Abstract
β-Blockers (metoprolol, bisoprolol, and carvedilol) are a cornerstone of heart failure (HF) treatment. However, it is well recognized that responses to a β-blocker are variable among patients with HF. Numerous studies now suggest that genetic polymorphisms may contribute to variability in responses to a β-blocker, including left ventricular ejection fraction improvement, survival, and hospitalization due to HF exacerbation. This review summarizes the pharmacogenetic data for β-blockers in patients with HF and discusses the potential implications of β-blocker pharmacogenetics for HF patients.
Keywords: Pharmacogenetics, β-Blockers, Heart failure, Metoprolol, ADRB1, ADRB2
Introduction
Heart failure (HF) is characterized by neurohormonal activation of the sympathetic nervous system (SNS) and renin–angiotensin–aldosterone system in response to a drop in cardiac output [1]. Excessive activation of these systems results in hemodynamic alterations such as systemic vasoconstriction and increases in sodium/water retention, and ventricular remodeling, the latter of which is primarily responsible for the progressive nature of the disease.
It was in the 1980s that the important role of neurohormonal activation in HF was first documented [2], with evidence supporting the role of β-blockers emerging in the 1990s [3]. Historically, β-blockers were contraindicated in patients with HF due to their negative inotropic effect. However, as the neurohormonal paradigm of HF was appreciated, there was increasing interest in the β-blockers. β-Blockers are competitive antagonists at the β-adrenergic receptors (β-AR), thereby reducing the level of SNS activation [4]. From mid-1990s to early 2000s, several landmark studies demonstrated that the β-blockers metoprolol, bisoprolol, and carvedilol reduce morbidity and mortality compared to placebo, when added to standard background therapy that included angiotensin–converting enzyme (ACE) inhibitor therapy (Table 1). Based on these trial data, these β-blockers are recommended by the consensus HF management guidelines as essential therapy for patients who have systolic HF and lack contraindications to β-blocker therapy [8].
Table 1.
Trial name | Treatments | Population | N | Primary outcomes |
Findings |
P-value for PO |
References |
---|---|---|---|---|---|---|---|
COPERNICUS | Carvedilol versus placebo | Severe HF | 2,289 | Death | Carvedilol ↓risk of PO by 35%, and ↓risk of death or hospitalization by 24% | 0.0014 | [2] |
CIBIS-II | Bisoprolol versus Placebo | NYHA III–IV | 2,647 | All-cause mortality | Bisoprolol ↓risk of PO by 34% | <0.0001 | [3] |
MERIT-HF | Metoprolol CR/XL versus Placebo | NYHA II–IV | 3,991 | Composite of death and hospitalization | Metoprolol ↓risk of PO by 19%, and ↓ risk of death or heart transplantation by 32% | <0.001 | [5] |
COMET | Carvedilol versus Metoprolol CR/XL | NYHA II–IV | 3,029 | All-cause mortality | Carvedilol ↓risk of PO by 17% | 0.0017 | [6] |
BEST | Bucindolol versus Placebo | NYHA III–IV | 2,708 | All-cause mortality | Early termination of the trial due to no significant difference between the treatments. However, CV death rate was lower in bucindolol group than placebo (HR, 0.86; 95% CI 0.74–0.99) | ns | [7] |
COPERNICUS, The Carvedilol Prospective Randomized Cumulative Survival; CIBIS-II, The Cardiac Insufficiency Bisoprolol Study II; MERIT-HF, The Metoprolol CR/XL Randomized Intervention Trial in Congestive Heart Failure; COMET, Carvedilol or Metoprolol European Trial; BEST, The Beta-blocker Evaluation of Survival Trial; CR/XL, controlled release/extended release; HF, heart failure; NYHA, New York Heart Association functional class; N, sample size; ↓, decreased; PO, primary outcomes; CV, cardiovascular; ns, not significant
However, not all β-blockers have shown beneficial effects in HF patients [8]. In addition, there is variability in responses to β-blockers among HF patients. β-Blocker therapy requires a careful monitoring and slow dose titration to minimize adverse events from the drug [9]. Despite careful titration, approximately 25% of patients require discontinuation of therapy due to intolerance to the drug [10]. Additionally, morbidity and mortality rates in symptomatic HF patients remain high [11], therefore, identifying causes of response variability to β-blockers may help optimize β-blocker therapy and HF regimens. Genetic differences may contribute to this variability in response to β-blockers. Pharmacogenetics is the study of genetic contributions to variable drug response, with the clinical potential to optimize therapy by identifying (predicting) the patients who will respond well (or poorly) to a given drug or are at high risk for adverse events from the drug.
The purpose of this review is to summarize the β-blocker pharmacogenetics literature in HF and to discuss the potential clinical implications of these data. Studies were identified in the Medline database from 1966 to January 2007 by combining MeSH search terms ‘heart failure, congestive, genetic polymorphism, ‘single nucleotide polymorphism,’ ‘pharmacogenetics,’ ‘adrenergic beta antagonists’ as well as individual β-blocker used for HF. We also checked the references of all identified papers. Only studies that tested pharmacogenetic hypotheses on β-blockers in HF or provided analyses data in a subgroup of patients who were on β-blockers were included in this review.
Genetic polymorphisms of β-AR
The pharmacological effects of β-blockers derive from their ability to antagonize the β-ARs. Thus, the genes for these receptors have been a primary focus in β-blocker pharmacogenetic studies. In the cardiovascular system, there are two β-ARs that β-blockers can antagonize: β1-AR and β2-AR, both of which are members of the G-protein coupled receptors superfamily. The β1-AR gene (ADRB1), consisting of 2,860 bp, is located in chromosome 10q24–26. It encodes a 51.3 kDa protein, with 477 amino acid residues. β1-ARs are primarily found in the heart, controlling contractility, and heart rate. β1-ARs are also expressed in kidney, vasculature, and adipose tissues. β2-ARs are more widely distributed including expression in the heart, vasculature, respiratory smooth muscle, kidney, adipose tissue and brain, among others. The gene consists of one exon with 2,033 bp located at chromosome 5q31–q32. The receptors play an important role in the regulation of a variety of the systems such as cardiac, vascular, respiratory, and endocrine systems. β-Blockers used for the treatment of HF possess different receptor specificity: Metoprolol and bisoprolol are β1-selective, while carvedilol is a non-selective β-blocker with additional antagonist effects at the α1-AR (Table 2).
