You are on page 1of 16

Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

Contents lists available at ScienceDirect

Journal of Cardiothoracic and Vascular Anesthesia


journal homepage: www.jcvaonline.com

Review Article

Perioperative β-Adrenergic Blockade in Noncardiac


and Cardiac Surgery: A Clinical Update
Adriana D. Oprea, MD*, Frederick W. Lombard, MBChB, FANZCA†,
Miklos D. Kertai, MD, PhD†,
1

*
Department of Anesthesiology, Yale School of Medicine, New Haven, CT

Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical
Center, Nashville, TN

Key Words: atrial fibrillation; beta-adrenergic blockers; cardiac surgery; mortality; noncardiac surgery; pharmacokinetics; pharmacodynamics

BETA-ADRENERGIC BLOCKERS (β-blockers) represent bronchospasm. Nevertheless, overwhelming evidence demon-


a heterogeneous class of cardiovascular drugs with different strated that β-blockers are well tolerated in patients with
characteristics regarding their pharmacologic properties and asthma or chronic obstructive pulmonary disease, and their
clinical effects. As a class, β-blockers have been around for use was associated with a significant decrease in morbidity and
more than 50 years, and their use in the chronic treatment of mortality due to cardiovascular diseases.7,8 In the elderly
ischemic heart disease and heart failure is well established.1 population with a history of myocardial infarction, β-blocker
Currently, β-blockers are indicated as long-term therapy in use also was associated with a significant mortality benefit;
patients with a history of prior myocardial infarction (asso- however, this mortality benefit was confined to elderly patients
ciated with a lower reinfarction and mortality rate) and in with pre-existing cognitive impairment.9
patients with systolic heart failure (survival benefit).2,3 Of Perioperative β-blocker use, however, has been a matter of
note, β-blockers are not indicated as first-line therapy in ongoing controversy. Initial small-scale clinical trials by
patients with uncomplicated hypertension unless there is Mangano et al., as well as Poldermans et al., observed
concomitant ischemic heart disease or heart failure.4 However, beneficial effects in noncardiac surgery patients who were at
a
their chronic use in patients with a history of myocardial moderate to high risk for cardiovascular complications. By
infarction has been challenged recently. Contemporary studies reducing episodes of intraoperative and postoperative tachy-
have demonstrated limited mortality benefit when used for cardia, perioperative β-blocker use resulted in a decreased risk
secondary prevention in patients who have suffered their first for major adverse cardiac events (MACE).10,11 The findings of
myocardial infarction unless there is evidence for heart these initial clinical trials prompted expert committees of the
failure.5,6 European Society of Cardiology and the American College of
Traditionally, severe asthma and chronic obstructive pul- Cardiology/American Heart Association (ACC/AHA) to
monary disease have been contraindications for β-blocker use, recommend prophylactic β-blocker use for all moderate- to
since β-blockers may increase airway reactivity and high-risk patients ( 4 2 Revised Cardiac Risk Index [RCRI]

1 a
Address reprint requests to Miklos D. Kertai, MD, PhD, Director of Dr. Poldermans’ work (including the DECREASE trials II-VI) has come
Perioperative Precision Medicine Program, Professor of Anesthesiology, under scrutiny for potential scientific misconduct and ethical concerns.
Division of Cardiothoracic Anesthesiology, Department of Anesthesiology, However, DECREASE I trial has never come under scrutiny, and since its
Vanderbilt University Medical Center, 1215 21st Ave South, Suite 5160, results are part of studies on the historical use of perioperative β-blockers, the
Nashville, TN 37232. authors decided to include and review it in this manuscript in an unbiased
E-mail address: miklos.kertai@vanderbilt.edu (M.D. Kertai). manner.

http://dx.doi.org/10.1053/j.jvca.2018.04.045
1053-0770/& 2018 Elsevier Inc. All rights reserved.

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
2 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

Table 1 bypass graft (CABG) surgery for the prevention of post-


Revised Cardiac Risk Index. operative atrial fibrillation (AF).14–17
Revised Cardiac Risk Index criteria60
To date, the controversy surrounding perioperative β-
History of ischemic heart disease blocker use remains with current guidelines giving limited
History of congestive heart failure guidance for their use only in specific patient populations,
History of cerebrovascular disease (stroke or transient ischemic attack) unique clinical scenarios, or special surgical settings. The
History of diabetes mellitus requiring preoperative insulin use purpose of this review is to evaluate the most recent evidence
Chronic kidney disease (creatinine 42 mg/dL)
Undergoing high-cardiac risk surgery (suprainguinal vascular, intraperitoneal,
on perioperative β-blocker use and to provide additional
or intrathoracic) recommendations for perioperative β-blocker use in patients
Estimated risk for cardiac death, nonfatal myocardial infarction, and nonfatal undergoing both cardiac and noncardiac surgery. Although
cardiac arrest: there is a wide array of β-blockers available on the market, the
0 risk factors: 0.4% primary focus will be on β-blockers commonly used in the
1 risk factor: 0.9%
2 risk factors: 6.6%
perioperative period (ie, metoprolol, atenolol, and bisoprolol).
3 or more risk factors: 411%

Pharmacokinetics of β-blockers

criteria) (Table 1) undergoing high-risk noncardiac surgical As a drug class, β-blockers have similar pharmacodynamic
procedures (eg, vascular surgery, thoracic, and major abdom- effects in terms of heart rate control, but individually they have
inal) in 2002.12,13 distinct and wide-ranging pharmacokinetic profiles. Intrave-
However, a subsequent large-scale clinical trial, Periopera- nous β-blockers such as esmolol, landiolol, propranolol, and
tive Ischemic Evaluation (POISE), challenged those initial metoprolol are instantly fully bioavailable. The bioavailability
observations and found that prophylactic β-blocker use of oral β-blockers, however, primarily depends on their lipid
initiated on the morning of a noncardiac surgical procedure solubility, and to a lesser degree on their extent of gastro-
was associated with a higher risk for bradycardia, stroke, and intestinal absorption (most oral β-blockers are well absorbed).
mortality. These findings led to a change and revision of the The lipophilic β-blockers such as propranolol, metoprolol, and
recommendations of the relevant European Society of Cardi- carvedilol tend to have shorter half-lives and often need to be
ology and American College of Cardiology/American Heart administered more than once a day (Table 2).1 To overcome
Association guidelines for prophylactic β-blocker use in this and in an attempt to ensure a more stable effect, extended-
noncardiac surgery; that is, preoperative initiation and prophy- release preparations have been formulated for some β-block-
lactic use of β-blockers is not recommended in the setting of ers, such as metoprolol. Unfortunately, lipophilic β-blockers,
noncardiac surgery.13 Nevertheless, prophylactic β-blocker use in particular metoprolol, are also subject to varying degrees of
still is recommended in patients undergoing coronary artery first-pass hepatic metabolism, resulting in less predictable

Table 2
Pharmacokinetics of β-Blockers.

β-Blocker Absorption (%) Bioavailability (%) Lipophilicity Half-life Degradation/Excretion

Acebutolol 90 40 Yes 3-4 h Hepatic metabolism 30-40%


Renal 50%-60%
Atenolol 50 40 No 6-7 h Renal 50%-80%
Betaxolol 90 89 Yes 14-22 h Hepatic metabolism 15%
Renal 80%
Bisoprolol 90 80-94 Yes 9-12 h Hepatic metabolism 50%
Renal 50%
Carvedilol 90 24 Yes 7-10 h Hepatic metabolism 90%
Esmolol n/a n/a N/A 9 min Plasma esterases
Labetalol 90 33 Yes 5-8 h Hepatic metabolism 50%
Renal 50%-60%
Landiolol n/a n/a N/A 2-4 min Pseudocholinesterase
Liver carboxyesterase
Metoprolol 90 65-70 Yes 3-4 h Hepatic metabolism 95%
Renal 5%
Nadolol 50 30 No 20-24 h Renal 100%
Nebivolol 90 12-96 Yes 12-19 h Hepatic metabolism
Pindolol 90 90 Yes 3-4 h Hepatic metabolism 60-65%
Renal 35%-40%
Propranolol 90 30-40 Yes 3-6 h Hepatic metabolism
Renal o1%

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 3

plasma concentrations and significant interpatient response drug-specific properties, such as intrinsic sympathomimetic
variability. The now-recognized genetic basis for this observed activity and direct vasodilator activity.
interpatient variability in extent of liver metabolism is dis-
cussed in more detail in the section on pharmacogenomics
below.18 Of note, though, some lipophilic β-blockers such as β1-receptor Selectivity
nebivolol demonstrate little or no difference in effect on blood
pressure between rapid and poor metabolizers, likely due to There are 3 different subtypes of β-adrenergic receptors
the fact the metabolites of these β-blockers are active and may (ARs) with varying effects on cardiovascular physiology
exert similar β-adrenergic receptor blocking properties.19 (Fig 1). β1 ARs are expressed mostly in the myocardium,
In contrast to the lipophilic β-blockers, water-soluble β- and their stimulation results in positive chronotropic and
blockers such as atenolol and nadolol demonstrate less inotropic effects. β2 and β3 ARs are present mostly in the
gastrointestinal absorption, depend mostly on renal excretion, vasculature, but also in the myocardium. Stimulation of β2
and rely less on hepatic metabolism.20 Therefore, typically ARs leads to vasodilation, increased heart rate, and contrac-
have longer half-lives and maintain more stable plasma tility, whereas stimulation of β3 ARs results in vasodilation
concentrations.21 Finally, ultra-short-acting intravenous β- and a negative inotropic effect.22
blocker metabolism also may depend on red blood cell cytosol β1 selective agents, such as acebutolol, betaxolol, bisoprolol,
esterases (esmolol) or pseudocholinesterase and hepatic esmolol, atenolol, nebivolol, and metoprolol, have the theore-
esterases (landiolol) (Table 2). tical advantage over nonselective agents since their use should
reduce the risk for inhibition of β2-mediated cerebral vasodila-
Pharmacodynamics of β-blockers tion, bronchodilation, and metabolic effects. However, at
higher doses, β1 selective agents may lose their receptor
The observed differences in cardiovascular effects among selectivity, which combined with anemia in the perioperative
different β-blockers are due predominantly to varying setting could increase the risk for cardiovascular complications
β-receptor selectivity, but they also are due to unique and stroke23 (Table 3). Finally, nebivolol is the only β-blocker

Fig 1. Presynaptic nerve terminal, cardiac adrenergic receptors, and adrenergic-receptor signaling in the heart. Localization of the common polymorphisms of the
adrenergic receptors with their functional consequences. A coupling of the β2 adrenergic receptor to stimulatory G protein (Gs) and inhibitory G protein (Gi) is
shown in the cardiomyocytes but not in the presynaptic nerve terminal. AC, adenyl cyclase; AR, adrenergic receptor; cAMP, cyclic adenosine monophosphate; Epi,
epinephrine; GRK5, G protein-coupled-receptor kinase; Nepi, norepinephrine (Adapted with permission from Kertai et al. J Cardiothorac Vasc Anesth
2012;26:1101-14).

