ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Acute myocardial infarction: Role of beta blocker therapy

Acute myocardial infarction: Role of beta blocker therapy
Literature review current through: Jan 2024.
This topic last updated: Dec 02, 2020.

INTRODUCTION — For patients with acute myocardial infarction (MI), beta blocker therapy reduces infarct size and early mortality when started early and lowers the risk of death when continued long term. (See "Overview of the acute management of ST-elevation myocardial infarction" and "Overview of the acute management of non-ST-elevation acute coronary syndromes" and "Prevention of cardiovascular disease events in those with established disease (secondary prevention) or at very high risk" and "Overview of the nonacute management of ST-elevation myocardial infarction" and "Overview of the nonacute management of unstable angina and non-ST-elevation myocardial infarction".)

This topic will discuss the use of beta blockers in patients with acute MI and focus on those without heart failure or reduced left ventricular systolic dysfunction [1]. Other relevant issues regarding beta blockers are discussed elsewhere. (See "Major side effects of beta blockers" and "Ventricular arrhythmias during acute myocardial infarction: Prevention and treatment" and "Primary pharmacologic therapy for heart failure with reduced ejection fraction".)

MECHANISM OF ACTION — Potentially beneficial effects of beta blockers in patients with acute myocardial infarction (MI) include [2]:

Decreased oxygen demand due to the reductions in heart rate, blood pressure, and contractility, and the consequent relief of ischemic chest pain.

Decreased risk of ventricular fibrillation as suggested by experimental studies demonstrating an increase in the ventricular fibrillation threshold and by clinical trials showing a relative risk reduction in sudden cardiac death (eg, 30 to 47 percent) [3-5].

Decreased automaticity, increased electrophysiologic threshold for activation, and slowing of conduction.

Bradycardia prolongs diastole and therefore improves coronary diastolic perfusion and reduces after-depolarizations and triggered activity.

Reduction in remodeling and improvement in left ventricular hemodynamic function, depending upon infarct size and the timing of treatment [6-8]. (See "Pharmacologic therapy of heart failure with reduced ejection fraction: Mechanisms of action", section on 'Beta blockers'.)

Improved left ventricular diastolic function with a less restrictive filling pattern [9]. (See "Treatment and prognosis of heart failure with preserved ejection fraction".)

Slowing of the yearly rate of progression of coronary atherosclerosis in patients with and without MI, as demonstrated by intravascular ultrasound evaluation of atheroma volume [10].

Inhibition of platelet aggregation and thromboxane synthesis [11].

One or more of the above mechanisms may contribute to a reduction in reperfusion injury. (See "Reperfusion injury of the heart", section on 'Definition'.)

INDICATIONS — We treat all patients with acute myocardial infarction (MI) with beta blocker therapy. The evidence supporting the benefit of beta blockers has been obtained primarily from randomized trials that included predominantly patients with ST-elevation myocardial infarction (STEMI). There have been no randomized trials specifically addressing the efficacy of these drugs in non-ST elevation MI (NSTEMI). Observational studies have attempted to evaluate patients with STEMI and those with NSTEMI.

Many of the relevant studies were performed before the routine use of long-term antiplatelet and statin therapy. Thus, it is possible that the absolute magnitude of the mortality benefit from beta blocker described below may be smaller due to the beneficial impact of these preventative medications as well as the use of reperfusion therapies.

Evidence from studies of STEMI patients — The use of beta blockers has been relatively well studied in patients with STEMI treated with no reperfusion, fibrinolytic therapy, or primary percutaneous coronary intervention.

No reperfusion – Randomized trials performed before the use of reperfusion therapy with either fibrinolysis or PCI consistently showed a reduction in cardiovascular mortality of 10 to 25 percent in patients treated with propranolol, metoprolol, or atenolol  (figure 1) [12-16]. A 1985 meta-analysis of these studies found a 25 percent reduction in mortality at one year [17]. These patients did not routinely receive statin therapy or a P2Y12 receptor blocker, both of which are now standard of care.

Fibrinolytic therapy – There is no high quality evidence demonstrating a beneficial impact of long-term beta blocker use in patients treated with fibrinolytic therapy. Thus, much of the evidence is indirect, coming from the benefit shown in patients not receiving reperfusion.

The individual studies that evaluated the long-term impact of beta blockade in patients treated with fibrinolysis were limited by small size [18,19], early termination [20], observational design [21], or relatively short duration follow-up [22]. A 1999 meta-analysis, which included trials of patients (n = 24,974) who did or did not receive fibrinolytic therapy, found that beta blockers reduced the odds of death by 23 percent (95% CI 15-31) when taken long term, a result comparable to that seen in patients who received no reperfusion [23].

Primary PCI – The evidence presented above, supporting long-term beta blocker use in patients treated with or without fibrinolysis, provides the basis for a similar recommendation in patients who undergo primary percutaneous coronary intervention (PCI). Some experts have suggested that the benefit of beta blockers in this subpopulation may be reduced by the improvement in outcome afforded by revascularization. There have been no randomized trials and few observational studies of long-term beta blocker therapy patients treated with primary PCI [24-26]. Three large observational studies have come to differing conclusions:

One observational study evaluated mortality in over 20,000 STEMI patients (2005 to 2010) [27]. In a propensity-matched analysis of 2650 patients who received and 1325 who did not receive a beta blocker at discharge, treatment was associated with a lower incidence of all-cause death (2.8 versus 4.1 percent; adjusted hazard ratio 0.46, 95% CI 0.27-0.78) during a median follow-up of about one year. In addition, the result was consistent across all subgroups, including those at relatively low risk, such as those with left ventricular ejection fraction >40 percent and those with single-vessel disease.

