Вы находитесь на странице: 1из 10

Feature Curbside Consult

Through the Decades: β-Blocker Use and Outcomes


in Acute Coronary Syndromes
Alina Kukin, Zachary R. Noel, PharmD, BCPS, Kristin Watson, PharmD, BCPS

Abstract: Beta-adrenergic receptor antagonists, or β-blockers, have been a β-BLOCKER USE IN THE PRETHROMBOLYTIC ERA
cornerstone of treatment in patients with acute coronary syndromes (ACS) 1960s: Trials of Propranolol in MI
for more than 4 decades. First studied in the 1960s, β-blockers in ACS
The first prospective, randomized trial to investigate the
have been shown to decrease the risk of death, recurrent ischemic events,
effect of β-blockade on clinical outcomes in acute myocardial
and arrhythmias by reducing catecholamine-mediated effects and reducing
infarction (AMI) was conducted in 1965.9 A total of 91 patients
myocardial oxygen demand. Through the decades, the β-blocker of choice,
who experienced symptoms suggestive of AMI within 24 hours
timing of initiation, duration of therapy, and dosing have evolved consider-
of presentation to the hospital were enrolled to receive proprano-
ably. Despite having clear benefits in certain patient populations (eg, patients
lol or no propranolol. Diagnosis was confirmed by the presence
with systolic dysfunction who are hemodynamically stable), the benefit of
of electrocardiogram (ECG) changes (ie, Q-waves or ST-segment
β-blockers in other populations (ie, in patients at low risk for complications
changes) and changes in biomarkers (eg, elevations in lactate
receiving modern revascularization therapies and optimal medical manage-
dehydrogenase or aspartate transaminase). Patients were random-
ment) remains unclear. This article provides a review of the landmark clinical
ized to treatment with oral propranolol 20 mg every 8 hours or no
trials of β-blockers in ACS and highlights the chronology and evolution of
treatment for 28 days. No exclusion criteria were noted in the pub-
guideline recommendations through the decades.
lished trial. The mean age of patients was 59 years, and 75% were
Key Words: acute coronary syndrome, beta-adrenergic receptor antagonists, male. Anticoagulation was administered to all patients unless con-
myocardial infarction traindicated, and supportive treatments such as digitalis, diuret-
ics, antihypertensives, and hydrocortisone were used as medically
(Cardiology in Review 2018;26: 157–166)
necessary. The rates of supportive treatment were not recorded, nor
was the median dose of propranolol. A 54% relative risk reduc-
tion (RRR) in mortality was observed in those who received pro-
pranolol (35% vs 16%; no P value provided). Fewer cardiac arrests

T he management of patients presenting with acute coronary syn-


dromes (ACS) has evolved dramatically over the last several
decades with the advent of coronary intervention procedures and
and episodes of cardiac pain were recorded in those receiving
propranolol; however, the sample size was too small to accurately
determine statistical significance. Adverse effects were insuffi-
the development of improved medical therapies, including anti- ciently tracked and disclosed, but the absolute number of patients
coagulants, 3-hydroxy-3-methyl-glutaryl-co-enzyme A reductase exhibiting symptoms of heart failure (HF) were similar between
inhibitors and more potent antiplatelet therapies.1–7 One treatment the propranolol and no treatment arms (2 patients vs 4 patients). A
option that has remained constant since the publication of the first subsequent publication would raise concerns for incomplete data
American College of Cardiology/American Heart Association Acute and poor study design, thus bringing into question the validity and
Myocardial Infarction guideline in 1990 is the use of β-adrenergic reliability of the results.10
receptor antagonists, or β-blockers, for select patients.1 β-Blockers, In light of the concerns with the data reported by Snow,9 mul-
which have been studied for the management of myocardial infarc- tiple prospective, randomized controlled trials ensued in an attempt
tion (MI) since the 1960s, reduce myocardial oxygen demand by to reproduce the observed effects. Balcon et al11 conducted a pro-
attenuating catecholamine stimulation of cardiac myocytes, thereby spective, single-center, randomized, placebo-controlled, double-
improving coronary perfusion, minimizing infarct size, and reduc- blind trial that evaluated the role of oral propranolol after an AMI.
ing arrhythmias.8 Through the decades, the β-blocker of choice, the A total of 114 patients who experienced symptoms within 24 hours
timing of administration, duration of therapy, dosing, and patient of presentation had ECG changes consistent with AMI and elevated
populations in which to initiate β-blocker therapy have evolved con- biomarkers were randomized to propranolol 20 mg every 6 hours or
siderably. This article will review landmark clinical trials regarding placebo. Treatment was administered as soon as possible follow-
the use of β-blocker therapy for the treatment of ACS and highlight ing admission and continued for 28 days. Patients were excluded
how therapy has changed over time (Fig. 1). This article focuses if they were unconscious or presented with complete heart block.
on the use of β-blockers within the first year after an ACS. A sum- The mean age of the patients was 60 years, and approximately two-
mary of the treatment strategies utilized in the trials discussed in this thirds were male. The mean time from onset of pain to first treatment
article is presented in Table 1. dose was 9.19 ± 0.85 hours and 9.95 ± 0.77 hours in the treatment and
placebo arms, respectively. Bed rest was mandated for 3 weeks in
all patients. Other cardiac medications were discouraged during the
trial period, but anticoagulation, diuretics, digitalis, and vasopressors
From the Department of Pharmacy Practice and Science, University of Maryland could be given as necessary. The rates of these supportive treatments
School of Pharmacy, Baltimore, MD.
Disclosure: The authors declare no conflicts of interest. were not provided. No difference was observed in 28-day mortality
Correspondence: Zachary R. Noel, PharmD, BCPS, Department of Pharmacy between those treated with propranolol or placebo (23.2% vs 24.1%;
Practice and Science, University of Maryland School of Pharmacy, 20 North no P value provided). The rates of HF and arrhythmias were similar
Pine Street, S427 Baltimore, MD. E-mail: znoel@rx.umaryland.edu.
Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.
between groups, but a significant increase in the rate of symptom-
ISSN: 1061-5377/18/2603-0157 atic hypotension with bradycardia was observed in those treated with
DOI: 10.1097/CRD.0000000000000197 propranolol (30% vs 14%; P < 0.05).

Cardiology in Review  •  Volume 26, Number 3, May/June 2018 www.cardiologyinreview.com  |  157

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Kukin et al Cardiology in Review  •  Volume 26, Number 3, May/June 2018

FIGURE 1.  Evolution of guideline recommendations for β-blocker use in acute coronary syndromes. AMI, acute myocardial
infarction; CI, contraindication; HF, heart failure; IV, intravenous; LV, left ventricular; MI, myocardial infarction; po, oral; UA,
unstable angina.

Similar to the trial by Balcon et al11, Clausen et al12 conducted a provided). Routine ECG monitoring was not performed, and thus no
prospective, single-center, randomized trial that examined the effects conclusions could be made regarding the incidence of arrhythmias.
of oral propranolol in the setting of AMI. A total of 110 patients who An additional, larger trial was conducted in 1968 to determine
experienced symptoms of AMI within 24 hours of presentation were the effect of propranolol on mortality and morbidity in the setting
randomized to propranolol 10 mg 4 times daily or no treatment for 14 of AMI.14 This prospective, multicenter, randomized, placebo-con-
days. Patients were observed for 22 days following randomization. trolled, double-blind trial enrolled patients presumed to have AMI
The only exclusion criterion was asthma. Routine care consisting of within the previous 3 days. Patients were randomized to treatment
digitalis, diuretics, and anticoagulation was administered upon phy- with 20 mg oral propranolol 4 times daily or placebo for 3 weeks.
sician discretion, but rates of use were not provided. At the conclu- Key exclusion criteria included shock, HF, heart block, and sinus
sion of the trial, no significant difference in mortality was observed bradycardia. A total of 454 patients were deemed to have certain or
in those who received propranolol versus those who did not (30% vs probable AMI, as defined by the presence of ECG changes, eleva-
33%; no P value provided), irrespective of age, gender, or severity tions in biomarkers, and clinical presentation, and were included in
of the current condition. No differences in the incidence and type the analysis. Of the patients with confirmed AMI, those who received
of arrhythmias or in the incidence of HF was noted between the 2 propranolol had similar mortality as those who received placebo
study arms. There was an increase in the absolute number of patients (14% vs 11%; no P value provided). The rates of HF, hypotension,
experiencing shock, defined as a systolic blood pressure < 90 mm Hg, and chest pain were also similar between groups; however, a signifi-
in those who received propranolol; however, this was not statistically cant reduction in arrhythmias due to ectopic beats was observed in
significant (47% vs 26%; 0.10 > P > 0.05). those receiving propranolol (12% vs 22%; P < 0.01). No significant
The Propranolol in Acute Myocardial Infarction trial was a difference in mortality was noted between the treatment and placebo
prospective, multicenter, randomized, placebo-controlled, double- groups based on age, previous ischemic history, status on admission,
blind trial that investigated the role of propranolol for the prevention administration of anticoagulants, or time of mobilization. Although
of arrhythmias and reduction in mortality following AMI.13 A total it was stated that patients were included if suspected of having AMI
of 226 patients with diagnostic ECG changes for MI who presented within 3 days, no further data were reported on the time frame of
within 24–48 hours of symptoms were randomized to oral propranolol symptom onset to propranolol administration.
20 mg every 6 hours or placebo for 28 days. Exclusion criteria included
bronchospasm, asthma, bradycardia, and hypotension. The average Summary of the 1960s
age was 58 years, 80% were men, and the overall acuity of patients The data published by Snow9 in 1965 were among the first to
was low as determined by the Peel’s Coronary Prognostic Index. The show a mortality reduction in AMI. The primitive treatment strategies
average time from onset of symptoms to treatment initiation was 16.2 for AMI at that time—which mainly consisted of anticoagulation and
hours in the propranolol group and 13.4 hours in the placebo group. bed rest for up to 3 weeks to reduce myocardial oxygen demand—
The use of anticoagulants was given based on physician’s practice. had proven to yield little benefit on outcomes. Although the results
When indicated, digitalis, diuretics, pain relievers, and antiarrhyth- were promising, they were met with great skepticism as flaws in
mic drugs such as quinidine and procainamide were administered, the study design were identified. Of the remaining trials conducted in
rates of which were not disclosed. Overall mortality was nonstatisti- the 1960s, none were able to reproduce the beneficial effects on mor-
cally significantly higher in those treated with propranolol compared tality. Three of the 4 trials in this era also tested the hypothesis that
with placebo (15% vs 12.6%; no P value provided). Patients receiv- propranolol would prevent arrhythmias. One trial noted more sinus
ing propranolol experienced more cardiac failure (31% vs 14.7%; no bradycardia in the propranolol group, another trial observed a reduc-
P value provided) and more hypotension (19% vs 10.5%; no P value tion in arrhythmias due to ectopic beats, and the third trial detected

