This systematic review and meta-analysis quantifies the difference in the antipsychotic and antidepressant exposure among patients with genetically determined CYP2C19 and CYP2D6 poor, intermediate, and normal metabolism.
Key Points
Question
What is the difference in the expected antipsychotic and antidepressant exposure between genetically associated CYP2C19 and CYP2D6 poor (PM), intermediate (IM), and normal (NM) metabolism?
Findings
A systematic review and meta-analysis of 94 unique studies and 8379 unique patients quantified the increases of risperidone, aripiprazole, and haloperidol exposure in patients with CYP2D6 PM and IM status and increases of escitalopram and sertraline exposure in patients with CYP2C19 PM and IM status as compared with patients with the NM group.
Meaning
The obtained results represent a scientific foundation for CYP2D6/CYP2C19 genotype-based dosing recommendations that could potentially lead to improved clinical outcome in drug treatment for patients with psychiatric disorders.
Abstract
Importance
Precise estimation of the drug metabolism capacity for individual patients is crucial for adequate dose personalization.
Objective
To quantify the difference in the antipsychotic and antidepressant exposure among patients with genetically associated CYP2C19 and CYP2D6 poor (PM), intermediate (IM), and normal (NM) metabolizers.
Data Sources
PubMed, Clinicaltrialsregister.eu, ClinicalTrials.gov, International Clinical Trials Registry Platform, and CENTRAL databases were screened for studies from January 1, 1990, to June 30, 2020, with no language restrictions.
Study Selection
Two independent reviewers performed study screening and assessed the following inclusion criteria: (1) appropriate CYP2C19 or CYP2D6 genotyping was performed, (2) genotype-based classification into CYP2C19 or CYP2D6 NM, IM, and PM categories was possible, and (3) 3 patients per metabolizer category were available.
Data Extraction and Synthesis
The Meta-analysis of Observational Studies in Epidemiology (MOOSE) guidelines were followed for extracting data and quality, validity, and risk of bias assessments. A fixed-effects model was used for pooling the effect sizes of the included studies.
Main Outcomes and Measures
Drug exposure was measured as (1) dose-normalized area under the plasma level (time) curve, (2) dose-normalized steady-state plasma level, or (3) reciprocal apparent total drug clearance. The ratio of means (RoM) was calculated by dividing the mean drug exposure for PM, IM, or pooled PM plus IM categories by the mean drug exposure for the NM category.
Results
Based on the data derived from 94 unique studies and 8379 unique individuals, the most profound differences were observed in the patients treated with aripiprazole (CYP2D6 PM plus IM vs NM RoM, 1.48; 95% CI, 1.41-1.57; 12 studies; 1038 patients), haloperidol lactate (CYP2D6 PM vs NM RoM, 1.68; 95% CI, 1.40-2.02; 9 studies; 423 patients), risperidone (CYP2D6 PM plus IM vs NM RoM, 1.36; 95% CI, 1.28-1.44; 23 studies; 1492 patients), escitalopram oxalate (CYP2C19 PM vs NM, RoM, 2.63; 95% CI, 2.40-2.89; 4 studies; 1262 patients), and sertraline hydrochloride (CYP2C19 IM vs NM RoM, 1.38; 95% CI, 1.27-1.51; 3 studies; 917 patients). Exposure differences were also observed for clozapine, quetiapine fumarate, amitriptyline hydrochloride, mirtazapine, nortriptyline hydrochloride, fluoxetine hydrochloride, fluvoxamine maleate, paroxetine hydrochloride, and venlafaxine hydrochloride; however, these differences were marginal, ambiguous, or based on less than 3 independent studies.
Conclusions and Relevance
In this systematic review and meta-analysis, the association between CYP2C19/CYP2D6 genotype and drug levels of several psychiatric drugs was quantified with sufficient precision as to be useful as a scientific foundation for CYP2D6/CYP2C19 genotype-based dosing recommendations.
Introduction
The efficacy of psychiatric drugs is suboptimal; however, because the development of new antipsychotics and antidepressants is slow, it is of paramount importance to use the currently available drugs as effectively as possible. An important aspect of effective use is dose personalization because, owing to interindividual differences in drug metabolism, the dose required to achieve optimal blood levels of antidepressants and antipsychotics varies substantially between patients.1 Recently published meta-analyses2,3 focused on dose-response curves for antipsychotics and antidepressants supported the claim that the appropriate dosing is important for maximizing the efficacy and tolerability of these drugs. In addition, according to recently published data on more than 5000 patients,4,5,6 when treated with escitalopram oxalate, 10 mg/d, sertraline hydrochloride, 100 mg/d, risperidone, 4 mg/d, or aripiprazole, 20 mg/d, more than one-third of the patients exhibit blood drug levels outside the therapeutic concentration window defined for these drugs.1 Therefore, although these daily doses fit an average patient well, there is an apparent need to personalize the dose and maximize the treatment response beyond population-based dosing.
Most antipsychotics and antidepressants are metabolized by the polymorphic CYP2C19 and CYP2D6 enzymes,1 and their capacity is genetically determined.7,8 First, normal metabolizers (NM category) have normal enzymatic capacity and carry homozygous wild-type (Wt) alleles; they may also carry other genotypes if the enzymatic capacity is not significantly different compared with Wt/Wt carriers. Second, CYP2C19/CYP2D6 genotype-determined poor metabolizers (PM category) carry homozygous loss-of-function alleles and do not possess the active enzyme. Third, CYP2C19/CYP2D6 genotype-determined intermediate metabolizers (IM category) carry genotypes connected with substantially reduced but not abolished enzymatic capacity. Finally, CYP2C19/CYP2D6 genotype-determined ultrarapid metabolizers (UM category) carry genotypes connected with higher-than-normal enzymatic capacity. All these phenotypes are present in substantial proportion worldwide (Table 1).9
Table 1. Allele Frequencies of Variant CYP2C19 and CYP2D6 Genes Among Different Populations Worldwidea.
Genotype-based phenotype by metabolism category | Population, % | ||||
---|---|---|---|---|---|
European | African | East Asian | South Asian | Admixed American | |
CYP2C19 | |||||
PM | 3.3 | 3.3 | 14.2 | 11.8 | 1.1 |
IM | 21.7 | 21.2 | 45.8 | 35.8 | 16.0 |
PM plus IM | 25.0 | 24.6 | 60.1 | 47.6 | 17.1 |
NM | 43.4 | 42.5 | 38.1 | 36.4 | 62.8 |
UM | 31.6 | 32.9 | 1.8 | 16.0 | 20.1 |
CYP2D6 | |||||
PM | 6.2 | 2.8 | 0.7 | 2.1 | 3.8 |
IM | 2.6 | 24.5 | 48.6 | 10.0 | 2.6 |
PM plus IM | 8.8 | 27.3 | 49.3 | 12.2 | 6.4 |
NM | 88.1 | 64.7 | 49.6 | 85.9 | 91.4 |
UM | 3.2 | 8.0 | 1.2 | 1.9 | 2.2 |
Abbreviations: IM, intermediate metabolizer; NM, normal metabolizer; PM, poor metabolizer; UM, ultrarapid metabolizer.
Data are based on Zhou et al.9 Notable variations also exist within the global regions.
Well-replicated clinical findings indicate that the patients in the PM and IM categories exhibit a substantial increase in the exposure and adverse drug reactions of certain psychotropic drugs,4,5,6,10,11 whereas those in the UM category most often have lower levels of response, owing to faster drug metabolism.4,5,12,13 In addition, recent studies4,5 found that those in the PM and UM categories are more prone to risperidone and escitalopram treatment failure, which was quantified as an increase in the incidence of switching to an alternative antipsychotic/antidepressant within 1 year. The recommended and maximum daily doses are originally designed to fit the mean genotype-weighted population. Thus, the official dosing recommendations for psychiatric drugs usually do not acknowledge the clinical relevance of CYP2C19/CYP2D6 metabolizer categories and do not distinguish between them. Investigators4,5,6 observed, however, that the daily doses of escitalopram, sertraline, risperidone, and aripiprazole, prescribed in naturalistic settings based on clinical observations alone, were lower in individuals in the PM compared with NM categories and that the observed dose reductions were insufficient to fully compensate for the increased drug exposure. In rare cases, as with aripiprazole treatment, relevant sources such as the US Food and Drug Administration, European Medicines Agency, CPIC (Clinical Pharmacogenetics Implementation Consortium), and DPWG (Dutch Pharmacogenetics Working Group) recommend dose reduction for patients in the CYP2D6 PM category; however, these sources offer conflicting information related to the magnitude of dose adjustment. In fact, most of the recommendations are based on underpowered studies, and insufficient data are available to allow the estimation of the difference in drug exposure between metabolizer categories with sufficient precision.14
Many previous studies, often of limited sample size, have investigated the effects of CYP2C19 and CYP2D6 genotype on the exposure of antipsychotic and antidepressant drugs, and recently published reports substantially increased the number of participants undergoing genotyping.4,5,6 Thus, the aim of this systematic review and meta-analysis of prospective and retrospective cohort studies was to quantify, with the best attainable precision, the increase of antidepressant and antipsychotic exposure in individuals in CYP2C19/CYP2D6 PM and IM categories compared with those in the NM category. Individuals in the UM category were not included in the analysis owing to the limited number of studies considering this phenotypic group.