In a meta-analysis of 10 observational studies of patients (n = 16,645) with preserved left ventricular ejection fraction who underwent PCI, there was no difference in one-year mortality group between those receiving or not receiving beta blocker [28].

A second study evaluated outcomes in 88,522 STEMI and 81,993 NSTEMI patients (2007 to 2013) who did not have heart failure or left ventricular systolic dysfunction [29]. The overall one-year mortality was 5.2 percent. After adjustment, there was no significant difference in mortality between those with and without beta blocker use in either STEMI or NSTEMI groups.

INITIAL THERAPY — For patients without contraindications, we treat all patients with an oral beta blocker within the first 24 hours after diagnosis. There is no consistent evidence of benefit from the use of intravenous beta blocker use prior to primary percutaneous coronary intervention. We reserve the intravenous route principally for patients with refractory angina and/or hypertension who have ongoing ischemia prior to percutaneous coronary intervention (PCI) in whom there is no hemodynamic instability, risk for shock, heart failure, or other contraindications.

Left ventricular dysfunction/heart failure — Some of the studies discussed directly below were performed before the availability of angiotensin converting enzyme inhibitors and generally did not assess patients with asymptomatic left ventricular dysfunction or heart failure [13,15,16]. They were also performed before the strong beneficial effects of beta blockers in heart failure were demonstrated. The use of beta blockers in patients with heart failure due to systolic dysfunction is discussed elsewhere. (See "Primary pharmacologic therapy for heart failure with reduced ejection fraction".)

Fibrinolytic therapy — While, in the aggregate, the available evidence does not support routine, early initiation of intravenous beta blockade in patients treated with fibrinolytic therapy, we do recommend oral therapy. This recommendation for these patients is based on the overall benefit of beta blockers. (See 'Indications' above.)

Three randomized trials (1989, 1991, and 1993) of ST-elevation myocardial infarction (STEMI) patients treated with fibrinolytic therapy compared early and deferred beta blocker therapy and found no clear evidence of a mortality benefit with early beta blocker [22,30,31]. In these studies, deferred therapy occurred within 24 hours to up to six days.

The findings were somewhat similar in the randomized COMMIT/CCS2 trial of almost 46,000 MI patients (93 percent with ST elevation or left bundle branch block), one-half of whom received fibrinolytic therapy [32]. In this trial, patients were randomly assigned to placebo or to three 5 mg intravenous boluses of metoprolol tartrate followed by oral metoprolol extended release (succinate) 200 mg/day for 30 days. There was no overall mortality benefit from early intravenous beta blocker therapy. However, hemodynamic stability appeared to be an important determinant of the response to beta blockers. There was a significant increase in mortality in patients who presented with hemodynamic compromise that was balanced by a trend toward reduced mortality in patients who were hemodynamically stable. Importantly, patients with hemodynamic compromise and who received beta blockers did not go through a period of up-titration of the drug, raising the possibility that a more careful use of early intravenous beta blocker may improve outcomes.

Some evidence of benefit of early intravenous administration comes from the following studies:

The TIMI-IIB trial randomly assigned 1434 patients to either early intravenous (IV) therapy (metoprolol tartrate, 15 mg IV given in three equal doses at two-minute intervals if tolerated, followed by 50 mg orally twice daily for the first day and 100 mg orally twice daily thereafter) or to delayed oral beta blockade (metoprolol, 50 mg twice daily on day six and 100 mg twice daily thereafter) [30].

Immediate beta blocker therapy may reduce the incidence of intracerebral hemorrhage [30,33]. This effect was suggested in a review of data from 60,329 patients treated with alteplase in the National Registry of Myocardial Infarction [33]. Immediate beta blocker use was associated with a lower incidence of intracerebral hemorrhage (0.67 versus 1 percent for no immediate beta blocker; odds ratio 0.69, 95% CI 0.57-0.84). This apparent benefit was seen in all age groups and in both men and women.

Pooled data from controlled trials involving over 29,000 patients undergoing early, short-term IV administration of a beta blocker showed a 13 percent relative risk reduction in acute mortality [34].

Primary PCI — The evidence presented below does not support the routine use of intravenous beta blocker prior to primary percutaneous coronary intervention (PCI). It should also be kept in mind that clinical experience has shown that intravenous therapy has worsened outcomes in patients with hemodynamic instability or evidence of Killip Class II or III (table 1) heart failure. It must be given cautiously or avoided in these patients.

Two randomized trials, each with significant limitations, have evaluated the role of intravenous beta blocker before primary PCI:

In METOCARD-CNIC, 270 patients with Killip class II or less (table 1) anterior STEMI were randomly assigned to receive intravenous metoprolol (up to three 5 mg boluses of metoprolol tartrate given two minutes apart) or not before reperfusion and all patients received oral metoprolol within 24 hours [35]. The primary end point of infarct size on magnetic resonance imaging performed five to seven days was significantly smaller in the group that was treated early (25.6 versus 32.0 grams; p = 0.012). In addition, left ventricular ejection fraction was higher in the treated group at five to seven days and at six months (adjusted treatment difference 2.67 percent; p = 0.045 and 3.49 percent; p = 0.025) [36]. There was no difference in the safety end point. Limitations of the study include lack of a placebo control and exclusion of patients with inferior MI.

