlund 2011) for continuous outcomes. We will use the standardised mean difference when the trials all assess the same outcome but measure it in a variety of ways (e.g. different scales) (Higgins 2011).

Dealing with missing data

We will contact all study authors for missing data.

Dichotomous outcomes

If included studies have used rigorous methodology (i.e. reporting on outcomes for all participants or multiple imputation to deal with missing data), we will use these data in our primary analysis (Sterne 2009), otherwise we will use the last observation carried forward to handle missing data or if the proportion of dropouts are less than 5%. We will not impute missing values for any outcomes in our primary analysis.

Continous outcomes

If included studies have used rigorous methodology (i.e. reporting on outcomes for all participants or multiple imputation to deal with missing data), we will use these data in our primary analysis (Sterne 2009), otherwise we will use the last observation carried forward to handle missing data or if the proportion of dropouts are less than 5%. We will not impute missing values for any outcomes in our primary analysis. If standard deviations (SD) are not reported, the SDs will be calculated using data from the trial if possible. We will not use intention‐to‐treat data if the original report did not contain such data.

In our sensitivity analysis for dichotomous and continuous outcomes, we will impute data, see below and Sensitivity analysis.

Best‐worst and worst‐best case scenarios

To assess the potential impact of the missing data for dichotomous outcomes, we will perform the two following sensitivity analyses.

  1. 'Best‐worst' case scenario: it will be assumed that all participants lost to follow‐up in the experimental group survived, had no serious adverse event, had no cardiovascular event, and had no adverse events; and all those with missing outcomes in the control group have not survived, had a serious adverse event, had a cardiovascular event, and had adverse events.

  2. 'Worst‐best' case scenario: it will be assumed that all participants lost to follow‐up in the experimental group did not survive, had a serious adverse event, had a cardiovascular event, and had adverse events; and all those with missing outcomes in the control group survived, had no serious adverse event, had no cardiovascular event, and had no adverse events.

Results from both scenarios will be presented in our publication.

To assess the potential impact of missing SDs for continuous outcomes, we will perform the following sensitivity analysis.

  1. Where SDs are missing and not possible to calculate, we will impute SDs from trials with similar populations and low risk of bias. If no such trials can be found, we will impute SDs from trials with a similar population. As the final option we will impute SDs from all trials.

Assessment of heterogeneity

We will primarily investigate forest plots to visually assess any sign of heterogeneity. We will secondly assess the presence of statistical heterogeneity by Chi2 test (threshold P < 0.10) and measure the quantities of heterogeneity by the I2 statistic (Higgins 2002; Higgins 2003).

We will follow the recommendations for threshold by the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011):

  • 0% to 40%: might not be important;

  • 30% to 60%: may represent moderate heterogeneity;

  • 50% to 90%: may represent substantial heterogeneity;

  • 75% to 100%: may represent considerable heterogeneity.

We will investigate possible heterogeneity through subgroup analyses. Ultimately, we may decide that a meta‐analysis should be avoided (Higgins 2011).

Assessment of reporting biases

We will use a funnel plot to assess reporting bias if ten or more trials are included. Using the asymmetry of the funnel plot we will assess the risk of bias. For dichotomous outcomes we will test asymmetry with the Harbord test (Harbord 2006) if Tau2 < 0.1 and with the Rücker test (Rücker 2008) if Tau2 > 0.1.

For continuous outcomes we will use the regression asymmetry test (Egger 1997).

Data synthesis

Meta‐analysis

We will accept both end scores and change from baseline scores analysing continuous outcomes. If both end scores and change from baseline scores are reported then we will use end scores. If only change values are reported the results will be analysed together with end scores (Higgins 2011a).

We will undertake this systematic review according to the recommendations stated in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2011) and according to Jakobsen 2014 and Keus 2010. We will use the statistical software Review Manager 5 (RevMan 2014) provided by Cochrane to meta‐analyse data. We will use Stata (Stata 2015) in case of zero‐event trials, where Review Manager 5 zero‐event handling (replacing 0 with a constant of 0.5) is not sufficient (e.g. in cases with skewed number of participants between groups, which we will handle with reciprocal zero‐event handling according to Sweeting 2004, and in case metaregression (post hoc) is needed).

Assessment of significance

We will assess our intervention effects with both random‐effects model meta‐analyses (DerSimonian 1986) and fixed‐effect model meta‐analyses (DeMets 1987). We will use the more conservative point estimate of the two (Jakobsen 2014). The more conservative point estimate is the estimate closest to no effect. If the two estimates are equal, the estimate with the widest confidence interval will be used. We use three primary outcomes and we will therefore consider a P value less than P ≤ 0.025 as statistically significant (Jakobsen 2014). We will use the eight‐step procedure to assess if the thresholds for significance are crossed or not (Jakobsen 2014). We use four secondary outcomes and we will therefore consider a P value less than P ≤ 0.02 for the secondary outcomes (Jakobsen 2014). We will use the eight‐step procedure to assess if the thresholds for significance are crossed or not (Jakobsen 2014).

We will include all studies in our analyses, and conduct a sensitivity analysis with studies at low risk of bias. If the results are similar we will base our primary conclusions at the time point closest to one month on the overall analysis. If they differ, we will base our primary conclusions on studies with a low risk of bias.

We will present a table describing the types of serious adverse events in each trial.