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
4 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

Table 3
Pharmacodynamic Properties of β-Blockers.

β-Blocker Relative β1Selectivity β1/β2 Affinity Ratio β3 ISA

Acebutolol þ 2.4 - þ
Atenolol þþ 4.7 - -
Betaxolol þþ 6.8 - -
Bisoprolol þþ 13.5 - -
Carvedilol - - -
Esmolol þþ 33 - -
Labetalol - - þ
Landiolol þþþ 255 - -
Metoprolol þþ 2.3 - - Fig 2. Cytochrome P-450 2D6 (CYP2D6) dependency of β-blockers. NOTE:
Nadolol - - - Extensive metabolizers are subjects with 2 copies of normal activity alleles of
Nebivolol þþ 46 þ - CYP2D6—no need for β-blocker dose change. Intermediate metabolizers are
Pindolol - - þþ patient subjects with only 1 copy of normal activity alleles of CYP2D6—β-
Propranolol - - - blocker dose reduction should be considered. Poor metabolizers are patients
without any copy of normal activity alleles of CYP2D6—further β-blocker
Abbreviations: ISA, intrinsic sympathomimetic activity. dose reduction should be considered. Ultrafast metabolizers are patients with
more than 2 copies of normal activity alleles of CYP2D6—higher β-blocker
dose should be considered.
with known β3-receptor agonist activity, which is beneficial in
patients with heart failure.24
dependent on CYP2D6 isoenzyme for its biotransformation.
β-blockers With Vasodilating Properties In fact, 70% to 80% of metoprolol metabolism is dependent on
this isoenzyme.32
Certain β-blockers, such as pindolol and carteolol, also have Previous studies indicated that subjects who were CYP2D6
partial intrinsic sympathomimetic activity, and their use is poor metabolizers, that is, subjects without any functional
associated with a decrease in peripheral vascular resistance and alleles of the CYP2D6 gene, had 3- to 10-fold higher
less heart rate reduction when compared with β-blockers metoprolol plasma concentrations compared to subjects who
without intrinsic sympathomimetic activity.1 Further, other β- were extensive metabolizers (2 functional alleles of the
blockers such as labetalol and carvedilol also block the alpha CYP2D6 gene resulting in a normal CYP2D6 isoenzyme
receptors in addition to β ARs, resulting in vasodilation and activity). Predictably, in poor metabolizers, the elimination
maintenance of the cardiac output.25 Additionally, nebivolol half-life of metoprolol increased to 7.5 hours as opposed to
has a direct vasodilating effect due to direct secretion of nitric 2.5 hours in extensive (normal) metabolizers.32,33 This
oxide, thought to confer myocardial protection26 (Table 3). decrease in metoprolol clearance and subsequent drug accu-
mulation in poor metabolizers has been shown previously to
Pharmacogenetics of β-blockers translate into significantly more pronounced and prolonged
decreases in heart rate during exercise.34 Several further
It is clear that genetic differences could explain some of the studies corroborated that poor metabolizers on metoprolol
interpatient variability in clinical responses to different β- had a significantly higher risk for bradycardia.35,36 In contrast,
blockers.27 These genetic differences could influence both the in a study of patients with acute myocardial infarction,
pharmacokinetic and the pharmacodynamic properties of β- ultrarapid metabolizers started on metoprolol had lower trough
blockers. One of the most extensively studied and best metoprolol plasma concentrations, higher mean heart rates,
characterized pharmacogenetic variations is the cytochrome and a higher incidence of ventricular rhythm disturbance
(CYP) P450 (CYP2D6) isoenzyme, which is involved in the compared with patients with other CYP2D6 genotypes.37
hepatic biotransformation of several of the lipophilic β- The findings of these studies suggest that CYP2D6 poly-
blockers (eg, metoprolol, propranolol, carvedilol, labetalol, morphisms significantly influence β-blocker pharmacokinetics,
and timolol). CYP2D6 is highly polymorphic, and at least 105 in particular with regard to metoprolol.
functional variants have been identified (www.pharmvar.org/ However, in the ambulatory setting, the potential impact of
htdocs/archive/cyp2d6.htm; accessed April 14, 2018).28 Based genotyping for these pharmacokinetic genetic variants prior to
on the presence of polymorphisms and the number of long-term cardiovascular risk management is diminished, since
functional alleles, patients can be stratified as ultrarapid in practice there is sufficient time to titrate β-blockers.38
metabolizers, extensive metabolizers (the normal phenotype), In contrast, studies on how CYP2D6 polymorphisms could
intermediate metabolizers, and poor metabolizers29(Fig 2). influence the efficacy of lipophilic β-blockers in preventing or
Further, the prevalence of different CYP2D6 enzyme poly- treating perioperative cardiovascular complications are com-
morphisms show a great variation by ethnicity.30,31 pletely lacking. As such, Badgett et al. recently proposed
Metoprolol, the most frequently prescribed β-blocker for several hypotheses about why some clinical trials were unable
either primary or secondary prevention of cardiovascular to demonstrate a beneficial effect of perioperative β-blockers in
complications, also happens to be the β-blocker most preventing cardiovascular complications.39 In their meta-

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 5

analysis of clinical trials in which patients received CYP2D6- discontinued from extended-release metoprolol. Moreover, in
dependent β-blockers in the perioperative period, where the patients with uncomplicated hypertension, perioperative treat-
drug titration period was limited, an increased risk for ment with β-blockers may be associated with an increased risk
mortality was observed. Furthermore, an interaction analysis for perioperative MACEs and all-cause mortality.45
between the duration of titration period and CYP2D6 meta- In a study of 140 patients undergoing vascular surgery,
bolism indicated that the highest mortality was observed in withdrawal of β-blockers was associated with increased odds
clinical trials that administered β-blockers dependent on for cardiovascular mortality and postoperative myocardial
CYP2D6 metabolism over a short titration period. infarction (odds ratio [OR] 17.7, 95% confidence interval
Thus, these findings support the concept that the efficacy of [CI], 2.6-113; p ¼ 0.003).46 Another trial on 711 patients
β-blockers metabolized via the CYP2D6 isoenzyme is affected undergoing vascular surgery indicated a higher 1-year mortal-
by relevant polymorphisms with potential implications for the ity in patients who had β-blockers discontinued before surgery
perioperative use of β-blockers. Indeed, when there is an as compared to nonusers (OR 2.7, 95% CI, 1.2-5.9; p o
indication for preoperative initiation of β-blockers, adminis- 0.001).47 In another cohort of 38,779 surgical patients
tration of β-blockers that do not require a long titration period (including cardiac and vascular surgical procedures), disconti-
and do not rely on hepatic biotransformation, such as atenolol, nuation of β-blockers was associated with an increase for 30-
should be considered.39 day (OR 3.93; 95% CI, 2.57-6.01; p o 0.0001) and 1-year
Several genetic variations involving the β1 AR have been mortality (OR 1.96; 95% CI, 1.49-2.58; p o 0.0001).48
described, and some of these are prevalent and potentially Similarly, data from 8,431 patients undergoing colorectal
could influence the efficacy and safety of β-blockers40–42 and bariatric procedures showed an increased risk for com-
(Table 4). However, as indicated in Table 4, the majority of bined adverse events at 30 days, 90 days, and 1 year in
studies evaluating the role of these polymorphisms are lacking patients with a higher cardiac risk profile who were discon-
a sufficient level of scientific evidence to meet the expectations tinued preoperatively from β-blockers.49 Recently, a large
for personalized perioperative β-blocker use in patients with an retrospective study in 19,145 patients demonstrated that with-
elevated risk for cardiovascular complications and mortality. drawal of β-blockers was associated with increased risk for
These studies were mostly underpowered and were conducted mortality within 48 hours after noncardiac surgery. However,
in ambulatory patients, and the replication of positive results β-blocker withdrawal also was associated with a decreased risk
still is lacking. Also, the majority of these studies only for need of vasopressor use during the early postoperative
examined associations between individual genetic polymorph- period and resulted in a shorter stay in the post-anesthesia care
isms and cardiovascular risk or long-term cardiovascular event unit. Although these studies were retrospective in nature, they
survival, and not with treatment response to β-blockers. provided important supporting evidence for not withdrawing
Therefore, it remains unknown how the effects of these genetic β-blockers in the perioperative period in patients who were on
polymorphisms may influence clinical therapeutic responses to chronic β-blocker therapy.50 In addition, the findings of these
β-blockers with different pharmacologic properties. studies also highlighted the potential risk for hemodynamic
perturbations (hypotension requiring use of vasopressors) and
Perioperative Use of β-blockers an increase in utilization of health care resources (ie, extended
post-anesthesia care) when β-blockers are continued periopera-
Patients With Chronic β-blocker Use tively.51 These specific concerns about the consequences of the
withdrawal of β-blockers were reflected in the latest 2014
Perioperative continuation of β-blockers in patients on ACC/AHA Guideline on Perioperative Cardiovascular Evalua-
chronic β-blockers for various cardiovascular indications (eg, tion and Management of Patients Undergoing Noncardiac
coronary artery disease, AF, hypertension) is a matter of less Surgery, which has a class I recommendation for continuing
controversy than compared to the initiation of therapy pre- perioperative β-blocker therapy in patients with chronic β-
operatively. Chronic β-blocker therapy leads to upregulation of blocker use.15 However, the current guidelines do not provide
β AR, therefore abrupt cessation of therapy leads to an recommendations as to how to modify regimens or potentially
increased number of unoccupied β AR being stimulated by discontinue perioperative β-blocker therapy when clinical
endogenous catecholamines. Withdrawal of β-blockers is circumstances dictate (eg, anticipated significant surgical
known to increase the risk for reflex tachycardia and hyperten- blood loss, or preoperative hypotension/bradycardia).
sion, with a potential for oxygen supply/demand mismatch due
to a shortened diastolic coronary perfusion time.43 This, in Preoperative Initiation of Preoperative β-blocker Use
turn, could lead to cardiac ischemia, subsequently increasing
the risk for perioperative cardiovascular events, including Patients Undergoing Noncardiac Surgery
mortality. Discontinuation of chronic β-blocker use in the
perioperative period or nonadherence to the perioperative Patients undergoing noncardiac surgery, especially patients
medication instructions is particularly concerning in patients with 1 or more RCRI risk factors, can be at increased risk for
taking agents with a shorter (metoprolol, propranolol) rather perioperative myocardial ischemia and infarction. Although
than a longer half-life (atenolol).44 To date, there are no studies results from previous small-scale trials on preoperative initia-
with data on risk of cardiovascular events in patients who were tion of atenolol and bisoprolol had led the committee of the
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
6
Table 4
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045

Common single nucleotide polymorphisms of the adrenergic receptors and their intracellular signaling system with their functional consequences.