158  |  www.cardiologyinreview.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


TABLE 1.  Key Trials of β-Blockers
Duration of
Study Agent/Initial Dose Timing of Initial Dose Maximal Dose Treatment Key Findings
Snow9 Propranolol PO 20 mg Within 24 hr of symptom onset Propranolol 20 mg 28 d Propranolol use associated with a reduced
every 8 hr PO every 8 hr risk of death; concerns with validity of
results.
Balcon et al11 Propranolol PO 20 mg Within 24 hr of symptom onset; Propranolol 20 mg 28 d No difference in the rate of death,
every 6 hr mean time from onset of pain PO every 6 hr arrhythmia, or HF.
to first treatment dose was
9.19 ± 0.85 hr in treatment group
and 9.95 ± 0.77 hr in placebo group
Clausen et al12 Propranolol PO 10 mg 4 times Within 24 hr of symptom onset Propranolol 10 mg 14 d No difference in the rate of death,
daily PO 4 times daily arrhythmia, or HF.
Propranolol in Acute Myocardial Propranolol 20 mg PO Within 24–48 hr of symptom onset; Propranolol 20 mg 28 d No difference in the rate of death.
Infarction: A Multicentre Trial every 6 hr average of 16.2 hr from symptom PO every 6 hr Cardiac failure higher in the propranolol
196613 onset in the propranolol group group.
and 13.4 hr in the placebo group
Norris et al14 Propranolol 20 mg PO Within 3 d of symptom onset Propranolol 20 mg 3 wk No difference in the rate of death or HF.
4 times daily PO 4 times daily Risk of arrhythmia lower with propranolol.
The Norwegian Multicenter Study Timolol 5 mg PO twice daily 7–28 d after MI; average of Timolol 10 mg PO 17 mo (median) Timolol use associated with lower risk of

© 2018 Wolters Kluwer Health, Inc. All rights reserved.


of Timolol after Myocardial 11.5 d from presentation twice daily all-cause death and reinfarction; reduction
Infarction17,18 persisted at 6 yr.
The Göteborg Metoprolol Trial19 Metoprolol 5 mg (up to 15 mg) Average of 11.3 hr from Metoprolol tartrate 90 d Metoprolol use associated with a lower risk
IV followed by metoprolol symptom onset PO 100 mg twice of death.
tartrate 50 mg PO every 6 hr daily
for 48 hr
Cardiology in Review  •  Volume 26, Number 3, May/June 2018

β-blocker Heart Attack Trial20 Propranolol 20 mg PO 3 times 5–21 d after myocardial infarction; Propranolol PO 60 25 mo (average) Propranolol use associated with lower risk
daily average of 14 d from presentation to 80 mg 3 times of all-cause death.
to treatment daily
The Metoprolol in Acute Metoprolol 5 mg (up to 15 mg) Within 24 hr of symptom onset; Metoprolol tartrate 16 d Overall, no difference in the risk of death.
Myocardial Infarction trial21 IV followed by metoprolol average of 6.7 hr from PO 100 mg twice Metoprolol use associated with lower risk of
tartrate 50 mg PO every symptom onset daily death in “high-risk” patients.
6 hr for 48 hr Metoprolol use associated with lower risk of
supraventricular arrhythmias.
Long-Term Treatment With Metoprolol tartrate PO 100 mg 1–2 wk after MI Metoprolol tartrate 36 mo Overall, no difference in the rate of death.
Metoprolol After Myocardial twice daily PO 100 mg twice Metoprolol use associated with lower risk of
Infarction Trial22 daily cardiac death in those with a large infarct.
The first international study of Atenolol 5–10 mg Average of 5 hr from Atenolol 100 mg 7d Atenolol use associated with lower risk of
infarct survival23 intravenously, depending on symptom onset PO daily vascular death at 7 d; no difference at 1 yr.
heart rate Atenolol associated with lower risk of
nonfatal cardiac arrest and reinfarction.
The Thrombolysis in Myocardial “Immediate” group: metoprolol “Immediate” group: average of Metoprolol tartrate Not reported No difference in LVEF.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Infarction Phase II trial27 5 mg (up to 15 mg) IV 3.3 hr from symptom onset PO 100 mg twice At 6 wk, metoprolol use associated with
followed by metoprolol “Delayed” group: 6 d after MI daily lower risk of nonfatal/fatal reinfarction;
tartrate 50 mg PO every 12 hr difference did not persist at 1 yr.
for the first 24 hr Overall, no difference in mortality. Mortality
“Delayed” group: metoprolol benefit observed with metoprolol in the “low-
25 mg PO every 12 hr risk” group who received immediate therapy
Atenolol use in the Global Atenolol 5 mg (up to 10 mg) Within 2 d of MI; 92% of patients Atenolol 50–100 mg 30 d Atenolol use associated with lower rate of
Utilization of Streptokinase and IV followed by atenolol who received both IV + oral and PO daily death, HF and ventricular arrhythmias.
Tissue Plasminogen Activator--I 50–100 mg atenolol PO daily 68% of patients who received
Trial28 oral only received first dose
within 1 d
(Continued )
β-Blocker Use and Outcomes in ACS

www.cardiologyinreview.com | 159
Kukin et al Cardiology in Review  •  Volume 26, Number 3, May/June 2018

no difference in incidence or type of arrhythmias.11,12,14 Supportive

risk of cardiovascular shock, especially in


Carvedilol use associated with lower risk of
all-cause death, cardiovascular death and
treatment strategies during this time drastically differed from cur-

those at high risk for developing shock.


Metoprolol use associated with increased

Primary outcome did not include clinical


No difference in the composite of death,
reinfarction, or cardiac arrest; or all-
rent practice and consisted of anticoagulation, digitalis, and in some
No difference in the composite of all-

instances select class I antiarrhythmics. It is possible that the use of


cause death or hospitalization.

these antiarrhythmic drugs actually increased the rates of arrhyth-

Metoprolol use associated with


Key Findings

mias based on currently available literature.15 Unfortunately, the rates

No difference in infarct size.

HF, heart failure; IV, intravenous; LVEF, left ventricular ejection fraction; MI, myocardial infarction; PCI, percutaneous coronary intervention; PO = oral; STEMI, ST-segment elevation myocardial infarction.
of utilization of these medications were often unreported. Not to be

improvement in LVEF.
underappreciated are the limitations in diagnosing AMI during this
era. Cardiac-specific biomarkers, such as creatinine kinase myo-
globin and troponins I or T, were not available until the 1970s and
nonfatal MI.

cause death.