Trial Sequential Analysis (TSA)

Cumulative meta‐analyses are at risk of producing random errors due to sparse data and multiple testing of accumulating data (Brok 2008; Brok 2009; Higgins 2011a; Pogue 1997; Thorlund 2009; Wetterslev 2008); therefore TSA (CTU 2011) can be applied to assess and control this risk (http://www.ctu.dk/tsa/) (Thorlund 2011). Similar to a sample size calculation in a randomised controlled trial, TSA calculates the required information size for the meta‐analysis (that is the number of participants needed in a meta‐analysis to detect or reject a certain intervention effect) in order to minimise random errors (Wetterslev 2009). The required information size takes into account the event proportion in the control group, the assumption of a plausible relative risk reduction, and the heterogeneity of the meta‐analysis (Turner 2013; Wetterslev 2009). TSA enables testing for significance to be conducted each time a new trial is included in the meta‐analysis. On the basis of the required information size, trial sequential monitoring boundaries can be constructed. This enables one to determine the statistical inference concerning cumulative meta‐analysis that has not yet reached the required information size (Wetterslev 2008).

Firm evidence for benefit or harms may be established if the trial sequential monitoring boundary is crossed before reaching the required information size, in which case further trials may turn out to be superfluous. In contrast, if the boundary is not surpassed one may conclude that it is necessary to continue with further trials before a certain intervention effect can be detected or rejected. Firm evidence for lack of the postulated intervention effect can also be assessed with TSA. This occurs when the cumulative Z‐score crosses the trial sequential monitoring boundaries for futility.

For dichotomous outcomes we will estimate the required information size based on the proportion of participants with an outcome in the control group, a relative risk reduction of 10%, an alpha of 2.5% for primary outcomes and 2.0% for secondary outcomes, a beta of 10%, and a variance suggested by the trials in a random‐effects meta‐analysis (diversity‐adjusted required information size) (Jakobsen 2014; Wetterslev 2009). In case there is some evidence of effect of the intervention, a supplementary TSA will use the limit of the confidence interval closest to 1.00 as the anticipated intervention effect (Jakobsen 2014). Additionally, we will calculate TSA‐adjusted confidence intervals.

For continuous outcomes we will estimate the required information size based on the standard deviation observed in the control group of trials with low risk of bias or lower risk of bias and a minimal relevant difference of SD / 2 for continuous outcomes, an alpha of 2.0%, a beta of 10%, and a diversity suggested by the trials in the meta‐analysis (Jakobsen 2014; Wetterslev 2009). In case there is some evidence of effect of the intervention, as a supplementary TSA we will use the limit of the confidence interval closest to 0.00 as the anticipated intervention effect (Jakobsen 2014). Additionally, we will calculate TSA‐adjusted confidence intervals.

Subgroup analysis and investigation of heterogeneity

We will perform the following subgroup analyses.

  • Comparison of the effect of beta‐blockers versus placebo or no intervention between trials where the participants commenced beta‐blockers at different time points.

    • Acute phase ‐ suspected of myocardial infarction.

    • Subacute phase ‐ diagnosed with myocardial infarction.

  • Comparison of the effect of beta‐blockers versus placebo or no intervention between trials where the participants received intervention for reperfusion (coronary artery bypass graft, percutaneous coronary intervention or thrombolytics) to that in trials where the participants did not receive intervention for reperfusion. Additionally, we will test the difference between the different reperfusion strategies.

  • Comparison of the effect of beta‐blockers versus placebo or no intervention between trials where the experimental group received different types of beta‐blockers.

  • Comparison of the effect of beta‐blockers versus placebo or no intervention between trials with different age of participants.

    • Age 0 to 18 years.

    • Age 19 to 75 years.

    • Age 76 years or above.

  • Comparision of the effect of beta‐blockers versus placebo or no intervention between trials with different clinical trial registration status.

    • Pre‐registration.

    • Post‐registration.

    • No registration.

  • Comparison of the effect of beta‐blockers versus placebo or no intervention between trials including different types of acute myocardial infarction.

    • NSTEMI.

    • STEMI.

    • Unstable angina pectoris

Sensitivity analysis

To assess the potential impact of bias, we will perform a sensitivity analysis where we exclude trials with overall 'high risk of bias'.

To assess the potential impact of the missing data for dichotomous outcomes, we will perform best‐worst and worst‐best case scenarios (see Dealing with missing data).

Summary of findings

We will use the GRADE system (Guyatt 2008) to assess the quality of the body of evidence associated with each of the primary outcomes (all‐cause mortality, major cardiovascular events, and serious adverse events), quality of life and angina in our review constructing 'Summary of findings' (SoF) tables using the GRADEpro GDT software (ims.cochrane.org/revman/other‐resources/gradepro) and the TSA‐adjusted CI for statements of precision or imprecision (Jakobsen 2014). The GRADE approach appraises the quality 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. The quality measure of a body of evidence considers within study risk of bias, the directness of the evidence, heterogeneity of the data, precision of effect estimates, and risk of publication bias. We will include all studies in our analyses, and conduct a sensitivity analysis with studies at low risk of bias. If the results are similar we will base our primary SoF tables and primary conclusions on the overall analysis. If they differ, we will base our primary SoF and primary conclusions on studies with a low risk of bias.