Adrenergic Gene Physiologic response SNP Allele frequency Functional effect Clinical studies
receptor or location
signaling

β1 AR 10q24-26  Expressed in the  Amino acid  Whites, 15%-22%  Ser49 resistance to receptor downregulation
myocardium substitution of  Blacks, 13%-22%
 Positive inotropic and serine (Ser) by
chronotropic effect glycine (Gly) at
residue 49

A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]
(Ser49Gly)
 Arginine (Arg)  Whites, 27%  Altered G-protein coupling  Swiss BB in Spinal Anesthesia Study: 1) Arg389
by glycine  Blacks, 42%  Arg389 genotype: 2-fold greater basal and 3-fold genotype, lower incidence of cardiovascular morbidity
(Gly) at residue greater agonist-mediated adenyl cyclase activities compared to Gly389 genotype98; 2) selective use of
389  Higher resting heart rate, diastolic blood pressure in bisoprolol had no differential effect in patients with the
(Arg389Gly) Arg389 homozygous subjects Arg389 or the Gly389 genotype compared with
 placebo98
Gluy389 genotype: a loss-of-function variant, atte-
nuated β1 AR signaling cascade  Parvez et al99 in ambulatory patients with atrial
fibrillation: 1) Gly389 genotype better response to rate
control therapy (BBs and calcium channel blockers)
compared to Arg389 genotype
 Liu et al,100 Gly389 variant increased risk for heart
failure in East Asians, but decreased risk for whites;
patients with Arg389 variant showed a better response
to BB use
β2 AR 5q31-q32  Expressed in the  Arginine to  Gly16 and  Gly16 and Glu27, enhanced effect on agonist-  Gly16
myocardium and blood glycine at Glu27, 40%- promoted receptor downregulation  Case-control studies in patients ischemic heart dis-
vessels residue 16 50% in whites  Ile164, defective receptor coupling to the stimulatory ease101,102: no association with cardiovascular mor-
 Mediates smooth mus- (Arg16Gly) and blacks G protein, impaired agonist-promoted sequestration, bidity and mortality
cle cell relaxation,  Glutamine to  Ile164, 1% in and lower agonist binding affinity, impaired vasodi-  No interaction with BB use on the risk of myocar-
increased contractility, glutamic acid at white and lation, increased peripheral vascular resistance dial infarction and ischemic stroke102
heart rate, antiapopto- residue 27 o2% in blacks  Swiss BB in Spinal Anesthesia Study98 intraopera-
tic effects (Gln27Glu) tive hypotension independent of bisoprolol use
 Threonine to  Patients with acute coronary syndrome,103 the
isoleucine at Gly16 variant compared to the Arg16 variant was
residue 164 associated with a lower long-term mortality in
(Thr164Ile) BB users
 Gln27
 Lower risk for heart failure, cardiac transplantation
and death from heart failure in patients with dilated
cardiomyopathy104
 Hypertensive patients, the Gln27 variant in the
presence of the Gly16 variant, a higher systolic
blood pressure, but no increased risk for myocardial
infarction105
 Patients with acute coronary syndrome,103 the
Gln27 variant was associated with a higher long-
term mortality in BB users; haplotypes of Arg16 and
Gln27, highest risk for mortality
 Ile164
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045

 Thomsen et al,106 in women this variant was


associated with the severity of blood pressure
elevation, the frequency of hypertension, increased
cardiovascular risk
 Liggett et al,107 in patients with heart failure: a
higher risk for death or cardiac transplantation
 Littlejohn et al,108 in patients with heart failure: no
association with death, but BB use negatively
impacted survival in the Ile164 group
 Piscione et al,109 in patients with percutaneous

A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]
coronary artery intervention: earlier onset of cor-
onary artery disease, multivessel disease, higher
incidence of myocardial infarction, repeat percuta-
neous coronary intervention, and cardiac mortality
β2 AR 8p11-p12  Heart and blood vessels  Tryptophan to  Whites, 8%  Candelore et al,110 no difference in agonist-binding  Zafarmand et al, [Ref64] in patients with coronary
 Smooth muscle and arginine at  Blacks, 12% characteristics between the Arg64 and Trp64 variants artery disease: no evidence of Arg64 in acute
endothelial cell residue 64  Mexican  Pietri-Rouxel et al,111 Arg64 variant is associated myocardial infarction
mediated vasodilation (Trp64Arg) Americans, 13% with attenuated agonist-promoted coupling, reduced  Nebivolol use in heart failure patients,112 a selective β1
 In the cardiomyocytes,  Pima intracellular cyclic adenosine monophosphate AR antagonist with β3 AR agonist properties was
a negative inotropic, Indians, 31% accumulation associated with improved survival
positive lusitropic, and
antihypertrophic
effects
GRK5 10q26.11-  Myocardium  Glutamine to  Whites, 1%-2.4%  Leu41 more effective in blunting the effects of β AR  Liggett et al,113 Leu41 variant protected against early
26.11  β AR desensitization leucine at  Blacks, 23%-35% agonists through enhanced desensitization113 death and cardiac transplantation in blacks with heart
through phosphorylation residue 41 Chinese, 29%-35%  Leu41 protective effect similar to BB use113 failure
of the receptor (Gln41Leu)  Cresci et al,114 Leu41 variant is associated with
 rs4752292 improved survival in blacks with heart failure
 rs11198893  Kang S et al,115 Leu41 variant was not found in a
Chinese population. Arg389 no association with sys-
tolic heart failure related morbidity
 4 intronic SNPs  Whites, 9%-26%  Kertai et al,86 minor alleles of these intronic SNPs were
associated with postoperative atrial fibrillation despite
perioperative BB use in patients who underwent
coronary artery bypass grafting surgery
GNAS 20q13-  Encodes the stimulatory  Haplotype pairs,  Whites, 11.3%  Increased heart rate and cardiac contractility117  Frey et al,118 improved survival after coronary artery
q32 G protein-alpha-subunit G(-1211) and  Blacks15.2%  Enhanced expression and activity of the G protein- bypass grafting surgery
 Myocardium T2291C116  Chinese, 48% alpha-subunit  Frey et al,116 haplotype in the GNAS gene associated
 rs7121  Whites, 51%
with stimulatory G protein-alpha-subunit expression
 rs12481583  Whites 37%
with altered intraoperative hemodynamics in patients
with coronary artery bypass grafting surgery; in
patients with chronic BB use cardiac performance
was dependent on stimulatory G protein-alpha-subunit
expression
 Wieneke H et al,119 increased risk for ventricular
tachycardia in patients with implantable cardioverter-
defibrillator

7
8
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045

Table 5
Guideline-Based and Best-Practice Approach Recommendations for Perioperative Use of β-Blockers.

Surgery Perioperative β- ACC/AHA Guideline-Based Best-Practice Approach


Type Blocker Status Recommendations Recommendations (Evidence-Based) (Not Evidence-Based)

Noncardiac On chronic β Continue perioperatively.15 Dose adjustment or temporary discontinuation suggested if systolic blood Transition from short-acting metoprolol to atenolol (less risk of
and blockade pressure o100, heart rate o50, or high-risk bleeding procedures51,70,95 withdrawal).
cardiac
Noncardiac Not on Consider starting β-blocker for  High-risk patients with known ischemia on stress testing before high-  Oral β-blocker regimen needs to be slowly titrated and started

A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]
β-blocker only specific conditions.15 risk surgery (ie, vascular) 41 week prior to surgery.
preoperatively  Intermediate or high-risk patients ( 42-3 RCRI criteria) before high-  Metoprolol may need to be avoided or dose reduced in
risk surgery
 Patients with clear indications for long-term β blockade (systolic heart patients with liver disease or known genetic varia-
failure/myocardial infarction)
tionsaffecting metabolism and response.
 Atenolol may be avoided or dose reduced in patients with renal
disease.
 Carvedilol, extended-release metoprolol, or bisoprolol may be
preferred in patients with heart failure.96
 Atenolol or extended-release metoprolol may be preferred after
myocardial infarction.97
Cardiac Not on β- Start oral β-blocker before  Carvedilol may be the preferred oral agent to prevent AF.89
blocker CABG/valve surgery to  In urgent/emergent procedures, intravenous β-blockers may be
preoperatively prevent AF84. considered (may worsen hemodynamics).
 Landiolol, an ultra-short- acting, intravenous β-blocker, may have
a safer profile and is efficacious in preventing postoperative AF.

Abbreviations: ACC/AHA, American College of Cardiology/American Heart Association.


