Volume 86, Issue 6 p. 1015-1033
SYSTEMATIC REVIEW AND META-ANALYSIS
Free Access

CYP2D6 polymorphism and its impact on the clinical response to metoprolol: A systematic review and meta-analysis

Maxime Meloche

Maxime Meloche

Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada

Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada

Montreal Heart Institute, Montreal, Quebec, Canada

Search for more papers by this author
Michael Khazaka

Michael Khazaka

Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada

Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada

Search for more papers by this author
Imad Kassem

Imad Kassem

Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada

Search for more papers by this author
Amina Barhdadi

Amina Barhdadi

Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada

Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada

Search for more papers by this author
Marie-Pierre Dubé

Marie-Pierre Dubé

Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada

Montreal Heart Institute, Montreal, Quebec, Canada

Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada

Search for more papers by this author
Simon de Denus

Corresponding Author

Simon de Denus

Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada

Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Quebec, Canada

Montreal Heart Institute, Montreal, Quebec, Canada

Correspondence

Simon de Denus, Montreal Heart Institute, 5000 Bélanger St., H1T IC8, Montréal, Québec. Canada.

Email: [email protected]

Search for more papers by this author
First published: 23 February 2020
Citations: 41

Abstract

Aims

CYP2D6 genetic polymorphisms are associated with metoprolol pharmacokinetics. Whether the clinical response to metoprolol is also affected remains uncertain.

Methods

We conducted a systematic review on the effects of CYP2D6 polymorphism on the clinical response to metoprolol. Searches were conducted using MEDLINE. Meta-analyses were performed on the impact of CYP2D6-inferred phenotypes on heart rate (HR) reduction, diastolic (DBP) and systolic (SBP) blood pressure reduction, average daily doses, all-type adverse events and bradycardia.

Results

Our qualitative assessment indicated inconsistent results in individual studies and endpoints, but CYP2D6 poor metabolizers (PM) generally presented a greater reduction in HR. The meta-analysis of 15 studies, including a total of 1146 individuals, found a reduction in HR of 3 beats/min (P = .017), and of SBP and DBP by 3 mmHg (P = .0048) for PM compared to non-PM individuals using similar metoprolol doses. Bradycardia appeared more frequent by 4-fold for PM, although significant heterogeneity was observed regarding bradycardia, which limits the scope of this finding.

Conclusion

Patients without any CYP2D6 metabolic capacities appear to have increased reduction in DBP, HR and SBP during metoprolol treatment and may be at a higher risk of bradycardia compared to patients with active CYP2D6 phenotypes. Further prospective data are required to determine whether CYP2D6 is associated with clinical events in patients treated with metoprolol, as well as to demonstrate the clinical utility of an individualized approach of prescribing metoprolol using CYP2D6-inferred phenotypes.

What is already known about this subject

  • CYP2D6 highly contributes to drug metabolism. It contains several genetic polymorphisms affecting its enzymatic activity.
  • Metoprolol pharmacokinetics are significantly affected between genotype-inferred phenotypes of CYP2D6, but the impacts on the clinical response of patients to patients have not been summarized.

What this study adds

  • This paper reviews the cumulative results of the effects of CYP2D6 genetic polymorphisms for patients on metoprolol treatment.
  • Patients possessing an inactive CYP2D6 phenotype have increased clinical effects and bradycardia with metoprolol, compared to those with an active CYP2D6 metabolic capacity.
  • More prospective data should be generated to suggest the implementation of a personalized approach to CYP2D6-mediated β-blocker therapy.

1 INTRODUCTION

The cytochrome P450 (CYP) 2D6 accounts for nearly 20% of all hepatic enzyme expression and is responsible for the metabolism of up to 25% of all registered drugs.1 The Pharmacogenomics Knowledge Base regroups growing evidence that demonstrates a causal relationship between allelic variants of the CYP2D6 gene and variable response to certain drugs.2 More than 100 variations, primarily single nucleotide polymorphisms and copy number variants, have been reported.1 Such sequence variations can confer a null to increased CYP2D6 metabolic activity, and the effects on drug metabolism and pharmacokinetic profiles have been repeatedly documented across multiple drug classes.3 Guidelines have been elaborated to classify the CYP2D6 metabolic activity of individuals into 4 genetically-inferred phenotypes: ultrarapid metabolizers (UM), extensive, or normal, metabolizers (EM), intermediate metabolizers (IM) and poor metabolizers (PM).3-6 The predicted prevalence of CYP2D6 genotype-predicted phenotype is estimated to be 1–21%, 67–90%, 0.4–11% and 0.4–5.4%, for UM, EM, IM and PM respectively, depending on the population studied.7

The selective β1-antagonist metoprolol is one of the most commonly used cardiovascular drugs to treat conditions such as heart failure, hypertension and atrial fibrillation.8-10 Approximately 80% of metoprolol is metabolized by CYP2D6,11 whose metabolites exert negligible pharmacological activity.12 An earlier meta-analysis has confirmed the significant effect of CYP2D6 genotype on the pharmacokinetics of metoprolol.13 However, whether this pharmacogenetic effect translates into differences in clinical response to metoprolol treatment is uncertain. This is largely attributable to the fact that previous studies only had small sample sizes. Hence, the objective of this article was to systematically review and summarize the impact of CYP2D6 polymorphisms on the clinical response to metoprolol.

2 METHODS

This systematic review was conducted in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement.14

2.1 Data sources and search strategy

A series of comprehensive searches in the MEDLINE database were conducted on articles written from 1946 to September 2018 to investigate the association between CYP2D6 and the pharmacodynamics of metoprolol. The searches were restricted to articles written in English or in French, while also including ahead-of-print, in-process and nonindexed records. The following terms were used to guide and refine the iterative search process: metoprolol; cytochrome P-450 2D6; genomics; genes; pharmacogenetics; polymorphism; genetic; single-stranded conformation; amplified fragment length polymorphism analysis; single nucleotide; restriction fragment length; phenotype; genotype (Table S1). Database updates were activated to identify newly published articles. The reference lists of the articles retrieved were manually searched to find studies that did not appear based on our strategy.

2.2 Eligibility criteria and study selection

Studies to be included were considered for this review irrespectively of study design, follow-up duration, metoprolol dosing regimen, and drug formulation. Studies evaluating drug interactions between metoprolol and other CYP2D6 substrates were excluded, unless patients took metoprolol without or before administration of a potentially interacting agent. In such cases, only the relevant baseline information was included. Previous reviews and meta-analyses were also rejected after having searched their reference lists.