Methods
Search Strategy and Selection Criteria
The list of antipsychotic and antidepressant drugs was based on the list of frequently used antidepressants15 and antipsychotics.16 The investigated antidepressants included escitalopram, sertraline, fluoxetine hydrochloride, fluvoxamine maleate, paroxetine hydrochloride, venlafaxine hydrochloride, amitriptyline hydrochloride, nortriptyline hydrochloride, mianserin, and mirtazapine; the antipsychotics included clozapine, quetiapine fumarate, olanzapine, risperidone, aripiprazole, and haloperidol lactate. Racemic citalopram hydrobromide was not investigated owing to stereoselective metabolism. The information on which CYP450 isoforms are involved in the metabolism of each drug were retrieved from the recent consensus guidelines.1 The search was performed in PubMed, ClinicalTrials.gov, Clinicaltrialsregister.eu, International Clinical Trials Registry Platform, and CENTRAL databases for reports published from January 1, 1990, to June 30, 2020. An independent literature survey was performed for each drug and the search terms *NameOfTheDrug* AND CYP2C19 OR CYP2D6 were used. During the initial screening step, all studies that did not deal with drug exposure were excluded, and the remaining studies were considered for inclusion based on the following criteria: (1) participants were genotyped for all known common functional CYP2C19 or CYP2D6 variant alleles with minor allele frequency of greater than 1% according to Zhou et al9; (2) adequate classification of participants into CYP2C19 and/or CYP2D6 NM, IM, and PM categories was possible based on genotyping; (3) the study included at least 3 participants per experimental group; and (4) drug exposure was measured in a representative way by (a) dose-normalized steady-state plasma levels, (b) dose-normalized area under the plasma level (time) curve, or (c) apparent total clearance of the drug (reciprocal value). The screening and scanning for eligibility were performed manually by 2 independent investigators (F.M. and N.B.). The decision on study inclusion was made by consensus with a third investigator (M.M.J.), with the final checkup made by consensus among 3 (E.M., M.I.-S., and M.M.J.). Six domains were assessed by using the standardized risk of bias in nonrandomized studies of interventions tool,17 and studies with the critical risk of bias were excluded. No restrictions were made regarding the study design, participant characteristics (race, ethnicity, sex, age, smoking status, and patient vs healthy cohort), treatment duration, drug interactions, and language.
Data Extraction
The procedures of data acquisition and extraction, as well as the situations when the authors were contacted to provide the data that were inaccessible, are described in full detail in eMethods 1 in the Supplement. If a drug possesses an active metabolite, the drug exposure was calculated by pooling the parent compound and active metabolite (active moiety) exposure.1 Participants were classified into PM, IM, and NM categories for CYP2C19 and CYP2D6 by using the previously described classification criteria (Table 1).18 Participants in the PM category were homozygous carriers of the 2 loss-of-function (null) alleles for both CYP2C19 and CYP2D6. For CYP2C19, participants in the IM category carried 1 null and 1 Wt allele, whereas those in the NM category carried the CYP2C19 Wt/Wt genotype. The CYP2D6 gene possesses alleles that reduce but do not abolish the enzymatic capacity (Red), and the CYP2D6 IM category consisted of participants carrying either CYP2D6 Red/Null or CYP2D6 Red/Red genotype, whereas the subpopulation in the CYP2D6 NM category carries 1 or 2 CYP2D6 Wt alleles. For the purpose of this study, only the individuals carrying the CYP2D6 Wt/Wt genotype represented the NM reference group, as suggested by the consensus guidelines.18
Statistical Analyses
Meta-analyses were performed in accordance with the Meta-analysis of Observational Studies in Epidemiology (MOOSE) guidelines,19 and the checklist is available in eMethods 2 in the Supplement. Meta-analyses for specific phenotypes/drugs were performed and represented graphically if 3 or more studies met the inclusion criteria. The effect size was the mean exposure of the PM, IM, or PM plus IM groups divided by the mean exposure of the NM group, that is, the ratio of means (RoM).20 For example, an IM:NM group RoM of 1.5 means a 1.5 times higher exposure (ie, a 50% higher exposure in the IM compared with the NM group). Standard mean differences (Hedges g) were also calculated and presented in eFigure 3 in the Supplement. Weighted RoM between subgroups was used in calculation of pooling effect between studies by fixed-effects meta-analysis model. Heterogeneity across the studies was assessed using the Cochran Q test at a given significance level; the percentage of total variability attributable to heterogeneity was quantified by the I2 value. A fixed-effects model was used because all the pooled studies represent the same genetic/biological construct; however, owing to considerable heterogeneity in certain analyses, a post hoc sensitivity analysis was performed by using the random-effects model, and the comparison between the fixed- and random-effects model analyses is presented in eTable 1 in the Supplement. Differences between the effect sizes of PM vs NM and IM vs NM groups were examined by using the subgroup test, and when no difference was observed, a post hoc comparison between the pooled PM plus IM and NM experimental groups was performed. For each individual study, the PM plus IM experimental group exposure was calculated by combining the PM and IM subgroups according to the Cochrane handbook formula (section 6.5.2.10 on combining groups).21
Small trial or publication bias was evaluated using the Egger test for funnel plot asymmetry,22 and funnel plots are presented in the eFigure 5 in the Supplement. Statistical analyses were performed with RevMan, version 5.4, software (Cochrane). Ratios of means for the individual studies were calculated using Excel, version 2013 (Microsoft Corporation), according to the previously published formula,20 and subsequently entered into RevMan with the generic inverse variance option. Two-sided α < .05 indicated statistical significance.
Interpretation of Changes in Drug Exposure
If a lower boundary of the 95% CI for the drug exposure increase of the PM, IM, or PM plus IM groups compared with the NM group was greater than 1.25-fold, such an effect was considered clinically relevant. If this was not the case for a statistically significant effect, such an effect was considered preliminary or marginal. This quantitative cutoff was based on (1) the US Food and Drug Administration limits for bioequivalence (RoM, 0.80-1.25), which are based on the general consideration that the intraindividual variability in drug exposure from oral drug intake to intake is 20%,23 and (2) the previous finding that changes of this magnitude are associated with an increased risk of therapeutic failure, measured by the drug switch rates in 2 recent studies4,5 on patient cohorts treated with escitalopram (n = 2087) and risperidone (n = 890).
Results
Of the 2103 initially screened references, 94 unique studies4,5,6,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114 on 8379 unique individuals met the inclusion criteria. Reasons for exclusion are presented in Table 2 and eTable 2 in the Supplement. eFigure 1 in the Supplement gives the PRISMA115 flow diagram. A list of included and excluded studies are presented in eMethods 3 in the Supplement.
Table 2. Overview of Search Process and Studies Incorporated Into Meta-analyses.
Analyzed drug | Enzyme | No. of studies | No. of excluded studies | No. of included studies | No. of individuals by metabolism categorya | ||||
---|---|---|---|---|---|---|---|---|---|
Not dealing with exposure | Incorrect genotyping | No usable data | NMb | IM | PM | ||||
Antipsychotics | |||||||||
Aripiprazole | CYP2D6 | 84 | 58 | 4 | 10 | 12 | 814 | 134 | 90 |
Clozapine | CYP2D6 | 86 | 78 | 3 | 0 | 5 | 33 | 15 | 4 |
CYP2C19 | 127 | 65 | 8 | ||||||
Haloperidol | CYP2D6 | 109 | 83 | 10 | 3 | 13 | 532 | 158 | 46 |
Quetiapine | CYP2D6 | 45 | 44 | 0 | 0 | 1 | 171 | 0 | 20 |
Risperidone | CYP2D6 | 221 | 163 | 9 | 26 | 23 | 1134 | 186 | 172 |
Antidepressants | |||||||||
Amitriptyline | CYP2D6 | 103 | 94 | 1 | 4 | 4 | 43 | 9 | 4 |
CYP2C19 | 34 | 18 | 6 | ||||||
Escitalopram | CYP2C19 | 147 | 135 | 4 | 4 | 4 | 1110 | 760 | 152 |
Fluoxetine | CYP2D6 | 313 | 305 | 4 | 1 | 3 | 8 | 0 | 3 |
CYP2C19 | 71 | 27 | 6 | ||||||
Fluvoxamine | CYP2D6 | 224 | 212 | 3 | 2 | 7 | 74 | 72 | 0 |
CYP2C19 | 6 | 6 | 6 | ||||||
Mirtazapine | CYP2D6 | 70 | 59 | 2 | 4 | 5 | 142 | 14 | 19 |
Nortriptyline | CYP2D6 | 97 | 87 | 3 | 3 | 4 | 28 | 14 | 4 |
Paroxetine | CYP2D6 | 335 | 318 | 8 | 4 | 5 | 89 | 14 | 11 |
Sertraline | CYP2C19 | 74 | 68 | 1 | 2 | 3 | 565 | 352 | 40 |
Venlafaxine | CYP2D6 | 195 | 170 | 5 | 12 | 8 | 509 | 87 | 120 |
CYP2C19 | 422 | 198 | 21 | ||||||
Total | 2103 | 1874 | 57 | 75 | 94 | 8379b |
Abbreviations: IM, intermediate metabolizer; NM, normal metabolizer; PM, poor metabolizer.