A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 9

2002 ACC/AHA guidelines for preoperative risk assessment studies on the efficacy and safety of perioperative β-blocker
and management to recommend the initiation of preoperative use by conducting meta-analyses. The most recent meta-
β-blocker therapy in patients not already on chronic β- analysis by Bouri and colleagues analyzed all randomized
blockers, subsequent trials and observational studies chal- trials (excluding the DECREASE trials) and found that
lenged that practice given efficacy and safety concerns initiation of β-blockers before surgery was associated with
associated with the initiation of β-blocker therapy in the an increased risk for hypotension (RR, 1.51; 95% CI, 1.37-
perioperative period.10–12,14 1.67; p o 0.00001), stroke (RR, 1.73; 95% CI, 1.00-2.99;
The Metoprolol after Vascular Surgery trial was the first p ¼ 0.05), and 30-day all-cause mortality (RR 1.27; 95% CI,
study that challenged those previous findings on the potential 1.01-1.6; p ¼ 0.04). In contrast, β-blocker use was associated
beneficial effect of β-blockers, recruiting 494 patients under- with a reduced risk for nonfatal myocardial infarction (RR,
going major vascular surgery. These patients were randomized 0.73; 95% CI, 0.61-0.88; p ¼ 0.001).56
to receive metoprolol versus placebo, starting 2 hours pre- There are several factors that may explain some of the
operatively until hospital discharge, for a maximum of 5 days differences in the efficacy and safety of β-blockers reported by
postoperatively. The results of the trial indicated that pre- these prior clinical trials and observational studies:
operative metoprolol was not effective in reducing 30-day or
6-month adverse cardiac events (nonfatal myocardial infarc- 1. Initiation of β-blockers immediately prior to the day of
tion, unstable angina, new congestive heart failure, new atrial surgery is associated with a higher risk for adverse events
or ventricular dysrhythmia requiring treatment, or cardiac and mortality: In support of this concern, a recent meta-
death). At the same time, patients receiving metoprolol had a analysis that included 16 randomized control trials with a
higher incidence of bradycardia and hypotension.52 total number of 12,043 patients examined the risk asso-
The Diabetic Postoperative Mortality and Morbidity trial, ciated with perioperative β-blocker use that was initiated
which included 921 patients with diabetes mellitus undergoing less than a day before noncardiac surgery.57 The results of
major nonvascular surgery, found that metoprolol started the study showed that perioperative β-blocker use initiated
before the day of surgery and continued for 8 days post- less than a day before noncardiac surgery was associated
operatively was not associated with a significant reduction in with a decreased risk for nonfatal myocardial infarction
the risk for mortality, but its use again was associated with (RR, 0.69; 95% CI, 0.58-0.82; p o 0.001), but it was
higher incidences of hypotension and bradycardia.53 associated with a higher risk for hypotension (RR, 1.47;
Metoprolol administrated twice daily initiated at the time of 95% CI, 1.34-1.60; p o 0.001), bradycardia (RR, 2.61;
admission and continued until 7 days postoperatively did not 95% CI, 2.18-3.12; p o 0.001), and nonfatal stroke (RR,
provide any mortality benefit in the Perioperative β Blockade 1.79; 95% CI, 1.09-2.95; p ¼ 0.02). When a sensitivity
trial that was conducted in 103 patients who underwent subanalysis was performed by excluding the DECREASE
infrarenal vascular surgery.54 trials, the results of the analysis indicated that perioperative
In contrast, a retrospective study of 3,062 patients who β-blocker use initiated less than a day before noncardiac
underwent vascular surgery demonstrated that a combination surgery also was associated with an increased risk for all-
of β-blocker (agent not specified) and statin use was associated cause mortality (RR, 1.30; 95% CI, 1.03-1.64; p ¼ 0.03).
with a 33% reduction in mortality at 2 years after surgery However, there is a paucity of data on how far in
(relative risk [RR] 0.33, p ¼ 0.0106).55 advance of noncardiac surgery perioperative β-blocker
These conflicting results about the efficacy and safety of therapy should be initiated to enable safe titration in an
preoperative β-blocker use for the prevention of cardiovascular effort to minimize these risks. Flu and colleagues, in a study
complications and mortality led the investigators of the of 940 vascular surgery patients, studied the association
Perioperative Ischemic Evaluation group to design and con- between 3 different regimens of perioperative β-blocker
duct a large-scale clinical trial (POISE) in patients who were initiation (started 0 to 1 weeks prior, 4 1 to 4 weeks prior,
undergoing noncardiac surgery. In the trial, 8,351 patients and 44 weeks prior to surgery) and 30-day cardiac events
were assigned randomly to perioperative β-blocker use (200 and long-term mortality.58 When β-blocker use was
mg of extended-dose metoprolol started 2-4 hours before initiated 4 1 to 4 weeks or 4 4 weeks compared to
surgery, continued for 30 days postoperatively) or placebo.14 treatment initiated o1 week prior to vascular surgery, its
The results of the trial showed that metoprolol use was use was associated with a lower incidence of 30-day cardiac
associated with a reduced risk for myocardial infarction, but events (OR 0.46, 95% CI, 0.27-0.76; and OR 0.48; 95% CI,
it also was associated with a higher risk for hypotension, 0.29-0.79) and long-term mortality (OR 0.52, 95% CI,
bradycardia, stroke, and mortality. Of note, metoprolol in this 0.21-0.67 and OR 0.50, 95% CI, 0.25-0.71). Based on the
trial was started in a higher dose, on the day of surgery, which findings of this and prior studies, recommendations on the
accounted for higher rates of bradycardia and hypotension, and timing of initiation of perioperative β-blocker therapy
a higher risk for stroke and mortality in patients randomized to before noncardiac surgery was revised in the 2014 ACC/
metoprolol. AHA guidelines for preoperative cardiac risk assessment
Following the publication of the findings of the POISE trial, and management, indicating that initiation of β-blocker use
several investigators attempted to address some of the sig- on the day of surgery is not recommended (class III).15
nificant limitations of the individual trials and observational More recently, these recommendations were corroborated

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
10 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

further by the findings of Chen and colleagues, who AHA guidelines for preoperative risk assessment and manage-
observed in a nationwide population-based cohort of ment for noncardiac surgery recommend careful perioperative
patients with diabetes mellitus who underwent noncardiac titration of β-blockers (class IIB).15
surgery that patients who were taking β-blockers for more Despite the current evidence pointing toward beneficial
than 30 days prior to noncardiac surgery were at lower risk effects of β-blockers only in the high-risk patients, it is plausible
for mortality compared to patients who were started on β- that the beneficial effects of lowering MACE may extend to the
blockers less than 30 days prior to noncardiac surgery.59 lower-risk patients. The incidence of myocardial injury after
2. Not all patient populations undergoing noncardiac surgery surgery is 8% in a nonselected, surgical population of 445
may benefit from perioperative β-blocker use: Patients with years of age, well above the number of troponin measurements
long-term indications for β-blocker use for either primary or performed. However, due to the practical restrictions of
secondary cardiovascular prevention, or high-risk patients measuring troponin T levels to detect only in symptomatic or
as defined by the RCRI criteria ( 4 3 risk factors), may high-risk patients, myocardial injury after surgery events likely
represent a particular subset of patients.50,60 The issue of are underdiagnosed. It is possible that a benefit of perioperative
perioperative β-blocker use in these patients has been β blockade could be detected with the widespread use of
addressed in several studies. In the DECREASE I trial, troponin measurements postoperatively. However, at present,
the most pronounced benefit from perioperative β-blocker these events would be difficult to detect under most of the
use was observed in a subset of patients who were at current observational studies.
intermediate risk for perioperative myocardial infarction 3. The efficacy and safety of perioperative β-blockers may be
and mortality.61 In a large retrospective study of 782,969 related to the type of β-blocker used: Interestingly, the trials
patients undergoing major noncardiac surgery, the relation- that were able to demonstrate a beneficial effect for
ship between perioperative β-blocker use and risk of death perioperative β-blockers in the prevention of cardiac
varied directly with cardiac risk.62 Among the 580,665 complications and mortality used atenolol or bisoprolol,
patients with an RCRI score of 0 to 1, treatment was whereas studies that used metoprolol demonstrated a great
associated with no benefit or possible harm. In contrast, variability in efficacy and safety.10,11,66 As such, Redelme-
among patients with an RCRI score of 2,3,4, or more, the ier and colleagues in a large-scale observational study of
adjusted ORs for in-hospital death were 0.88 (95% CI, 0.80 37,151 patients who either received perioperative atenolol
to 0.98), 0.71 (95% CI, 0.63 to 0.80), and 0.58 (95% CI, or metoprolol found that the incidence of myocardial
0.50 to 0.67), respectively.62 infarction or death was significantly lower in patients who
In another large-scale observational study of 28,263 received atenolol compared to patients who received
patients undergoing noncardiac surgery, patients with a metoprolol.44 Similarly, Wallace and colleagues observed
history of stable ischemic heart disease benefited from in a cohort of 38,779 patients who underwent noncardiac
perioperative β-blocker use in terms of an overall reduction and cardiac surgery that patients who received atenolol
in both MACE and 30-day mortality.63 Of note, the most compared to patients who received metoprolol had a
pronounced benefit from perioperative β-blocker use was significantly lower incidence of 30-day mortality (1% v
observed in a subset of patients with heart failure.63 3%) and 1-year mortality (7% v 13%).67 London and
In a more recent meta-analysis that included studies with a colleagues also noted a significant association of type of
total of 55,138 VA patients (undergoing noncardiac surgical β-blocker with stroke in a cohort of 55,128 patients
procedures including vascular surgery), it was observed that undergoing major noncardiac surgery (atenolol v metopro-
patients with more than 2 RCRI risk factors who received β- lol, OR 0.63, 95% CI, 0.45-0.89; p ¼ 0.008).64
blockers prior to noncardiac surgery were at a lower risk for Recently, Badgett and colleagues published an elegant
nonfatal Q-wave infarction or cardiac arrest (RR, 0.67; 95% CI, summary of the pharmacologic properties of β-blockers that
0.57-0.79; p o 0.001) and 30-day mortality (RR, 0.73; 95% could explain the often contradictory findings of observa-
CI, 0.65-0.83; p o 0.001) compared to patients who did not tional studies and clinical trials on the efficacy and safety of
receive β-blockers prior to noncardiac surgery.64 perioperative β-blocker use.39 According to their summary,
Similarly, in a retrospective observational study of 314,114 genetic variations in CYP2D6, the timing of initiation of
patients undergoing noncardiac surgery, it was found that perioperative β-blocker use, and the degree of selectivity of
perioperative β-blocker use was associated with a reduced 30- different β-blockers for β1/β2 ARs could explain some of
day risk for mortality in patients with 3 to 4 RCRI criteria (OR the variability in efficacy and safety of perioperative β-
0.63, 95% CI, 0.43-0.93).65 In the same study, perioperative β- blockers for the prevention of perioperative cardiac com-
blocker use in patients with 1 to 2 RCRI criteria was not plications and mortality. Indeed, these observations suggest
associated with a significant reduction in postoperative mortal- that metoprolol, which has the least β1 AR selectivity and
ity, whereas in patients without cardiac risk factors, periopera- also is metabolized extensively by the highly polymorphic
tive β-blocker use was associated with a significantly higher CYP2D6 (and therefore requires more prolonged and
risk for mortality (OR 1.19, 95% CI, 1.06-1.35).65 individualized dose titration both for safety and adequacy
Consequently, in patients with a long-term indication for β- of effect), may not be an ideal β-blocker for use in the
blocker use (eg, ischemia identified on preoperative stress perioperative setting.39 On the other hand, while a lower
testing or patients with 43 RCRI criteria), the 2014 ACC/ reduction in resting heart rate may be obtained with
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 11