In terms of subject inclusion, participants who were healthy, as well as patients with cardiovascular and noncardiovascular diseases, were included. Studies involving special populations such as twins, or pregnant and/or lactating women were excluded. No constraints were applied on the use of polypharmacy regimens. Publications where participants had not been genotyped, or studies that had classified patients primarily according to their pharmacokinetic profiles using probe drugs, or to drug responses without consideration to genetic information, were also excluded.

2.3 Data collection and data items

Eligibility assessment and data collection was performed independently by the first and second authors. Differences were resolved through discussions and consensus, and any remaining issues were settled with the senior author. Results from the database searches were ordered alphabetically according to the names of the first author to easily identify duplicates, which were removed afterwards. A primary screening with the titles and abstracts of all the articles was made. Articles considered eligible for review were entirely assessed and the data were collected in a MS Excel spreadsheet. The following details were included: year of publication; first author; title of study; presence of duplicates; study design; sample size and ethnicity; patients' diseases; investigated medications and concomitant drugs; dosing regimens; follow-up; outcome measures; effects on reviewed outcomes; genotyped single nucleotide polymorphisms; and phenotype assignment.

The reviewed parameters between all phenotypes were limited to changes from baseline at study enrolment of heart rate (ΔHR), diastolic (ΔDBP) and systolic (ΔSBP) blood pressure, mean daily metoprolol doses, and frequency of all-type adverse events (AE) and bradycardia. End-of-study values for each endpoint were collected. Mortality and hospitalizations, or other composite criteria reflecting death and morbidity were also reviewed.

2.4 Genotype-inferred phenotype assignment

We used the inferred phenotype assigned by the authors, according to their genotype classification. In studies having reported genotypes only, we attributed inferred phenotypes in accordance with the current Dutch Pharmacogenetics Working Group (DPWG) recommendations for groups which had not been assigned one. The DPWG annotations on metoprolol and CYP2D6 are referred by the Pharmacogenomics Knowledgebase website. Thus, subjects were categorized as UM if they carried >2 fully functional alleles (*1, *2, *33, *35, *39, *53), EM if they possessed 2 fully functional alleles or 1 fully functional and a semi-active allele (*9, *10, *17, *29, *36, *41, *53, *54, *59, *72, *84), IM if they had only 1 fully functional allele and a null allele (*3–*8, *11, *12, *14–*16, *18–*21, *38, *40- *42, *44, *51, *56, *57, *60, *62, *68, *69, *92, *96, *99–*101, *114) or 2 semi-functional alleles, and PM were determined to have 2 null alleles. All known CYP2D6 polymorphisms are indicated on the Pharmacogene Variation Consortium online database.15

2.5 Statistical methods and meta-analyses

Statistical analyses were carried using primarily with the meta package on the R software version 3.5.1 (The R Foundation for Statistical Computing). Random-effects models for meta-analyses were chosen. Irrespective of their designs, studies having reported quantitative values for the assessed parameters were included in our meta-analyses. For continuous variables (i.e. average daily doses, ΔDBP, ΔHR, ΔSBP), data were reported as mean difference, alongside confidence intervals (CI) set at 95% of the distribution (Table S2). For such parameters, random-effects models were conducted using the Hartung–Knapp adjustment.16 For binary outcomes (i.e. all-type AE, bradycardia), odds ratios (OR) were retrieved or calculated (Table S3). Summary effects sizes were generated by means of a restricted maximum likelihood method with an inverse-variance estimator. Other models have recently been brought forward to increase probability coverage for binary outcomes.17, 18 Recent simulations have shown that they do not considerably outperform restricted maximum likelihood estimations in random-effects meta-analyses with few studies.19 Nevertheless, considering the criticism that has been made surrounding the application of the Hartung–Knapp adjustment,20, 21 we performed sensitivity analyses to compare the results to the conventional DerSimonian–Laird approach in the case of continuous variables.

Other statistical analyses were made via Comprehensive Meta-Analysis, version 2.2.064 (Biostat, Englewood, NJ, USA). The various ways of reporting results from the included studies required that certain values be pooled. For example, if individuals carrying different genotypes but having the same inferred phenotype were reported separately in some articles, their respective values were weighed and aggregated together, according to the DPWG guidelines, generating single values of mean and standard deviation (SD) for each phenotype. The same weighing process was applied to calculate means and SDs for pooled phenotypes. Whenever needed, reported values were transformed into common units of measure. In some studies, only medians, interquartile ranges, and/or full ranges with sample sizes were reported. To transform those parameters into values that could be standardized and properly integrated into our models, we used the formulas proposed by Wan et al.22 Briefly, this approach utilizes the sample size, median, maximum and minimum values, or the interquartile range of a given distribution to estimate the mean and SD of a dataset. Values obtained from Wan et al.’s method22 are displayed in Table S4 (Supporting Information), and all values used in our meta-analyses for daily doses are listed in Table S5. If warranted, the estimated parameters were pooled as described earlier. Even though our primary comparison of interest was to compare PM individuals with those presenting any CYP2D6 activity (i.e. UM, EM, IM), we performed pairwise comparisons between single phenotypes as well.

2.6 Quality and risk of bias

The first and second authors (M.M., M.K.) performed an evaluation on the quality and risk of bias in each study. Multiple critical evaluation tools for study biases were available from different consortia and institutes, but their qualitative assessment methods were only partially adapted for pharmacogenetic studies. Thus, the following quality components were inspected without referring to general critical evaluation tools: study design, randomization, appropriateness of group comparisons, baseline characteristics, patients' diseases and medications, dosing regimens and concomitant medications, and inferred phenotype assignment from genotype. However, we did not exclude studies based on these criteria.

2.7 Ethical review

Ethical approval was unnecessary for this systematic review and meta-analysis, because only previously published data were retrieved. No recruitment of human participants was done, and personal data were not needed to perform this review.

3 RESULTS

3.1 Summary of systematic review

Figure 1 presents the article selection process. The comprehensive search generated 75 results from the MEDLINE database. Seven additional record from reference lists were added, along with 5 from manual searches, totalling 87 results. Of these, 2 articles were removed due to language barriers (German, Chinese), along with 1 off-topic systematic review, 2 earlier meta-analyses, and 1 editor correspondence. After removal of duplicates (n = 10), 13 articles were excluded based on their titles and 11 on their abstracts. The remaining 47 articles were completely assessed for eligibility. Twenty-five additional articles were then excluded because they did not meet all the eligibility criteria: 5 studies did not perform any genotyping, 4 studies did not present any reviewed parameters, 1 study was a retrospective assessment of 2 prior studies, and 1 study did not classify subjects according to their CYP2D6 genotype-inferred phenotype. Fifteen articles were rejected for presenting pharmacokinetic parameters only. The complete list of fully assessed and excluded articles is available as Supporting Information (Table S6). In total, 21 studies were included for presenting clinical parameters, 15 of which that were used in our meta-analyses.