The total number of patients is less than the sum of patients for all phenotypes/drugs owing to the fact that CYP2C19 and CYP2D6 genotyping was performed in certain studies.
Indicates reference category.
Association Between CYP2D6 Metabolizer Status and Drug Exposure
The CYP2D6 genotype was associated with significant exposure increases for aripiprazole5,25,26,27,28,29,30,31,32,33,115,116 (eFigure 2 in the Supplement) (PM plus IM vs NM RoM, 1.48; 95% CI, 1.41-1.57; 12 studies; 1038 patients), haloperidol26,34,35,36 (eFigure 2 in the Supplement) (PM vs NM RoM, 1.68; 95% CI, 1.40-2.02; 9 studies; 423 patients), and risperidone5,26,35,37,38,39,40,41,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57 (eFigure 2 in the Supplement) (PM plus IM vs NM RoM, 1.36; 95% CI, 1.28-1.44; 23 studies; 1492 patients). Nortriptyline exposure58,59,60 (RoM, 2.36; 95% CI, 2.10-2.65; 3 studies; 37 patients) (eFigure 2 in the Supplement) and paroxetine exposure61,62,63 (RoM, 3.50; 95% CI, 2.52-4.85; 3 studies; 41 patients) (eFigure 2 in the Supplement) were significantly increased in the CYP2D6 IM compared with the NM groups; however, after removing the studies associated with serious risk of bias (eResults in the Supplement), these differences were based on fewer than 3 independent studies. It is uncertain whether the exposure increases observed in the fluvoxamine IM group64,65,66,67,68,69 and mirtazapine PM group70,71,72,73 (eFigure 2 in the Supplement) compared with the NM groups are outside the bioequivalence (1.25) limit. Compared with the CYP2D6 NM group, marginal exposure increases were observed in the haloperidol IM group74,75,76,77,78,79,80,81,82 (RoM, 1.14; 95% CI, 105-125; 9 studies; 423 patients) (eFigure 2 in the Supplement) and venlafaxine IM plus PM group (RoM, 1.19; 95% CI, 1.09-1.29; 8 studies; 716 patients)83,84,85,86,87,88,89,90 (eFigure 2 in the Supplement). Statistically significant exposure increases based on less than 3 independent studies compared with the CYP2D6 NM group were observed in the quetiapine-treated PM (RoM, 1.32; 95% CI, 1.10-1.58; 1 study; 191 patients), amitriptyline-treated IM (RoM, 1.50; 95% CI, 1.23-1.84; 2 studies; 35 patients), mirtazapine-treated IM (RoM, 1.39; 95% CI, 1.23-1.57; 4 studies; 144 patients), paroxetine-treated PM (RoM, 5.13; 95% CI, 3.82-6.87; 2 studies; 73 patients), nortriptyline-treated PM (RoM, 3.32; 95% CI, 2.08-5.29; 1 study; 9 patients), and fluoxetine-treated PM (RoM, 2.26; 95% CI, 1.68-2.83; 1 study; 11 patients) groups (Table 3).
Table 3. Detailed Statistical Report of the Association of Metabolism Status With Antipsychotic and Antidepressant Exposure.
Drug | Enzyme | No. of studies | No. of patients by metabolism group | RoM (95% CI) | P value | I2 value, % | |
---|---|---|---|---|---|---|---|
Reference | Comparator | ||||||
Antipsychotics | |||||||
Aripiprazole | CYP2D6 | 5 | 693 NM | 90 PM | 1.51 (1.38-1.65) | <.001 | 0 |
CYP2D6 | 9 | 664 NM | 134 IM | 1.47 (1.38-1.57) | <.001 | 65 | |
CYP2D6 | 12 | 814 NM | 224 PM plus IM | 1.48 (1.41-1.56) | <.001 | 56 | |
Clozapine | CYP2D6 | 1 | 22 NM | 4 PM | 1.00 (0.43-2.32) | >.99 | NA |
CYP2D6 | 2 | 33 NM | 15 IM | 1.22 (0.79-1.88) | .51 | 0 | |
CYP2C19 | 2 | 70 NM | 8 PM | 1.92 (1.32-2.79) | .008 | 0 | |
CYP2C19 | 4 | 127 NM | 65 IM | 1.00 (0.84-1.19) | .84 | 10 | |
Haloperidol | CYP2D6 | 4 | 267 NM | 46 PM | 1.68 (1.40-1.91) | <.001 | 21 |
CYP2D6 | 9 | 265 NM | 158 IM | 1.14 (1.05-1.25) | .003 | 0 | |
Quetiapine | CYP2D6 | 1 | 171 NM | 20 PM | 1.32 (1.10-1.58) | <.001 | NA |
Risperidone | CYP2D6 | 13 | 937 NM | 172 PM | 1.40 (1.30-1.50) | <.001 | 17 |
CYP2D6 | 11 | 469 NM | 186 IM | 1.31 (1.20-1.43) | <.001 | 44 | |
CYP2D6 | 23 | 1134 NM | 358 PM plus IM | 1.36 (1.28-1.44) | <.001 | 34 | |
Antidepressants | |||||||
Amitriptyline | CYP2D6 | 1 | 17 NM | 4 PM | 1.04 (0.65-1.68) | .86 | NA |
CYP2D6 | 2 | 26 NM | 9 IM | 1.50 (1.23-1.84) | <.001 | 0 | |
CYP2C19 | 1 | 4 NM | 6 PM | 1.07 (0.81-1.41) | .58 | NA | |
CYP2C19 | 1 | 30 NM | 18 IM | 1.06 (0.89-1.25) | .50 | NA | |
Escitalopram | CYP2C19 | 4 | 1110 NM | 152 PM | 2.63 (2.40-2.89) | <.001 | 84 |
CYP2C19 | 4 | 1110 NM | 760 IM | 1.38 (1.28-1.48) | <.001 | 86 | |
Fluvoxamine | CYP2D6 | 6 | 74 NM | 72 IM | 1.52 (1.23-1.89) | <.001 | 0 |
CYP2C19 | 1 | 6 NM | 6 IM | 0.87 (0.31-2.45) | .77 | NA | |
CYP2C19 | 1 | 6 NM | 6 PM | 0.90 (0.31-2.65) | .84 | NA | |
Fluoxetine | CYP2D6 | 1 | 8 NM | 3 PM | 2.26 (1.68-2.83) | <.001 | NA |
CYP2C19 | 1 | 4 NM | 6 PM | 2.94 (2.36-3.67) | <.001 | NA | |
CYP2C19 | 2 | 71 NM | 27 IM | 1.48 (1.24-1.76) | <.001 | 13 | |
Mirtazapine | CYP2D6 | 4 | 125 NM | 19 PM | 1.39 (1.23-1.57) | <.001 | 64 |
CYP2D6 | 1 | 17 NM | 14 IM | 1.51 (1.20-1.91) | .010 | NA | |
Nortriptyline | CYP2D6 | 1 | 5 NM | 4 PM | 3.32 (2.08-5.29) | <.001 | NA |
CYP2D6 | 3 | 23 NM | 14 IM | 2.36 (2.10-2.65) | <.001 | 74 | |
Paroxetine | CYP2D6 | 2 | 62 NM | 11 PM | 5.13 (3.82-6.87) | <.001 | 85 |
CYP2D6 | 3 | 27 NM | 14 IM | 3.50 (2.52-4.85) | <.001 | 0 | |
Sertraline | CYP2C19 | 3 | 565 NM | 352 IM | 1.38 (1.27-1.51) | <.001 | 0 |
CYP2C19 | 2 | 537 NM | 40 PM | 2.70 (2.15-3.39) | <.001 | 0 | |
Venlafaxine | CYP2D6 | 6 | 486 NM | 120 PM | 1.18 (1.04-1.33) | .01 | 50 |
CYP2D6 | 3 | 436 NM | 87 IM | 1.14 (1.03-1.26) | .009 | 70 | |
CYP2D6 | 8 | 509 NM | 207 PM plus IM | 1.19 (1.09-1.29) | <.001 | 40 | |
CYP2C19 | 1 | 422 NM | 21 PM | 2.13 (1.54-2.93) | <.001 | NA | |
CYP2C19 | 1 | 422 NM | 247 IM | 1.19 (1.11-1.31) | <.001 | NA |
Abbreviations: IM, intermediate metabolizer; NA, not applicable; NM, normal metabolizer; PM, poor metabolizer; RoM, ratio of means.