atenolol (-19 beats per minute, 95% CI, -20.6 to -17.6) preoperative β-blocker use remained consistent in the majority
versus metoprolol (-13.2 beats per minute, 95% CI, -14.7 to of patient subgroups, including patients of advanced age;
-11.7), an appropriate titration period may decrease the women; and patients with chronic lung disease, diabetes, or
variability in response and the choice of β-blocker may not moderately depressed ventricular function. In contrast, an
matter.39,68 elevated mortality risk, albeit statistically nonsignificant, was
4. The perioperative use of some β-blockers may confer a observed in patients with β-blocker use who had a left
higher risk for postoperative stroke, which could offset ventricular ejection fraction of less than 30% (OR 1.13, 95%
their beneficial effects in the prevention of cardiac compli- CI, 0.96-1.33, p ¼ 0.23).72 Nevertheless, the findings of this
cations and mortality. A meta-analysis by Bangalore and study resulted in a recommendation for preoperative β-blocker
colleagues found an increased risk of nonfatal stroke, use, which eventually also became a national quality metric for
hypotension, and bradycardia with the use of β-blockers patients undergoing CABG surgery.16,73
in patients undergoing noncardiac surgery, with the POISE In contrast, subsequent large-scale observational studies
trial carrying the greatest weight in the analysis.14,69 were not able to validate the benefit of perioperative β- blocker
Although there is an association between β-blocker use, use in patients undergoing CABG surgery. A retrospective
hypotension, and stroke, this cannot be assumed to be a study of 12,855 patients undergoing CABG surgery found that
direct cause-and-effect relationship. It is possible that acute preoperative β-blocker use was not associated with a decreased
anemia precipitated by surgical bleeding also could con- risk for morbidity and mortality. Nevertheless, a small benefit
tribute to the risk of stroke, as patients on β-blockers cannot was detected as far as the rates of deep sternal wound infection
compensate for a significant drop in hematocrit (30%- (0.3% v 0.5%, p ¼ 0.032), pneumonia (1.9% v 2.4%, p ¼
35%).70 0.039), and intraoperative blood usage (37.2% v 34.1%, p o
In a single-center study of 44,092 consecutive patients 0.001) compared with patients who did not receive preopera-
who underwent noncardiac, non-neurologic surgery, the use tive β-blockers.73
of bisoprolol was associated with a lower risk for post- Similarly, a retrospective analysis of 43,747 patients from
operative stroke compared to metoprolol or atenolol use the Society of Thoracic Surgeons Adult Cardiac Surgery
(OR 0.2, 95% CI, 0.04-0.99). The investigators suggested Database observed that after risk adjustment, preoperative β-
that the higher risk for postoperative stroke with metoprolol blocker use was not associated with a reduced risk for
or atenolol use was due to these β-blockers being less mortality, morbidity, length of stay (LOS), or hospital read-
selective for β1 ARs and their effect on β2 ARs impairing mission, but patients who received preoperative β-blockers had
cerebral vasodilation.23 Mashour and colleagues also found greater intraoperative blood usage.74 Of note, the lack of
in 57,218 consecutive patients who underwent noncardiac benefit of perioperative β-blockers in this study led the
surgery that routine use of preoperative metoprolol (OR investigators to challenge their use as a quality metric in
4.2, 95% CI, 2.2-8.1, p o 0.001) compared to atenolol was patients with CABG surgery.74
associated with a higher risk for postoperative stroke after Further, another recent large-scale retrospective analysis of the
noncardiac surgery. Further, intraoperative use of metopro- Society of Thoracic Surgeons Adult Cardiac Surgery Database of
lol (OR 3.3, 95% CI, 1.4-7.8, p ¼ 0.003) compared to 506,110 patients, who were free from recent myocardial infarction
esmolol or labetalol was associated with an increased risk and underwent CABG surgery, found that preoperative β-blocker
for perioperative stroke.71 use was not associated with a lower risk for operative mortality
(OR 0.96, 95% CI, 0.87-1.06, p ¼ 0.38), permanent stroke (OR
Patients Undergoing Cardiac Surgery 0.99, 95% CI, 0.89-1.10, p ¼ 0.81), prolonged ventilation (OR
1.02, 95% CI, 0.98-1.07, p ¼ 0.26), any reoperation (OR 0.97,
Perioperative β-blocker use in patients undergoing cardiac 95% CI, 0.92-1.03, p ¼ 0.35), renal failure (OR 1.04, 95% CI,
surgery is similarly controversial since β-blockers may affect 0.97-1.11, p ¼ 0.3), or deep sternal wound infection (OR 0.86,
certain outcomes (eg, AF) more than others (eg, mortality). 95% CI, 0.71-1.04, p ¼ 0.12 ) compared to patients without
Initial evidence from an observational study showed a small preoperative β-blocker use.75
but consistent survival benefit in patients who received β- A recent observational study corroborated the findings of
blockers prior to CABG surgery. Patients who received these previous studies and also found in 4,076 patients who
preoperative β-blockers had a lower incidence of mortality underwent elective CABG surgery, CABG surgery with valve
(2.8% v 3.4%, OR 0.80, 95% CI, 0.78-0.82; p o 0.001). surgery, or valve surgery that preoperative β-blocker use was
Unadjusted morbidity rates in patients who received β- not associated with a lower risk for postoperative delirium,
blockers compared to patients who did not receive β- stages I and III of acute kidney injury, stroke, AF, mortality, or
blockers were also significantly lower for stroke, 1.6% versus prolonged LOS.76
1.7% (OR 0.93, 95% CI, 0.89-0.96); prolonged ventilation, In summary, as proposed in the section on perioperative β-
5.6% versus 6.6% (OR 0.85, 95% CI, 0.83-0.87); reoperation, blocker use in noncardiac surgery, the different pharmacologic
4.9% versus 5.3% (OR 0.94, 95% CI, 0.92-0.96); and renal properties of β-blockers, such as their dependence on CYP2D6
failure, 3.4% versus 3.8% (OR 0.87, 95% CI, 0.85-0.89); but metabolism, genetic variations in β1 ARs, and receptor
not for deep sternal wound infection, 0.62% versus 0.64% (OR selectivity, could explain some of the heterogeneities in the
0.98, 95% CI, 0.92-1.04).72 The beneficial effect of outcomes reported in these studies.
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
12 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

In support of these observations, a retrospective study of protein-coupled receptor kinase 5 was associated with post-
5,248 patients who underwent urgent or elective CABG operative AF despite perioperative β-blocker therapy.86 A
surgery demonstrated that among these patients, preoperative more recent genome-wide association study identified genetic
use of β-blockers not dependent on CYP2D6 biotransforma- variation in the lymphocyte 96 antigen receptor, an essential
tion (OR 0.33, 95% CI, 0.13-0.83, p ¼ 0.02), but not coreceptor of toll-like receptor 4 signaling, which plays a role
preoperative use of β-blockers dependent on CYP2D6 bio- in initiating the innate immune response to cardiac surgery, to
transformation (OR 0.44, 95% CI, 0.16-1.07, p ¼ 0.06), was be associated with a decreased risk for postoperative AF.78
associated with a decreased risk of operative mortality.77 These latter findings indicated not only that innate immune
One of the most frequent complications after cardiac surgery responses play a central role in the development of post-
is postoperative AF, which occurs in 25% to 40% of operative AF, but also why β-blockers in a subset of patients
patients.78 This potentially life-threatening complication is undergoing cardiac surgery may not prevent postoperative AF.
associated with increased risks for adverse neurologic events, This study illustrates the importance of mechanisms unrelated
congestive heart failure, myocardial infarction, perioperative to β AR signaling as potential targets for therapeutic interven-
mortality, prolonged LOS, and increased hospital costs.79 tion in AF after cardiac surgery. Although there are no
Sympathetic activation or an exaggerated response to adrener- pharmacogenomic studies with a specific focus on the phar-
gic stimulation is an important trigger for postoperative AF.80 macokinetics of β-blockers in the prevention of postoperative
β-Blockers are a mainstay in the prevention and treatment of AF, when orally administered carvedilol (less dependent on
postoperative AF.81 CYP2D6 biotransformation) was compared to metoprolol
Indeed, an earlier meta-analysis of 58 studies that included a (highly dependent on CYP2D6 biotransformation), patients
total of 8,565 patients who underwent cardiac surgery also who received carvedilol had a significantly lower risk of
found that β-blocker use was associated with the greatest postoperative AF than patients who received metoprolol (OR
reduction in the risk of postoperative AF.82 Based on these and 0.50, 95% CI, 0.32-0.80, I2 ¼ 15%).89
earlier findings, the 2011 American College of Cardiology Intravenous β-blockers also have been compared to orally
Foundation/AHA Guideline for Coronary Artery Bypass Graft administered β-blockers in the prevention of AF after cardiac
Surgery recommends perioperative β-blockers to prevent post- surgery. In a small study of 50 CABG surgery patients,
operative AF after CABG surgery, a class I designated intravenous esmolol compared to orally administered meto-
recommendation.16 prolol was less well tolerated hemodynamically, and its
A more recent meta-analysis of 33 trials that included a total administration provided no additional benefit in preventing
of 4,698 patients who underwent cardiac surgery reconfirmed postoperative AF.90 Another moderate-size study of 240
the findings of the previous meta-analysis and supported the patients undergoing CABG surgery, aortic valve replacement,
2011 American College of Cardiology Foundation/AHA or combined aortic valve replacement and CABG surgery
Guideline for Coronary Artery Bypass Graft Surgery recom- randomized patients to either a 48-hour infusion of metoprolol
mendations. This study demonstrated that β-blocker use was or orally administered metoprolol (both regimens titrated to
associated with a significant reduction in the risk for post- heart rate) started on the morning of the first postoperative day.
operative AF compared to placebo (OR 0.33, 95% CI, 0.26- The results of the study showed that intravenously adminis-
0.43, I2 ¼ 55%).83 tered metoprolol was more effective than orally administered
A systematic review and meta-analysis of 5 randomized metoprolol in the prevention of postoperative AF.91 More
controlled trials with a total number of 688 patients also recently, the introduction of landiolol (an ultra-short-acting
demonstrated that preoperative β-blocker administration prior selective intravenous β-blocker) in Japan and Europe led to
to elective cardiac surgery was associated with a significant several investigators studying the efficacy of landiolol in the
reduction in the risk for postoperative AF (OR 0.43, 95% CI, prevention of AF after cardiac surgery. Similar to esmolol,
0.30-0.61, I2 ¼ 0%) without significantly increasing the risk landiolol is administered intravenously and has a short half-life
of ischemic stroke, nonfatal myocardial infarction, overall of 4 minutes, but compared to esmolol, it is highly selective
mortality, or LOS.84 for β1 ARs (esmolol 33  selective v landiolol 233 
Nevertheless, approximately 20% of patients undergoing selective), which makes landiolol a potentially ideal intrave-
cardiac surgical procedures such as CABG surgery develop nous β-blocker for rapid intraoperative and postoperative
postoperative AF despite β-blocker use.81,82 Several patient- titration and prevention of postoperative AF.92 However, since
specific and procedure-related risk factors associated with landiolol is ultra-short-acting, ideally a combination with a
postoperative AF despite β-blocker use have been identified longer-acting β-blocker should be considered. In this respect,
in cardiac surgery patients.79,85,86 As the authors described in Sezai and colleagues recently showed that intravenous land-
more detail above, genes coding for β ARs and hepatic iolol in combination with oral bisoprolol was more effective in
metabolism of several β-blockers are highly polymorphic, preventing postoperative AF than intravenous landiolol
and thus potentially could affect pharmacodynamic and alone.93 A subsequent meta-analysis of 6 trials that included
pharmacokinetic responses of β-blockers.80,87,88 These poly- a total of 560 patients concluded that intravenous landiolol was
morphisms affect the risk for cardiovascular complications associated with a significant reduction in the incidence of
during β-blocker therapy.36,88 Indeed, a candidate-gene study postoperative AF after cardiac surgery (OR 0.26, 95% CI,
in CABG surgery patients found that genetic variation in G 0.17-0.40, p o 0.0001).94
Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 13