Details are in the caption following the image
Flow chart of the review process

Table 1 contains the main characteristics of the included studies with regards to their clinical parameters. Study designs, types of populations and diseases, medications and dosing regimens, durations and follow-ups, inferred phenotype classification, and the summarized results of every parameter reviewed are indicated. Values in the table were all explicitly reported in the original articles.

Table 1. Summary of included studies evaluating the effects of CYP2D6 phenotypes on the clinical response to metoprolol
Authors (y) Study design Population and diseases Follow-up Genotype-inferred phenotypes Medication and dosing regimens Results of reviewed markers
Batty et al. (2014)28

Retrospective substudy of the metoprolol CR/XL randomized

Intervention trial in congestive heart failure (MERIT-HF)

718 patients from a subpopulation of the MERIT-HF trial with NYHA II-IV, and with LVEF≤40%

313 patients included in the analyses

Mean 339 d

Patients taking metoprolol:

189 EM: *1/*1

112 IM: *1/*4

12 PM: *4/*4

Low initial dose metoprolol CR/XL (12.5 mg/d for patients NYHA III-IV, 25 mg/d for patient NYHA II); progressive, bi-weekly, upward titration for a target dose of 200 mg/d

Individual dosing regimens freely adjustable to the judgement of the investigator

• No change in dosing profiles and mean daily doses between all genotypes (P = .29)
No differences between all genotypes in achieving a 200 mg/d target dose (P = .25)
• Risks of discontinuation nonsignificant for the 3 groups compared to placebo:
- EM: OR = 0.55, 95%CI = [0.25–1.19], P = .12
- IM: OR = 0.96, 95%CI = [0.49–1.91]; P = .92
- PM: OR = 0.59, 95%CI = [0.05–7.43]; P = .81
• Significant decreases in resting HR at wk 4, 6 and 8 for IM and PM compared to EM from their respective baseline values (all P < .05), but not at 2 wk and 3 mo
• No difference of resting HR between IM and PM during all follow-up period, but greater differences in HR decreases from baseline for IM compared to EM (P = .02), and for PM compared to EM (P = .04) at maximum doses. No differences after 6 mo, which corresponded to the longest time of measurement.
• No difference for SBP between all genotypes (P = .56)
• Significant decreases of DBP for IM compared to EM after 4 wk and 3 mo (P = .03; P = .004), but not at wk 2, 6, 8, and after 6 mo, and between PM and EM after wk 2 and 4, and 3 mo (P = .04; P = .04; P = .03), not at wk 6 and 8, and at 6 mo
• Significant decreases of DBP for IM and PM compared to EM at maximum doses (P = .04; P = .01)
• Genotype conferred no effect on all-cause mortality and hospitalization; combined endpoints were similar in all 3 groups compared to placebo:
- EM: OR = 0.62; 95%CI = [0.37–1.06], P = .08
- IM: OR = 0.71; 95%CI = [0.36–1.45], P = .36
- PM: OR = 0.80; 95%CI = [0.11–5.77], P = .83
Baudhuin et al. (2010)30 Retrospective, nonrandomized cohort study 93 patients with CHF for ≥6 mo, NYHA I-IV, LVEF ≤45% Total 14 mo

Patients taking metoprolol:

5 UM: *1/*2A,

*1/*2AxN

1 EM: *1/*1

10 IM: *1/*4, *1/*5,

*2/*4, *2/*9, *4/*9

3 PM: *3/*10, *4/*4, *4/*6

25 mg/d metoprolol IR, doubling every 10 d until MTD achieved; target dose 200 mg/d, or 6.25 mg/d carvedilol, doubling every 10 d until MTD achieved; target dose of 25 mg/d if BW ≤ 85 kg, and 50 mg/d if BW > 85 kg Phenotype comparisons for patients taking metoprolol:
• Nonsignificant difference in metoprolol dosage and MTD between all genotypes (data not shown)
19 patients treated with metoprolol
Bijl et al. (2009)37 Prospective, population-based cohort study (Rotterdam study) 6348 β-blocker users at baseline (2878 at final follow-up); 199, 243, 205, and 196 metoprolol users at baseline, first, second and third follow-up Total 14 y

ΔHR measurements:

451 EM: *1/*1,

255 IM: *1/*4,

34 PM: *4/*4

ΔDBP/ΔSBP

measurements (same

genotypes):

496 EM

276 IM

37 PM

Phenotypes attributed

according to DPWG

guidelines

Dosage titrated on hypertensive effect and dependant on CYP2D6 genotype (data not shown)

Comedications tolerated during the study, including strong and weak CYP2D6 inhibitors

Metoprolol formulation not mentioned

• HR, adjusted for age and sex, was significantly decreased in PM compared to EM (P < .0001), and in IM compared to EM (P = .013)
• Metoprolol conveyed more risks of bradycardia for PM compared to EM (P = .0014)
• Mean difference in HR significantly decreased for IM and PM compared to EM that were all β-blocker users (P = .012; P < .001)
• Mean difference in DBP was significant for PM compared to EM that were metoprolol users (P = .045), yet no changes were seen for SBP
• Within-person change for HR decreased significantly only for PM compared to EM (P = .019), but not between IM and EM (P = .06). Discontinuation did not lead to significant differences between any phenotype.
• DBP did not change for every phenotype when starting (n = 315) or discontinuing (n = 161) metoprolol therapy
• Differences in mean dose was observed for CYP2D6-mediated β-blockers, which consisted mainly of metoprolol (P = .03)
Fedorinov et al. (2018)31 Prospective, observational cohort study 201 patients of Russian or Yakut ethnicity with CHD on routine β-blocker therapy (atenolol, bisoprolol, metoprolol) Specific duration not mentioned

63 EM: *1/*1, *1/*10

16 IM: *1/*4

Patients receiving metoprolol:

Titrated dose between 12 and 150 mg/d

Metoprolol formulation not mentioned

• Titrated doses between the 39 patients genotyped as *1/*1 carriers and the 16 other *1/*4 patients did not vary significantly, although a trend was observed (P = .071)
• The differences in titrated doses were even less significant when comparing the 39 *1/*1 with the 24 *1/*10 carriers (P = .240)
79 patients receiving metoprolol
Fux et al. (2005)32 Multicentre, prospective, open-label, nonrandomized study 121 subjects with various cardiovascular and noncardiovascular diseases (HT, CHD, arrhythmias, migraine, anxiety disorder) not previously treated with metoprolol 6 wk