Association Between CYP2D6 Metabolizer Status and Drug Exposure
The CYP2C19 genotype was associated with significant exposure increases for escitalopram (eFigure 2 in the Supplement) (PM vs NM RoM, 2.63; 95% CI, 2.40-2.89; 4 studies; 1262 patients)4,91,92,93 and sertraline (eFigure 2 in the Supplement) (IM vs NM RoM, 1.38; 95% CI, 1.27-1.51; 3 studies; 917 patients).6,94,95 Considerable heterogeneity was observed in the escitalopram meta-analyses, and the elevation in escitalopram exposure in the CYP2C19 IM group was not observed if the random-effect model was used (eFigure 4 in the Supplement). The CYP2C19 IM and NM groups did not exhibit statistically significant difference in clozapine exposure.96,97,98,99 Statistically significant exposure increases based on less than 3 independent studies compared with the CYP2C19 NM group were observed in the clozapine-treated PM (RoM, 1.92; 95% CI, 1.32-2.79; 2 studies; 78 patients), fluoxetine-treated IM (RoM, 1.48; 95% CI, 1.24-1.76; 2 studies; 98 patients) and PM (RoM, 2.94; 95% CI, 2.36-3.67; 1 study; 10 patients), sertraline-treated PM (RoM, 2.70; 95% CI, 2.15-3.39; 2 studies; 577 patients), and venlafaxine-treated IM (RoM, 1.19; 95% CI, 1.11-1.31; 1 study; 669 patients) and PM (RoM, 2.13; 95% CI, 1.54-2.93; 1 study; 443 patients) groups (Table 3).
Heterogeneity, Small Trial or Publication Bias, and Risk of Bias Assessment
Significant heterogeneity was observed in the aripiprazole IM and IM plus PM, escitalopram PM and IM, mirtazapine PM, nortriptyline IM, and venlafaxine IM group meta-analyses. No small trial or publication bias was observed in the meta-analyses related to risperidone and aripiprazole (eResults in the Supplement), whereas asymmetry could not be assessed in other meta-analyses owing to the insufficient number of included studies (n < 10).
According to the standardized risk of bias in nonrandomized studies of interventions tool, 23 studies were associated with a serious risk of bias,24,32,36,38,41,44,46,49,56,58,59,61,62,70,74,79,81,82,86,103,105,107,112 and 71 studies4,5,6,25,26,27,28,29,30,31,33,34,35,37,39,40,42,43,45,47,48,50,51,52,53,54,55,57,60,63,64,65,66,67,68,69,71,72,73,75,76,77,78,80,83,84,85,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,104,106,108,109,110,111,113,114 were associated with moderate risk of bias (ie, the analysis is comparable with a well-performed nonrandomized study). The sensitivity analysis results performed for the studies with moderate risk of bias is presented in eTable 3 and eFigure 6 in the Supplement.
Discussion
The results obtained in this systematic review and meta-analysis provide precise quantifications of the differences in antipsychotic and antidepressant drug exposure between patients with PM or IM vs NM CYP2C19/CYP2D6 phenotypes. These results represent scientific foundations for CYP2D6/CYP2C19 genotype-based dosing recommendations, which could lead to improved clinical outcomes in drug treatment of patients with psychiatric disorders.
Although many studies show that CYP2C19 and CYP2D6 PM and IM groups exhibit a significant increase in drug exposure compared with NM groups, the power of these studies was insufficient to quantify these exposure increases with sufficient precision and to evaluate their prospective clinical relevance. The present set of meta-analyses, which incorporates 8379 CYP2C19 and CYP2D6 genotyped individuals with exposure measurements for 16 frequently used psychiatric drugs, allowed (1) validation of whether CYP2C19 and CYP2D6 PM or IM phenotypes significantly increase the drug exposure compared with the NM phenotype, (2) differentiation between marginal changes and clinically relevant drug exposure increases caused by specific phenotypes, and (3) precise estimation of the magnitude of increase in drug exposure for the clinically relevant exposure changes. High precision of clinically relevant estimates is important for the clinical implementation of appropriate dose recommendations for subpopulations defined by CYP2C19 or CYP2D6 genotype.
There is a consensus in the field about the relevance of the CYP2C19 and CYP2D6 polymorphism for interindividual variability in drug metabolism and clinical response,117,118 and CYP2C19/CYP2D6 genotyping is already included in all currently commercially available pharmacogenetic tests.119 Pharmacogenomic recommendations on drug labels offer a tool by which knowledge of the specific genotype can be translated to the clinical setting in a quantitative manner. However, the dosing recommendations are usually not uniform among the relevant sources,14 and the dosing recommendations on the US Food and Drug Administration–approved drug labels120,121,122,123,124,125 clearly do not comply on many points with the findings summarized herein. The results suggest that there is a need to distinguish between CYP2D6 metabolism categories when deciding on aripiprazole, haloperidol, and risperidone doses and to distinguish between CYP2C19 metabolism categories when deciding on escitalopram and sertraline dose. Furthermore, unlike the PM phenotype, the IM phenotype is seldom considered a relevant factor for drug dosing and treatment, which is noteworthy in relation to results and the fact that more than half of the East Asian population and a considerable amount of other populations have the CYP2C19 or CYP2D6 IM phenotype.9
To approach the question of whether preemptive CYP2C19 and CYP2D6 genotyping can improve the drug treatment outcome of patients with psychiatric disorders, one must (1) demonstrate the effect of serum concentration on adverse effects and efficacy and (2) quantify the effect of genotype on serum concentration. The former has been demonstrated by a series of pharmacokinetic, clinical, and positron emission tomography studies1,126,127 and to an extent by 2 recent meta-analyses on dose-response curves for antidepressants and antipsychotics.2,3 The present report addresses the latter, because it quantifies the effect of PM and IM CYP2C19/CYP2D6 phenotypes on blood levels. Therapeutic drug monitoring can be used as a tool in personalized dosing because it directly measures drug blood levels and encompasses all sources of variability in drug exposure, including CYP2D6/CYP2C19 genotype. However, therapeutic drug monitoring becomes applicable only when the drug level reaches a steady state and is therefore not a suitable tool for preventing the suboptimal response or adverse effects during the initial weeks, or sometimes months, of psychiatric drug treatment. This period is critical for rapid symptom control, patients’ treatment belief, and adherence; in a therapeutic field characterized by a substantial degree of trial and error, preemptive genotyping has a potential to improve dose personalization and subsequently the drug treatment outcome as well. Overall, the optimal dose stabilization would be obtained in an ideal clinical situation, in which a psychiatrist would know the patients’ CYP2D6/CYP2C19 genotype before the drug treatment initiation to make the best possible initial dosing decisions. These decisions can be checked by therapeutic drug monitoring after the steady state is achieved. However, although several industry-sponsored clinical trials128,129,130 advocate the advantage of genotype-guided over usual treatment in psychiatry, a well-designed trial is still necessary to validate and quantify the clinical utility of preemptive CYP2C19/CYP2D6 genotyping.
Limitations
The most important limitation of this report is the potential presence of confounding factors, which arise from the nature of the studies incorporated into meta-analyses. Most of the studies were performed in naturalistic settings, and the factors that are known to affect drug metabolism are seldom completely controlled for. Next, the inclusion and exclusion criteria were designed in a way to eliminate the possibility of erroneous classification into metabolism categories, and this revealed the apparent scarcity of representative studies for many gene-drug interactions. In addition, approximately one-third of the studies that dealt with drug exposure did not measure exposure representatively, and the data were therefore not usable. Although CYP2C19/CYP2D6 UM status may also affect the exposure of certain drugs, and although the CYP2C19 and CYP2D6 PM and/or IM status significantly affect drug exposures of most of the analyzed drugs, more studies and larger cohorts are needed to ascertain the relevance of many gene-drug interactions (eFigure 8 in the Supplement). Also, in some cases, the number of usable studies was relatively low and heterogeneity was considerable; the most notable example is the analysis of CYP2C19-escitalopram interaction with 4 representative studies for each comparison4,91,92,93 and I2 > 80%. Although the directionality of the effect is apparent, more representative studies on this interaction are needed to precisely quantify the effect size of the exposure increase. Next, it was possible to address the presence of small trial or publication bias only for several comparisons owing to the small number of studies (n < 10) for many gene-drug interactions. Although the test result was negative for the analyzed comparisons, we cannot exclude the possibility that the publication bias is present in some of the gene-drug interaction comparisons to a degree. Finally, we were able to compare the effect of ethnicity in several comparisons by the subgroup test only, and these post hoc tests are presented in the eFigure 7 in the Supplement. Although these test results were negative, we cannot completely exclude the possibility that the exposure increases of certain drugs may be ethnicity dependent to a degree.