Conclusion undergoing vascular surgery. Dutch Echocardiographic Cardiac Risk


Evaluation Applying Stress Echocardiography Study Group. N Engl J
Med 1999;341:1789–94.
In summary, supporting evidence for the perioperative use
12 Eagle KA, Berger PB, Calkins H, et al. ACC/AHA guideline update for
of β-blockers to decrease perioperative morbidity and mortality perioperative cardiovascular evaluation for noncardiac surgery—executive
remains inconclusive. Over the past 2 decades, a plethora of summary: A report of the American College of Cardiology/American Heart
studies on perioperative β-blocker use, both in patients already Association Task Force on Practice Guidelines (Committee to Update the
on β-blockers and in patients who are candidates for β-blocker 1996 Guidelines on Perioperative Cardiovascular Evaluation for Noncar-
diac Surgery). Circulation 2002;105:1257–67.
therapy, have thus far failed to provide definitive answers
13 Durham J, Mackey WC. Perioperative β-blockade in noncardiac surgery: A
regarding the type of β-blocker, timing of drug initiation, and cautionary tale of over-reliance on small randomized prospective trials.
effective yet safe dose adjustments. The evidence that emerged Clin Ther 2016;38:2302–16.
from these studies suggests that when it comes to perioperative 14 Devereaux PJ, Yang H, Yusuf S, et al. Effects of extended-release
β-blocker use, one type and dose of β-blocker will not fit all metoprolol succinate in patients undergoing non-cardiac surgery (POISE
patients. As reviewed above, several studies suggest that an trial): A randomised controlled trial. Lancet 2008;371:1839–47.
15 Fleisher LA, Fleischmann KE, Auerbach AD, et al. 2014 ACC/AHA
individualized approach to perioperative β-blocker use should guideline on perioperative cardiovascular evaluation and management of
be adopted (Table 5). However, studies that investigated the patients undergoing noncardiac surgery: A report of the American College
impact of individualized perioperative β-blocker protocols in of Cardiology/American Heart Association Task Force on practice guide-
the prevention of cardiac complications and mortality after lines. J Am Coll Cardiol 2014;64:e77–137.
surgery are still lacking. Until such protocols are validated and 16 Hillis LD, Smith PK, Anderson JL, et al. 2011 ACCF/AHA Guideline for
Coronary Artery Bypass Graft Surgery: Executive summary: A report of
become available, a rational approach to perioperative β- the American College of Cardiology Foundation/American Heart Associa-
blocker use should take into account several factors, based tion Task Force on Practice Guidelines. Circulation 2011;124:2610–42.
on information from conventional observational studies, ran- 17 Kristensen SD, Knuuti J, Saraste A, et al. 2014 ESC/ESA Guidelines on
domized clinical trials, and expert opinion, including clinical- non-cardiac surgery: Cardiovascular assessment and management: The
Joint Task Force on non-cardiac surgery: Cardiovascular assessment and
and procedure-related factors, impact on desired outcomes,
management of the European Society of Cardiology (ESC) and the European
and genetic variations with potential influence on pharmaco- Society of Anaesthesiology (ESA). Eur J Anaesthesiol 2014;31:517–73.
kinetics and pharmacodynamics of specific β-blockers. 18 Kirchheiner J, Heesch C, Bauer S, et al. Impact of the ultrarapid
metabolizer genotype of cytochrome P450 2D6 on metoprolol pharmaco-
kinetics and pharmacodynamics. Clin Pharmacol Ther 2004;76:302–12.
References 19 Lefebvre J, Poirier L, Poirier P, et al. The influence of CYP2D6 phenotype
on the clinical response of nebivolol in patients with essential hyperten-
1 Frishman WH. β-Adrenergic blockade in cardiovascular disease. J Cardi- sion. Br J Clin Pharmacol 2007;63:575–82.
ovasc Pharmacol Ther 2013;18:310–9. 20 Mann SJ. Redefining beta-blocker use in hypertension: Selecting the right
2 Teo KK, Yusuf S, Furberg CD. Effects of prophylactic antiarrhythmic drug beta-blocker and the right patient. J Am Soc Hypertens 2017;11:54–65.
therapy in acute myocardial infarction. An overview of results from 21 Frishman WH, Saunders E. β-adrenergic blockers. J Clin Hypertens
randomized controlled trials. JAMA 1993;270:1589–95. 2011;13:649–53.
3 Brophy JM, Joseph L, Rouleau JL. Beta-blockers in congestive heart 22 Gauthier C, Rozec B, Manoury B, et al. Beta-3 adrenoceptors as new
failure. A Bayesian meta-analysis. Ann Intern Med 2001;134:550–60. therapeutic targets for cardiovascular pathologies. Curr Heart Fail Rep
4 Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ 2011;8:184–92.
ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Pre- 23 Ashes C, Judelman S, Wijeysundera DN, et al. Selective β1-antagonism
vention, Detection, Evaluation, and Management of High Blood Pressure with bisoprolol is associated with fewer postoperative strokes than atenolol
in Adults: A Report of the American College of Cardiology/American or metoprolol: A single-center cohort study of 44,092 consecutive patients.
Heart Association Task Force on Clinical Practice Guidelines. Hyperten- Anesthesiology 2013;119:777–87.
sion 2017. [epub ahead of print]. 24 Cannavo A, Koch WJ. Targeting β3-adrenergic receptors in the heart:
5 Bangalore S, Steg G, Deedwania P, et al. β-Blocker use and clinical Selective agonism and β-blockade. J Cardiovasc Pharmacol 2017;69:71–8.
outcomes in stable outpatients with and without coronary artery disease. 25 Frishman WH. Cardiovascular pharmacotherapeutics, 3rd ed. Minneapolis,
JAMA 2012;308:1340–9. MN: Cardiotext Inc; 57–86.
6 Abbasi J. Do all patients need β-blockers after a heart attack?. JAMA 26 Mangrella M, Rossi F, Fici F. Pharmacology of nebivolol. Pharmacol Res
2018;319:853–5. 1998;38:419–31.
7 Minor DS, Meyer AM, Long RC, et al. β-Blockers and chronic obstructive 27 Johnson JA, Cavallari LH, Beitelshees AL, et al. Pharmacogenomics:
pulmonary disease: Inappropriate avoidance?. J Clin Hypertens 2013;15: Application to the management of cardiovascular disease. Clin Pharmacol
925–30. Ther 2011;90:519–31.
8 Malerba M, Montuschi P, Radaeli A, et al. Role of beta-blockers in 28 Available at: 〈http://www.pharmvar.org/htdocs/archive/cyp2d6.htm〉.
patients with COPD: Current perspective. Drug Discov Today 2015;20: Accessed April 11, 2018.
129–35. 29 Zhou SF. Polymorphism of human cytochrome P450 2D6 and its clinical
9 Steinman MA, Zullo AR, Lee Y, et al. Association of β-blockers with significance: Part I. Clin Pharmacokinet 2009;48:689–723.
functional outcomes, death, and rehospitalization in older nursing home 30 Ingelman-Sundberg M, Sim SC, Gomez A, et al. Influence of cytochrome
residents after acute myocardial infarction. JAMA Intern Med 2017;177: P450 polymorphisms on drug therapies: Pharmacogenetic, pharmacoepi-
254–62. genetic and clinical aspects. Pharmacol Ther 2007;116:496–526.
10 Mangano DT, Layug EL, Wallace A, et al. Effect of atenolol on mortality 31 Aklillu E, Persson I, Bertilsson L, et al. Frequent distribution of ultrarapid
and cardiovascular morbidity after noncardiac surgery. Multicenter Study metabolizers of debrisoquine in an Ethiopian population carrying dupli-
of Perioperative Ischemia Research Group. N Engl J Med 1996;335: cated and multiduplicated functional CYP2D6 alleles. J Pharmacol Exp
1713–20. Ther 1996;278:441–6.
11 Poldermans D, Boersma E, Bax JJ, et al. The effect of bisoprolol on 32 Shin J, Johnson JA. Pharmacogenetics of beta-blockers. Pharmacotherapy
perioperative mortality and myocardial infarction in high-risk patients 2007;27:874–87.