In the EARLY-BAMI trial, 683 patients with STEMI were randomly assigned to intravenous metoprolol (two sequential 5 mg boluses) or placebo before PCI [37]. The first bolus was given in the ambulance. The primary end point of myocardial infarct size (percent of LV), as assessed by cardiac magnetic resonance imaging at 30 days, was performed in 342 patients. There was no difference in infarct size between the two groups (15.3 versus 14.9 percent, respectively). Supporting this finding of no difference was a lack of difference in the secondary end point of peak and area under the creatinine kinase curve. Limitations of the study include failure to achieve the necessary sample size for the primary endpoint, the possibility that the groups with and without measurement of the primary end point were not well balanced, and a smaller than estimated infarct size than predicted, which may have made it harder to find a true difference. There was no difference in the rate of adverse events between the two groups.

Observational studies have also not shown convincing evidence of benefit from early intravenous beta blocker use:

An apparent benefit from preprocedural intravenous administration of a beta blocker was also shown in a retrospective analysis from the CADILLAC trial of 2082 patients [38]. At 30 days, patients who had received a preprocedural beta blocker had a significantly lower mortality than those who had not (1.5 versus 2.8 percent); the lower mortality was limited to patients who had not been receiving an oral beta blocker before admission. (See "Primary percutaneous coronary intervention in acute ST elevation myocardial infarction: Determinants of outcome".)

The optimal timing of beta blocker therapy was evaluated in a study of patients enrolled in the TIMI II trial [31]. A subset of 1390 patients who were eligible for intravenous beta blockade were randomly assigned to 15 mg of IV metoprolol tartrate (followed by oral metoprolol) or oral metoprolol begun on day six. There was no significant difference between the two groups in the in-hospital left ventricular ejection fraction or in mortality at 6 and 42 days. However, by day six, the early therapy group had significant reductions in nonfatal reinfarction (16 versus 31 patients) and recurrent ischemic episodes (107 versus 147 patients).

Patients who do not receive a beta blocker during the first 24 hours because of early contraindications should be re-evaluated for beta blocker candidacy for subsequent therapy.

Choice of drug and route of administration — For most patients, we prefer an oral rather than intravenous beta blocker. A cardioselective oral beta blocker, such as metoprolol or atenolol, is preferred in the setting of an acute myocardial infarction (MI). We use oral metoprolol tartrate 25 to 50 mg every 6 to 12 hours or atenolol 25 to 50 mg twice daily, initially, titrating upward as needed. Short-acting beta blockers are preferred early to allow for more rapid adjustment of dose based on the patient’s blood pressure and heart rate response. Near the time of discharge, we prefer to switch to longer-acting beta blockers.

For the uncommon patient in whom intravenous therapy may be chosen, such as those with ongoing ischemia prior to PCI in whom there is no hemodynamic instability including heart failure, we suggest the following regimens [16] (see 'Primary PCI' above):

Intravenous metoprolol tartrate can be given in 5 mg increments by slow intravenous administration (5 mg over one to two minutes), repeated every five minutes for a total initial dose of 15 mg (three doses). Patients who tolerate this regimen should then receive early oral therapy with either metoprolol succinate 25 to 50 mg daily (dependent on heart rate and blood pressure post-IV infusion) with careful up-titration to as much as 200 mg daily or metoprolol tartrate 25 to 50 mg 2 to 4 times daily beginning 15 to 30 min after the last intravenous dose. We attempt to achieve a discharge heart rate of approximately 70 beats per minute.

Intravenous atenolol can be given in a 5 mg dose followed by another 5 mg five minutes later. Patients who tolerate this regimen should then receive maintenance oral therapy with atenolol 50 mg every 12 hours daily beginning one to two hours after the last intravenous dose. Intravenous atenolol is not available in the United States or Canada.

Esmolol (50 mcg/kg per min increasing to a maximum of 200 to 300 mcg/kg per min), an ultra-short-acting beta blocker, can be given to assess tolerance to beta blockade in patients with borderline or questionable left ventricular function.

Bradycardia and hypotension are the most common limitations to achieving the full dose. In this setting, the rate of administration should be slowed or oral therapy initiated. However, a rigid "cookbook" regimen should not be used since there is a variable sympathetic response to acute MI.

Patients with hypertension — Some experts have recommended the use of intravenous beta blocker therapy for patients with acute MI and hypertension. Due to our concerns regarding this application of intravenous beta blockade, we prefer intravenous nitroglycerin, starting at a low dose, for such patients. (See 'Primary PCI' above.)

Special situations (eg, malignant hypertension or dissecting aortic aneurysm) may require the judicious use of intravenous beta blockade in such patients.

LONG-TERM THERAPY — While all patients with acute myocardial infarction (MI) should receive long-term beta blocker therapy, the optimal duration, dose, and agent are not known. We recommend the use of a long-acting, once daily beta blocker to improve treatment adherence.