Table 2.
Name | β-blocking property |
α1-blocking property |
Elimination routea | References |
---|---|---|---|---|
Carvedilol | Nonselective | Yes | Oral bioavailability: About 25–35% | [4, 12–15] |
Both CYP2D6 and CYP2C9 are major metabolizing enzymes | ||||
CYP2D6 is responsible for 30–50% of metabolism of carvedilol | ||||
CYP2C9 is responsible for 5–20% of metabolism of carvedilol | ||||
Metoprolol | β1-selective | No | Oral bioavailability: | [4, 12, 16, 17] |
Immediate release form: About 50% | ||||
Controlled release form: About 25–30% | ||||
CYP2D6 is a major hepatic enzyme responsible for 60–70% of metoprolol metabolism. | ||||
Bisoprolol | β1-selective | No | Oral bioavailability: 80–90% | [4, 12, 18] |
Approximately 50% of oral dose is recovered as parent drug in urine | ||||
Bucindolol | Nonselective | Yes but weak | Oral bioavailability: About 30% | [4, 12, 19] |
Extensively metabolized by liver enzymes |
Elimination of parent drug
CYP, cytochrome P450
There are 13 validated SNPs in the ADRB1, which have been reported to the National Center for Biotechnology Information Single Nucleotide Polymorphism database (dbSNP). An additional six validated SNPs located near the ADBR1 gene region are also found in dbSNP. Of these SNPs, two polymorphisms have been extensively studied both in vitro and in vivo: Ser49Gly (nt 34552562 A>G on NT_030059, rs1801252) and Arg389Gly (nt 34553582 C>G on NT_030059, rs1801253). The Ser49Gly polymorphism is located in the N-terminus and the Arg389Gly polymorphism is located in a putative G-protein binding. The functional changes these polymorphisms cause are summarized in Table 3. Interestingly, a recent report using fluorescent resonance energy transfer (FRET)-based approach compared β1-AR binding affinity of the three β-blockers (bisoprolol, metoprolol, and carvedilol) between Arg389 and Gly389 receptors [28]. Although Gly-389-β1-AR showed a comparable degree of inhibitory effects by the antagonists on its activation and cAMP production, carvedilol had significantly higher degree of inhibitory effects than the other two β-blockers in Arg-389-β1-AR. These data suggest that effects of β-blockers may differ by β1-AR 389 allele.
Table 3.
Gene | Polymorphism | Minor allele | MAF | Functional consequences | References |
---|---|---|---|---|---|
ADRB1 | Ser49Gly | Gly | Caucasians (12–16%) | Gly49 allele has greater receptor down-regulation with agonist treatment | [20] |
African-Americans (23–28%) | |||||
Hispanics (20–21%) | Gly49-β1-AR is more sensitive to the inhibitory effects of metoprolol than Ser49- β1-AR | [21] | |||
Asians (14%) | |||||
Arg389Gly | Gly | Caucasians (24–34%) | Arg389 allele has higher basal and agonist-stimulated | [22] | |
African-Americans (39–46%) | AC activity | ||||
Hispanics (31–33%) | Lower AC activity upon agonist simulation in heart samples from HF patients with Arg389 allele than with Gly389 allele | [23] | |||
Asians (20–30%) | |||||
ADRB2 | Gly16Arg | Arg | Caucasians (39%) | Gly16 allele has greater receptor down-regulation with agonist stimulation | [24] |
African-Americans (49%) | |||||
Asians (51%) | |||||
Gln27Glu | Glu | Caucasians (25%) | Glu27 allele is resistant to receptor down-regulation | [24] | |
African-Americans (19%) | |||||
Asians (9%) | |||||
Thr164Ile | Ile | Caucasians (1–2%) | Ile164 allele causes defective coupling with G protein | [25] | |
African-Americans (0%) | |||||
Asians (0%) | |||||
ADRA2C | Insertion/Deletion | D | Caucasians (4%) | D allele causes loss of auto-inhibitory function of the receptor | [26] |
African-Americans (41%) | |||||
Asians (Unknown) | |||||
ACE | Insertion/Deletion | Ia | Caucasians (40–48%) | D allele is associated with higher plasma ACE level | [27] |
African-Americans (37–43%) | |||||
Asians (58–70%) |
Insertion allele is a major allele in Asians
ADRB1, β1-adrenergic receptor gene; ADRB2, β2-adrenergic receptor gene; ADRA2C, α2c-adrenergic receptor gene; ACE, angiotensin-converting enzyme gene; Arg, arginine; Gly, glycine; Ser, serine; Glu, glutamate; Gln, glutamine; Thr, threonine; Ile, isoleucine; I, insertion, D, deletion; MAF, minor allele frequency; AC, adenylyl cyclase; AR, adrenergic receptors; HF, heart failure
Of the 13 validated SNPs in the ADRB2 gene and 15 additional validated SNPs in the region close to the gene that have been reported to dbSNP (accessed on March 15, 2007), two polymorphisms have been widely investigated both in vivo and in vitro. The Gly16Arg (nt 9369376 G>A on NT 029289, rs1042713) and the Gln27Glu (nt 9369409 C>G on NT_29289, rs1042714) polymorphisms, both of which are located in the extracelluar portion of N-terminus, did not influence the receptors’ ability to couple to G-protein and to activate adenylyl cyclase [24]. Rather, the polymorphisms altered the extent to which the receptors underwent down-regulation (Table 3). In an in vivo study with healthy volunteers, the Arg16 and the Gln27 alleles have been associated with increased desensitization mediated by isoproterenol infusion [29]. Receptor expression and functions by common haplotypes of β2-AR have also been studied [30]. Of the 11 haplotypes constructed from 13 SNPs in the 5′ untranslated region and the coding sequence, there were two haplotypes that represented over 80 and 30% of the observed haplotypes in Caucasians and African-Americans, respectively. In an HEK293 cell transfection experiment, the amount of β2-AR and its mRNA were significantly different between the two haplotypes [30].