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
14 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

33 Kertai MD, Fontes M, Podgoreanu MV. Pharmacogenomics of β-blockers 55 Barrett TW, Mori M, De Boer D. Association of ambulatory use of statins
and statins: Possible implications for perioperative cardiac complications. J and beta-blockers with long-term mortality after vascular surgery. J Hosp
Cardiothorac Vasc Anesth 2012;26:1101–14. Med 2007;2:241–52.
34 Lennard MS, Silas JH, Freestone S, et al. Oxidation phenotype — a major 56 Bouri S, Shun-Shin MJ, Cole GD, et al. Meta-analysis of secure
determinant of metoprolol metabolism and response. N Engl J Med randomised controlled trials of β-blockade to prevent perioperative death
1982;307:1558–60. in non-cardiac surgery. Heart 2014;100:456–64.
35 Bijl MJ, Visser LE, van Schaik RH, et al. Genetic variation in the CYP2D6 57 Wijeysundera DN, Duncan D, Nkonde-Price C, et al. Perioperative beta
gene is associated with a lower heart rate and blood pressure in beta- blockade in noncardiac surgery: A systematic review for the 2014 ACC/
blocker users. Clin Pharmacol Ther 2009;85:45–50. AHA guideline on perioperative cardiovascular evaluation and manage-
36 Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6 genotype on ment of patients undergoing noncardiac surgery: A report of the American
the clinical effects of metoprolol: A prospective longitudinal study. Clin College of Cardiology/American Heart Association Task Force on practice
Pharmacol Ther 2009;85:269–72. guidelines. J Am Coll Cardiol 2014;64:2406–25.
37 Goryachkina K, Burbello A, Boldueva S, et al. CYP2D6 is a major 58 Flu WJ, van Kuijk JP, Chonchol M, et al. Timing of pre-operative beta-
determinant of metoprolol disposition and effects in hospitalized Russian blocker treatment in vascular surgery patients: Influence on post-operative
patients treated for acute myocardial infarction. Eur J Clin Pharmacol outcome. J Am Coll Cardiol 2010;56:1922–9.
2008;64:1163–73. 59 Chen RJ, Chu H, Tsai LW. Impact of beta-blocker initiation timing on
38 Fux R, Mörike K, Pröhmer AM, et al. Impact of CYP2D6 genotype on mortality risk in patients with diabetes mellitus undergoing noncardiac
adverse effects during treatment with metoprolol: A prospective clinical surgery: A nationwide population-based cohort study. J Am Heart Assoc
study. Clin Pharmacol Ther 2005;78:378–87. 2017;6. [epub ahead of print].
39 Badgett RG, Lawrence VA, Cohn SL. Variations in pharmacology of beta- 60 Lee TH, Marcantonio ER, Mangione CM, et al. Derivation and prospective
blockers may contribute to heterogeneous results in trials of perioperative validation of a simple index for prediction of cardiac risk of major
beta-blockade. Anesthesiology 2010;113:585–92. noncardiac surgery. Circulation 1999;100:1043–9.
40 Mason DA, Moore JD, Green SA, et al. A gain-of-function polymorphism 61 Boersma E, Poldermans D, Bax JJ, et al. Predictors of cardiac events after
in a G-protein coupling domain of the human beta1-adrenergic receptor. J major vascular surgery: Role of clinical characteristics, dobutamine
Biol Chem 1999;274:12670–4. echocardiography, and beta-blocker therapy. JAMA 2001;285:1865–73.
41 Rathz DA, Brown KM, Kramer LA, et al. Amino acid 49 polymorphisms 62 Lindenauer PK, Pekow P, Wang K, et al. Perioperative beta-blocker therapy and
of the human beta1-adrenergic receptor affect agonist-promoted trafficking. mortality after major noncardiac surgery. N Engl J Med 2005;353:349–61.
J Cardiovasc Pharmacol 2002;39:155–60. 63 Andersson C, Mérie C, Jørgensen M, et al. Association of β-blocker
42 Sandilands A, Yeo G, Brown MJ, et al. Functional responses of human therapy with risks of adverse cardiovascular events and deaths in patients
beta1 adrenoceptors with defined haplotypes for the common 389R G and with ischemic heart disease undergoing noncardiac surgery: A Danish
49S G polymorphisms. Pharmacogenetics 2004;14:343–9. nationwide cohort study. JAMA Intern Med 2014;174:336–44.
43 Frishman WH. Beta-adrenergic blocker withdrawal. Am J Cardiol 64 London MJ, Hur K, Schwartz GG, et al. Association of perioperative β-
1987;59:26–32F. blockade with mortality and cardiovascular morbidity following major
44 Redelmeier D, Scales D, Kopp A. Beta blockers for elective surgery in elderly noncardiac surgery. JAMA 2013;309:1704–13.
patients: Population based, retrospective cohort study. BMJ 2005;331:932. 65 Friedell ML, Van Way CW, Freyberg RW, et al. β-Blockade and operative
45 Jørgensen ME, Hlatky MA, Køber L, et al. β-Blocker-associated risks in mortality in noncardiac surgery: Harmful or helpful?. JAMA Surg 2015;150:
patients with uncomplicated hypertension undergoing noncardiac surgery. 658–63.
JAMA Intern Med 2015;175:1923–31. 66 Beattie WS, Wijeysundera DN, Karkouti K, et al. Does tight heart rate
46 Shammash JB, Trost JC, Gold JM, et al. Perioperative beta-blocker control improve beta-blocker efficacy? An updated analysis of the
withdrawal and mortality in vascular surgical patients. Am Heart J noncardiac surgical randomized trials. Anesth Analg 2008;106:1039–48.
2001;141:148–53. 67 Wallace AW, Au S, Cason BA. Perioperative β-blockade: Atenolol is
47 Hoeks SE, Scholte Op Reimer WJ, van Urk H, et al. Increase of 1-year associated with reduced mortality when compared to metoprolol. Anesthe-
mortality after perioperative beta-blocker withdrawal in endovascular and siology 2011;114:824–36.
vascular surgery patients. Eur J Vasc Endovasc Surg 2007;33:13–9. 68 Cucherat M, Borer JS. Reduction of resting heart rate with antianginal
48 Wallace AW, Au S, Cason BA. Association of the pattern of use of drugs: Review and meta-analysis. Am J Ther 2012;19:269–80.
perioperative β-blockade and postoperative mortality. Anesthesiology 69 Bangalore S, Wetterslev J, Pranesh S, et al. Perioperative beta blockers in
2010;113:794–805. patients having non-cardiac surgery: A meta-analysis. Lancet 2008;372:
49 Kwon S, Thompson R, Florence M, et al. β-blocker continuation after 1962–76.
noncardiac surgery: A report from the surgical care and outcomes 70 Beattie WS, Wijeysundera DN, Karkouti K, et al. Acute surgical anemia
assessment program. Arch Surg 2012;147:467–73. influences the cardioprotective effects of beta-blockade: A single-center,
50 Karam D, Arora R. Perioperative β-blockers in patients undergoing propensity-matched cohort study. Anesthesiology 2010;112:25–33.
noncardiac surgery-Scientific misconduct and clinical guidelines. Am J 71 Mashour GA, Sharifpour M, Freundlich RE, et al. Perioperative metoprolol and
Ther 2017;24:e435–41. risk of stroke after noncardiac surgery. Anesthesiology 2013;119:1340–6.
51 Kertai MD, Cooter M, Pollard RJ, et al. Is compliance with Surgical Care 72 Ferguson TB, Coombs LP, Peterson ED, et al. Preoperative beta-blocker
Improvement Project Cardiac (SCIP-Card-2) measures for perioperative β- use and mortality and morbidity following CABG surgery in North
blockers associated with reduced incidence of mortality and America. JAMA 2002;287:2221–7.
cardiovascular-related critical quality indicators after noncardiac surgery?. 73 Brinkman WT, Herbert MA, Prince SL, et al. Preoperative beta-blocker
Anesth Analg 2017. [epub ahead of print]. usage: Is it really worthy of being a quality indicator?. Ann Thorac Surg
52 Yang H, Raymer K, Butler R, et al. The effects of perioperative beta- 2011;92:788–95. discussion 795-6.
blockade: Results of the Metoprolol after Vascular Surgery (MaVS) study, 74 LaPar DJ, Crosby IK, Kron IL, et al. Preoperative beta-blocker use should
a randomized controlled trial. Am Heart J 2006;152:983–90. not be a quality metric for coronary artery bypass grafting. Ann Thorac
53 Juul AB, Wetterslev J, Gluud C, et al. Effect of perioperative beta Surg 2013;96:1539–44. discussion 1544-5.
blockade in patients with diabetes undergoing major non-cardiac surgery: 75 Brinkman W, Herbert MA, O’Brien S, et al. Preoperative β-blocker use in
Randomised placebo controlled, blinded multicentre trial. BMJ 2006;332: coronary artery bypass grafting surgery: National database analysis. JAMA
1482. Intern Med 2014;174:1320–7.
54 Brady AR, Gibbs JS, Greenhalgh RM, et al. Perioperative beta-blockade 76 O’Neal JB, Billings FT, Liu X, et al. Effect of preoperative beta-blocker
(POBBLE) for patients undergoing infrarenal vascular surgery: Results of use on outcomes following cardiac surgery. Am J Cardiol 2017;120:
a randomized double-blind controlled trial. J Vasc Surg 2005;41:602–9. 1293–7.