Duration — The evidence summarized below supports the use of beta blockers for as long as three years after acute MI. The evidence supporting a longer duration or indefinite therapy is limited. Multiple randomized trials evaluating the optimal duration of beta blocker after acute MI are ongoing.

It should be kept in mind that many patients with prior MI have a clear indication for continued beta blocker therapy such as heart failure with reduced ejection fraction, asymptomatic left ventricular systolic dysfunction, hypertension, or angina. (See "Choice of drug therapy in primary (essential) hypertension" and "Beta blockers in the management of chronic coronary syndrome", section on 'Summary and recommendations' and "Primary pharmacologic therapy for heart failure with reduced ejection fraction".)

We suggest continuing beta blocker indefinitely in patients who had high-risk features at presentation such as cardiogenic shock, heart failure, or chronic kidney disease. For patients without these high-risk features, we suggest that practitioners discuss the potential benefits and risks of continued therapy beyond three years with patients and have them participate in decision making. (See 'Contraindications' below.)

In addition, it is reasonable to discontinue beta blocker therapy, using a tapering protocol carried out over a few weeks, in patients with unacceptable side effects, for whom the financial burden is unacceptable, or in those for whom the use of multiple medications is problematic (polypharmacy). There are no known life-threatening side effects (such as proarrhythmia or malignancy) of long-term beta blocker therapy [39].

Older studies with follow-up of as long as four years suggested a mortality benefit from long-term beta blocker therapy but also raised the possibility that the benefits declined with time [21,23,25,40].

A 2020 observational study confirmed benefit for up to three years but not longer [41]. In this study, nearly 29,000 patients who underwent coronary revascularization for acute MI, who were discharged on beta blocker, and who were event free from death, recurrent MI, or heart failure for one year were followed long term. Mortality was compared in patients who continued beta blocker for one year or longer with those who received therapy for less than one year. All-cause death was lower in patients who continued with beta blocker for one year or longer (adjusted hazard ratio [HR] 0.81, 95% CI 0.72-0.91). The lower risk of all-cause death in those treated for longer than one year lost statistical significance beyond three years after MI (adjusted HR 0.87, 95% CI 0.73-1.03).

Dose — The optimal long-term dose for each beta blocker is not known. Our approach is to attempt to prescribe doses used in randomized trials.

While it makes sense to recommend the doses used in the randomized trials, that approach has not been adopted widely by the medical community. The practice of using a lower dose than used in the trials is supported by a 2015 multicenter registry (OBTAIN) study that evaluated the relationship between dose and survival in 6682 acute MI survivors who were discharged on a beta blocker [42]. Discharge beta blocker dose was compared with target doses used in the randomized trials, grouped as >0 to 12.5 percent, 12.5 to 25 percent, >25 to 50 percent, and >50 percent of target dose. The following was noted after 2.1 years median follow-up:

91.5 percent of all patients were discharged on a beta blocker, with a mean dose 38.1 percent of the target dose. Metoprolol and carvedilol accounted for 92 percent of the drugs used, with target doses of 200 mg/day and 50 mg per day, respectively.

Lower mortality was observed with all doses compared with non-beta blocker therapy.

The primary end point of the study was mortality at two years. Compared with the >50 percent dose, the adjusted hazard ratios for the mortality for the 0 to 12.5, 12.5 to 25, and 25 percent groups were 0.86, 0.80, and 0.96, respectively. None of these were statistically significant.

This study raises the possibility that doses lower than those used in clinical trials may lead to similar or better outcomes. However, the study has significant limitations and we recommend doses uses in randomized trials.

Agent — For long-term administration, we believe that the benefit from beta blockers is a class effect and that agents without intrinsic sympathomimetic activity (ISA) are preferred [43-45]. In a 1999 meta-analysis of randomized controlled trials, there was no significant difference in benefit with cardioselective compared to nonselective drugs, but there was an almost significant trend toward less benefit with drugs that have ISA [23].

We start with a beta-1-selective agent in most patients. Usual initial doses are metoprolol tartrate (immediate release preparation) 25 to 50 mg two to four times daily or metoprolol succinate (extended release preparation) 25 to 200 mg daily or atenolol 50 to 100 mg divided twice daily. When possible, we use the longer-acting preparation. For patients in whom there is a concern for intolerance to these doses, lower initial doses should be used; in this setting, up-titration should occur as promptly as possible.

For patients who are initially treated with metoprolol tartrate, we give the drug twice daily long term or switch to metoprolol succinate.

Patients with heart failure with reduced ejection fraction (HFrEF) should be treated long-term with carvedilol, extended-release preparation metoprolol succinate, or bisoprolol. (See "Primary pharmacologic therapy for heart failure with reduced ejection fraction".)

Heart rate goal — We suggest reducing the heart rate to between 55 and 70 beats per minute while maintaining a systolic pressure above 90 mmHg, based on a small amount of published evidence and our clinical experience.

An inverse relationship between short-term and long-term mortality and the degree of heart rate reduction with beta blockade after MI was suggested in the 1980s [46]. This hypothesis was confirmed in a 2007 meta-regression analysis of 17 trials including nearly 31,000 MI patients who were placed on either a beta blocker (14 trials) or a rate slowing calcium channel blocker (three trials) [47]. There was a statistically significant and progressive reduction in the odds ratio for all-cause and cardiac death and non-fatal MI as the resting heart rate was slowed from 5 to 15 beats per minute compared to baseline. However, the optimal degree of heart rate lowering is unknown.