In an in vitro experiment with ventricle tissues isolated from patients with HF, β1-Arg-389 homozygote had 4.3-fold greater contractility than β1-Gly-389 carrier upon agonist stimulation [31]. In failing hearts, the ratio of the amount between β1-AR and β2-AR is reduced due to down-regulation of β1-AR compared to that in normal hearts [32]. In addition, there is evidence that β2-AR plays an important role in regulation of contractility and apoptosis induced by activation of β1-AR in failing hearts [33]. However, it is not currently clear whether ADRB2 genotype/haplotype influences regulation of contractility and apoptosis in failing hearts and how it differs by ADRB1 genotype.
β-Blockers pharmacogenetics in HF
Genetic associations with β-blocker-induced changes in left ventricular ejection fraction
Left ventricular ejection fraction (LVEF) is regarded as a surrogate marker to predict the adverse outcomes in systolic HF [34]. The degree of improvement in LVEF is well correlated with survival of the patients in HF [34]. As such, many studies have looked at associations of genetic polymorphisms of β-AR genes with improvement in LVEF after a β-blocker (Table 4). However, the results have been somewhat inconclusive. Two studies (one with metoprolol succinate and the other with carvedilol) reported significant associations between ADRB1 genotype and LVEF, with patients homozygous for Arg389 demonstrating the greatest improvement in LVEF [23, 35] Conversely, two other studies (one with carvedilol or bisoprolol, the other with bucindolol) found no such association [31, 36]. These mixed results may have arisen from many factors such as use of different β-blockers, duration of therapy, characteristics of the study population, among others.
Table 4.
Study population | Study type | B-blocker | N | Duration | Gene | Polymorphism | Results | P-value | References |
---|---|---|---|---|---|---|---|---|---|
Systolic | P | Metoprolol | 54 | >5 months | ADRB1 | Arg389Gly | Greater LVEF improvement in Arg389Arg than Gly carriers | 0.008 | [35] |
HF | CR/XL | Ser49Gly | (From 23 ± 5 to 29 ± 10% vs. from 22 ± 9 to 23 ± 11%) | ||||||
Systolic | R | Carvedilol | 224 | >6 months | ADRB1 | Arg389Gly | Greater LVEF improvements in Arg389Arg than Gly389Gly | 0.02 | [23] |
HF | (8.7 ± 1.1 vs. 0.93 ± 1.7%) | ||||||||
Systolic | R | Bisoprolol | 199 | 3 months | ADRB1 | 2 SNPs | No associations | ns | [36] |
HF | Carvedilol | ADRB2 | 3 SNPs | ||||||
Systolic | P | Bucindolol | 1040 | Median | ADRB1 | Arg389Gly | No associations | ns | [31] |
HF | 2 years | ||||||||
Systolic | R | Carvedilol | 80 | 4 months | ADRB2 | 2SNPs | Glu27 carriers had more good responders than Gln27Gln | 0.003 | [37] |
HF | (63 vs. 26%) | ||||||||
Systolic | R | Metoprolol | 54 | >5 months | ADRB1 | Arg389Gly | Greatest improvement in LVEF those who were ADRB1 Arg389Arg and ADRA2C deletion carriers | <0.02 | [38] |
HF | CR/XL | ADRA2C | I/D 322–325 | (12 vs. 0–2% in others) |
CR/XR, controlled release/extended release; N, sample size; SNPs, single nucleotide polymorphisms; Δ, change before and after treatment; LVEF, left ventricular ejection fraction; LVEDD, left ventricular end diastolic diameter; ns, not significant; Arg, arginine; Gly, glycine; Ser, serine; Glu, glutamate; Gln, glutamine; C, cytosine; T, thymidine, I/D, insertion/deletion; ADRB1, β1-adrenergic receptor gene; ADRB2, β2-adrenergic receptor gene; ADRA2C, α2c- adrenergic receptor gene; HF, heart failure
P, prospective study, which was designed specifically to test pharmacogenetics hypotheses, or where the primary outcome studied in the pharmacogenetic study was the primary outcome in the clinical trial
R, retrospective study, which was conducted on an existing data set
Two studies evaluated the association of ADRB2 genotype and LVEF response to carvedilol [36, 37]. Again, the results are mixed: one found that compared to Glu27 carriers, Gln27 homozygotes had significantly lower proportion of good responders defined as LVEF improvement by 10% in 3 months (63vs. 26%, P = 0.003) [37]. However, a study where response to either bisoprolol or carvedilol was assessed did not observe the same association [36]. A possible explanation for this is that bisoprolol, used by 65% of subjects in this study, is β1-AR selective, and so ADRB2 SNPs might be expected to have a less important role for this drug. Therefore, it is not possible to draw clear conclusions about the relationship among change in LVEF and β1-AR and β2-AR genotype at present.
The α2c-adrenergic receptor (α2c-AR) is an autoreceptor regulating presynaptic norepinephrine release. The gene (ADRA2C) located at chromosome 8p21-p11.2 has a 12-nucleotide deletion polymorphism which results in loss of four consecutive amino acid residues at 322–325 [26]. The deletion allele has been shown to reduce the auto-inhibitory function of the receptor [26]. This insertion/deletion and the ADRB1 Arg89Gly polymorphisms have been shown to synergistically increased the risk for HF in African-Americans in a case control study [39]. One study tested whether the ADRA2C and ADRB1 polymorphisms synergistically influenced the LVEF response to metoprolol in 54 HF patients [38]. In this study, patients with ADRB1 Arg389Arg and ADRA2C deletion carriers had the greatest improvement of LVEF (12 vs. 0–2% in others). However, large studies are needed to determine the effects of the two polymorphisms on clinical outcomes in HF patients.