1980s, respectively; thus, less sensitive biomarkers such as lactate

outcomes.
dehydrogenase and aspartate aminotransferase were used to aid in
diagnosing AMI. The lack of specificity for AMI of these biomarkers
may have contributed to incorrect diagnoses of AMI.16 Study design
is also a significant limitation of data generated from this era. Pla-
cebo-controlled blinded studies were not routinely performed, thus
earlier; average
whichever was

introducing potential bias and confounders that, for many, brought


Duration of

1.3 yr (average)
Treatment

into question their validity. In summation, the evidence provided by


or hospital
discharge,

1d

1d

these trials was not sufficient to recommend β-blockade as a standard


Up to 28 d

of 15 d

therapy following AMI during this time.


The 1980s: Trials of Short and Long-Term β-blocker
Therapy and Impact on Mortality
200 mg PO daily
Maximal Dose

The Norwegian Multicenter Study of Timolol after Myocardial


PO twice daily
Carvedilol 25 mg

Infarction trial was a prospective, multicenter, randomized, placebo-


succinate

15 mg IV

10 mg IV

controlled, double-blind trial that examined the effect of timolol on


Metoprolol

Metoprolol

Metoprolol

mortality and reinfarction following MI.17 A total of 1884 clinically


stable post-MI patients were randomized to receive oral timolol 10 mg
or placebo twice daily. Patients who met inclusion criteria had 2 of
the following: (1) chest pain, cardiogenic shock, or acute pulmonary
edema; (2) characteristic ECG changes; or (3) 2 elevated aspartate
median of 10 min after STEMI
Within 4.5 hr of symptom onset;

aminotransferase levels. Key exclusion criteria included contraindi-


Within 12 hr of symptom onset
Timing of Initial Dose

cations for a β-blocker (eg, bradycardia, uncontrolled cardiac failure,


onset; average of 10.3 hr
Within 24 hr of symptom

severe lung disease), serious illness that would impact follow-up or


from symptom onset

affect the end-point (eg, alcoholism, malignancy), and an alterna-


tive indication for a β-blocker. The average age of enrollees was 61
3–21 d after MI

years, with 80% being male, 20% having a previous infarction, and
33% having HF. Although treatment with digitalis and diuretics was
diagnosis

permitted, the use of antiarrhythmic agents was prohibited. Random-


ization occurred at a median of 11.5 days following presentation, and
treatment duration was a median of 17 months. Patients were clas-
sified into 3 risk groups based on the presence of high risk factors,
including left ventricular (LV) failure, enlarged heart, atrial fibrilla-
Metoprolol 5 mg (up to 15 mg)

tartrate 50 mg PO every 6 hr


IV followed by metoprolol

tion or flutter, transient fall in systolic blood pressure to < 100 mm


Agent/Initial Dose

Hg, and serum aspartate aminotransferase levels exceeding 4 times


Carvedilol 6.25 mg PO

the upper limit of normal. Risk group I was identified as the highest
(up to 15 mg) IV

(up to 10 mg) IV

risk group, whereas risk group III was the lowest risk group. Overall,
for the first day

Metoprolol 5 mg

Metoprolol 5 mg

those treated with timolol had a 39.3% RRR in all-cause mortality


twice daily

(21.9 % vs 13.3%; P = 0.0003), a 28.4% RRR in reinfarction rates


(20.1% vs 14.4%; P = 0.0006), and 50% RRR in cardiac death (12%
vs 6%; P < 0.001). The impact on mortality appeared to persist for
at least 24 months, whereas the impact on reinfarction appeared to
diminish after 6 months. When analyzed by level of risk, the effects
Cardioprotection During an Acute
The Clopidogrel and Metoprolol in

on mortality were driven by a reduction in death in risk group II


with STEMI (EARLY-BAMI)35
Ventricular Dysfunction Trial30

before Primary PCI in Patients


Early β-Blocker Administration
Myocardial Infarction Trial31

(13.6% vs 7.1%; P < 0.01), which consisted of those who experi-


Myocardial Infarction trial32
TABLE 1.  (Continued )

enced their first MI and had a high risk condition (ie, LV failure,
The Carvedilol Post-Infarct
Survival Control in Left-

enlarged heart on chest x-ray, transient hypotension, or atrial fibril-


Effect of Metoprolol in

lation). No differences in mortality were noted among age groups.


Adverse effects that were more common in those treated with timolol
included bradycardia (5% vs 0%; P < 0.001), hypotension (3.1% vs
1.6%; P < 0.05), fatigue (4.8% vs 1.2%; P < 0.001), and cold extrem-
ities (7.7% vs 0.6%; P < 0.001). The most common reasons for study
Study

drug discontinuation were requirement for a β-blocker, need for an


antiarrhythmic, bradycardia, and hypotension.

160  |  www.cardiologyinreview.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Cardiology in Review  •  Volume 26, Number 3, May/June 2018 β-Blocker Use and Outcomes in ACS

A follow-up of the Norwegian Multicenter Study of Timo- metoprolol tartrate was administered every 6 hours for 48 hours fol-
lol after Myocardial Infarction trial found a persistent benefit of lowed by 100 mg twice daily thereafter until day 16. Exclusion crite-
β-blockade in patients who continued to take timolol for 6 years ria included current β-blocker or calcium channel blocker use, heart
after the index infarction.18 Of the surviving patients who completed rate < 60 beats per minute, systolic blood pressure < 105 mm Hg, and
the double-blind period and participated in the open-label follow-up LV failure. The average age was 60 years, and 78% of the patients
study, 87.2% of timolol patients continued on treatment, and 88.1% were male. Before treatment, glycosides and diuretics were used in
of the placebo patients continued without a β-blocker. At 6 years, a 3% and 11% of patients, respectively. The average time from onset of
significant reduction in mortality (26.4% vs 32.3%; P = 0.0028) was symptoms to randomization was 6.7 hours. A nonstatistically signifi-
sustained in the timolol group. cant 13% (95% CI, ˗8 to 33) decrease in mortality was observed in
The Göteborg Metoprolol Trial was a prospective, multicenter, those who received metoprolol (4.3% vs 4.9%; P = 0.29). Subgroup
randomized, placebo-controlled, double-blind trial that assessed the analyses showed a statistically significant 29% RRR in mortality
impact of early β-blockade with metoprolol on mortality.19 Unlike the in high-risk patients, defined by having 3 or more of the following
Norwegian Multicenter Study of Timolol after MI trial, β-blockade characteristics: age > 60 years, ECG changes consistent with MI,
was administered soon after presentation for an AMI. A total of 1395 history of MI, hypertension, angina pectoris, congestive HF, diabe-
patients were randomized to receive metoprolol 15 mg intravenously, tes, and treatment with diuretics or cardiac glycosides. A reduction
administered as 5 mg every 2 minutes for 3 doses, followed by 50 mg in supraventricular arrhythmias and chest pain, as demonstrated by
oral metoprolol tartrate every 6 hours for 48 hours, and then 100 mg a significantly lower calcium channel blocker and narcotic require-
metoprolol tartrate twice daily, or matching placebo for 90 days. ment, was observed in the metoprolol arm. The rates of hypotension
Major exclusion criteria included contraindication to β-blocker ther- and bradyarrhythmias were 9.8% and 4.8% for the placebo group and
apy (eg, cardiogenic shock) and the presence of an alternative indica- 18.4% and 12.9% for the treatment group, respectively (P < 0.001).
tion for receiving a β-blocker. Approximately two-thirds of included The long-term treatment with Metoprolol after Myocardial
patients were younger than 64 years, and 76% were male. At baseline Infarction Trial was a prospective, multicenter, randomized, placebo-
and prior to treatment, about 25% of patients in each treatment group controlled, double-blind study that aimed to determine the effect of
were already on a β-blocker. An average of 11.3 hours elapsed from metoprolol on long-term morbidity and mortality in patients for 36
onset of pain to treatment initiation. The rate of withdrawal from months following MI.22 The study consisted of 301 patients, all of
the study was equal between the 2 groups (19.1%); however, more whom were hospitalized within 48 hours of the onset of symptoms.
patients receiving metoprolol withdrew due to hypotension (4.2% vs Patients were randomized to a study treatment of oral metoprolol tar-
1.9%; P = 0.018) and bradycardia (2.6% vs 0.7%; P = 0.011). At the trate 100 mg twice daily or placebo, which was initiated 1–2 weeks
conclusion of the 90-day follow-up period, a significant 36% RRR after the acute infarction. Key exclusion criteria included HF, bundle
in mortality was observed in the metoprolol arm (8.9% vs 5.7%; P branch block, a need for β-blocker therapy, and atrial fibrillation. The
< 0.03). Subgroup analyses suggested a benefit regardless of patient average age was 60 years, and 80% of the patients were male. Digitalis
age, previous MI, or prior β-blockade use. and diuretics were prescribed in 24% and 45% of patients, respec-
The β-blocker Heart Attack Trial (BHAT) was a prospective, tively. Approximately 6% of patients in each group were on antiar-
multicenter, randomized, placebo-controlled, double-blind trial eval- rhythmic medications. The withdrawal rates were 18% for each arm;
uating the impact of propranolol on mortality following MI.20 Nearly however, more patients in the metoprolol arm required withdrawal
4000 patients were randomized between 1978 and 1980 to proprano- for HF. The dose was reduced to 50 mg twice daily in 25% of patients
lol 20 mg 3 times daily or placebo within 5–21 days of MI. Doses receiving metoprolol and in 10% of patients receiving placebo, pri-
were titrated to 40 mg 3 times daily as tolerated, and at 4 weeks, the marily due to symptomatic bradycardia. The intent-to-treat analysis
dose was increased to 60 or 80 mg 3 times daily depending on serum showed a nonstatistically significant decrease in all-cause mortality
propranolol concentrations. Key exclusion criteria included severe (21.1% vs 16.2%; P-value = non-significant) and cardiac mortality
HF, bradycardia, and asthma. The mean age was 55 years, and about (19.7% vs 13%; P-value = NS) in those receiving metoprolol. In those
85% of the patients were male. Aspirin was administered to 21% with a large infarct, as determined by lactate dehydrogenase levels,
of patients in both groups, and antiarrhythmic drugs were used in a significant reduction in cardiac mortality was observed (32.1%
21% of patients in the treatment group and in 25% in the placebo vs 12.5%; P < 0.05) as well as a reduction in sudden cardiac death
group. The average time from presentation to randomization was 14 (13 events vs 1 event; P < 0.001). A significant reduction in non-
days. Eighty-two percentage of patients received propranolol 60 mg fatal reinfarction was also observed in the metoprolol arm (21.1%
3 times daily. The RRR in the primary endpoint, all-cause mortality, vs 11.7%; P < 0.05), which was consistent among all risk groups.
was 26% in the propranolol group (9.8% vs 7.2%; P < 0.005) with a Patients in the metoprolol arm experienced more symptomatic
number needed to treat of 38 over a mean of 25 months. Patients were bradycardia (12.3% vs 2.7%; P < 0.01) and HF (5.2% vs 1.4%;
stratified into risk groups, similar to those used in the Norwegian P < 0.05), but less uncontrolled angina than those in the placebo
Multicenter Study of Timolol after MI study.17 An analysis based on group (3.9% vs 10.9%; P < 0.05). The benefits of long-term metopro-
these groups revealed that the beneficial effects of propranolol were lol treatment appeared to persist throughout the study period.
consistent, regardless of risk factors; however, post hoc subgroup The First International Study of Infarct Survival (ISIS-1)
analyses suggested a greater benefit in those who had electrical or was a prospective, multicenter, randomized, placebo-controlled,
mechanical complications (eg, pulmonary edema or hypotension) open-label trial designed to assess the impact of early, intravenous
during their hospitalization. β-blocker therapy on cardiovascular mortality in patients with sus-
The Metoprolol in Acute Myocardial Infarction (MIAMI) trial pected MI.23 Over 16,000 patients were randomized to atenolol 5 mg
was a prospective, multicenter, randomized, placebo-controlled, dou- intravenously, plus an additional 5 mg 10 minutes after the initial
ble-blind trial conducted in 17 countries, which evaluated the signifi- injection if the heart rate remained > 60 beats per minute, followed
cance of early intravenous metoprolol on short-term mortality.21 A by 100 mg of oral atenolol daily for an additional 6 days or match-
total of 5778 patients under 75 years of age with suspected AMI were ing placebo. Exclusion criteria included patients with a clear con-
included. Patients were administered 15 mg of intravenous metopro- traindication to β-blocker therapy, including persistent bradycardia
lol, given as 5 mg every 2 minutes for 3 doses, within 24 hours of or hypotension, second- or third-degree heart block, severe HF,
onset of symptoms. Following the intravenous metoprolol, 50 mg or bronchospasm. The average age was 60 years, and 77% of the

© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.cardiologyinreview.com | 161

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Kukin et al Cardiology in Review  •  Volume 26, Number 3, May/June 2018

patients were male. Other drugs administered during the treatment β-BLOCKER USE IN THE REPERFUSION ERA
period included diuretics and antiarrhythmics in similar proportions
among both groups, as well as calcium antagonists, which were The 1990s: Timing of β-Blocker Initiation After
utilized more by the control group (17% vs 9%; no P-value pro- Thrombolysis
vided). Treatment was initiated an average of 5 hours after the onset The Thrombolysis in Myocardial Infarction Phase II (TIMI
of symptoms. The mean intravenous dose achieved was 8.1 mg, and II-B) trial was the first large trial to assess the role of β-blockers in the
37% of patients required a dose reduction of intravenous or oral setting of reperfusion therapy among patients with MI.28 In the over-
atenolol at some point during treatment. A statistically significant all prospective, multicenter TIMI II trial, 2948 patients presenting
15% RRR (95% CI, 0.01–0.27) in vascular mortality was observed with an AMI were randomized to an invasive (routine angiography
in the atenolol group during the 7-day treatment period (3.89% vs and percutaneous transluminal coronary angiogram between 18 and
4.57%; P < 0.04), with the benefit being limited to days 0–1. During 48 hours after presentation) or a conservative strategy (angiography
the 1-year follow-up, no significant difference in vascular mortal- to evaluate for percutaneous transluminal coronary angiogram in set-
ity was observed outside of the initial 7-day treatment period. The ting of recurrent ischemia or a positive submaximal exercise study)
atenolol group was observed to have an increased use of inotropic after treatment with recombinant tissue-type plasminogen activator.
agents (5.0% vs 3.4%; P < 0.0001). The authors did not find sig- All patients also received aspirin and unfractionated heparin. Of the
nificant differences among outcomes between higher risk patients enrolled patients, 1434 received β-blocker therapy with either “imme-
(eg, older age, electrocardiographic signs of MI, diabetes, previous diate” treatment consisting of 3 doses of 5 mg intravenous metopro-
MI, first-degree block, atrial fibrillation, and systolic blood pressure lol or “delayed” treatment with 25 mg oral metoprolol tartrate every
< 120 mm Hg combined with heart rate > 90 bpm). 12 hours on day 6. Placebo controls were not utilized. Notable exclu-
sion criteria included a history of a cerebrovascular event, previous
Summary coronary artery bypass surgery, prosthetic heart valve replacement,
Although many β-blocker trials were conducted during the dilated cardiomyopathy, asthma, and chronic obstructive lung dis-
1980s, results were inconsistent and left clinicians with no clear con- ease. The average age was 55 years, and 85% were male. Twice
sensus of how or when to incorporate β-blockade into the manage- daily oral metoprolol tartrate was initiated in 94.7% of the “imme-
ment of AMI. Among trials that studied long-term treatment with diate” treatment group who did not meet hemodynamic goals with
β-blockers, ranging from 25 to 36 months, the BHAT and Norwegian intravenous metoprolol. Treatment was initiated an average of 3.3
Multicenter Study of Timolol after MI trials demonstrated a reduc- hours from symptom onset in the immediate group. In the delayed
tion in mortality.17,24 The trial conducted by Olsson et al22, however, group, treatment was initiated at the scheduled time in 73.2% of
found no difference in mortality between the metoprolol-treated and patients. No difference was observed in the primary end point, global
placebo groups. In all these trials, β-blockade was started multiple LV ejection fraction (EF) at discharge, between the immediate and
days after infarction (ie, 5–28 days). One consistency across these delayed groups (51.0% vs 50.1%; P = 0.22). At 6 days, there was a
trials is the distinction of low-risk patients (eg, good exercise toler- significant reduction in nonfatal or fatal reinfarction (2.7% vs 5.1%;
ance, normal LV function, no arrhythmias) garnering little, if any, P = 0.02) and recurrent chest pain (18.8% vs 24.1%; P < 0.02) among
benefit from β-blockade, in contrast to moderate high risk patients those who were in the immediate treatment group. At 6 weeks, the
(eg, symptoms of HF or arrhythmias), which had long-term benefits. lower risk of nonfatal or fatal reinfarction persisted (4.5% vs 7.3%;
The Göteborg Metoprolol trial was the first trial to show mor- P = 0.03) in the immediate treatment group, but these differences did
tality reduction with early β-blockade, initiated within 24 hours of not persist at 1 year. There was no difference in mortality between the
infarction. A few years later, the ISIS-I trial corroborated these find- groups. The subgroup of patients with low-risk features (ie, younger
ings, demonstrating a 15% reduction in vascular death at 7 days with age, no history of MI, not presenting with an anterior MI) appeared
immediate atenolol administration. The MIAMI trial contradictorily to derive a mortality benefit from the immediate treatment compared
found no mortality benefit with early metoprolol. ISIS-I noted no with deferred treatment (0.4% vs 3.6%; P = 0.009). Patients were
differences among risk groups, but a retrospective analysis of the identified as low risk if they did not possess any of the following
MIAMI trial revealed a statistically significant reduction of mortal- characteristics: history of MI, ST-segment elevation of anterior elec-
ity in high-risk patients. A large, comprehensive meta-analysis pub- trocardiographic leads, rales extending upward to include more than
lished in 1985 analyzed 65 trials and concluded that published data one-third of the lung fields, hypotension and sinus tachycardia, atrial
at the time did not support early β-blockers therapy, but did support fibrillation or flutter, age 70 years or higher, pulmonary edema, or
long-term benefit, citing a relative reduction in mortality of 20%.25 cardiogenic shock.
An editorial by Griggs et al26 suggested that the published data at The Global Utilization of Streptokinase and Tissue Plasmino-
the time did not support the need for β-blockers in low-risk patients. gen Activator for Occluded Coronary Arteries (GUSTO-I) trial was
Low-risk patients, estimated to have a 1–2% risk of death per year, a prospective, multicenter, randomized, open-label trial that com-
were defined as those with good exercise tolerance, normal LV func- pared 4 thrombolytic strategies in MI patients.29 The protocol recom-
tion, and no ventricular arrhythmia. The authors criticized the BHAT mended 2 doses of 5 mg intravenous atenolol followed by 50–100 mg
and Norwegian studies for not incorporating ambulatory and elec- oral atenolol daily, during hospitalization, in patients without hypo-
trocardiographic monitoring or exercise testing into their criteria tension, bradycardia, or HF. All patients were to receive aspirin
for determining risk levels of patients in their studies, speculating therapy. A prospectively planned, observational post hoc analysis of
that these trials could have underestimated the risk level of the “low- the GUSTO-I dataset was conducted to assess the 30-day mortal-
risk” patients, leading to trends indicating survival benefit among all ity among patients who did or did not receive atenolol.30 The aver-
groups. age age was 60 years, and 75% of the patients were male. Of the
It is important to consider that treatment strategies for AMI 41,021 patients enrolled in GUSTO-1, 24.8% did not receive ateno-
during this time were still quite limited. Aspirin was not routinely lol; 44.5% only received the intravenous form, and 30.7% received
used until after 1988, and reperfusion strategies (ie, thrombolysis) only the oral form. Ninety-two percentage of those who received
were not available until after these trials were completed.27 In addi- both forms of atenolol received the first oral dose within the first
tion, statins and P2Y12 inhibitors would not become available until day of enrollment, and 68% of those who received only oral atenolol
the late 1990s and early 2000s. received it within the first day of enrollment. A lower rate of 30-day