5 UM: More than 2 fully

functional alleles

91 EM: *1/*3-*10, *1/*41

21 IM: *8-*10/*8-*10,

*8-*10/*3-*8, *8-*10/*41,

*8-*10/*3-*8

4 PM: *3-*8/*3-*8

Any daily dose allowed, with any dosing scheme of metoprolol CR/XL

Individual dosage freely adjustable based on clinical grounds

• Mean daily doses did not differ between all genotypes (P = .78)
• Tendency toward more frequent cases of cold extremities observed in the PM + IM vs EM + UM (P = .56, CI95% = [1.03–14.3]). Sexual dysfunction lower for PM + IM vs EM + UM (P < .05)
• EM and IM had significant SBP, DBP, and HR reductions compared to baseline (all P < .01), not UM or PM
• Mean reductions between all phenotypes were nonsignificant for SBP (P = .92), DBP (P = .88), and HR (P = .30)
Goryachkina et al. (2008)23 Prospective, nonrandomized, open-label study 187 Caucasians hospitalized after AMI occurrence 10 d

Patients kept for clinical

parameter analyses:

5 UM: *1/*1xN

108 EM: *1/*1, *1/*3,

*1/*4, *1/*4xN, *1/*4,

*1/*10, *10/*4

2 PM: *4/*4

75 mg/d ±38 mean metoprolol IR or CR/XL • Median resting HR at discharge (15–20 d after admission) were different between groups with 0, 1, 2, or 3 functional alleles (P = .01). Changes occurred between d 7–10
115 patients genotyped and included for analysis, including the 17 depressed AMI patients used for the previous study; Goryachkina et al. (2008a) Use of concomitant drugs accepted and considered, with multiple drug classes coadministered other than β-blockers
Goryachkina et al. (2008)34 Prospective, nonrandomized, open-label, study 34 patients taking metoprolol at baseline, with 17 depressed AMI patients, and 17 nondepressed control patients 10 d 17 EM: *1/*1, *1/*3, *1/*4 Titrated metoprolol IR or CR/XL on d 1 Baseline measurements:
Coadministration of 20 mg/d paroxetine on d 2–8, or placebo • Nonsignificant differences in metoprolol dose allocation between homozygous and heterozygous genotypes before administration of paroxetine (not shown)
Use of concomitant drugs accepted
Hamadeh et al. (2014)26 Multicentre, prospective, open-label, sequential monotherapy, randomized trial (PEAR2 -Pharmacogenomic evaluation of antihypertensive responses 2) 218 patients with uncomplicated HT starting metoprolol therapy 8 wk

8 UM: *1/*1xN, *1xN/*2,

*1/2*N, *2xN/*10, *2xN/*2

184 EM: *1/*1, *1/*2,

*1/*3, *1/*4, *1/*5, *1/*6,

*1/*10, *1/*17, *1/*41,

*1/*4xN, *1/*10xN,

*4/*17xN, *2/*2, *2/*4,

*2/*4xN, *2/*5, *2/*10,

*2/*17, *2/*41, *10/*17,

*10xN/*41, *17/*17,

*17/*41, *17/*41

15 IM: *3/*17, *4/*17,

*4/*10, *4/*41, *5/*10,

*5/*17

11 PM: *3/*4, *4/*4, *4/*5,

*4/*6

Starting dose 100 mg/d metoprolol IR for 2 wk, titration up to 200 mg/d or MTD for 6 wk if blood pressure > 120/70 mmHg and HR > 55 BPM

Treatment interrupted with 50 mg decreases every 2 d until stopped

• Significant differences in changes for HR from baseline values when comparing all 4 phenotypes (P < .0001), but not for SBP (P = .91) and DBP (P = .37)
• No difference in AE frequencies between all genotypes, including bradycardia (P = .87). No difference in the type of AE observed (P = .41)
• No difference in metoprolol mean doses between all genotypes (P = .77). Only 5% of all subjects (9 EM, 1 IM) did not reach the maximum recommended dose of 200 mg/d
Hamelin et al. (2000)42 Prospective, randomized, double-blind, placebo-controlled, 2-period crossover study 16 healthy male subjects

Blood and urine samplings collected 48 h after metoprolol dose

10 EM: *1/*1, *1/*5

6 PM: *4/*4, *4/*5

100 mg single dose metoprolol IR on d 3

Coadministration of 50 mg/d diphenhydramine, or placebo for 5 d, for 2 periods

Baseline measurements:
• Exercise HR profiles were more decreased for PM than for EM (P < .0009) and persisted from 3 to 12 h postdose. After 12 h, HR was decreased by 14% ± 2 for PM taking only metoprolol, compared to 5% ± 7 for EM.
• Decrease in SBP during exercise initially more pronounced for PM than for EM compared to baseline (P < .0001). After 12 h, PM still had a decrease of 14–17%, whereas EM returned to baseline values.
• Decrease in HR more pronounced between 8 and 12 h for PM than for EM (P < .05)
Kirchheiner et al. (2004)41 Prospective, nonrandomized, controlled study 29 heathy volunteers Blood samples collected for 24 h after single dose

12 UM: *1-*2/*2x1, *1-

*2/*2x2, *1-*2/*2x35, *9-

*10/*2x1, *9-*10/*2x2, *9

-*10/*2x35, *35/*2x1,

*35/*2x2, *35/*2x35,

*41/*2x1, *41/*2x2,

*41/*2x35

13 EM: 2 any combination

of alleles *1, *2, *9, *10,

*35, and *41

4 PM: *3-*6/*3-*6

100 mg single dose metoprolol IR • Marginal effect on resting HR between all phenotypes, but significant (P = .03)
• Significant reductions for PM compared to EM (P = .027) and UM (P = .017) when evaluating exercise-induced heart rate differences, but not between EM and UM phenotypes
• No significant effect on resting DBP and SBP between any phenotypes
Koytchev et al. (1998)39 Prospective, randomized, 3-period cross-over, bioequivalence study 36 healthy Caucasians Total 15 d

16 EM: *1/*1

6 IM: 1/*4

200 mg/d metoprolol CR/XL • Resting HR decreased more significantly for IM than for EM compared to baseline (P < .05)
22 subjects included for pharmacogenetic analyses

Phenotypes attributed

according to DPWG

guidelines

• No significant differences for resting SBP and DBP between IM and EM, even though a trend was observed.
Luzum et al. (2017)27 Retrospective chart-review from the Ohio State University-Coriell personalized medicine collaborative

102 patients with systolic HF

33 patients treated with metoprolol

Median 5.3 y

Median 1.7 y for the duration of treatment for patients taking metoprolol

Comparison of 3

classification schemes for

genotype-inferred

phenotypes;

*4 allele carriers: 23 EM, 9

IM, 1 PM

CPIC: 28 EM, 4 IM, 1 PM

DPWG: 21 EM, 11 IM, 1

PM

Patients taking metoprolol:

Titration of normalized* metoprolol CR/XL (n = 32) or IR (n = 1)