Genetic associations with clinical outcomes during β-blocker therapy in randomized controlled trials
ADRB1 genotype may be a prognostic factor for adverse clinical outcomes, such as death and hospitalization in HF [31]. Bucindolol is an investigational non-selective β-blocker, which in the BEST trial did not show overall survival benefit versus placebo, although most secondary endpoints showed significant benefit from bucindolol [8]. In a post hoc analysis, ADRB1 Arg389Arg was significantly associated with fewer adverse outcomes in patients receiving bucindolol versus placebo [hazard ratio (HR) = 0.66, 95% CI 0.50–0.88] [31]. Interestingly, the benefit of bucindolol was not observed in Gly389 carriers. Additionally, the genotype associated with survival benefit from bucindolol is the same genotype associated with improved LVEF in some studies. MERIT-HF is the other large clinical trial that has reported genetic results [5]. In examining the relationship between the ADRB1 genotype and the trial outcomes (death and hospitalization), they found no significant association [40]. However, this analysis has been criticized because it combined treatment (metoprolol CR/XR) and placebo groups as one group and tested the association between outcomes and genotype. This analysis approach makes it difficult to assess the pharmacogenetic associations with adverse outcomes. It is similarly difficult to assess the effect of the polymorphism on the natural history of the disease, which would have been possible through consideration of the placebo-only patients. Thus, the data from this study are probably not informative regarding the role of the ADRB1 polymorphism and outcomes with metoprolol therapy.
Genetic associations with clinical outcomes during β-blocker therapy in retrospective cohort studies
Given that data from randomized-controlled trials are limited, insights might also be gained from non-interventional cohort studies. These studies are summarized in Table 5. For example, data from one such analysis suggest that high dose of a β-blocker may improve the unfavorable clinical outcomes associated with an ADRB1 genotype in HF. In a study with idiopathic dilated cardiomyopathy patients, 5-year mortality was lower in Gly49 carriers in ADRB1 than Ser49Ser (RR = 0.24, 95% CI0.07–0.80) in patients who received a low dose of β-blocker (≤50% of target dose) [41]. In the same study, Gly389 carriers had higher 5-year mortality rate than Arg389Arg in those who were on a low dose of β-blocker (RR = 2.42, 95% CI 1.04–5.63) [41]. However, these genotypes were not associated with the adverse outcomes in those who were on high dose of a β-blocker [41].
Table 5.
Study population | β-blocker | N | Duration | Gene | Polymorphism | Outcomes | Results | P-value | References |
---|---|---|---|---|---|---|---|---|---|
DCM | Various | 375 | 37–60 months | ADRB1 | Ser49Gly | Death or heart transplantation | Among patients on <50% of the target dose for the given β-blocker, Gly49 carrier was associated with the longer survival than Ser49Ser (HR = 0.24, 95% CI 0.07–0.80) | 0.014 | [41] |
Arg389Gly | ns | ||||||||
Among patients on a high dose of a β-blocker, no genetic association was detected | |||||||||
Systolic | Various | 328 | Median | ACE | I/D | Death or heart transplantation | D allele associated with higher risk for outcomes than I/I (HR = 1.80) | 0.04 | [42] |
HF | 21 months | No association in patients on a β-blocker | ns | ||||||
Various | Various | 220 | Median | Seven genes | Eight polymorphisms | Death or heart transplantation | Homozygotes for both Arg16Arg and Gln27Gln in the ADRB2 gene were associated with increased risk (HR = 1.91) | 0.02 | [43] |
34 months | Among patients on a β-blocker, none of the genetic polymorphisms were associated with the outcomes | ns |
HF, heart failure; DCM, dilated cardiomyopathy; N, sample size; ns, not significant; Arg, arginine; Gly, glycine; Ser, serine; Glu, glutamate; Gln, glutamine; I/D, insertion/deletion; ADRB1, β1-adrenergic receptor gene; ADRB2, β2-adrenergic receptor gene; ACE, angiotensin-converting enzyme gene; CI, confidence interval
β-Blocker therapy may also influence the risk associated with other genes in HF patients. For example, the ACE gene has a 287-bp insertion/deletion (I/D) polymorphism in intron 16. D/D genotype has been associated with elevated plasma ACE levels and higher rates of the adverse outcomes in HF [27, 42]. In a study with systolic HF patients, McNamara et al. [42] reported that although they saw significantly higher adverse events (death or heart transplantation) in patients with the D/D genotype, the association was not detected in patients who received a β-blocker at baseline. Importantly, the result was subsequently replicated in an independent study [44].
In a recently published study, a β2-AR haplotype (Arg16Arg26/Gln27Gln) was associated with increased risk for death or heart transplantation in 220 patients, 95 and 80% of whom were on an ACE inhibitor/angiotensin receptor blocker and a β-blocker at baseline [43]. When considered relative to β-blocker use, this association was most strongly driven by those not on a β-blocker (HR of 3.52 vs. HR of 1.55). Although these findings came from subgroup analyses, the results suggest that certain genotypes/haplotypes may be at increased risk of adverse outcomes and that β-blockers may attenuate the risk associated with that genotype/haplotype. Interestingly, these findings are consistent with those from an acute coronary syndrome population, in which the Arg16Gln27 haplotype was also associated with adverse outcomes, even among those treated with a β-blocker [45]. Collectively, these data may suggest that the Arg16Gln27 haplotype of the ADRB2 may be a high-risk haplotype group deserving of more aggressive therapy.
Genetic associations with β-blocker tolerability in HF
Although the benefits of β-blocker therapy are well documented, it has been reported that β-blockers are often not titrated to optimal doses, in part due to concerns about decompensation during titration [2, 10]. One study determined whether ADRB1 genotypes were associated with initial tolerability of metoprolol CR/XL [46]. The study found that there were significant differences by genotype in tolerability of the initiation of β-blocker therapy, as assessed by less need for increased diuretic doses during the titration period. Specifically, Gly49 carriers showed better tolerance to β-blocker initiation than Ser49Ser. Similarly, those who were Arg389Arg tolerated β-blocker initiation the best. An analysis by ADRB1 haplotype showed that 52% of patients who were Ser49Ser and Arg389Gly required an increase in diuretics, while none of the patients who was Ser49Gly and Arg389Arg were required to increase diuretic dose during β-blocker titration. Patients with other genotype combinations had need for diuretic dose titration that was intermediate between these two groups. If these findings were replicated, it might provide a mechanism for identifying those patients who will need careful attention and close follow up during the β-blocker titration period.