162  |  www.cardiologyinreview.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Cardiology in Review  •  Volume 26, Number 3, May/June 2018 β-Blocker Use and Outcomes in ACS

mortality was observed among those who received any atenolol The Clopidogrel and Metoprolol in Myocardial Infarction
treatment (3.8%) versus no atenolol (16.5%), and the median time Trial (COMMIT)/Second Chinese Cardiac Study was a prospective,
to death was longer among those who received atenolol (79.1 hours multicenter, randomized, placebo-controlled trial that evaluated the
vs 18.8 hours). Patients who received atenolol had lower risk features addition of early metoprolol to standard therapy in those presenting
than those who did not, including younger age, higher systolic blood within 24 hours of an acute MI.32 This study utilized a 2 × 2 factorial
pressure, and lower Killip class, but a significant mortality difference design and also evaluated the use of clopidogrel in addition to aspirin
persisted after adjustment for differences in baseline characteristics in those with a suspected AMI. A total of 45,852 patients were ran-
(odds ratio [OR], 0.33 [95% CI, 0.31–0.36], P < 0001). The adjusted, domized to receive placebo or a combination of intravenous and oral
30-day risk of death was increased in those who received the com- metoprolol. Those randomized to the metoprolol group received 5 mg
bination of oral and intravenous therapy versus oral atenolol alone intravenously, for up to 3 doses, depending on their hemodynamics,
(OR, 1.2 [1.1–1.5]; P = 0.0001). Patients who received any form of followed by metoprolol tartrate 50 mg every 6 hours for the first day
atenolol vs no atenolol also had a lower incidence of shock (3.9% and then metoprolol succinate 200 mg daily thereafter. Therapy was
vs 12.4%), HF (14.1% vs 22.7%), ventricular tachycardia (5.1% vs continued until 28 days, death, or hospital discharge, whichever
9.6%), and ventricular fibrillation (5.7% vs 9.9%). When compared occurred first. Key exclusion criteria included primary percutaneous
with oral atenolol administered within 24 hours, intravenous atenolol coronary intervention (PCI), hypotension, bradycardia, heart block,
was associated with a significantly higher incidence of congestive and cardiogenic shock. Moderate HF of Killip class II or III was not
HF (14.3% vs 10.7%; P < 0.01), cardiogenic shock (3.3% vs 2.2%; an exclusion, unlike many previous trials. The average age was 61
P < 0.01), recurrent ischemia (22.1% vs 18.9%; P < 0.01), and need years; 72% of the patients were male, and over 50% of the patients
for pacing (6% vs 4%; P < 0.01). Because this was an observational received thrombolytic agents. The majority of patients received anti-
study, it is limited by the fact that patients were not randomized with coagulation therapy; 68.2% received an ACEi, and 11.8% received a
respect to atenolol treatment, but rather reperfusion treatment. calcium channel blocker. An average of 10.3 hours elapsed between
the onset of symptoms and randomization, and mean treatment dura-
Summary tion was 15 days. Of those allocated to metoprolol, 90% received all
The 1990s included the first trials evaluating the usefulness 3 intravenous doses, and 86.2% received the scheduled oral treat-
of β-blockers in the setting of AMI and treatment with reperfusion ment. Of those who completed the oral regimen, 1.9% required a per-
therapy. The TIMI II-B and GUSTO-I trials both evaluated timing manent dose reduction. Reinfarction (2.0% vs 2.5%; OR, 0.82 [95%
of β-blocker administration in the setting of thrombolytic therapy in CI, 0.72–0.92]; P = 0·001) and ventricular fibrillation (2.5% vs 3.0%;
addition to aspirin. There was no difference in mortality observed OR, 0.83 [95% CI, 0.75–0.93]; P = 0·001) occurred less frequently
among those treated with metoprolol immediately after presenta- in the metoprolol group. However, there was no difference in the
tion or on day 6 in TIMI II-B. In the GUSTO-I, the use of atenolol composite primary outcome (death, reinfarction, or cardiac arrest;
decreased the risk of cardiovascular events, but immediate intrave- OR, 0.96 [95% CI, 0.90–1.01; P = 0.10) or the co-primary outcome
nous β-blockade was associated with an increased risk of harm com- of death (OR, 0.99 [95% CI, 0.92–1.05]; P = 0.69) between the 2
pared with oral atenolol therapy. The timing and dosing of initiation groups. Significantly more patients in the metoprolol arm developed
of β-blocker were therefore further explored. cardiogenic shock (5.0% vs 3.9%; OR, 1.30 [95% CI, 1.19–1.41]; P
< 0·0001), which predominately occurred on days 0–1. In particular,
THE 2000s: ADDITIONAL TRIALS OF β-BLOCKERS patients identified as being at high risk of developing shock, such as
those with Killip class III HF (absolute increase of 56.9 [standard
WITH THROMBOLYSIS error (SE), 14.4] deaths per 1000), systolic blood pressure < 120 mm
The Carvedilol Post-Infarct Survival Control in Left-ventricu- Hg (absolute increase of 23.3 [SE, 4.0] deaths per 1000), or heart
lar Dysfunction (CAPRICORN) trial was a prospective, multicenter, rate > 110 bpm at presentation (absolute increase of 34.6 [SE, 11.5]
randomized, placebo-controlled, double-blind trial evaluating the use deaths per 1000), experienced a notable net hazard.
of carvedilol in patients with LV dysfunction with or without signs
of HF following MI.31 A total of 1959 patients were enrolled and Summary
followed for a mean of 1.3 years. Three to 21 days after infarction, The CAPRICORN trial demonstrated a clear benefit of
patients were started on carvedilol 6.25 mg twice daily or placebo, β-blockade in those with concurrent MI and LV dysfunction. It
and the dose was titrated over 4–6 weeks to a goal of 25 mg twice is important to note that the β-blocker was not started until 3–21
daily. Background therapy with an angiotensin-converting enzyme days after infarction, titrated slowly (eg, every 2 weeks), and that
inhibitor (ACEi) was required. Notable exclusion criteria were unsta- all patients in this trial were on an ACEi. In COMMIT, early intra-
ble angina, indication for β-blocker therapy, and insulin-dependent venous metoprolol did not result in a difference in mortality com-
diabetes. The average age was 63 years, 75% were male, and the pared with placebo. Patients at higher risk of developing cardiogenic
average EF was 32.8%. At baseline, 86% were receiving aspirin shock experienced greater mortality, implying that β-blocker initia-
therapy, 97% were receiving an ACEi, 33% were receiving intrave- tion should be reserved for hemodynamically stable patients. These
nous diuretic therapy, and 46% underwent reperfusion predominately findings are similar to findings in GUSTO-1, which suggested that
using thrombolytic therapies. Treatment discontinuation was simi- a delay in β-blocker therapy may be optimal. It is also important
lar between those receiving carvedilol and placebo (20% vs 18%, to recognize that the majority of patients who received reperfusion
respectively). The maximum dose was achieved by 74% of those in therapy in CAPRICORN and COMMIT included few patients who
the carvedilol group. There was no difference in primary endpoint of underwent PCI, as thrombolysis was the preferred and most readily
composite all-cause mortality or hospital admission due to cardiovas- accessible treatment at the time.
cular causes between those receiving carvedilol or placebo (35% vs
37%; hazard ratio [HR] 0.92 [95% CI, 0.80–1.07]; P = 0.296). There
was, however, a reduction in the risk of all-cause mortality among THE 2010s: β-BLOCKER USE IN PATIENTS
those who received carvedilol (12% vs 15%; HR, 0.77 [95% CI, UNDERGOING PCI
0.6–0.98]; P = 0.031). This difference appeared to be most apparent The Effect of Metoprolol in Cardioprotection During an
after 6 months of therapy. The use of carvedilol was also associated Acute Myocardial Infarction (METOCARD-CNIC) trial was a pro-
with reductions in the risk of cardiovascular death and nonfatal MI. spective, multicenter, randomized, controlled, single-blinded trial

© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.cardiologyinreview.com | 163

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Kukin et al Cardiology in Review  •  Volume 26, Number 3, May/June 2018

that evaluated the effect of metoprolol, administered before PCI, on A reductase inhibitors were higher in those discharged on a
infarct size in patients with an anterior ST elevation MI (STEMI) and β-blocker. Numerous multivariable analyses, which incorporated
Killip class II or less symptoms.33 A total of 270 patients undergoing adjustments for differences in baseline characteristics among dose
PCI within 6 hours of symptom onset were enrolled and followed groups, revealed no significant association between higher doses and
for an average of 2 years. Patients randomized to prereperfusion increased survival (HR, 0.889 [95% CI, 0.754–1.048]; P = 0.16).
β-blockade received up to 3 doses of 5 mg intravenous metopro- A subgroup analysis determined that patients who underwent revas-
lol during transfer to the PCI center. All patients except those who cularization conferred more benefit from lower doses than doses >
developed contraindications received oral metoprolol within 12–24 50% of the target dose (HR, 0.649 [95% CI, 0.472–0.891], 0.546
hours of infarction. The control group did not receive a placebo. [95% CI, 0.403–0.740], and 0.768 [95% CI, 0.563–1.048]; P = 0.037
Cardiac magnetic resonance (CMR) imaging was scheduled for 5–7 for the > 0% to 12.5%, > 12.5% to 25%, and > 25% to 50% doses,
days after the index event to evaluate infarct size. Exclusion criteria respectively). No differences in benefit were observed among differ-
included systolic blood pressure < 120 mm Hg, type II–III atrioven- ent doses in nonrevascularized patients. Subgroup analyses found no
tricular block, heart rate < 60 beats/min, prior infarction, or active interaction with the effect of β-blocker dose and survival on the basis
treatment with β-blockers. The average age was 58 years, and 86% of β-blocker agent (metoprolol vs carvedilol), type of MI (STEMI vs
were male. Over 95% received heparin, aspirin, and a thienopyridine non-STEMI), or LVEF (> vs < 40%).
at the time of PCI. Grade 2–3 TIMI flow was achieved in 93.7% after The Early Beta-blocker Administration before Primary
PCI. Patients randomized to prereperfusion β-blockade received PCI in Patients with ST-elevation Myocardial Infarction (EARLY-
metoprolol a median of 10 minutes after STEMI diagnosis, and 67% BAMI) trial was a prospective, international, multicenter, random-
of these patients received all 3 boluses. All patients received meto- ized, placebo-controlled, double-blind trial that studied the role of
prolol tartrate within 12–24 hours after the MI, unless a contraindica- intravenous β-blockade before PCI in 683 patients presenting with
tion to use developed, mean infarct size, the primary end point, was a STEMI.36 The primary objective of the study was 30-day infarct
smaller in the group treated with intravenous metoprolol a week after size evaluated through CMR. Patients presenting within 12 hours of
infarction (25.6 g vs 32.0 g; adjusted treatment effect ˗6.52 [95% symptom onset were randomized to intravenous metoprolol or pla-
CI, ˗11.39 to ˗1.78]; P = 0.012). There was a statistically significant cebo. Of note, the results of METOCARD-CINC were not available
increase in mean left ventricular ejection fraction (LVEF) 1 week at the time this study was designed. Patients in the metoprolol group
after STEMI, in those who received intravenous metoprolol (46.1% received 5 mg of intravenous metoprolol during ambulance transit
vs 43.4%; adjusted treatment effect, 2.67 [95% CI, 0.09–5.21; and another 5 mg immediately before PCI initiation in the catheter-
P = 0.045). There was no difference in the rate of major cardiovascu- ization laboratory. A 10 mg dose was selected due to the increased
lar adverse events (composite of death, malignant ventricular arrhyth- risk of cardiogenic shock observed in the COMMIT trial with 15 mg
mia, cardiogenic shock, atrioventricular block, and reinfarction) at of intravenous metoprolol. All participants were to receive oral meto-
24 hours between groups (7.1% metoprolol group vs 12.3% control); prolol, at a dose recommended by their physician, 12 hours post-
however, this study was not designed to determine a difference in PCI and upon discharge. Participants also received unfractionated
clinical outcomes. Six months after enrollment, there was a statisti- heparin, aspirin, and clopidogrel or ticagrelor. Key exclusion criteria
cally significant higher mean LVEF in the metoprolol arm (48.7% included, but were not limited to, Killip class III and IV, systolic BP
vs 45.0%; adjusted treatment effect, 3.49 [95% CI, 0.44–6.55]; < 100 mm Hg, heart rate < 60 beats per minute, type II and III atrio-
P = 0.025) and a significantly lower occurrence of severely depressed ventricular block, previous MI, and asthma. Baseline characteristics
LVEF of ≤ 35% (11% vs 27%; P = 0.006).34 Because this study was were similar between groups with the exception of more patients in
not developed to show effects on clinical endpoints, its application to the placebo group undergoing coronary artery bypass graft surgery
clinical practice is limited. (7% vs 3.6%; P = 0.049) and a lower heart rate in the metoprolol
The Outcomes of Beta-blocker Therapy After Myocardial group (74.35 ± 13.71 vs 78.68 ± 15.69; P < 0.001). The average age
Infarction (OBTAIN) study was an observational multicenter regis- was 62.4 years, and 74.8% of the patients were male. One-half of the
try that analyzed the effect of β-blocker dose on survival post-MI.35 patients presented with an anterior MI. At baseline, 18.8% of partici-
The authors’ hypothesis was that higher β-blocker dose would be pants were on a β-blocker, and 73% were discharged on a β-blocker.
associated with improved survival. The primary endpoint was the dif- Mean time from onset of symptoms to first medical contact was
ference in time to all-cause death right-censored at 2 years. Doses similar between the 2 groups (135.5 minutes vs 147.8 minutes;
were categorized as no β-blocker, > 0% to 12.5%, > 12.5% to 25%, P = 0.880). Of the patients randomized to intravenous metoprolol,
> 25% to 50%, and > 50% of the target dose. Target doses were defined 81.1% received the 10 mg target dose. CMR was performed in 66%
as atenolol 100 mg/day, bisoprolol 10 mg/day, carvedilol immediate of those enrolled. No differences were noted in mean (± SD) infarct
release 50 mg/day or carvedilol controlled release 80 mg/day, meto- size (15.3 ± 11% vs 14.9 ± 11.5%; P = 0.616) or LVEF (51.0 ± 10.9%
prolol 200 mg/day, propranolol 180 mg/day, and timolol 20 mg/day. vs 51.7 ± 10.8%; P = 0.683) between those receiving metoprolol or
Data were collected from a registry of patients admitted with an acute placebo, respectively. There was no difference in the rate of major
MI. A total of 6682 patients who were discharged from the hospi- adverse cardiac events (ie, cardiac death, nonfatal reinfarction, or
tal post-MI, with a median follow-up of 2.1 years, were included in target vessel revascularization) at 30 days between the metoprolol
the analysis. The average age of patients studied was 64 years, and (6.2%) and placebo groups (6.9%; P = 0.72). Subgroup analyses
approximately 65% of the patients were male. The majority of patients were performed based on the location of infarct, time to presenta-
presented with a non-STEMI. Upon discharge, 91.5% were receiving tion, and the presence of an occluded vessel, but these were not deter-
a β-blocker; the most commonly prescribed agent was metoprolol fol- mined to affect infarct size. This study was limited by the larger than
lowed by carvedilol. The average length of stay was 5 days, and 13.4% anticipated dropout rate of 34% before CMR imaging. Patients who
of patients were discharged on > 50% of the target dose β-blocker died before undergoing CMR could have led to a selection bias of
dose. The mean administered dose was 38.1% of the target. The tim- patients with smaller infarcts.
ing of β-blocker initiation relative to MI symptoms was not reported. Huang et al37 conducted a meta-analysis to evaluate the role
The use of aspirin, clopidogrel, or the combination of the 2 was simi- of β-blockers in patients post-MI who underwent PCI. Data from 10
lar between groups. Discharge prescriptions for an ACEi or angioten- observational studies and from over 40,000 patients were included.
sin II receptor blocker and 3-hydroxy-3-methyl-glutaryl-co-enzyme The duration of follow-up varied between studies and ranged from