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]] 15

77 Kertai MD, Esper SA, Akushevich I, et al. Preoperative CYP2D6 Myocardial Infarction: A report of the American College of Cardiology/
metabolism-dependent β-blocker use and mortality after coronary artery American Heart Association Task Force on Practice Guidelines (Writing
bypass grafting surgery. J Thorac Cardiovasc Surg 2014;147: Committee to Revise the 2002 Guidelines for the Management of
1368–75. e1363. Patients With Unstable Angina/Non-ST-Elevation Myocardial Infarction):
78 Kertai MD, Li YJ, Ji Y, et al. Genome-wide association study of new-onset Developed in collaboration with the American College of Emergency
atrial fibrillation after coronary artery bypass grafting surgery. Am Heart J Physicians, the Society for Cardiovascular Angiography and Interventions,
2015;170:580–90. e528. and the Society of Thoracic Surgeons: Endorsed by the American
79 Mathew JP, Fontes ML, Tudor IC, et al. A multicenter risk index for atrial Association of Cardiovascular and Pulmonary Rehabilitation and the
fibrillation after cardiac surgery. JAMA 2004;291:1720–9. Society for Academic Emergency Medicine. J Am Coll Cardiol 2007;50:
80 Parvez B, Chopra N, Rowan S, et al. A common β1-adrenergic receptor e1–157.
polymorphism predicts favorable response to rate-control therapy in atrial 98 Zaugg M, Bestmann L, Wacker J, et al. Adrenergic receptor genotype but not
fibrillation. J Am Coll Cardiol 2012;59:49–56. perioperative bisoprolol therapy may determine cardiovascular outcome in at-
81 Fuster V, Rydén LE, Cannom DS, et al. 2011 ACCF/AHA/HRS focused risk patients undergoing surgery with spinal block: the Swiss Beta Blocker in
updates incorporated into the ACC/AHA/ESC 2006 guidelines for the Spinal Anesthesia (BBSA) study: a double-blinded, placebo-controlled, multi-
management of patients with atrial fibrillation: A report of the American center trial with 1-year follow-up. Anesthesiology. 2007;107:33–44.
College of Cardiology Foundation/American Heart Association Task Force 99 Parvez B, Chopra N, Rowan S, et al. A common β1-adrenergic receptor
on practice guidelines. Circulation 2011;123:e269–367. polymorphism predicts favorable response to rate-control therapy in atrial
82 Crystal E, Garfinkle MS, Connolly SS, et al. Interventions for preventing fibrillation. J Am Coll Cardiol 2012;59:49–56.
post-operative atrial fibrillation in patients undergoing heart surgery. 100 Liu WN, Fu KL, Gao HY, et al. β1 adrenergic receptor polymorphisms
Cochrane Database Syst Rev 2004;1. CD003611. and heart failure: a meta-analysis on susceptibility, response to β-blocker
83 Arsenault KA, Yusuf AM, Crystal E, et al. Interventions for preventing therapy and prognosis. PLoS One 2012;7:e37659.
post-operative atrial fibrillation in patients undergoing heart surgery. 101 Tseng ZH, Aouizerat BE, Pawlikowska L, et al. Common beta-adrenergic
Cochrane Database Syst Rev 2013;1. CD003611. receptor polymorphisms are not associated with risk of sudden cardiac death in
84 Thein PM, White K, Banker K, et al. Preoperative use of oral beta- patients with coronary artery disease. Heart Rhythm. 2008;5:814–21.
adrenergic blocking agents and the incidence of new-onset atrial fibrillation 102 Lemaitre RN, Heckbert SR, Sotoodehnia N, et al. beta1- and beta2-
after cardiac surgery. A systematic review and meta-analysis. Heart Lung adrenergic receptor gene variation, beta-blocker use and risk of myocar-
Circ 2018;27:310–21. dial infarction and stroke. Am J Hypertens 2008;21:290–6.
85 Kertai MD, Qi W, Li YJ, et al. Gene signatures of postoperative atrial 103 Lanfear DE, Jones PG, Marsh S, et al. Beta2-adrenergic receptor
fibrillation in atrial tissue after coronary artery bypass grafting surgery in genotype and survival among patients receiving beta-blocker therapy
patients receiving β-blockers. J Mol Cell Cardiol 2016;92:109–15. after an acute coronary syndrome. JAMA. 2005;294:1526–33.
86 Kertai MD, Li YW, Li YJ, et al. G protein-coupled receptor kinase 5 gene 104 Forleo C, Resta N, Sorrentino S, et al. Association of beta-adrenergic
polymorphisms are associated with postoperative atrial fibrillation after receptor polymorphisms and progression to heart failure in patients with
coronary artery bypass grafting in patients receiving β-blockers. Circ idiopathic dilated cardiomyopathy. Am J Med 2004;117:451–8.
Cardiovasc Genet 2014;7:625–33. 105 Wallerstedt SM, Eriksson AL, Ohlsson C, et al. Haplotype association analysis
87 Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism that inhibits of the polymorphisms Arg16Gly and Gln27Glu of the adrenergic beta2
beta-adrenergic receptor signaling is protective in heart failure. Nat Med receptor in a Swedish hypertensive population. J Hum Hypertens. 19 2005:
2008;14:510–7. 705–8.
88 Johnson JA, Liggett SB. Cardiovascular pharmacogenomics of adrenergic 106 Thomsen M, Dahl M, Tybjaerg-Hansen A, et al. β2 -adrenergic receptor
receptor signaling: Clinical implications and future directions. Clin Thr164IIe polymorphism, blood pressure and ischaemic heart disease in
Pharmacol Ther 2011;89:366–78. 66 750 individuals. J Intern Med 2012;271:305–14.
89 DiNicolantonio JJ, Beavers CJ, Menezes AR, et al. Meta-analysis 107 Liggett SB, Wagoner LE, Craft LL, et al. The Ile164 beta2-adrenergic
comparing carvedilol versus metoprolol for the prevention of postoperative receptor polymorphism adversely affects the outcome of congestive heart
atrial fibrillation following coronary artery bypass grafting. Am J Cardiol failure. J Clin Invest 1998;102:1534–9.
2014;113:565–9. 108 Littlejohn MD, Palmer BR, Richards AM, et al. Ile164 variant of beta2-
90 Balcetyte-Harris N, Tamis JE, Homel P, et al. Randomized study of early adrenoceptor does not influence outcome in heart failure but may interact
intravenous esmolol versus oral beta-blockers in preventing post-CABG with beta blocker treatment. Eur J Heart Fail 2008;10:55–9.
atrial fibrillation in high risk patients identified by signal-averaged ECG: 109 Piscione F, Iaccarino G, Galasso G, et al. Effects of Ile164 polymorphism
Results of a pilot study. Ann Noninvasive Electrocardiol 2002;7:86–91. of beta2-adrenergic receptor gene on coronary artery disease. J Am Coll
91 Halonen J, Hakala T, Auvinen T, et al. Intravenous administration of Cardiol 2008;52:1381–8.
metoprolol is more effective than oral administration in the prevention of 110 Candelore MR, Deng L, Tota LM, et al. Pharmacological characterization
atrial fibrillation after cardiac surgery. Circulation 2006;114:I1–4. of a recently described human beta 3-adrenergic receptor mutant.
92 Oprea AD. Do we need another short-acting beta-blocker? A definite Endocrinology. 1996;137:2638–41.
maybe…. J Clin Anesth 2017;39:96–7. 111 Piétri-Rouxel F, St John Manning B, Gros J, et al. The biochemical effect
93 Sezai A, Nakai T, Hata M, et al. Feasibility of landiolol and bisoprolol for of the naturally occurring Trp64–4Arg mutation on human beta3-
prevention of atrial fibrillation after coronary artery bypass grafting: A adrenoceptor activity. Eur J Biochem 1997;247:1174–9.
pilot study. J Thorac Cardiovasc Surg 2012;144:1241–8. 112 van Veldhuisen DJ, Cohen-Solal A, Böhm M, et al. Beta-blockade with
94 Sakamoto A, Hamasaki T, Kitakaze M. Perioperative landiolol adminis- nebivolol in elderly heart failure patients with impaired and preserved left
tration reduces atrial fibrillation after cardiac surgery: A meta-analysis of ventricular ejection fraction: Data From SENIORS (Study of Effects of
randomized controlled trials. Adv Ther 2014;31:440–50. Nebivolol Intervention on Outcomes and Rehospitalization in Seniors
95 Feringa HH, Bax JJ, Boersma E, et al. High-dose beta-blockers and tight With Heart Failure). J Am Coll Cardiol 2009;53:2150–8.
heart rate control reduce myocardial ischemia and troponin T release in 113 Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism that inhibits
vascular surgery patients. Circulation 2006;114:I344–9. beta-adrenergic receptor signaling is protective in heart failure. Nat Med.
96 Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for 2008;14:510–7.
the management of heart failure: A report of the American College of 114 Cresci S, Kelly RJ, Cappola TP, et al. Clinical and genetic modifiers of
Cardiology Foundation/American Heart Association Task Force on Prac- long-term survival in heart failure. J Am Coll Cardiol 2009;54:432–44.
tice Guidelines. J Am Coll Cardiol 2013;62:e147–239. 115 Kang S, Hong X, Ruan CW, et al. Effects of GRK5 and ADRB1
97 Anderson JL, Adams CD, Antman EM, et al. ACC/AHA 2007 Guidelines polymorphisms influence on systolic heart failure. J Transl Med 2015;
for the Management of Patients With Unstable Angina/Non-ST-elevation 13:44.

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045
16 A.D. Oprea et al. / Journal of Cardiothoracic and Vascular Anesthesia ] (]]]]) ]]]–]]]

116 Frey UH, Adamzik M, Kottenberg-Assenmacher E, et al. 118 Frey UH, Kottenberg E, Kamler M, et al. Genetic interactions in the β-
A novel functional haplotype in the human GNAS gene alters adrenoceptor/G-protein signal transduction pathway and survival after coronary
Galphas expression, responsiveness to beta-adrenoceptor stimulation, artery bypass grafting: a pilot study. Br J Anaesth 2011;107:869–78.
and peri-operative cardiac performance. Eur Heart J 2009;30: 119 Wieneke H, Svendsen JH, Lande J, et al. Polymorphisms in the GNAS
1402–10. Gene as Predictors of Ventricular Tachyarrhythmias and Sudden Cardiac
117 Iwase M, Uechi M, Vatner DE, et al. Cardiomyopathy induced by cardiac Death: Results From the DISCOVERY Trial and Oregon Sudden
Gs alpha overexpression. Am J Physiol 1997;272:H585–589. Unexpected Death Study e 003905. J Am Heart Assoc 2016;5.

Please cite this article as: Oprea AD, et al. (2018), http://dx.doi.org/10.1053/j.jvca.2018.04.045

You might also like