CONTRAINDICATIONS

Absolute contraindications — Potential absolute contraindications to the immediate use of beta blocker therapy include the following groups:

Hemodynamically compromised individuals, including those with hypotension with or without shock [43,44].

Patients with active bronchospasm, heart block greater than first degree (unless the patient has a permanent pacemaker), those with first degree heart block and a PR interval >0.30 seconds, or severe bradycardia. The rate at which a patient is labelled as having severe bradycardia is somewhat age dependent, as heart rate tends to decrease normally with age. While there is little evidence upon which to guide recommendations for the use of beta blockers in patients with bradycardia, we do not start such therapy in patients with a heart rate <40 beats per minute (bpm). Above 40 BPM, our threshold for starting beta blockers is lower in younger patients. Some of our experts use a higher threshold than 40 bpm.

Patients with overt heart failure, including pulmonary edema. However, there is a strong indication for carefully initiated oral beta blocker therapy (beginning at very low doses) in such patients whose heart failure has been brought under control prior to discharge. (See "Primary pharmacologic therapy for heart failure with reduced ejection fraction".)

The role of beta blocker therapy in patients with myocardial infarction (MI) precipitated by cocaine use is controversial. (See "Cocaine: Acute intoxication".)

Patients with comorbidities — A survival benefit from beta blockade is seen in patients with relative or potential contraindications to such therapy such as chronic obstructive pulmonary disease (COPD)/asthma, reduced left ventricular (LV) ejection fraction, treated heart failure, diabetes mellitus, and peripheral artery disease [21,48-55].

COPD/asthma — Beta blockers are safe and effective in MI patients with mild to moderate pulmonary disease, as illustrated by the following data from observational studies:

In a study of 54,962 MI patients over the age of 65 years who had no contraindications to beta blocker therapy, beta blockers were associated with a lower one-year mortality in patients with COPD or asthma who were not on beta agonist therapy (adjusted relative risk 0.85 compared to no beta blockers); the lower mortality was similar to that seen in patients without COPD or asthma [51]. In contrast, beta blockers had no difference in survival in patients who were using a beta agonist or who had severe COPD or asthma.

In a review of over 200,000 MI patients from the Cooperative Cardiovascular Project, survival among patients with COPD was significantly higher in those treated with a beta blocker (83 versus 72 percent without a beta blocker) at two years (figure 2) [21].

In a cohort study of 1063 patients with mild or moderate COPD who sustained a first MI, mortality was lower in those who started or were previously taking a beta blocker (adjusted hazard ratio 0.50, 95% CI 0.36-0.69 and 0.59, 95% CI 0.44-0.79, respectively) during nearly three years of follow-up [56].

Diabetes mellitus — Concern about the possibility of masking hypoglycemic symptoms or worsening glycemic control has made some physicians reluctant to prescribe beta blockers to patients with diabetes during an acute MI. However, these concerns have been overstated, and analysis of outcomes of diabetic subgroups in several postinfarction beta blocker trials documents an overall benefit from the use of beta blockers that is at least equivalent to and may be greater than that seen in patients without diabetes. (See "Acute myocardial infarction: Patients with diabetes mellitus", section on 'Beta blockers'.)

Peripheral artery disease — Although there has been a concern involving the use of beta blockers in patients with intermittent claudication, there appears to be no adverse effect of beta-1 selective blockers on claudication symptoms [57-59]. The use of beta blockers in patients with peripheral artery disease, including abdominal aortic aneurysm, is discussed elsewhere. (See "Management of claudication due to peripheral artery disease".)

Patients treated with antiarrhythmic drugs — Beta blockers are effective in patients treated with antiarrhythmic drugs (figure 3 and figure 4) [34,39,60]. This was illustrated in a pooled analysis of patients in the CAMIAT and EMIAT trials of amiodarone in acute MI [60]. The relative risks for all-cause mortality, cardiac death, arrhythmic deaths, and resuscitated cardiac arrest were lower for patients receiving beta blockers along with amiodarone than for those without a beta blocker, with or without amiodarone. This interaction was significant for cardiac death and for arrhythmic death or resuscitated cardiac arrest (relative risk 0.59 and 0.39) (figure 5A-B) [60]. Antiarrhythmic drugs other than amiodarone have not been similarly studied. (See "Ventricular arrhythmias during acute myocardial infarction: Prevention and treatment".)

In patients treated with beta blocker therapy in whom amiodarone is initiated, there is an increased risk of bradycardia. We advise close monitoring of the heart rate in these patients. For patients with HR <70 beats per minute, we recommend lowering the beta blocker dose at the time of initiation of amiodarone. (See "Amiodarone: Adverse effects, potential toxicities, and approach to monitoring", section on 'Adverse cardiac effects' and "Amiodarone: Clinical uses", section on 'Dose adjustment'.)

RECOMMENDATIONS OF OTHERS — Our approach to the use of beta blockers in patients with acute myocardial infarction (MI) is similar to that made in the 2015 European Society of Cardiology guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation [61].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Non-ST-elevation acute coronary syndromes (non-ST-elevation myocardial infarction)" and "Society guideline links: ST-elevation myocardial infarction (STEMI)".)