Many β-blockers including metoprolol and carvedilol are substrates for the cytochrome (CYP) P450 2D6 enzyme [12]. In particular, metoprolol is most highly dependent on CYP2D6 with 60–70% of its metabolism via this pathway, whereas 20–30% of metabolism of carvedilol is via CYP2D6 [12]. The CYP2D6 gene is highly polymorphic with about 80 alleles reported to date [47]. Due to the polymorphic drug metabolizing enzyme, pharmacokinetics of metoprolol vary among individuals [48, 49]. This may result in variable responses to the drug, particularly at therapy initiation, given the need to initiate β-blockers at low doses. Several studies also have determined whether the pharmacokinetic differences caused by the CYP2D6 polymorphisms are translated into variable adverse effects or efficacies in different diseases [46, 50–52]. In HF, only one study has tested this question [46]. In this study, CYP2D6 genotype was not associated with tolerability to metoprolol CR/XL upon initiation of therapy. Together with the study results in other diseases [51, 52], it appears that CYP2D6 genotype does not play a role in causing variability in responses (adverse or efficacious) to metoprolol. While other β-blockers have not been tested in this regard in HF, given that they are all less dependent on CYP2D6 for their metabolism than metoprolol, it seems unlikely that CYP2D6 variability would importantly affect their efficacy or tolerability.
Summary of pharmacogenetic findings for β-blockers in HF
The majority of the β-blocker pharmacogenetic studies in HF have focused on the β1-AR gene, the primary target for the β-blockers. Within this gene, Arg389Gly polymorphism is particularly interesting. Although LVEF response to a β-blocker with regards to the Arg389Gly polymorphism is not conclusive [23, 31, 35, 36], it was the Arg389 homozygous genotype that has been associated with better LVEF response in the studies that reported a positive association [23, 36]. The same genotype has also been associated with better clinical outcomes (survival and hospitalization) in a sub-study from a large clinical trial [36]. Importantly, this genotype has been associated with better response to β-blockers in different settings, such as hypertension (blood pressure lowering) [53, 54] and glaucoma (intraocular pressure lowering) [55]. Interestingly, while not all studies reported a positive association, studies with a positive association always showed Arg389Arg is a predictor of better response. On the other hand, the direction of the association with many other polymorphisms, such as ACE I/D and the common ADRB2 SNPs, has not been consistent, even in studies with a significant association. Finally, the functional studies on codon 389 polymorphism in ADBR1 support a logical biological mechanism for the clinical findings [22]. As such, the ADRB1 codon 389 polymorphism may be a starting point for individualized β-blocker therapy in the future. Data on the other genes have not been sufficiently consistent to draw clear messages on their role. The Revpresence of mechanistic corroboration for ADRB1 Arg389-Gly polymorphism that has been associated with clinical outcomes in patients with HF, and the lack thereof for other variants, also supports this conclusion.
Potential clinical implications of β-blocker pharmacogenetics
There are several potential clinical implications for β-blocker pharmacogenetics, based on the current literature and for the future. These are summarized below under specific categories.
Determining who receives β-blocker therapy or which β-blocker
Data from several studies suggest that there is a genotype group that responds less favorably to β-blocker therapy [43, 45]. However, given the consensus-guideline driven use of β-blockers, it is hard to envision withholding β-blocker therapy based on genetic information. The level of evidence required to take such action would probably come from a randomized-controlled trial (or two) and these are not likely to be done. Thus, it is unlikely that in the future we will withhold β-blocker therapy based on genotype. However, based on results in a large clinical trial with over 1,000 patients enrolled in an adrenergic receptor polymorphism sub-study [31], a small pharmaceutical company is seeking FDA approval of bucindolol, with the therapy targeted at patients with specific genotypes. They suggest that the patients with the genotypes of interest may have better outcomes with bucindolol than the currently used β-blockers, whereas for those with alternative genotypes, the current β-blockers would be preferred. The FDA submission will be of great interest to the pharmacogenetics research community as it will represent the first request for labeling in a specific genotype group during the drug approval process, outside of cancer. FDA approval of bucindolol, with an indication in specific genetic groups, may also sound the beginning of pharmacogenetic-guided therapy in cardiovascular disease. The recent study using FRET to study receptor activation, and showing that carvedilol inhibited Arg389Arg more effectively than metoprolol or bisoprolol also lends support to the concept that certain genotypes might respond more favorably to certain drugs [28].
Identifying those perhaps not fully benefitting from therapy or given dose
Another potential application of β-blocker pharmacogenetics is identifying those patients whose genotype potentially places them at risk for responding less than optimally to a β-blocker, or for whom a low dose is insufficient. Specifically, several studies suggest that ADRB1 Gly389 carriers might respond less favorably to β-blockers than Arg389Arg patients [23, 36]. As discussed above, it is unlikely that one would withhold therapy from this former group; however, this might be a genotype group that should be targeted for more aggressive HF therapy.
Data from several studies also suggest that patients of certain genotypes may require high doses of β-blocker therapy to offset the risk associated with a given genotype [42, 44]. While it is preferable for all patients to receive target doses of β-blocker, such genetic information might provide insights into those patients in whom it is especially important for them to be on target doses. Use of genetic information for the above examples could be supported based on the current literature, and could be implemented in practice with minimal controversy.
Identifying target populations for drug development
It might be possible to use genetic information to identify high-risk populations, whom might be best suited for new drug development. In the past decade, there have been numerous HF drugs that failed to show efficacy in late Phase III clinical trials, at a point when the pharmaceutical company has invested hundreds of millions in the drug. These include endothelin blockers, vasopeptidase inhibitors, phosphodiesterase inhibitors, and TNF receptor blockers. As a result of these failures, pharmaceutical industry is now much less interested in developing new drugs for HF. Given that most of the studies did not pre-select patients for risk or poor outcomes with current therapy, it is not surprising that they were unable to show benefit in addition to standard therapy of an ACE inhibitor, β-blocker, diuretic, and often digoxin. However, it is quite likely that some of the drugs that made it this far in the drug development process would be of benefit in a subset of the HF population. Thus, if genetic data could be utilized to identify a high-risk population to enroll in the study, efficacy might be more easily documented. Based on the current data, the best candidate in this category is the ADRB2 Arg16Gln27 haplotype. Two different studies (one in HF and one in post-acute coronary syndrome patients) have shown that carriers of this haplotype are at increased risk of adverse outcomes, even in the face of treatment with standard drug therapy [43, 45].