164  |  www.cardiologyinreview.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Cardiology in Review  •  Volume 26, Number 3, May/June 2018 β-Blocker Use and Outcomes in ACS

0.5 to 4 years. Of the studies that reported which agent was used, conducted before the widespread implementation of high potency
carvedilol was the most commonly prescribed. β-Blocker use was statins and P2Y12 inhibitors. High intensity statins, which have been
associated with a significant reduction in all-cause death (unadjusted shown to reduce cardiovascular mortality, recurrent MI, and stroke
relative risk, 0.58, 95% CI, 0.48–0.71; adjusted HR, 0.76, 95% CI, in patients with an ACS, may also reduce rates of sudden cardiac
0.62–0.94). This benefit was only observed in those with a reduced death.41,42 Third generation P2Y12 inhibitors (ie, ticagrelor and pra-
EF, with low rates of other secondary prevention drugs (eg, ACEIs/ sugrel) seem to reduce infarct size and microvascular obstruction
angiotensin II receptor blockers and antiplatelet therapy), or pre- more than clopidogrel.43 One can only assume that as medical thera-
sentation with non-STEMI. Authors concluded that the diminished pies continue to advance (eg, proprotein convertase subtilisin/kexin
effect of β-blockers in the general MI population may be due to type 9 [PCSK9] inhibitors, anti-inflammatory therapies, etc.), similar
advancement in other treatment options. trends may be observed. Taken altogether, the beneficial effects of
β-blockers in ACS may be blunted in the context of faster, more com-
Summary plete reperfusion strategies and better medical therapies. In support
During this decade, many trials investigated the role of of this, a meta-analysis of patients with ACS but without LV dysfunc-
early intravenous β-blockade in patients undergoing PCI.33,36,38 The tion compared trials in the prereperfusion era and postreperfusion
METOCARD-CNIC and EARLY-BAMI trials both randomized era, and showed no mortality benefit of β-blockers in the reperfusion
STEMI patients to intravenous metoprolol or control group before era.44 In addition, a national cohort study of over 160,000 patients
PCI, but found conflicting outcomes.33,36 Variations between the 2 tri- presenting with an AMI between 2007 and 2013 failed to show a dif-
als should be considered. A lower intravenous metoprolol dose was ference in 1-year mortality in patients receiving a β-blocker without
administered in EARLY-BAMI, and patients were permitted to be on LV dysfunction or HF.45
β-blocker therapy before enrollment. On the contrary, prior β-blocker
use was not permitted in METOCARD-CNIC. METOCARD-CNIC
also only included those with anterior wall MI, and thus, many of CONCLUSION
these patients had a larger infarct size. METOCARD-CNIC was not β-blocker use in ACS has evolved considerably throughout the
double-blinded or placebo-controlled and had a relatively small sam- decades. The role of β-blocker therapy in ACS is to reduce myocardial
ple size (n = 270), but EARLY-BAMI utilized a double-blinded, pla- oxygen demand and arrhythmias by blunting catecholamine-mediated
cebo-controlled design and studied over twice as many patients. It is adverse effects. Although early trials of β-blockers showed a mortality
imperative to note that METOCARD-CNIC did not include patients benefit, more recent trials have failed to replicate these results.
with Killip Class III to IV, so the results of this trial should not be
extrapolated to this population, especially considering the net hazard REFERENCES
experienced by patients with Killip Class III in the early metoprolol 1. Gunnar RM, Bourdillon PD, Dixon DW, et al. ACC/AHA guidelines for the
group of COMMIT. early management of patients with acute myocardial infarction. A report of the
American College of Cardiology/American Heart Association Task Force on
The trials in the 2010s are among the most recent trials evalu- Assessment of Diagnostic and Therapeutic Cardiovascular Procedures (sub-
ating β-blockers in ACS and include standard medical therapies such committee to develop guidelines for the early management of patients with
as ACEi, statins, and P2Y12 inhibitors. Unfortunately, the data are acute myocardial infarction). Circulation. 1990;82:664–707.
limited by study design, which was aimed at determining changes in 2. Anderson JL, Adams CD, Antman EM, et al. 2012 ACCF/AHA focused
infarct size rather than patient outcomes. update incorporated into the ACCF/AHA 2007 guidelines for the manage-
ment of patients with unstable angina/non-ST-elevation myocardial infarc-
tion: a report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol.
THROUGH THE DECADES: WHERE WE’VE COME 2013;61:e179–e347.
FROM AND WHERE WE ARE TODAY 3. Amsterdam EA, Wenger NK, Brindis RG, et al. 2014 AHA/ACC guideline for
Context is vitally important in any science to accurately inter- the management of patients with non-ST-elevation acute coronary syndromes:
pret and apply clinical data. Contextualizing β-blocker therapy for a report of the American College of Cardiology/American Heart Association
Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;64:e139–e228.
the treatment of ACS is no exception—in fact, failure to do so can
result in gross misinterpretation and misapplication of β-blockers 4. Anderson JL, Adams CD, Antman EM, et al.; American College of
Cardiology; American Heart Association Task Force on Practice Guidelines
in AMI in current medical practice. Although certain patient pop- (Writing Committee to Revise the 2002 Guidelines for the Management of
ulations undoubtedly benefit from long-term β-blocker therapy Patients With Unstable Angina/Non-ST-Elevation Myocardial Infarction);
(eg, reduced LV function), others remain in question and require American College of Emergency Physicians; Society for Cardiovascular
patient-specific decision making. It is evident from available trial data Angiography and Interventions; Society of Thoracic Surgeons; American
Association of Cardiovascular and Pulmonary Rehabilitation; Society for
that in the setting of hemodynamic instability or cardiogenic shock, Academic Emergency Medicine. ACC/AHA 2007 guidelines for the man-
signs of HF, or bradycardia, early β-blocker therapy has consistently agement of patients with unstable angina/non-ST-elevation myocardial
proven to be potentially harmful.3,30,39 On the contrary, patients with infarction: a report of the American College of Cardiology/American Heart
AMI with a reduced EF who do not have signs of HF have a clear Association Task Force on Practice Guidelines (writing committee to revise
the 2002 guidelines for the management of patients with unstable angina/
mortality benefit from long-term β-blocker therapy (class I level A non-ST-elevation myocardial infarction) developed in collaboration with the
recommendation in current guidelines).3 American College of Emergency Physicians, the Society for Cardiovascular
Although the use of β-blockers in ACS is uniformly recog- Angiography and Interventions, and the Society of Thoracic Surgeons
nized and accepted as a standard of care, the benefits of β-blockers endorsed by the American Association of Cardiovascular and Pulmonary
Rehabilitation and the Society for Academic Emergency Medicine. J Am Coll
in the current era of ACS management remain in question in patients Cardiol. 2007;50:e1–e157.
without LV dysfunction or ventricular arrhythmias. Large, random- 5. Ryan TJ, Anderson JL, Antman EM, et al. ACC/AHA guidelines for the man-
ized, placebo-controlled trials, such as COMMIT, TIMI-IIB, and agement of patients with acute myocardial infarction. A report of the American
GUSTO-I, which showed benefit with β-blockers in the reperfusion College of Cardiology/American Heart Association Task Force on Practice
era, included few patients who received PCI, which has been shown Guidelines (Committee on Management of Acute Myocardial Infarction).
J Am Coll Cardiol. 1996;28:1328–1428.
to reduce infarct size and blunt the sympathetic response more so
6. Antman EM, Anbe DT, Armstrong PW, et al. ACC/AHA guidelines for the
than thrombolytic therapy, subsequently reducing the incidence of management of patients with ST-elevation myocardial infarction; A report
catecholamine-mediated adverse effects.40 In addition, many of the of the American College of Cardiology/American Heart Association Task
β-blocker trials that we base current recommendations on were Force on Practice Guidelines (committee to revise the 1999 guidelines for the

© 2018 Wolters Kluwer Health, Inc. All rights reserved. www.cardiologyinreview.com | 165

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.