SUMMARY AND RECOMMENDATIONS

Beta blockers should be given to all myocardial infarction (MI) patients with heart failure (HF) and a left ventricular ejection fraction ≤40 percent. However, in patients with active HF, initiation should be delayed until the HF is under control. (See 'Left ventricular dysfunction/heart failure' above.)

For all patients who have sustained an acute MI without HF and preserved left ventricular systolic function and who have been treated with standard-of-care risk factor interventions, as well as dual antiplatelet therapy, we suggest treatment with an oral beta blocker (Grade 2C). (See 'Indications' above.)

For all patients with acute MI, we recommend initiation of oral beta blockers within the first 24 hours, as long as no contraindications are present (Grade 1B). (See 'Indications' above and 'Absolute contraindications' above.)

Patients who do not receive a beta blocker during the first 24 hours because of early contraindications should be reevaluated for beta blocker candidacy prior to discharge. If on reevaluation the preexisting contraindication is no longer present, a beta blocker should be administered. (See 'Primary PCI' above.)

The optimal duration of beta blocker therapy is not known. We treat most patients for a minimum of three years. In high risk patients, such as those who present with cardiogenic shock, heart failure, or chronic kidney disease, we treat for longer than three years. (See 'Long-term therapy' above.)

We attempt a target heart rate of between 55 and 70 beats per minute while maintaining a systolic pressure above 90 mmHg. (See 'Heart rate goal' above.)

Absolute contraindications to the initiation of beta blocker therapy include cardiogenic shock, active bronchospasm, severe bradycardia or heart block greater than first degree (unless the patient has a permanent pacemaker), and patients with overt heart failure. Patients with treated heart failure or bronchospastic lung disease, peripheral artery disease, or diabetes mellitus should receive beta blocker therapy; however, these individuals should be monitored for the development of adverse side effects. (See 'Contraindications' above.)