*normalization of metoprolol daily maintenance dose defined as [carvedilol dose x 4 = metoprolol dose]

Comedications permitted, including CYP2D6-interacting drugs

• CYP2D6 genotype-inferred phenotype significantly associated with decreased maintenance dose of metoprolol for both single and double CYP2D6*4 allele carriers compared to noncarriers (P = .023, OR = 0.13; 95% CI = [0.02–0.75])
• Significant trend also observed for the DPWG classification (P = .014, OR = 0.18; 95%CI = [0.03–0.66])
• Nonsignificant differences in dosing regimens observed for the CIPC classification (P = .12, OR = 0.19; 95% CI = [0.24–1.58])
• Regardless of the method of phenotype classification (DPWG, CIPC, *4 polymorphism), none of the PM or IM subjects achieved the recommended target metoprolol dose for treating systolic HF
Nozawa et al. (2005)33 Prospective, nonrandomized, comparative study

72 patients with various cardiovascular diseases (AF, IHD, HT, hypertrophic cardiomyopathy) on routine treatment with metoprolol or bisoprolol for ≥2 wk

38 patients treated with metoprolol

Baseline values taken 3 and 4 h after dosing

30 EM: *1/*1, *1/*10

8 IM: *10/*10

Homozygous and

heterozygous EM analyzed

separately

Patients taking routine metoprolol IR

Initiation of metoprolol or bisoprolol treatment at least 2 wk before the beginning of study

Dosing regimens not titrated during study

Baseline measurements for patients taking metoprolol:

• Dosing regimen differences nonsignificant between all 3 phenotypes
Rau et al. (2002)25 Prospective, cross-sectional study 91 Caucasians with various cardiovascular diseases (HT, CAD, HF, MI), ranging from NYHA I-III Median 12.6 mo

52 EM: *1/*1, *1/*2, *1/*9,

*1/*41, *2/*2, *2/*41

31 IM: *1/*0, *2/*0, *9/*0,

*10/*0, *41/*0, *41/*41

8 PM: *0/*0

*0 includes null alleles *3,

*4, and *6

Phenotypes attributed according to DPWG guidelines

47.5 mg/d median metoprolol CR/XL

Dose titration based on clinical grounds, individual daily drug doses constant for at least 2 wk before blood sampling

78 patients treated with concomitant cardiovascular drugs

• No difference and wide variability between groups in daily drug doses (P = .79)
Rau et al. (2009)24 Prospective, nonrandomized, longitudinal study

91 naïve β-blocker patients with various cardiovascular diseases (AF, CAD, HF, HT), all treated with metoprolol

Different cohort from Rau et al. (2002)25

90 d

63 EM: 2 fully functional, or

1 fully and 1 partly

functional alleles

8 IM: *0/*10, *0/*41

17 PM: *0/*0

*0 includes null alleles *3,

*4, and *6

Patients regrouped as PM

(17) and non-PM (71)

47.5 mg/d mean dose of metoprolol CR/XL for PM and non-PM

Titration allocated on medical grounds

Other cardiovascular comedications allowed during study

• Dose considered too low to witness any disparities in side effects. However, 14 PM out of 71 non-PM subjects (20%) had doses increases during treatment (P < .004), compared to 1 in 17 (6%) for PM subjects (P = .99)
• Average daily dose was not significantly different for the 2 groups (P = .73)
• PM had significantly higher decreases compared to non-PM in resting HR (P = .013), and resting DBP (P = .007), but no significant differences observed for resting SBP (P = .36)
• No difference of all-type AE between groups (P = .55), but significantly more occurrences of bradycardia among PM (log rank P = .0001)
• More PM reduced their resting HR to <60 BPM and had bradycardia compared to non-PM (P < .001)
• Differences in plasma concentrations were associated with significantly and persistently enhanced drug effects between phenotypes
Sharma et al. (2005)43 Prospective, randomized, double-blind, placebo-controlled, 2-period crossover study 20 healthy premenopausal women Total 2 mo

16 EM: *1/*1, *1/*3, *1/*4,

*1/*5

4 PM: *4/*4, *4/*5

100 mg single dose metoprolol IR on d 3

Coadministration of 50 mg/d diphenhydramine, or placebo for 5 d, for 2 periods

Baseline measurements:
• No significant difference between the profile of PM and EM in terms of stroke volume index (P > 0.05). However, the exercise cardiac index appeared different (P = .009)
• Exercise HR and exercise rate-pressure products were significantly different between PM and EM (P < .05)
Sharp et al. (2009)29 Multi-centre, prospective, nonrandomized, open-label study 52 patients with systolic HF (NYHA II-IV, LVEF ≤45%) not previously on metoprolol therapy

Blood sampling performed at least 2 wk after maximum tolerated dose achieved

Specific duration not mentioned

27 EM: *1/*1

22 IM: *1/*4

3 PM: *4/*4

Phenotypes attributed

according to DPWG

guidelines

23.75 mg/d median initial metoprolol CR/XL, doubled every 2–4-wk intervals titration occurred until MTD of 190 mg/d was achieved (median MTD 95 mg/d)

Other treatments permitted during the study

• Nonsignificant differences between EM and IM for percentage change of HR (P = .71), and between all 3 phenotypes for percentage change of SBP (P = .79), and DBP (P = .40)
• Nonsignificant AE-related withdrawals between all phenotypes (P = .79)
• Nonsignificant difference in the number of patients who achieved MTD between all phenotypes (P = .26)
• Proportions not statistically different between patients with 1 or 2 functional alleles that improved condition with metoprolol therapy (P = .71)
Terra et al. (2005)35 Multi-centre, prospective study 61 naïve β-blocker users, with symptomatic ischaemic or nonischaemic HF (NYHA II-III, LVEF ≤40%) 51 patients genotyped 10 wk

37 EM: *1-*2/*1-*2, *1-

*2/*3, *1-*2/*4, *1-*2/*6,

*1-*2/*9, *1-*2/*10, *1-

*2/*17, *1-*2/*29, *1-

*2/*41

10 IM: Any combination of

2 reduced function alleles

*9, *10, *17, *29, *41, or 1

reduced function allele with

another null allele *3, *4, *6

4 PM: *3-*4/*3-*4, *3-

*4/*6, *6/*6

25 mg/d for patients NYHA II, and 12.5 mg/d metoprolol CR/XL for patients NYHA III as starting dose. Titration biweekly until 200 mg dose achieved, or MTD

Other comedications allowed during the study

• No differences between all groups in cardiac decompensation rates, which was the composite criteria of death, HF hospitalization, increase in other HF medications, or need to discontinue metoprolol (P = .61)
• Final daily dose differences nonsignificant (P = .45)
Wuttke et al. (2002)36 Retrospective, pharmacovigilance census by means of standardized questionnaires 24 German subjects treated with metoprolol 22 mo for recruitment