Conclusions
In this systematic review and meta-analysis, the association between CYP2C19/CYP2D6 genotype and drug levels of aripiprazole, haloperidol, risperidone, escitalopram, and sertraline was quantified with sufficient precision as to be useful as a scientific foundation for CYP2D6/CYP2C19 genotype-based dosing recommendations. In addition, there was an indication that the CYP2C19/CYP2D6 genotype is associated with changes in drug levels of clozapine, quetiapine, amitriptyline, fluvoxamine, fluoxetine, mirtazapine, nortriptyline, paroxetine, and venlafaxine. However, more representative studies focused on these specific gene-drug associations are necessary for an adequate quantification of the magnitude of drug level changes and for representative evaluation of the relevance of these changes.
References
- 1.Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51(1-02):9-62. doi: 10.1055/s-0043-116492 [DOI] [PubMed] [Google Scholar]
- 2.Furukawa TA, Cipriani A, Cowen PJ, Leucht S, Egger M, Salanti G. Optimal dose of selective serotonin reuptake inhibitors, venlafaxine, and mirtazapine in major depression: a systematic review and dose-response meta-analysis. Lancet Psychiatry. 2019;6(7):601-609. doi: 10.1016/S2215-0366(19)30217-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Leucht S, Crippa A, Siafis S, Patel MX, Orsini N, Davis JM. Dose-response meta-analysis of antipsychotic drugs for acute schizophrenia. Am J Psychiatry. 2020;177(4):342-353. doi: 10.1176/appi.ajp.2019.19010034 [DOI] [PubMed] [Google Scholar]
- 4.Jukić MM, Haslemo T, Molden E, Ingelman-Sundberg M. Impact of CYP2C19 genotype on escitalopram exposure and therapeutic failure: a retrospective study based on 2,087 patients. Am J Psychiatry. 2018;175(5):463-470. doi: 10.1176/appi.ajp.2017.17050550 [DOI] [PubMed] [Google Scholar]
- 5.Jukic MM, Smith RL, Haslemo T, Molden E, Ingelman-Sundberg M. Effect of CYP2D6 genotype on exposure and efficacy of risperidone and aripiprazole: a retrospective, cohort study. Lancet Psychiatry. 2019;6(5):418-426. doi: 10.1016/S2215-0366(19)30088-4 [DOI] [PubMed] [Google Scholar]
- 6.Bråten LS, Haslemo T, Jukic MM, Ingelman-Sundberg M, Molden E, Kringen MK. Impact of CYP2C19 genotype on sertraline exposure in 1200 Scandinavian patients. Neuropsychopharmacology. 2020;45(3):570-576. doi: 10.1038/s41386-019-0554-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gaedigk A, Sangkuhl K, Whirl-Carrillo M, Klein T, Leeder JS. Prediction of CYP2D6 phenotype from genotype across world populations. Genet Med. 2017;19(1):69-76. doi: 10.1038/gim.2016.80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fricke-Galindo I, Céspedes-Garro C, Rodrigues-Soares F, et al. Interethnic variation of CYP2C19 alleles, “predicted” phenotypes and “measured” metabolic phenotypes across world populations. Pharmacogenomics J. 2016;16(2):113-123. doi: 10.1038/tpj.2015.70 [DOI] [PubMed] [Google Scholar]
- 9.Zhou Y, Ingelman-Sundberg M, Lauschke VM. Worldwide distribution of cytochrome P450 alleles: a meta-analysis of population-scale sequencing projects. Clin Pharmacol Ther. 2017;102(4):688-700. doi: 10.1002/cpt.690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fabbri C, Tansey KE, Perlis RH, et al. Effect of cytochrome CYP2C19 metabolizing activity on antidepressant response and side effects: Meta-analysis of data from genome-wide association studies. Eur Neuropsychopharmacol. 2018;28(8):945-954. doi: 10.1016/j.euroneuro.2018.05.009 [DOI] [PubMed] [Google Scholar]
- 11.de Leon J, Susce MT, Pan RM, Fairchild M, Koch WH, Wedlund PJ. The CYP2D6 poor metabolizer phenotype may be associated with risperidone adverse drug reactions and discontinuation. J Clin Psychiatry. 2005;66(1):15-27. doi: 10.4088/JCP.v66n0103 [DOI] [PubMed] [Google Scholar]
- 12.Jukić MM, Opel N, Ström J, et al. Elevated CYP2C19 expression is associated with depressive symptoms and hippocampal homeostasis impairment. Mol Psychiatry. 2017;22(8):1155-1163. doi: 10.1038/mp.2016.204 [DOI] [PubMed] [Google Scholar]
- 13.Rahikainen AL, Vauhkonen P, Pett H, et al. Completed suicides of citalopram users-the role of CYP genotypes and adverse drug interactions. Int J Legal Med. 2019;133(2):353-363. doi: 10.1007/s00414-018-1927-0 [DOI] [PubMed] [Google Scholar]
- 14.Ingelman-Sundberg M. Translation of pharmacogenomic drug labels into the clinic: current problems. Pharmacol Res. 2020;153:104620. doi: 10.1016/j.phrs.2019.104620 [DOI] [PubMed] [Google Scholar]
- 15.Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Lancet. 2018;391(10128):1357-1366. doi: 10.1016/S0140-6736(17)32802-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Leucht S, Cipriani A, Spineli L, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet. 2013;382(9896):951-962. doi: 10.1016/S0140-6736(13)60733-3 [DOI] [PubMed] [Google Scholar]
- 17.Sterne JA, Hernán MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919. doi: 10.1136/bmj.i4919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Caudle KE, Sangkuhl K, Whirl-Carrillo M, et al. Standardizing CYP2D6 genotype to phenotype translation: consensus recommendations from the Clinical Pharmacogenetics Implementation Consortium and Dutch Pharmacogenetics Working Group. Clin Transl Sci. 2020;13(1):116-124. doi: 10.1111/cts.12692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting: Meta-analysis Of Observational Studies in Epidemiology (MOOSE) group. JAMA. 2000;283(15):2008-2012. doi: 10.1001/jama.283.15.2008 [DOI] [PubMed] [Google Scholar]
- 20.Friedrich JO, Adhikari NKJ, Beyene J. The ratio of means method as an alternative to mean differences for analyzing continuous outcome variables in meta-analysis: a simulation study. BMC Med Res Methodol. 2008;8(1):32. doi: 10.1186/1471-2288-8-32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Higgins JPT TJ, Chandler J, Cumpston M, Li T, Page MJ, Welch VA. Cochrane Handbook for Systematic Reviews of Interventions, version 6.1 (updated September 2020). Cochrane; 2020. [Google Scholar]
- 22.Sterne JA, Sutton AJ, Ioannidis JP, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ. 2011;343:d4002. doi: 10.1136/bmj.d4002 [DOI] [PubMed] [Google Scholar]
- 23.US Food and Drug Administration. Guidance for Industry Bioavailability and Bioequivalence Studies for Orally Administered Drug Products—General Considerations. Center for Drug Evaluation and Research; 2003. [Google Scholar]
- 24.Lisbeth P, Vincent H, Kristof M, Bernard S, Manuel M, Hugo N. Genotype and co-medication dependent CYP2D6 metabolic activity: effects on serum concentrations of aripiprazole, haloperidol, risperidone, paliperidone and zuclopenthixol. Eur J Clin Pharmacol. 2016;72(2):175-184. doi: 10.1007/s00228-015-1965-1 [DOI] [PubMed] [Google Scholar]
- 25.Belmonte C, Ochoa D, Román M, et al. Influence of CYP2D6, CYP3A4, CYP3A5 and ABCB1 polymorphisms on pharmacokinetics and safety of aripiprazole in healthy volunteers. Basic Clin Pharmacol Toxicol. 2018;122(6):596-605. doi: 10.1111/bcpt.12960 [DOI] [PubMed] [Google Scholar]
- 26.van der Weide K, van der Weide J. The influence of the CYP3A4*22 polymorphism and CYP2D6 polymorphisms on serum concentrations of aripiprazole, haloperidol, pimozide, and risperidone in psychiatric patients. J Clin Psychopharmacol. 2015;35(3):228-236. doi: 10.1097/JCP.0000000000000319 [DOI] [PubMed] [Google Scholar]