  1. Anderson JL, Morrow DA. Acute Myocardial Infarction. N Engl J Med 2017; 376:2053.
  2. López-Sendón J, Swedberg K, McMurray J, et al. Expert consensus document on beta-adrenergic receptor blockers. Eur Heart J 2004; 25:1341.
  3. Rydén L, Ariniego R, Arnman K, et al. A double-blind trial of metoprolol in acute myocardial infarction. Effects on ventricular tachyarrhythmias. N Engl J Med 1983; 308:614.
  4. Nuttall SL, Toescu V, Kendall MJ. beta Blockade after myocardial infarction. Beta blockers have key role in reducing morbidity and mortality after infarction. BMJ 2000; 320:581.
  5. Friedman LM, Byington RP, Capone RJ, et al. Effect of propranolol in patients with myocardial infarction and ventricular arrhythmia. J Am Coll Cardiol 1986; 7:1.
  6. Hu K, Gaudron P, Ertl G. Long-term effects of beta-adrenergic blocking agent treatment on hemodynamic function and left ventricular remodeling in rats with experimental myocardial infarction: importance of timing of treatment and infarct size. J Am Coll Cardiol 1998; 31:692.
  7. Galcerá-Tomás J, Castillo-Soria FJ, Villegas-García MM, et al. Effects of early use of atenolol or captopril on infarct size and ventricular volume: A double-blind comparison in patients with anterior acute myocardial infarction. Circulation 2001; 103:813.
  8. Doughty RN, Whalley GA, Walsh HA, et al. Effects of carvedilol on left ventricular remodeling after acute myocardial infarction: the CAPRICORN Echo Substudy. Circulation 2004; 109:201.
  9. Poulsen SH, Jensen SE, Egstrup K. Effects of long-term adrenergic beta-blockade on left ventricular diastolic filling in patients with acute myocardial infarction. Am Heart J 1999; 138:710.
  10. Sipahi I, Tuzcu EM, Wolski KE, et al. Beta-blockers and progression of coronary atherosclerosis: pooled analysis of 4 intravascular ultrasonography trials. Ann Intern Med 2007; 147:10.
  11. Mak IT, Weglicki WB. Protection by beta-blocking agents against free radical-mediated sarcolemmal lipid peroxidation. Circ Res 1988; 63:262.
  12. Norwegian Multicenter Study Group. Timolol-induced reduction in mortality and reinfarction in patients surviving acute myocardial infarction. N Engl J Med 1981; 304:801.
  13. Hjalmarson A, Herlitz J, Holmberg S, et al. The Göteborg metoprolol trial. Effects on mortality and morbidity in acute myocardial infarction. Circulation 1983; 67:I26.
  14. Chadda K, Goldstein S, Byington R, Curb JD. Effect of propranolol after acute myocardial infarction in patients with congestive heart failure. Circulation 1986; 73:503.
  15. Metoprolol in acute myocardial infarction (MIAMI). A randomised placebo-controlled international trial. The MIAMI Trial Research Group. Eur Heart J 1985; 6:199.
  16. Randomised trial of intravenous atenolol among 16 027 cases of suspected acute myocardial infarction: ISIS-1. First International Study of Infarct Survival Collaborative Group. Lancet 1986; 2:57.
  17. Yusuf S, Peto R, Lewis J, et al. Beta blockade during and after myocardial infarction: an overview of the randomized trials. Prog Cardiovasc Dis 1985; 27:335.
  18. Green BK, Gordon GD, Horak AR, et al. Safety of combined intravenous beta-adrenergic blockade (atenolol or metoprolol) and thrombolytic therapy in acute myocardial infarction. Am J Cardiol 1992; 69:1389.
  19. Risenfors M, Herlitz J, Berg CH, et al. Early treatment with thrombolysis and beta-blockade in suspected acute myocardial infarction: results from the TEAHAT Study. J Intern Med Suppl 1991; 734:35.
  20. Boissel JP, Leizorovicz A, Picolet H, Peyrieux JC. Secondary prevention after high-risk acute myocardial infarction with low-dose acebutolol. Am J Cardiol 1990; 66:251.
  21. Gottlieb SS, McCarter RJ, Vogel RA. Effect of beta-blockade on mortality among high-risk and low-risk patients after myocardial infarction. N Engl J Med 1998; 339:489.
  22. Van de Werf F, Janssens L, Brzostek T, et al. Short-term effects of early intravenous treatment with a beta-adrenergic blocking agent or a specific bradycardiac agent in patients with acute myocardial infarction receiving thrombolytic therapy. J Am Coll Cardiol 1993; 22:407.
  23. Freemantle N, Cleland J, Young P, et al. beta Blockade after myocardial infarction: systematic review and meta regression analysis. BMJ 1999; 318:1730.
  24. Kernis SJ, Harjai KJ, Stone GW, et al. Does beta-blocker therapy improve clinical outcomes of acute myocardial infarction after successful primary angioplasty? J Am Coll Cardiol 2004; 43:1773.
  25. Nakatani D, Sakata Y, Suna S, et al. Impact of beta blockade therapy on long-term mortality after ST-segment elevation acute myocardial infarction in the percutaneous coronary intervention era. Am J Cardiol 2013; 111:457.
  26. Ozasa N, Kimura T, Morimoto T, et al. Lack of effect of oral beta-blocker therapy at discharge on long-term clinical outcomes of ST-segment elevation acute myocardial infarction after primary percutaneous coronary intervention. Am J Cardiol 2010; 106:1225.
  27. Yang JH, Hahn JY, Song YB, et al. Association of beta-blocker therapy at discharge with clinical outcomes in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. JACC Cardiovasc Interv 2014; 7:592.
  28. Huang BT, Huang FY, Zuo ZL, et al. Meta-Analysis of Relation Between Oral β-Blocker Therapy and Outcomes in Patients With Acute Myocardial Infarction Who Underwent Percutaneous Coronary Intervention. Am J Cardiol 2015; 115:1529.
  29. Dondo TB, Hall M, West RM, et al. β-Blockers and Mortality After Acute Myocardial Infarction in Patients Without Heart Failure or Ventricular Dysfunction. J Am Coll Cardiol 2017; 69:2710.
  30. Roberts R, Rogers WJ, Mueller HS, et al. Immediate versus deferred beta-blockade following thrombolytic therapy in patients with acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) II-B Study. Circulation 1991; 83:422.
  31. TIMI Study Group. Comparison of invasive and conservative strategies after treatment with intravenous tissue plasminogen activator in acute myocardial infarction. Results of the thrombolysis in myocardial infarction (TIMI) phase II trial. N Engl J Med 1989; 320:618.
  32. Chen ZM, Pan HC, Chen YP, et al. Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366:1622.
  33. Barron HV, Rundle AC, Gore JM, et al. Intracranial hemorrhage rates and effect of immediate beta-blocker use in patients with acute myocardial infarction treated with tissue plasminogen activator. Participants in the National Registry of Myocardial Infarction-2. Am J Cardiol 2000; 85:294.