The approach of utilizing genetic information to target patients for inclusion of Phase II or, more likely, Phase III clinical trials has been advocated as an approach that might substantially reduce the drug development cost, and salvage drugs that might not otherwise be able to meet the efficacy standards of the FDA. The pharmaceutical industry, however, has generally been unenthusiastic about this approach, since it would segment the market for their drug. However, HF seems to be a perfect case where there are examples of drugs that might be salvaged through a pharmacogenetic-driven drug development approach.
In conclusion, the data on β-blocker pharmacogenetics provide evidence for the influence of genetic variability in drug response, and provide insights into the potential role of using genetic information to improve drug therapy outcomes. Based on the literature to date, it seems that HF may represent one of the first areas where genetic information is used to guide therapy, particularly as it relates to β-blockers, and those who may require additional therapy due to predicted sub-optimal response to β-blockade.
Acknowledgments
This work was supported in part by NIH grants HL68834, HL74730, GM74492 Bethesda, MD, and American Heart Association postdoctoral fellowship grant 0525474B, St. Petersburg, FL.
Contributor Information
Jaekyu Shin, Department of Pharmacy Practice, College of Pharmacy, Center for Pharmacogenetics, University of Florida, P.O. Box 100486, Gainesville, FL 32610-0486, USA.
Julie A. Johnson, Email: johnson@cop.ufl.edu, Department of Pharmacy Practice, College of Pharmacy, Center for Pharmacogenetics, University of Florida, P.O. Box 100486, Gainesville, FL 32610-0486, USA.
References
- 1.Jessup M, Brozena S. Heart failure. N Engl J Med. 2003;348:2007–2018. doi: 10.1056/NEJMra021498. [DOI] [PubMed] [Google Scholar]
- 2.Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a Veterans Administration Cooperative Study. N Engl J Med. 1986;314:1547–1552. doi: 10.1056/NEJM198606123142404. [DOI] [PubMed] [Google Scholar]
- 3.The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II) A randomised trial Lancet. 1999;353:9–13. [PubMed] [Google Scholar]
- 4.Reiter MJ. Cardiovascular drug class specificity: beta-blockers. Prog Cardiovasc Dis. 2004;47:11–33. doi: 10.1016/j.pcad.2004.04.004. [DOI] [PubMed] [Google Scholar]
- 5.Hjalmarson A, Goldstein S, Fagerberg B, et al. MERIT-HF Study Group. Effects of controlled-release metoprolol on total mortality, hospitalizations, and well-being in patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure (MERIT-HF) JAMA. 2000;283:1295–1302. doi: 10.1001/jama.283.10.1295. [DOI] [PubMed] [Google Scholar]
- 6.Poole-Wilson PA, Swedberg K, Cleland JG, et al. Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial. Lancet. 2003;362:7–13. doi: 10.1016/S0140-6736(03)13800-7. [DOI] [PubMed] [Google Scholar]
- 7.A trial of the beta-blocker bucindolol in patients with advanced chronic heart failure. N Engl J Med. 2001;344:1659–1667. doi: 10.1056/NEJM200105313442202. [DOI] [PubMed] [Google Scholar]
- 8.Hunt SA, Abraham WT, Chin MH, et al. ACC/AHA 2005 Guideline update for the diagnosis and management of chronic heart failure in the adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Management of Heart Failure): developed in collaboration with the American College of Chest Physicians and the International Society for Heart and Lung Transplantation: endorsed by the Heart Rhythm Society. Circulation. 2005;112:e154–e235. doi: 10.1161/CIRCULATIONAHA.105.167586. [DOI] [PubMed]
- 9.Basile JN. Titration of beta-blockers in heart failure. How to maximize benefit while minimizing adverse events. Postgrad Med. 2003;113:63–70. doi: 10.3810/pgm.2003.03.1389. [DOI] [PubMed] [Google Scholar]
- 10.Tandon P, McAlister FA, Tsuyuki RT, et al. The use of beta-blockers in a tertiary care heart failure clinic: dosing, tolerance, and outcomes. Arch Intern Med. 2004;164:769–774. doi: 10.1001/archinte.164.7.769. [DOI] [PubMed] [Google Scholar]
- 11.Khand A, Gemmel I, Clark AL, et al. Is the prognosis of heart failure improving? J Am Coll Cardiol. 2000;36:2284–2286. doi: 10.1016/s0735-1097(00)00995-5. [DOI] [PubMed] [Google Scholar]
- 12.Brodde OE, Kroemer HK. Drug-drug interactions of beta-adrenoceptor blockers. Arzneimittelforschung. 2003;53:814–822. doi: 10.1055/s-0031-1299835. [DOI] [PubMed] [Google Scholar]
- 13.Oldham HG, Clarke SE. In vitro identification of the human cytochrome P450 enzymes involved in the metabolism of R(+)- and S(−)- carvedilol. Drug Metab Dispos. 1997;25:970–977. [PubMed] [Google Scholar]
- 14.Frishman WH. Carvedilol. N Engl J Med. 1998;339:1759–1765. doi: 10.1056/NEJM199812103392407. [DOI] [PubMed] [Google Scholar]
- 15.von Mollendorff E, Reiff K, Neugebauer G. Pharmacokinetics and bioavailability of carvedilol, a vasodilating beta-blocker. Eur J Clin Pharmacol. 1987;33:511–513. doi: 10.1007/BF00544245. [DOI] [PubMed] [Google Scholar]
- 16.Gattis WA. Metoprolol CR/XL in the treatment of chronic heart failure. Pharmacotherapy. 2001;21:604–613. doi: 10.1592/phco.21.6.604.34551. [DOI] [PubMed] [Google Scholar]