- 27.Tveito M, Molden E, Høiseth G, Correll CU, Smith RL. Impact of age and CYP2D6 genetics on exposure of aripiprazole and dehydroaripiprazole in patients using long-acting injectable versus oral formulation: relevance of poor and intermediate metabolizer status. Eur J Clin Pharmacol. 2020;76(1):41-49. doi: 10.1007/s00228-019-02768-0 [DOI] [PubMed] [Google Scholar]
- 28.Suzuki T, Mihara K, Nakamura A, et al. Effects of the CYP2D6*10 allele on the steady-state plasma concentrations of aripiprazole and its active metabolite, dehydroaripiprazole, in Japanese patients with schizophrenia. Ther Drug Monit. 2011;33(1):21-24. doi: 10.1097/FTD.0b013e3182031021 [DOI] [PubMed] [Google Scholar]
- 29.Suzuki T, Mihara K, Nakamura A, et al. Effects of genetic polymorphisms of CYP2D6, CYP3A5, and ABCB1 on the steady-state plasma concentrations of aripiprazole and its active metabolite, dehydroaripiprazole, in Japanese patients with schizophrenia. Ther Drug Monit. 2014;36(5):651-655. doi: 10.1097/FTD.0000000000000070 [DOI] [PubMed] [Google Scholar]
- 30.Nagai G, Mihara K, Nakamura A, et al. Prolactin concentrations during aripiprazole treatment in relation to sex, plasma drugs concentrations and genetic polymorphisms of dopamine D2 receptor and cytochrome P450 2D6 in Japanese patients with schizophrenia. Psychiatry Clin Neurosci. 2012;66(6):518-524. doi: 10.1111/j.1440-1819.2012.02391.x [DOI] [PubMed] [Google Scholar]
- 31.Azuma J, Hasunuma T, Kubo M, et al. The relationship between clinical pharmacokinetics of aripiprazole and CYP2D6 genetic polymorphism: effects of CYP enzyme inhibition by coadministration of paroxetine or fluvoxamine. Eur J Clin Pharmacol. 2012;68(1):29-37. doi: 10.1007/s00228-011-1094-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kubo M, Koue T, Inaba A, et al. Influence of itraconazole co-administration and CYP2D6 genotype on the pharmacokinetics of the new antipsychotic aripiprazole. Drug Metab Pharmacokinet. 2005;20(1):55-64. doi: 10.2133/dmpk.20.55 [DOI] [PubMed] [Google Scholar]
- 33.Kubo M, Koue T, Maune H, Fukuda T, Azuma J. Pharmacokinetics of aripiprazole, a new antipsychotic, following oral dosing in healthy adult Japanese volunteers: influence of CYP2D6 polymorphism. Drug Metab Pharmacokinet. 2007;22(5):358-366. doi: 10.2133/dmpk.22.358 [DOI] [PubMed] [Google Scholar]
- 34.Desai M, Tanus-Santos JE, Li L, et al. Pharmacokinetics and QT interval pharmacodynamics of oral haloperidol in poor and extensive metabolizers of CYP2D6. Pharmacogenomics J. 2003;3(2):105-113. doi: 10.1038/sj.tpj.6500160 [DOI] [PubMed] [Google Scholar]
- 35.Gassó P, Papagianni K, Mas S, et al. Relationship between CYP2D6 genotype and haloperidol pharmacokinetics and extrapyramidal symptoms in healthy volunteers. Pharmacogenomics. 2013;14(13):1551-1563. doi: 10.2217/pgs.13.150 [DOI] [PubMed] [Google Scholar]
- 36.Brockmöller J, Kirchheiner J, Schmider J, et al. The impact of the CYP2D6 polymorphism on haloperidol pharmacokinetics and on the outcome of haloperidol treatment. Clin Pharmacol Ther. 2002;72(4):438-452. doi: 10.1067/mcp.2002.127494 [DOI] [PubMed] [Google Scholar]
- 37.Troost PW, Lahuis BE, Hermans MH, et al. Prolactin release in children treated with risperidone: impact and role of CYP2D6 metabolism. J Clin Psychopharmacol. 2007;27(1):52-57. doi: 10.1097/JCP.0b013e31802e68d5 [DOI] [PubMed] [Google Scholar]
- 38.Jovanović N, Božina N, Lovrić M, Medved V, Jakovljević M, Peleš AM. The role of CYP2D6 and ABCB1 pharmacogenetics in drug-naïve patients with first-episode schizophrenia treated with risperidone. Eur J Clin Pharmacol. 2010;66(11):1109-1117. doi: 10.1007/s00228-010-0850-1 [DOI] [PubMed] [Google Scholar]
- 39.Hendset M, Molden E, Refsum H, Hermann M. Impact of CYP2D6 genotype on steady-state serum concentrations of risperidone and 9-hydroxyrisperidone in patients using long-acting injectable risperidone. J Clin Psychopharmacol. 2009;29(6):537-541. doi: 10.1097/JCP.0b013e3181c17df0 [DOI] [PubMed] [Google Scholar]
- 40.Mas S, Gassó P, Torra M, et al. ; PEPs Group . Intuitive pharmacogenetic dosing of risperidone according to CYP2D6 phenotype extrapolated from genotype in a cohort of first episode psychosis patients. Eur Neuropsychopharmacol. 2017;27(7):647-656. doi: 10.1016/j.euroneuro.2017.03.012 [DOI] [PubMed] [Google Scholar]
- 41.Mannheimer B, Holm J, Koukel L, Bertilsson L, Osby U, Eliasson E. Risperidone metabolic ratio as a biomarker of individual CYP2D6 genotype in schizophrenic patients. Eur J Clin Pharmacol. 2014;70(6):695-699. doi: 10.1007/s00228-014-1664-3 [DOI] [PubMed] [Google Scholar]
- 42.Gassó P, Mas S, Papagianni K, et al. Effect of CYP2D6 on risperidone pharmacokinetics and extrapyramidal symptoms in healthy volunteers: results from a pharmacogenetic clinical trial. Pharmacogenomics. 2014;15(1):17-28. doi: 10.2217/pgs.13.204 [DOI] [PubMed] [Google Scholar]
- 43.Scordo MG, Spina E, Facciolà G, Avenoso A, Johansson I, Dahl ML. Cytochrome P450 2D6 genotype and steady state plasma levels of risperidone and 9-hydroxyrisperidone. Psychopharmacology (Berl). 1999;147(3):300-305. doi: 10.1007/s002130051171 [DOI] [PubMed] [Google Scholar]
- 44.De Leon J, Susce MT, Pan RM, Wedlund PJ, Orrego ML, Diaz FJ. A study of genetic (CYP2D6 and ABCB1) and environmental (drug inhibitors and inducers) variables that may influence plasma risperidone levels. Pharmacopsychiatry. 2007;40(3):93-102. doi: 10.1055/s-2007-973836 [DOI] [PubMed] [Google Scholar]
- 45.Novalbos J, López-Rodríguez R, Román M, Gallego-Sandín S, Ochoa D, Abad-Santos F. Effects of CYP2D6 genotype on the pharmacokinetics, pharmacodynamics, and safety of risperidone in healthy volunteers. J Clin Psychopharmacol. 2010;30(5):504-511. doi: 10.1097/JCP.0b013e3181ee84c7 [DOI] [PubMed] [Google Scholar]
- 46.Cabaleiro T, Ochoa D, López-Rodríguez R, et al. Effect of polymorphisms on the pharmacokinetics, pharmacodynamics, and safety of risperidone in healthy volunteers. Hum Psychopharmacol. 2014;29(5):459-469. doi: 10.1002/hup.2420 [DOI] [PubMed] [Google Scholar]
- 47.Bondolfi G, Eap CB, Bertschy G, Zullino D, Vermeulen A, Baumann P. The effect of fluoxetine on the pharmacokinetics and safety of risperidone in psychotic patients. Pharmacopsychiatry. 2002;35(2):50-56. doi: 10.1055/s-2002-25026 [DOI] [PubMed] [Google Scholar]
- 48.Xiang Q, Zhao X, Zhou Y, Duan JL, Cui YM. Effect of CYP2D6, CYP3A5, and MDR1 genetic polymorphisms on the pharmacokinetics of risperidone and its active moiety. J Clin Pharmacol. 2010;50(6):659-666. doi: 10.1177/0091270009347867 [DOI] [PubMed] [Google Scholar]
- 49.Jung SM, Kim KA, Cho HK, et al. Cytochrome P450 3A inhibitor itraconazole affects plasma concentrations of risperidone and 9-hydroxyrisperidone in schizophrenic patients. Clin Pharmacol Ther. 2005;78(5):520-528. doi: 10.1016/j.clpt.2005.07.007 [DOI] [PubMed] [Google Scholar]
- 50.Yagihashi T, Mizuno M, Chino B, et al. Effects of the CYP2D6*10 alleles and co-medication with CYP2D6-dependent drugs on risperidone metabolism in patients with schizophrenia. Hum Psychopharmacol. 2009;24(4):301-308. doi: 10.1002/hup.1025 [DOI] [PubMed] [Google Scholar]
- 51.Yasui-Furukori N, Mihara K, Kondo T, et al. Effects of CYP2D6 genotypes on plasma concentrations of risperidone and enantiomers of 9-hydroxyrisperidone in Japanese patients with schizophrenia. J Clin Pharmacol. 2003;43(2):122-127. doi: 10.1177/0091270002239819 [DOI] [PubMed] [Google Scholar]