8 EM: *1/*1, *1/*2, *1/*41

7 IM: *1/*4, *1/*5, *2/*3,

*2/*41, *41/*4

9 PM: *3–5/*4–6

47.5 mg/d median metoprolol IR dose at time of AE

Dosing regimens titrated and based on clinical grounds

• Patients who had AE tended to be 5 times more likely to be PM, yet the till AE occurrence, or the time required for dose reduction were nonsignificant between PM and non-PM (P = .29; P = .26)
Patients divided into PM (9) and non-PM (15) • Nonsignificant differences in daily metoprolol doses and between EM and PM (P = .51)
• Trend observed for bradycardia occurrences when comparing PM to non-PM, but nonsignificant (P = .089)
Yuan et al. (2008)40 Prospective, observational, randomized study

300 Chinese Han patients with essential HT and homozygous Arg389 carriers for the ADRB1 gene

276 patients included in the analyses

8 wk

Group A:

40 EMA: *1/*1, *1/*2,

*2/*2

43 IMA: *1/*5, *2/*5,

*1/*10, *2/*10

60 PMA: *5/*5, *5/*10,

*10/10

Group B (same genotypes):

38 EMB

27 IMB

68 PMB

Subgroups A (3), fixed dose: 100 mg/d metoprolol (PMA, IMA, EMA)

Subgroups B (3), ascending doses: PM: 25 mg/d (PMB) IM: 50 mg/d (IMB) EM: 100 mg/d (EMB)

• Significant difference in resting SBP observed between IMA and EMA (P = .027), and between PMA and EMA (P = .046)
• Differences in resting DBP decreased more for PMA than for EMA compared to their respective baseline values (P = .022)
• No difference of resting SBP and DBP between EMB, IMB and PMB (statistical analysis not shown)
• Resting HR for all 6 groups were significantly decreased compared to before treatment; differences between phenotypes not assessed, but values of HR reduction and final HR were all similar. Ascending doses were given according to CYP2D6 phenotype.
Zineh et al. (2004)38 Prospective, nonrandomized study 50 patients with uncomplicated HT 4 wk

42 EM: Any combination

of alleles *1, *2, *9, *10,

*17, *29, *41, *45, and

*46

4 IM: *3-*6/*9, *3-

*6/*10 ¸*3-*6/*17 ¸*3-

*6/*29 ¸*3-*6/*41 ¸*3-

*6/*45, *3-*6/*46

4 PM: *3-*6/*3-*6

EM patients initially

divided into subgroups

(high EM, medium EM,

low EM), but

subsequently regrouped to

create a common general

phenotype for quantitative

analyses.

Start 100 mg/d metoprolol, titration doubled doses weekly until 1 of the following criteria was achieved: Clinical DBP was reduced to ≤90 mmHg or till patient had side effects that precluded upward titration of drug, or 400 mg/d achieved

Other comedications allowed during study, including CYP2D6 inhibitors

Metoprolol formulation not mentioned

• Based on quartile data of S-metoprolol AUCs and CYP2D6 activity scores, no difference was observed for general and dose-limiting AE
• No differences for changes in DBP between all S-metoprolol AUC quartiles, but differences in DBP not reported directly for any phenotype
• Total daily metoprolol doses not significantly different between phenotypes (P = .74)
  • AE; all-type adverse events AF, atrial fibrillation; AUC, area under the concentration–time curve; AMI, acute myocardial infarction; BPM, beats/min; CAD, coronary artery diseases; CHF, congestive heart failure; CPIC, Clinical Pharmacogenomics Implementation Consortium; CR/XL, controlled-release/extended-release metoprolol formulation; DBP, diastolic blood pressure; DPWG, Dutch Pharmacogenomics Working Group; EM, extensive metabolizers; HF, heart failure; HR, heart rate; HT, hypertension; IHD, ischaemic heart diseases; IM, intermediate metabolizers; IR, immediate-release metoprolol formulation; LVEF, left ventricular ejection fraction; MI, myocardial infarction; MTD, maximum tolerated dose; NYHA, New York Heart Association; PM, poor metabolizers.

3.2 Meta-analyses of clinical parameters

3.2.1 Average daily doses

Fifteen studies were available to review the qualitative differences between the daily titrated metoprolol dosing across phenotypes.23-37 Titration schemes varied and starting doses differed based on diagnosis. Forced titration by ascending doses was initiated according to fluctuating time periods and on patients' needs until a maximum tolerated dose or the final target dose had been achieved, which averaged between 47.5 and 200 mg daily. Generally, studies observed that PM participants did not reach target dose as often as non-PM, but those differences were not consistent across studies.

Our meta-analyses of daily metoprolol drug dosing differences between CYP2D6 inferred phenotypes included 8 studies and 789 individuals (Figure 2).24, 25, 28, 29, 32, 35, 36, 38 Average daily dose values were calculated using Wan et al.’s formulas for 5 studies.24, 25, 29, 32, 36 The difference in means (mg/d) was nonsignificant when comparing PM and non-PM (mean difference = −2.12 [95%CI: −14.26 to 10.03]; P = .202). No significant heterogeneity was observed in our main analysis, with I2 reaching 28% (P = .20). For pairwise comparisons, all phenotypes had similar dosages.

Details are in the caption following the image
Meta-analysis of daily drug doses between poor metabolizers (PM) and non-PM24, 25, 28, 29, 32, 35, 36, 38

3.2.2 Heart rate reduction

A total of 9 studies assessed the HR reduction at rest between the genotype-inferred phenotypes.24, 26, 28, 29, 32, 37, 39-41 Almost all studies showed significant differences between at least 2 phenotypes. We also found 3 studies in which exercise-induced HR differences had been measured,41-43 all of which showed significant differences (Table 1).

A total of 5 studies comprising 750 individuals were included in the meta-analysis (Figure 3).24, 26, 28, 32, 41 We observed that PM had a significantly greater reduction compared to non-PM when assessing the mean differences in beats/min (mean difference = 3.16 [95%CI: 0.94 to 5.37]; P = .017). After removal of 1 fixed-dose study, the same conclusions were obtained when comparing PM against non-PM (P = .044). No heterogeneity was observed in our main analysis (P = .89). The only 2-by-2 comparisons that reached significant differences were EM vs PM (P = .029), and UM vs PM (P = .048).