- 17.Johnson JA, Burlew BS. Metoprolol metabolism via cytochrome P4502D6 in ethnic populations. Drug Metab Dispos. 1996;24:350–355. [PubMed] [Google Scholar]
- 18.Lancaster SG, Sorkin EM. Bisoprolol. A preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in hypertension and angina pectoris. Drugs. 1988;36:256–285. doi: 10.2165/00003495-198836030-00002. [DOI] [PubMed] [Google Scholar]
- 19.Meredith PA, Kelman AW, McSharry DR, Vincent J, Reid JL. The pharmacokinetics of bucindolol and its major metabolite in essential hypertension. Xenobiotica. 1985;15:979–985. doi: 10.3109/00498258509045050. [DOI] [PubMed] [Google Scholar]
- 20.Levin MC, Marullo S, Muntaner O, et al. The myocardium-protective Gly-49 variant of the beta 1-adrenergic receptor exhibits constitutive activity and increased desensitization and down-regulation. J Biol Chem. 2002;277:30429–30435. doi: 10.1074/jbc.M200681200. [DOI] [PubMed] [Google Scholar]
- 21.Rathz DA, Brown KM, Kramer LA, et al. Amino acid 49 polymorphisms of the human beta1-adrenergic receptor affect agonist-promoted trafficking. J Cardiovasc Pharmacol. 2002;39:155–160. doi: 10.1097/00005344-200202000-00001. [DOI] [PubMed] [Google Scholar]
- 22.Mason DA, Moore JD, Green SA, et al. A gain-of-function polymorphism in a G-protein coupling domain of the human beta1-adrenergic receptor. J Biol Chem. 1999;274:12670–12674. doi: 10.1074/jbc.274.18.12670. [DOI] [PubMed] [Google Scholar]
- 23.Mialet Perez J, Rathz DA, Petrashevskaya NN, et al. Beta 1-adrenergic receptor polymorphisms confer differential function and predisposition to heart failure. Nat Med. 2003;9:1300–1305. doi: 10.1038/nm930. [DOI] [PubMed] [Google Scholar]
- 24.Green SA, Turki J, Innis M, et al. Amino-terminal polymorphisms of the human beta 2-adrenergic receptor impart distinct agonist-promoted regulatory properties. Biochemistry. 1994;33:9414–9419. doi: 10.1021/bi00198a006. [DOI] [PubMed] [Google Scholar]
- 25.Green SA, Cole G, Jacinto M, et al. A polymorphism of the human beta 2-adrenergic receptor within the fourth transmembrane domain alters ligand binding and functional properties of the receptor. J Biol Chem. 1993;268:23116–231121. [PubMed] [Google Scholar]
- 26.Small KM, Forbes SL, Rahman FF, et al. A four amino acid deletion polymorphism in the third intracellular loop of the human alpha 2C-adrenergic receptor confers impaired coupling to multiple effectors. J Biol Chem. 2000;275:23059–23064. doi: 10.1074/jbc.M000796200. [DOI] [PubMed] [Google Scholar]
- 27.Tiret L, Rigat B, Visvikis S, et al. Evidence, from combined segregation and linkage analysis, that a variant of the angiotensin I-converting enzyme (ACE) gene controls plasma ACE levels. Am J Hum Genet. 1992;51:197–205. [PMC free article] [PubMed] [Google Scholar]
- 28.Rochais F, Vilardaga JP, Nikolaev VO, et al. Real-time optical recording of beta(1)-adrenergic receptor activation reveals supersensitivity of the Arg389 variant to carvedilol. J Clin Invest. 2007;117:229–235. doi: 10.1172/JCI30012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dishy V, Sofowora GG, Xie HG, et al. The effect of common polymorphisms of the beta2-adrenergic receptor on agonist-mediated vascular desensitization. N Engl J Med. 2001;345:1030–1035. doi: 10.1056/NEJMoa010819. [DOI] [PubMed] [Google Scholar]
- 30.Drysdale CM, McGraw DW, Stack CB, et al. Complex promoter and coding region beta 2-adrenergic receptor haplotypes alter receptor expression and predict in vivo responsiveness. Proc Natl Acad Sci USA. 2000;97:10483–10488. doi: 10.1073/pnas.97.19.10483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Liggett SB, Mialet-Perez J, Thaneemit-Chen S, et al. A polymorphism within a conserved beta(1)-adrenergic receptor motif alters cardiac function and beta-blocker response in human heart failure. Proc Natl Acad Sci USA. 2006;103:11288–11293. doi: 10.1073/pnas.0509937103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhu W, Zeng X, Zheng M, et al. The enigma of beta2-adrenergic receptor Gi signaling in the heart: the good, the bad, and the ugly. Circ Res. 2005;97:507–509. doi: 10.1161/01.RES.0000184615.56822.bd. [DOI] [PubMed] [Google Scholar]
- 33.He JQ, Balijepalli RC, Haworth RA, et al. Crosstalk of beta-adrenergic receptor subtypes through Gi blunts beta-adrenergic stimulation of L-type Ca2+ channels in canine heart failure. Circ Res. 2005;97:566–573. doi: 10.1161/01.RES.0000181160.31851.05. [DOI] [PubMed] [Google Scholar]
- 34.Curtis JP, Sokol SI, Wang Y, et al. The association of left ventricular ejection fraction, mortality, and cause of death in stable outpatients with heart failure. J Am Coll Cardiol. 2003;42:736–742. doi: 10.1016/s0735-1097(03)00789-7. [DOI] [PubMed] [Google Scholar]
- 35.Terra SG, Hamilton KK, Pauly DF, et al. Beta1-adrenergic receptor polymorphisms and left ventricular remodeling changes in response to beta-blocker therapy. Pharmacogenet Genomics. 2005;15:227–234. doi: 10.1097/01213011-200504000-00006. [DOI] [PubMed] [Google Scholar]
- 36.de Groote P, Helbecque N, Lamblin N, et al. Association between beta-1 and beta-2 adrenergic receptor gene polymorphisms and the response to beta-blockade in patients with stable congestive heart failure. Pharmacogenet Genomics. 2005;15:137–142. doi: 10.1097/01213011-200503000-00001. [DOI] [PubMed] [Google Scholar]