- 52.Kang RH, Jung SM, Kim KA, et al. Effects of CYP2D6 and CYP3A5 genotypes on the plasma concentrations of risperidone and 9-hydroxyrisperidone in Korean schizophrenic patients. J Clin Psychopharmacol. 2009;29(3):272-277. doi: 10.1097/JCP.0b013e3181a289e0 [DOI] [PubMed] [Google Scholar]
- 53.Roh HK, Kim CE, Chung WG, Park CS, Svensson JO, Bertilsson L. Risperidone metabolism in relation to CYP2D6*10 allele in Korean schizophrenic patients. Eur J Clin Pharmacol. 2001;57(9):671-675. doi: 10.1007/s002280100372 [DOI] [PubMed] [Google Scholar]
- 54.Suzuki Y, Fukui N, Tsuneyama N, et al. Effect of the cytochrome P450 2D6*10 allele on risperidone metabolism in Japanese psychiatric patients. Hum Psychopharmacol. 2012;27(1):43-46. doi: 10.1002/hup.1260 [DOI] [PubMed] [Google Scholar]
- 55.Yoo HD, Lee SN, Kang HA, Cho HY, Lee IK, Lee YB. Influence of ABCB1 genetic polymorphisms on the pharmacokinetics of risperidone in healthy subjects with CYP2D6*10/*10. Br J Pharmacol. 2011;164(2b):433-443. doi: 10.1111/j.1476-5381.2011.01385.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mihara K, Kondo T, Yasui-Furukori N, et al. Effects of various CYP2D6 genotypes on the steady-state plasma concentrations of risperidone and its active metabolite, 9-hydroxyrisperidone, in Japanese patients with schizophrenia. Ther Drug Monit. 2003;25(3):287-293. doi: 10.1097/00007691-200306000-00006 [DOI] [PubMed] [Google Scholar]
- 57.Cho HY, Lee YB. Pharmacokinetics and bioequivalence evaluation of risperidone in healthy male subjects with different CYP2D6 genotypes. Arch Pharm Res. 2006;29(6):525-533. doi: 10.1007/BF02969428 [DOI] [PubMed] [Google Scholar]
- 58.Morita S, Shimoda K, Someya T, Yoshimura Y, Kamijima K, Kato N. Steady-state plasma levels of nortriptyline and its hydroxylated metabolites in Japanese patients: impact of CYP2D6 genotype on the hydroxylation of nortriptyline. J Clin Psychopharmacol. 2000;20(2):141-149. doi: 10.1097/00004714-200004000-00005 [DOI] [PubMed] [Google Scholar]
- 59.Lee SY, Sohn KM, Ryu JY, Yoon YR, Shin JG, Kim JW. Sequence-based CYP2D6 genotyping in the Korean population. Ther Drug Monit. 2006;28(3):382-387. doi: 10.1097/01.ftd.0000211823.80854.db [DOI] [PubMed] [Google Scholar]
- 60.Yue QY, Zhong ZH, Tybring G, et al. Pharmacokinetics of nortriptyline and its 10-hydroxy metabolite in Chinese subjects of different CYP2D6 genotypes. Clin Pharmacol Ther. 1998;64(4):384-390. doi: 10.1016/S0009-9236(98)90069-8 [DOI] [PubMed] [Google Scholar]
- 61.Chen R, Wang H, Shi J, Shen K, Hu P. Cytochrome P450 2D6 genotype affects the pharmacokinetics of controlled-release paroxetine in healthy Chinese subjects: comparison of traditional phenotype and activity score systems. Eur J Clin Pharmacol. 2015;71(7):835-841. doi: 10.1007/s00228-015-1855-6 [DOI] [PubMed] [Google Scholar]
- 62.Sawamura K, Suzuki Y, Someya T. Effects of dosage and CYP2D6-mutated allele on plasma concentration of paroxetine. Eur J Clin Pharmacol. 2004;60(8):553-557. doi: 10.1007/s00228-004-0792-6 [DOI] [PubMed] [Google Scholar]
- 63.Yoon YR, Cha IJ, Shon JH, et al. Relationship of paroxetine disposition to metoprolol metabolic ratio and CYP2D6*10 genotype of Korean subjects. Clin Pharmacol Ther. 2000;67(5):567-576. doi: 10.1067/mcp.2000.106128 [DOI] [PubMed] [Google Scholar]
- 64.Ohara K, Tanabu S, Ishibashi K, Ikemoto K, Yoshida K, Shibuya H. CYP2D6*10 alleles do not determine plasma fluvoxamine concentration/dose ratio in Japanese subjects. Eur J Clin Pharmacol. 2003;58(10):659-661. doi: 10.1007/s00228-002-0529-3 [DOI] [PubMed] [Google Scholar]
- 65.Sugahara H, Maebara C, Ohtani H, et al. Effect of smoking and CYP2D6 polymorphisms on the extent of fluvoxamine-alprazolam interaction in patients with psychosomatic disease. Eur J Clin Pharmacol. 2009;65(7):699-704. doi: 10.1007/s00228-009-0629-4 [DOI] [PubMed] [Google Scholar]
- 66.Gerstenberg G, Aoshima T, Fukasawa T, et al. Effects of the CYP 2D6 genotype and cigarette smoking on the steady-state plasma concentrations of fluvoxamine and its major metabolite fluvoxamino acid in Japanese depressed patients. Ther Drug Monit. 2003;25(4):463-468. doi: 10.1097/00007691-200308000-00008 [DOI] [PubMed] [Google Scholar]
- 67.Watanabe J, Suzuki Y, Fukui N, et al. Dose-dependent effect of the CYP2D6 genotype on the steady-state fluvoxamine concentration. Ther Drug Monit. 2008;30(6):705-708. doi: 10.1097/FTD.0b013e31818d73b3 [DOI] [PubMed] [Google Scholar]
- 68.Katoh Y, Uchida S, Kawai M, et al. Effects of cigarette smoking and cytochrome P450 2D6 genotype on fluvoxamine concentration in plasma of Japanese patients. Biol Pharm Bull. 2010;33(2):285-288. doi: 10.1248/bpb.33.285 [DOI] [PubMed] [Google Scholar]
- 69.Suzuki Y, Sugai T, Fukui N, et al. CYP2D6 genotype and smoking influence fluvoxamine steady-state concentration in Japanese psychiatric patients: lessons for genotype-phenotype association study design in translational pharmacogenetics. J Psychopharmacol. 2011;25(7):908-914. doi: 10.1177/0269881110370504 [DOI] [PubMed] [Google Scholar]
- 70.Sirot EJ, Harenberg S, Vandel P, et al. Multicenter study on the clinical effectiveness, pharmacokinetics, and pharmacogenetics of mirtazapine in depression. J Clin Psychopharmacol. 2012;32(5):622-629. doi: 10.1097/JCP.0b013e3182664d98 [DOI] [PubMed] [Google Scholar]
- 71.Lind AB, Reis M, Bengtsson F, et al. Steady-state concentrations of mirtazapine, N-desmethylmirtazapine, 8-hydroxymirtazapine and their enantiomers in relation to cytochrome P450 2D6 genotype, age and smoking behaviour. Clin Pharmacokinet. 2009;48(1):63-70. doi: 10.2165/0003088-200948010-00005 [DOI] [PubMed] [Google Scholar]
- 72.Kirchheiner J, Henckel HB, Meineke I, Roots I, Brockmöller J. Impact of the CYP2D6 ultrarapid metabolizer genotype on mirtazapine pharmacokinetics and adverse events in healthy volunteers. J Clin Psychopharmacol. 2004;24(6):647-652. doi: 10.1097/01.jcp.0000145341.30547.f0 [DOI] [PubMed] [Google Scholar]
- 73.González-Vacarezza N, Abad-Santos F, Carcas-Sansuan A, et al. Use of pharmacogenetics in bioequivalence studies to reduce sample size: an example with mirtazapine and CYP2D6. Pharmacogenomics J. 2013;13(5):452-455. doi: 10.1038/tpj.2012.29 [DOI] [PubMed] [Google Scholar]
- 74.Park JY, Shon JH, Kim KA, et al. Combined effects of itraconazole and CYP2D6*10 genetic polymorphism on the pharmacokinetics and pharmacodynamics of haloperidol in healthy subjects. J Clin Psychopharmacol. 2006;26(2):135-142. doi: 10.1097/01.jcp.0000203199.88581.c3 [DOI] [PubMed] [Google Scholar]
- 75.Suzuki A, Otani K, Mihara K, et al. Effects of the CYP2D6 genotype on the steady-state plasma concentrations of haloperidol and reduced haloperidol in Japanese schizophrenic patients. Pharmacogenetics. 1997;7(5):415-418. doi: 10.1097/00008571-199710000-00013 [DOI] [PubMed] [Google Scholar]
- 76.Mihara K, Suzuki A, Kondo T, et al. Effects of the CYP2D6*10 allele on the steady-state plasma concentrations of haloperidol and reduced haloperidol in Japanese patients with schizophrenia. Clin Pharmacol Ther. 1999;65(3):291-294. doi: 10.1016/S0009-9236(99)70108-6 [DOI] [PubMed] [Google Scholar]