Kukin et al Cardiology in Review  •  Volume 26, Number 3, May/June 2018

management of patients with acute myocardial infarction). J Am Coll Cardiol. infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) II-B
2004;44:E1–E211. Study. Circulation. 1991;83:422–437.
7. Braunwald E, Antman EM, Beasley JW, et al. ACC/AHA guidelines for the 29. GUSTO investigators. An international randomized trial comparing four

management of patients with unstable angina and non-ST-segment eleva- thrombolytic strategies for acute myocardial infarction. N Engl J Med.
tion myocardial infarction. A report of the American College of Cardiology/ 1993;329:673–682.
American Heart Association Task Force on Practice Guidelines (committee 30. Pfisterer M, Cox JL, Granger CB, et al. Atenolol use and clinical outcomes
on the management of patients with unstable angina). J Am Coll Cardiol. after thrombolysis for acute myocardial infarction: the GUSTO-I experience.
2000;36:970–1062. Global utilization of streptokinase and TPA (alteplase) for occluded coronary
8. Mueller HS, Ayres SM, Religa A, et al. Propranolol in the treatment of acute arteries. J Am Coll Cardiol. 1998;32:634–640.
myocardial infarction. Effect on myocardial oxygenation and hemodynamics. 31. Dargie HJ. Effect of carvedilol on outcome after myocardial infarction in
Circulation. 1974;49:1078–1087. patients with left-ventricular dysfunction: the CAPRICORN randomised trial.
9. Snow PJ. Effect of propranolol in myocardial infarction. Lancet. Lancet. 2001;357:1385–1390.
1965;2:551–553. 32. Chen ZM, Pan HC, Chen YP, et al.; COMMIT (ClOpidogrel and Metoprolol in
10. Snow PJ. Treatment of acute myocardial infarction with propranolol. Am J Myocardial Infarction Trial) collaborative group. Early intravenous then oral
Cardiol. 1966;18:458–462. metoprolol in 45,852 patients with acute myocardial infarction: randomised
11. Balcon R, Jewitt DE, Davies JP, et al. A controlled trial of propranolol in acute placebo-controlled trial. Lancet. 2005;366:1622–1632.
myocardial infarction. Lancet. 1966;2:918–920. 33. Ibanez B, Macaya C, Sánchez-Brunete V, et al. Effect of early metoprolol on
12. Clausen J, Felsby M, Jorgensen FS, et al. Absence of prophylactic effect of infarct size in ST-segment-elevation myocardial infarction patients undergo-
propranolol in myocardial infarction. Lancet. 1966;2:920–924. ing primary percutaneous coronary intervention: the effect of Metoprolol
in Cardioprotection During an Acute Myocardial Infarction (METOCARD-
13. Bath J, Blake S, Bloom R, et al. Propranolol in acute myocardial infarction. A CNIC) trial. Circulation. 2013;128:1495–1503.
multicentre trial. Lancet. 1966;2:1435–1438.
34. Pizarro G, Fernández-Friera L, Fuster V, et al. Long-term benefit of early pre-
14. Norris RM, Caughey DE, Scott PJ. Trial of propranolol in acute myocardial reperfusion metoprolol administration in patients with acute myocardial infarction:
infarction. Br Med J. 1968;2:398–400. results from the METOCARD-CNIC trial (effect of metoprolol in cardioprotection
15. Ruskin JN. The cardiac arrhythmia suppression trial (CAST). N Engl J Med. during an acute myocardial infarction). J Am Coll Cardiol. 2014;63:2356–2362.
1989;321:386–388. 35. Goldberger JJ, Bonow RO, Cuffe M, et al.; OBTAIN Investigators. Effect of
16. Danese E, Montagnana M. An historical approach to the diagnostic biomark- beta-blocker dose on survival after acute myocardial infarction. J Am Coll
ers of acute coronary syndrome. Ann Transl Med. 2016;4:194. Cardiol. 2015;66:1431–1441.
17. von der Lippe G, Hansen H, Lund-Johansen P: Timolol-induced reduction in 36. Roolvink V, Ibáñez B, Ottervanger JP, et al.; EARLY-BAMI Investigators.
mortality and reinfarction in patients surviving acute myocardial infarction. Early intravenous beta-blockers in patients with ST-segment elevation myo-
N Engl J Med. 1981;304:801–807. cardial infarction before primary percutaneous coronary intervention. J Am
Coll Cardiol. 2016;67:2705–2715.
18. Pedersen TR. Six-year follow-up of the Norwegian Multicenter Study
on timolol after acute myocardial infarction. N Engl J Med. 1985;313: 37. Huang BT, Huang FY, Zuo ZL, et al. Meta-analysis of relation between oral
1055–1058. β-blocker therapy and outcomes in patients with acute myocardial infarc-
tion who underwent percutaneous coronary intervention. Am J Cardiol.
19. Hjalmarson A, Elmfeldt D, Herlitz J, et al. Effect on mortality of metopro-
2015;115:1529–1538.
lol in acute myocardial infarction. A double-blind randomised trial. Lancet.
1981;2:823–827. 38. Er F, Erdmann E, Nia AM, et al. Esmolol for tight heart rate control in patients
with STEMI: design and rationale of the beta-blocker in acute myocardial
20. The beta-blocker heart attack trial. beta-Blocker Heart Attack Study Group.
infarction (BEAT-AMI) trial. Int J Cardiol. 2015;190:351–352.
JAMA. 1981;246:2073–2074.
39. Kaatz S, Ahmad D, Spyropoulos AC, et al.; Subcommittee on Control of
21. Metoprolol in acute myocardial infarction. Mortality. The MIAMI Trial

Anticoagulation. Definition of clinically relevant non-major bleeding in stud-
Research Group. Am J Cardiol. 1985;56:15G–22G. ies of anticoagulants in atrial fibrillation and venous thromboembolic disease
22. Olsson G, Rehnqvist N, Sjögren A, et al. Long-term treatment with metoprolol in non-surgical patients: communication from the SSC of the ISTH. J Thromb
after myocardial infarction: effect on 3 year mortality and morbidity. J Am Coll Haemost. 2015;13:2119–2126.
Cardiol. 1985;5:1428–1437. 40. Bonnemeier H, Hartmann F, Wiegand UK, et al. Heart rate variability in
23. Randomised trial of intravenous atenolol among 16 027 cases of suspected patients with acute myocardial infarction undergoing primary coronary angio-
acute myocardial infarction: ISIS-1. First International Study of Infarct plasty. Am J Cardiol. 2000;85:815–820.
Survival Collaborative Group. Lancet. 1986;2:57–66. 41. Mitchell LB, Powell JL, Gillis AM, et al.; AVID Investigators. Are lipid-lower-
24. A randomized trial of propranolol in patients with acute myocardial infarction. ing drugs also antiarrhythmic drugs? An analysis of the Antiarrhythmics versus
I. Mortality results. JAMA. 1982;247:1707–1714. Implantable Defibrillators (AVID) trial. J Am Coll Cardiol. 2003;42:81–87.
25. Yusuf S, Peto R, Lewis J, et al. Beta blockade during and after myocardial 42. Levantesi G, Scarano M, Marfisi R, et al. Meta-analysis of effect of statin
infarction: an overview of the randomized trials. Prog Cardiovasc Dis. treatment on risk of sudden death. Am J Cardiol. 2007;100:1644–1650.
1985;27:335–371. 43. Khan JN, Greenwood JP, Nazir SA, et al: Infarct size following treatment with
26. Griggs T, Adams K, Narvarte H, et al. Beta-blocker therapy after myocardial second- versus third-generation P2Y12 antagonists in patients with multives-
infarction: secondary prevention in all patients?. Zeitschrift Fur Kardiologie. sel coronary disease at ST-segment elevation myocardial infarction in the
1985;74;6:173–178. CvLPRIT study. J Am Heart Assoc. 2016;5:10.1161/JAHA.116.003403.
27. Randomised trial of intravenous streptokinase, oral aspirin, both, or neither 44. Bangalore S, Makani H, Radford M, et al. Clinical outcomes with β-blockers
among 17,187 cases of suspected acute myocardial infarction: ISIS-2. ISIS-2 for myocardial infarction: a meta-analysis of randomized trials. Am J Med.
(Second International Study of Infarct Survival) Collaborative Group. Lancet. 2014;127:939–953.
1988;2:349–360. 45. Dondo TB, Hall M, West RM, et al. β-Blockers and mortality after acute myo-
28. Roberts R, Rogers WJ, Mueller HS, et al. Immediate versus deferred beta- cardial infarction in patients without heart failure or ventricular dysfunction.
blockade following thrombolytic therapy in patients with acute myocardial J Am Coll Cardiol. 2017;69:2710–2720.

166  |  www.cardiologyinreview.com © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved.

Вам также может понравиться