Details are in the caption following the image
Meta-analysis of reduction in heart rate between poor metabolizers (PM) and non-PM24, 26, 28, 32, 41

3.2.3 Blood pressure reduction

Eight studies published data on resting ΔSBP and ΔDBP between CYP2D6 genotype-inferred phenotypes.24, 26, 28, 29, 32, 37, 39-41 In all the studies reviewed, patients were initially β-blocker naïve. Our systematic review showed inconsistent results across studies for both parameters. Only 1 of the studies that had examined exercise-induced HR variations also reported values for BP reduction after exercise.42 In that study, 12 hours after a single 100 mg metoprolol dose, PM still had a decrease of 14 to 17%, whereas EM had returned to baseline.

Five studies including 842 individuals suitable for meta-analysis were analysed to determine the effect of CYP2D6 genotype-derived phenotypes on resting ΔSBP (Figure 4).24, 26, 28, 32, 40 Our results indicated a significantly greater reduction (mmHg) for PM against non-PM (mean difference = 2.88 [95%CI: 1.47 to 4.29]; P = .0048), which was also observed for titrated-dose studies only (P = .031). No significant heterogeneity was observed in this meta-analysis (P = .99). EM against PM was the only pairwise comparison where statistical significance was achieved (mean difference = 4.15 [95%CI: 0.18 to 8.12]; P = .030), which was preserved when removing the fixed-dose study (P = .037).

Details are in the caption following the image
Meta-analysis of reduction in systolic blood pressure between poor metabolizers (PM) and non-PM24, 26, 28, 32, 40

As for ΔDBP, the same 5 studies were included in our meta-analyses (Figure 5). Significant results were observed between non-PM and PM (mean difference = 2.93 [95%CI: 1.53 to 4.32]; P = .0043). Again, no significant heterogeneity in this meta-analysis, with I2 = 0% (P = .91). By excluding the 1 fixed-dose study, results remained significant (P = .021), with PM showing greater reduction than other phenotypes. IM and PM showed significantly greater reductions compared to EM when including all studies (EM vs IM: P = .014; EM vs PM: P = .014). Titrated-dose studies also showed that the phenotype with less metabolic capacity had a greater ΔDBP, in both comparisons. For fixed-dose studies, comparisons involving EM and IM showed no statistical differences (P = .150). As was the case for ΔSBP, our quantitative analysis also showed no differences in ΔDBP between UM and other phenotypes.

Details are in the caption following the image
Meta-analysis of reduction in diastolic blood pressure between poor metabolizers (PM) and non-PM24, 26, 28, 32, 40

3.2.4 All-type adverse events and bradycardia

In total, 6 studies reported the frequency or the number of all-cause adverse reactions, either dose-limiting or not.24, 26, 29, 32, 36, 38 Five studies specifically monitored the frequency of bradycardia,24, 26, 28, 36, 37 which was uniformly defined as a HR < 60 beats/min. Two studies reported symptomatic bradycardia and all bradycardia events, while 2 reported bradycardia without specifying whether these events were symptomatic. All studies reported the use of multiple doses.

Only 3 studies including 356 individuals were pooled to evaluate the frequency of all-type AE.24, 26, 38 Considerable variability was observed for each individual study regarding confidence intervals. Neither PM or non-PM were more prone to adverse reactions due to their CYP2D6 phenotype (OR: 0.76 [95%CI: 0.33 to 1.74]; P = .514; Figure 6). No heterogeneity was found in this analysis (I2 = 0%, P = .59) Pairwise comparisons also did not yield significant results for any group.

Details are in the caption following the image
Meta-analysis of all-type adverse events between poor metabolizers (PM) and non-PM24, 26, 38

Values from 4 studies regrouping 643 individuals were used for meta-analyses of bradycardia.24, 26, 28, 36 Given that only 2 studies reported symptomatic bradycardia, and that the number of events was low in the 2 studies, we limited the meta-analyses to all cases of bradycardia. The frequency of events was significantly reduced for non-PM compared to PM (OR: 0.27 [95%CI: 0.08 to 0.89]; P = .032; Figure 7) The effect was more important in the 2 smaller studies, as opposed to the 2 larger studies, resulting in significant heterogeneity. As opposed to our previous comparisons, heterogeneity was high in this analysis (I2 = 68%, P = .03). Two-by-two meta-analyses showed that only EM were significantly less prone to developing bradycardia during treatment compared to IM (OR: 0.62 [95%CI: 0.40 to 0.96]; P = .032), but no other pairwise comparisons indicated a significant reduction of bradycardia for any phenotype. Again, UM were excluded from our meta-analyses for lack of values.

Details are in the caption following the image
Meta-analysis of bradycardia between poor metabolizers (PM) and non-PM24, 26, 28, 36

3.2.5 Mortality and hospitalizations

Hospitalizations, mortality and other composite criteria reflecting outcome events were only evaluated once in the retrospective substudy of the MERIT-HF trial.28 Thus, no meta-analysis was done for this parameter. As part of this study, no effect on all-cause mortality and hospitalizations was observed, and early discontinuation was similar between EM, IM and PM.

3.2.6 Sensitivity analyses

The comparisons of the results for the meta-analyses performed with both the Hartung–Knapp and the DerSimonian–Laird methods are presented in Table S7. For the PM vs non-PM meta-analyses, the effects were consistent across all analyses, although the ΔSBP meta-analysis was not significant using the DerSimonian–Laird method (P = .093). We initially sought to assess publication bias by means of funnel plot and asymmetry,44 yet no parameter reached the minimum requirement of 10 included studies for such analyses to be conducted.

4 DISCUSSION

We performed a qualitative systematic review and meta-analyses to assess the effects of CYP2D6 polymorphism on metoprolol pharmacodynamics. Our meta-analyses suggest that CYP2D6 PM have a greater reduction in HR compared to non-PM by >3 beats/min when treated with metoprolol. This enhanced β-blocking effect may also lead to PM being approximately 4 times more at risk of bradycardia, although this result should be interpreted with caution because of significant heterogeneity observed for this endpoint, suggesting an overestimated effect size in smaller studies. Also, given that the doses used in the reviewed studies were similar between phenotypes, these results are therefore consistent with a previous meta-analysis,13 indicating that PM present higher metoprolol plasma concentrations than non-PM when treated with a fixed metoprolol dose, resulting in greater reductions in HR and blood pressure. Both systolic and diastolic blood pressures were also significantly more reduced for PM, with average decreases both greater than non-PM by ~3 mmHg. Larger populations than those included in our meta-analyses will be required to investigate the impact of CYP2D6 phenotypes on other AE and reductions in cardiovascular endpoints across all 4 inferred phenotypes.