- 37.Kaye DM, Smirk B, Williams C, et al. Beta-adrenoceptor genotype influences the response to carvedilol in patients with congestive heart failure. Pharmacogenetics. 2003;13:379–382. doi: 10.1097/00008571-200307000-00002. Medline. [DOI] [PubMed] [Google Scholar]
- 38.Lobmeyer MT, Gong Y, Terra SG, et al. Synergistic polymorphisms of β1 and α2c-adrenergic receptors and the influence on left ventricular ejection fraction response to β-blocker therapy in heart failure. Pharmacogenet Genomics. 2007;17(4):277–282. doi: 10.1097/FPC.0b013e3280105245. [DOI] [PubMed] [Google Scholar]
- 39.Small KM, Wagoner LE, Levin AM, et al. Synergistic polymorphisms of beta1- and alpha2C-adrenergic receptors and the risk of congestive heart failure. N Engl J Med. 2002;347:1135–1142. doi: 10.1056/NEJMoa020803. [DOI] [PubMed] [Google Scholar]
- 40.White HL, Maqbool A, McMahon AD, et al. An evaluation of the beta-1 adrenergic receptor Arg389Gly polymorphism in individuals at risk of coronary events. A WOSCOPS substudy. Eur Heart J. 2002;23:1087–1092. doi: 10.1053/euhj.2001.3037. [DOI] [PubMed] [Google Scholar]
- 41.Magnusson Y, Levin MC, Eggertsen R, et al. Ser49Gly of beta1-adrenergic receptor is associated with effective beta-blocker dose in dilated cardiomyopathy. Clin Pharmacol Ther. 2005;78:221–231. doi: 10.1016/j.clpt.2005.06.004. [DOI] [PubMed] [Google Scholar]
- 42.McNamara DM, Holubkov R, Janosko K, et al. Pharmacogenetic interactions between beta-blocker therapy and the angiotensin-converting enzyme deletion polymorphism in patients with congestive heart failure. Circulation. 2001;103:1644–1648. doi: 10.1161/01.cir.103.12.1644. [DOI] [PubMed] [Google Scholar]
- 43.Shin J, Lobmeyer MT, Gong Y, et al. Relation of beta(2)-Adrenoceptor Haplotype to Risk of Death and Heart Transplantation in Patients With Heart Failure. Am J Cardiol. 2007;99:250–255. doi: 10.1016/j.amjcard.2006.08.020. [DOI] [PubMed] [Google Scholar]
- 44.de Groote P, Helbecque N, Lamblin N, et al. Beta-adrenergic receptor blockade and the angiotensin-converting enzyme deletion polymorphism in patients with chronic heart failure. Eur J Heart Fail. 2004;6:17–21. doi: 10.1016/j.ejheart.2003.09.006. [DOI] [PubMed] [Google Scholar]
- 45.Lanfear DE, Jones PG, Marsh S, et al. Beta2-adrenergic receptor genotype and survival among patients receiving beta-blocker therapy after an acute coronary syndrome. JAMA. 2005;294:1526–1533. doi: 10.1001/jama.294.12.1526. [DOI] [PubMed] [Google Scholar]
- 46.Terra SG, Pauly DF, Lee CR, et al. Beta-adrenergic receptor polymorphisms and responses during titration of metoprolol controlled release/extended release in heart failure. Clin Pharmacol Ther. 2005;77:127–137. doi: 10.1016/j.clpt.2004.10.006. [DOI] [PubMed] [Google Scholar]
- 47.Zanger UM, Raimundo S, Eichelbaum M. Cytochrome P450 2D6: overview and update on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch Pharmacol. 2004;369:23–37. doi: 10.1007/s00210-003-0832-2. [DOI] [PubMed] [Google Scholar]
- 48.Taguchi M, Nozawa T, Mizumaki K, et al. Nonlinear mixed effects model analysis of the pharmacokinetics of metoprolol in routinely treated Japanese patients. Biol Pharm Bull. 2004;27:1642–1648. doi: 10.1248/bpb.27.1642. [DOI] [PubMed] [Google Scholar]
- 49.Nozawa T, Taguchi M, Tahara K, et al. Influence of CYP2D6 genotype on metoprolol plasma concentration and beta-adrenergic inhibition during long-term treatment: a comparison with bisoprolol. J Cardiovasc Pharmacol. 2005;46:713–720. doi: 10.1097/01.fjc.0000184117.76188.68. [DOI] [PubMed] [Google Scholar]
- 50.Wuttke H, Rau T, Heide R, et al. Increased frequency of cytochrome P450 2D6 poor metabolizers among patients with metoprolol-associated adverse effects. Clin Pharmacol Ther. 2002;72:429–437. doi: 10.1067/mcp.2002.127111. [DOI] [PubMed] [Google Scholar]
- 51.Zineh I, Beitelshees AL, Gaedigk A, et al. Pharmacokinetics and CYP2D6 genotypes do not predict metoprolol adverse events or efficacy in hypertension. Clin Pharmacol Ther. 2004;76:536–544. doi: 10.1016/j.clpt.2004.08.020. [DOI] [PubMed] [Google Scholar]
- 52.Fux R, Morike K, Prohmer AM, et al. Impact of CYP2D6 genotype on adverse effects during treatment with metoprolol: a prospective clinical study. Clin Pharmacol Ther. 2005;78:378–387. doi: 10.1016/j.clpt.2005.07.004. [DOI] [PubMed] [Google Scholar]
- 53.Johnson JA, Zineh I, Puckett BJ, et al. Beta 1-adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin Pharmacol Ther. 2003;74:44–52. doi: 10.1016/S0009-9236(03)00068-7. [DOI] [PubMed] [Google Scholar]
- 54.Wang SL, Huang JD, Lai MD, et al. Molecular basis of genetic variation in debrisoquin hydroxylation in Chinese subjects: polymorphism in RFLP and DNA sequence of CYP2D6. Clin Pharmacol Ther. 1993;53:410–418. doi: 10.1038/clpt.1993.44. [DOI] [PubMed] [Google Scholar]
- 55.Schwartz SG, Puckett BJ, Allen RC, et al. Beta1-adrenergic receptor polymorphisms and clinical efficacy of betaxolol hydrochloride in normal volunteers. Ophthalmology. 2005;112:2131–2136. doi: 10.1016/j.ophtha.2005.08.014. [DOI] [PubMed] [Google Scholar]