  34. Teo KK, Yusuf S, Furberg CD. Effects of prophylactic antiarrhythmic drug therapy in acute myocardial infarction. An overview of results from randomized controlled trials. JAMA 1993; 270:1589.
  35. Ibanez B, Macaya C, Sánchez-Brunete V, et al. Effect of early metoprolol on infarct size in ST-segment-elevation myocardial infarction patients undergoing primary percutaneous coronary intervention: the Effect of Metoprolol in Cardioprotection During an Acute Myocardial Infarction (METOCARD-CNIC) trial. Circulation 2013; 128:1495.
  36. Pizarro G, Fernández-Friera L, Fuster V, et al. Long-term benefit of early pre-reperfusion metoprolol administration in patients with acute myocardial infarction: results from the METOCARD-CNIC trial (Effect of Metoprolol in Cardioprotection During an Acute Myocardial Infarction). J Am Coll Cardiol 2014; 63:2356.
  37. Roolvink V, Ibáñez B, Ottervanger JP, et al. Early Intravenous Beta-Blockers in Patients With ST-Segment Elevation Myocardial Infarction Before Primary Percutaneous Coronary Intervention. J Am Coll Cardiol 2016; 67:2705.
  38. Halkin A, Grines CL, Cox DA, et al. Impact of intravenous beta-blockade before primary angioplasty on survival in patients undergoing mechanical reperfusion therapy for acute myocardial infarction. J Am Coll Cardiol 2004; 43:1780.
  39. Kennedy HL, Brooks MM, Barker AH, et al. Beta-blocker therapy in the Cardiac Arrhythmia Suppression Trial. CAST Investigators. Am J Cardiol 1994; 74:674.
  40. Bangalore S, Steg G, Deedwania P, et al. β-Blocker use and clinical outcomes in stable outpatients with and without coronary artery disease. JAMA 2012; 308:1340.
  41. Kim J, Kang D, Park H, et al. Long-term β-blocker therapy and clinical outcomes after acute myocardial infarction in patients without heart failure: nationwide cohort study. Eur Heart J 2020; 41:3521.
  42. Goldberger JJ, Bonow RO, Cuffe M, et al. Effect of Beta-Blocker Dose on Survival After Acute Myocardial Infarction. J Am Coll Cardiol 2015; 66:1431.
  43. Antman EM, Anbe DT, Armstrong PW, et al. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction. www.acc.org/qualityandscience/clinical/statements.htm (Accessed on August 24, 2006).
  44. www.acc.org/qualityandscience/clinical/statements.htm (Accessed on September 18, 2007).
  45. Shivkumar K, Schultz L, Goldstein S, Gheorghiade M. Effects of propanolol in patients entered in the Beta-Blocker Heart Attack Trial with their first myocardial infarction and persistent electrocardiographic ST-segment depression. Am Heart J 1998; 135:261.
  46. Kjekshus JK. Importance of heart rate in determining beta-blocker efficacy in acute and long-term acute myocardial infarction intervention trials. Am J Cardiol 1986; 57:43F.
  47. Cucherat M. Quantitative relationship between resting heart rate reduction and magnitude of clinical benefits in post-myocardial infarction: a meta-regression of randomized clinical trials. Eur Heart J 2007; 28:3012.
  48. Spargias KS, Hall AS, Greenwood DC, Ball SG. beta blocker treatment and other prognostic variables in patients with clinical evidence of heart failure after acute myocardial infarction: evidence from the AIRE study. Heart 1999; 81:25.
  49. Dargie HJ. Effect of carvedilol on outcome after myocardial infarction in patients with left-ventricular dysfunction: the CAPRICORN randomised trial. Lancet 2001; 357:1385.
  50. Lichstein E, Hager WD, Gregory JJ, et al. Relation between beta-adrenergic blocker use, various correlates of left ventricular function and the chance of developing congestive heart failure. The Multicenter Diltiazem Post-Infarction Research Group. J Am Coll Cardiol 1990; 16:1327.
  51. Chen J, Radford MJ, Wang Y, et al. Effectiveness of beta-blocker therapy after acute myocardial infarction in elderly patients with chronic obstructive pulmonary disease or asthma. J Am Coll Cardiol 2001; 37:1950.
  52. Vantrimpont P, Rouleau JL, Wun CC, et al. Additive beneficial effects of beta-blockers to angiotensin-converting enzyme inhibitors in the Survival and Ventricular Enlargement (SAVE) Study. SAVE Investigators. J Am Coll Cardiol 1997; 29:229.
  53. Exner DV, Dries DL, Waclawiw MA, et al. Beta-adrenergic blocking agent use and mortality in patients with asymptomatic and symptomatic left ventricular systolic dysfunction: a post hoc analysis of the Studies of Left Ventricular Dysfunction. J Am Coll Cardiol 1999; 33:916.
  54. Gundersen T, Kjekshus J. Timolol treatment after myocardial infarction in diabetic patients. Diabetes Care 1983; 6:285.
  55. Malmberg K, Herlitz J, Hjalmarson A, Rydén L. Effects of metoprolol on mortality and late infarction in diabetics with suspected acute myocardial infarction. Retrospective data from two large studies. Eur Heart J 1989; 10:423.
  56. Quint JK, Herrett E, Bhaskaran K, et al. Effect of β blockers on mortality after myocardial infarction in adults with COPD: population based cohort study of UK electronic healthcare records. BMJ 2013; 347:f6650.
  57. Califf RM, Lokhnygina Y, Velazquez EJ, et al. Usefulness of beta blockers in high-risk patients after myocardial infarction in conjunction with captopril and/or valsartan (from the VALsartan In Acute Myocardial Infarction [VALIANT] trial). Am J Cardiol 2009; 104:151.
  58. Radack K, Deck C. Beta-adrenergic blocker therapy does not worsen intermittent claudication in subjects with peripheral arterial disease. A meta-analysis of randomized controlled trials. Arch Intern Med 1991; 151:1769.
  59. Solomon SA, Ramsay LE, Yeo WW, et al. beta blockade and intermittent claudication: placebo controlled trial of atenolol and nifedipine and their combination. BMJ 1991; 303:1100.
  60. Boutitie F, Boissel JP, Connolly SJ, et al. Amiodarone interaction with beta-blockers: analysis of the merged EMIAT (European Myocardial Infarct Amiodarone Trial) and CAMIAT (Canadian Amiodarone Myocardial Infarction Trial) databases. The EMIAT and CAMIAT Investigators. Circulation 1999; 99:2268.
  61. Roffi M, Patrono C, Collet JP, et al. 2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task Force for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation of the European Society of Cardiology (ESC). Eur Heart J 2016; 37:267.
Topic 96 Version 50.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