The clinical significance of our findings requires further investigation. Indeed, although our results suggest that PM could be at a higher risk of bradycardia, one could also argue that the greater HR reduction in PM (3 beats/min) could result in a greater benefit from metoprolol. Indeed, a previous meta-analysis of 17 randomized, placebo-controlled trials of β-blocker combining 17 831 patients with heart failure, indicated a decrease in relative risk of death by 18% per 5 beats/min (95%CI: 6 to 29%) for patients undergoing β-blocker treatment,45 which is slightly above the differences in reduction between non-PM and PM. The clinical benefits in resting HR reduction have also been demonstrated during post-myocardial infarction intervention, notably through β-blocker use.46 Hence, PM patients would benefit more than other phenotypes from receiving metoprolol treatment at similar doses, as long as symptomatic bradycardia is avoided. One could also speculate that using higher metoprolol doses in non-PM, or using other non-CYP2D6 metabolized β-blockers, could lead to comparable efficacy. For example, in the pharmacogenetic sub-study of the MERIT-HF trial, the authors concluded that to achieve a 1-beat reduction of HR, 7.7, 5.9 and 5.3 mg of metoprolol CR/XL were needed for EM, IM and PM, respectively, which was significantly different between phenotypes (P = .04). Thus, our results are consistent with the DPWG recommending initiation of metoprolol at lower doses for PM, given the apparent greater response and potentially higher risk of bradycardia. Nevertheless, since a greater HR response could not only be associated with a greater risk of bradycardia, but also with a greater clinical benefit,45 the optimal metoprolol dosing algorithm based on CYP2D6 genotyping and its clinical utility remains to be established.

We found a statistically significant increase of bradycardia amongst PM compared to non-PM, although significant heterogeneity was observed in the meta-analysis. However, these results are consistent with those from a report by Bijl et al37 in which PM were more likely to suffer from bradycardia than EM (OR: 3.86 [95%CI: 1.68 to 8.86]; P = .0014). Unfortunately, we could not include their data in the current meta-analysis because specific sample sizes and values in each group were not reported. Still, the 4-fold difference in the risk of bradycardia is consistent with the risk observed in the current meta-analysis. Given that only 2 studies specifically reported symptomatic bradycardia, we could not determine whether the differences in frequencies of bradycardia would warrant changes in medication or dosing for PM. Thus, particularly given the low incidence of symptomatic bradycardia in the 2 studies reporting it, future pharmacogenetic investigations on this gene–drug pair should precisely document the occurrence of symptomatic bradycardia.

Beta-blockers, including metoprolol, are known to have modest blood pressure lowering effect.8 Nonetheless, the results of this meta-analysis indicate that CYP2D6 genotype-inferred phenotypes could be useful to guide the selection of β-blocker in the treatment of hypertension. Indeed, even if individual studies did not consistently show significant differences between inferred phenotypes, our meta-analysis showed that both reductions in SBP and DBP are greater in PM compared to other phenotypes by approximately 3 mmHg. Although such a difference might appear modest, evidence suggests that from a populational standpoint, a DBP reduction of only 2 mmHg would lead to a decrease of stroke by 15%.47 According to recent estimates, an equally small SBP reduction would also significantly reduce the annual incidence of coronary heart disease, stroke and heart failure events.48

5 STUDY LIMITATIONS

Our meta-analysis contains some limitations. For this systematic review, we used the DPWG guidelines to assign a phenotype to patients in studies for which only their CYP2D6 genotypes had been mentioned. However, others could have favoured the classification proposed by the Clinical Pharmacogenomics Implementation Consortium. Since we focused our attention on comparisons between PM and non-PM, this would have minimal impact on our conclusions because discrepancies between the 2 groups are mainly related to EM and IM.49

In addition, many of the included studies only investigated a limited number of CYP2D6 polymorphisms. As different polymorphisms may be present within specific populations,7 limited genotyping assays could prove problematic with this highly polymorphic gene. These technical issues have been addressed prior to this review.4, 50 However, such shortcomings would have added heterogeneity to our analyses. Therefore, this limitation should not restrain the scope of our findings.

From a statistical perspective, an important limitation in our review is the small number of studies, and the small and unbalanced number of patients within the compared groups. Thus, whether CYP2D6 inferred phenotypes has an impact of the overall risk of AE or cardiovascular events remains to be established. A greater number of patients would also be required to perform adequately powered comparisons between all 4 phenotypes.

A strength of our study is that our sensitivity analyses for continuous variables show that results generally remain consistent between the 2 methods for calculating confidence intervals. The Hartung–Knapp adjustment has been shown to be a useful tool in making better inferences and refining measurements when performing meta-analyses.20 Despite those improvements, confidence intervals for random-effects models can be overly conservative in some scenarios, while becoming too restrained in others.20, 21 This could explain the fact that reductions in SBP were not statistically significant using the DerSimonian–Laird method, as confidence intervals calculated were wider. Although we were able to perform meta-analyses on almost all parameters reviewed, comparisons where ≤3 studies were included may contain considerable uncertainty, regardless of low heterogeneity. Indeed, some small studies with neutral results have not reported all relevant data, thus potentially overestimating the effects observed.38 While our systematic review shows certain trends that seem likely to be confirmed, careful assessment of our results is required.

To conclude, this systematic review provides a comprehensive assessment of the literature regarding the genetic influence of CYP2D6 polymorphisms on metoprolol response. Our meta-analyses suggest that patients with a PM phenotype have greater HR, SBP and DBP reductions compared to non-PM, and may be more subject to bradycardia. This latter finding should be interpreted with caution, given the heterogeneity observed in the analysis of this specific end point. Further prospective data are required to determine whether CYP2D6 is associated with clinical events in patients treated with metoprolol. Ultimately, the clinical utility of CYP2D6-guided β-blocker use should be investigated in a prospective clinical trial.

ACKNOWLEDGEMENTS

Maxime Meloche has received scholarships from the Université de Montréal Faculty of Pharmacy and Faculty of Higher Education.

Simon de Denus holds the Université de Montréal Beaulieu-Saucier Chair in Pharmacogenomics. This study was also supported by the Montreal Heart Institute Foundation.

COMPETING INTERESTS

M.-P.D. is a coauthor on a patent pertaining to pharmacogenomics-guided CETP inhibition and has minor equity interest in DalCor. M.-P.D. has received honoraria from Dalcor and research support (access to samples and data) from AstraZeneca, Pfizer, Servier, Sanofi and GlaxoSmithKline. S.D. has received research support through grants from Pfizer, AstraZeneca, Roche Molecular Science, and DalCor.

CONTRIBUTORS

M.M. performed the systematic review, generated the statistical meta-analyses, and wrote parts of the manuscript. M.K. was the second reviewer and wrote parts of the manuscript. I.K. developed the computer code used in the R software to create the meta-analyses. A.B. contributed to elaborate the statistical models and revised the manuscript. M.-P.D. Dubé contributed to the design of the analyses and revised the manuscript. S.D. designed the project, wrote parts of and revised the manuscript.