- 77.Roh HK, Chung JY, Oh DY, et al. Plasma concentrations of haloperidol are related to CYP2D6 genotype at low, but not high doses of haloperidol in Korean schizophrenic patients. Br J Clin Pharmacol. 2001;52(3):265-271. doi: 10.1046/j.0306-5251.2001.01437.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Someya T, Suzuki Y, Shimoda K, et al. The effect of cytochrome P450 2D6 genotypes on haloperidol metabolism: a preliminary study in a psychiatric population. Psychiatry Clin Neurosci. 1999;53(5):593-597. doi: 10.1046/j.1440-1819.1999.00611.x [DOI] [PubMed] [Google Scholar]
- 79.Ohara K, Tanabu S, Yoshida K, Ishibashi K, Ikemoto K, Shibuya H. Effects of smoking and cytochrome P450 2D6*10 allele on the plasma haloperidol concentration/dose ratio. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(6):945-949. doi: 10.1016/S0278-5846(03)00154-4 [DOI] [PubMed] [Google Scholar]
- 80.Shimoda K, Morita S, Yokono A, et al. CYP2D6*10 alleles are not the determinant of the plasma haloperidol concentrations in Asian patients. Ther Drug Monit. 2000;22(4):392-396. doi: 10.1097/00007691-200008000-00005 [DOI] [PubMed] [Google Scholar]
- 81.Inada T, Senoo H, Iijima Y, Yamauchi T, Yagi G. Cytochrome P450 II D6 gene polymorphisms and the neuroleptic-induced extrapyramidal symptoms in Japanese schizophrenic patients. Psychiatr Genet. 2003;13(3):163-168. doi: 10.1097/00041444-200309000-00005 [DOI] [PubMed] [Google Scholar]
- 82.Someya T, Shimoda K, Suzuki Y, et al. Effect of CYP2D6 genotypes on the metabolism of haloperidol in a Japanese psychiatric population. Neuropsychopharmacology. 2003;28(8):1501-1505. doi: 10.1038/sj.npp.1300213 [DOI] [PubMed] [Google Scholar]
- 83.Hermann M, Hendset M, Fosaas K, Hjerpset M, Refsum H. Serum concentrations of venlafaxine and its metabolites O-desmethylvenlafaxine and N-desmethylvenlafaxine in heterozygous carriers of the CYP2D6*3, *4 or *5 allele. Eur J Clin Pharmacol. 2008;64(5):483-487. doi: 10.1007/s00228-007-0453-7 [DOI] [PubMed] [Google Scholar]
- 84.Whyte EM, Romkes M, Mulsant BH, et al. CYP2D6 genotype and venlafaxine-XR concentrations in depressed elderly. Int J Geriatr Psychiatry. 2006;21(6):542-549. doi: 10.1002/gps.1522 [DOI] [PubMed] [Google Scholar]
- 85.Nichols AI, Focht K, Jiang Q, Preskorn SH, Kane CP. Pharmacokinetics of venlafaxine extended release 75 mg and desvenlafaxine 50 mg in healthy CYP2D6 extensive and poor metabolizers: a randomized, open-label, two-period, parallel-group, crossover study. Clin Drug Investig. 2011;31(3):155-167. doi: 10.2165/11586630-000000000-00000 [DOI] [PubMed] [Google Scholar]
- 86.Shams ME, Arneth B, Hiemke C, et al. CYP2D6 polymorphism and clinical effect of the antidepressant venlafaxine. J Clin Pharm Ther. 2006;31(5):493-502. doi: 10.1111/j.1365-2710.2006.00763.x [DOI] [PubMed] [Google Scholar]
- 87.Preskorn S, Patroneva A, Silman H, et al. Comparison of the pharmacokinetics of venlafaxine extended release and desvenlafaxine in extensive and poor cytochrome P450 2D6 metabolizers. J Clin Psychopharmacol. 2009;29(1):39-43. doi: 10.1097/JCP.0b013e318192e4c1 [DOI] [PubMed] [Google Scholar]
- 88.Kringen MK, Bråten LS, Haslemo T, Molden E. The influence of combined CYP2D6 and CYP2C19 genotypes on venlafaxine and O-desmethylvenlafaxine concentrations in a large patient cohort. J Clin Psychopharmacol. 2020;40(2):137-144. doi: 10.1097/JCP.0000000000001174 [DOI] [PubMed] [Google Scholar]
- 89.Fukuda T, Nishida Y, Zhou Q, Yamamoto I, Kondo S, Azuma J. The impact of the CYP2D6 and CYP2C19 genotypes on venlafaxine pharmacokinetics in a Japanese population. Eur J Clin Pharmacol. 2000;56(2):175-180. doi: 10.1007/s002280050737 [DOI] [PubMed] [Google Scholar]
- 90.Jiang F, Kim HD, Na HS, et al. The influences of CYP2D6 genotypes and drug interactions on the pharmacokinetics of venlafaxine: exploring predictive biomarkers for treatment outcomes. Psychopharmacology (Berl). 2015;232(11):1899-1909. doi: 10.1007/s00213-014-3825-6 [DOI] [PubMed] [Google Scholar]
- 91.Tsai MH, Lin KM, Hsiao MC, et al. Genetic polymorphisms of cytochrome P450 enzymes influence metabolism of the antidepressant escitalopram and treatment response. Pharmacogenomics. 2010;11(4):537-546. doi: 10.2217/pgs.09.168 [DOI] [PubMed] [Google Scholar]
- 92.Hodgson K, Tansey K, Dernovsek MZ, et al. Genetic differences in cytochrome P450 enzymes and antidepressant treatment response. J Psychopharmacol. 2014;28(2):133-141. doi: 10.1177/0269881113512041 [DOI] [PubMed] [Google Scholar]
- 93.Tsuchimine S, Ochi S, Tajiri M, et al. Effects of cytochrome P450 (CYP) 2C19 genotypes on steady-state plasma concentrations of escitalopram and its desmethyl metabolite in Japanese patients with depression. Ther Drug Monit. 2018;40(3):356-361. doi: 10.1097/FTD.0000000000000506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Rudberg I, Hermann M, Refsum H, Molden E. Serum concentrations of sertraline and N-desmethyl sertraline in relation to CYP2C19 genotype in psychiatric patients. Eur J Clin Pharmacol. 2008;64(12):1181-1188. doi: 10.1007/s00228-008-0533-3 [DOI] [PubMed] [Google Scholar]
- 95.Saiz-Rodríguez M, Belmonte C, Román M, et al. Effect of polymorphisms on the pharmacokinetics, pharmacodynamics and safety of sertraline in healthy volunteers. Basic Clin Pharmacol Toxicol. 2018;122(5):501-511. doi: 10.1111/bcpt.12938 [DOI] [PubMed] [Google Scholar]
- 96.Lesche D, Mostafa S, Everall I, Pantelis C, Bousman CA. Impact of CYP1A2, CYP2C19, and CYP2D6 genotype- and phenoconversion-predicted enzyme activity on clozapine exposure and symptom severity. Pharmacogenomics J. 2020;20(2):192-201. doi: 10.1038/s41397-019-0108-y [DOI] [PubMed] [Google Scholar]
- 97.Sirot EJ, Knezevic B, Morena GP, et al. ABCB1 and cytochrome P450 polymorphisms: clinical pharmacogenetics of clozapine. J Clin Psychopharmacol. 2009;29(4):319-326. doi: 10.1097/JCP.0b013e3181acc372 [DOI] [PubMed] [Google Scholar]
- 98.Vasudev K, Choi YH, Norman R, Kim RB, Schwarz UI. Genetic determinants of clozapine-induced metabolic side effects. Can J Psychiatry. 2017;62(2):138-149. doi: 10.1177/0706743716670128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Tóth K, Csukly G, Sirok D, et al. Potential role of patients’ CYP3A-status in clozapine pharmacokinetics. Int J Neuropsychopharmacol. 2017;20(7):529-537. doi: 10.1093/ijnp/pyx019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Koller D, Saiz-Rodríguez M, Zubiaur P, et al. The effects of aripiprazole and olanzapine on pupillary light reflex and its relationship with pharmacogenetics in a randomized multiple-dose trial. Br J Clin Pharmacol. 2020;86(10):2051-2062. Published online April 6, 2020. doi: 10.1111/bcp.14300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Akamine Y, Sugawara-Kikuchi Y, Uno T, Shimizu T, Miura M. Quantification of the steady-state plasma concentrations of clozapine and N-desmethylclozapine in Japanese patients with schizophrenia using a novel HPLC method and the effects of CYPs and ABC transporters polymorphisms. Ann Clin Biochem. 2017;54(6):677-685. doi: 10.1177/0004563216686377 [DOI] [PubMed] [Google Scholar]
- 102.Ryu S, Park S, Lee JH, et al. A Study on CYP2C19 and CYP2D6 polymorphic effects on pharmacokinetics and pharmacodynamics of amitriptyline in healthy Koreans. Clin Transl Sci. 2017;10(2):93-101. doi: 10.1111/cts.12451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Halling J, Weihe P, Brosen K. The CYP2D6 polymorphism in relation to the metabolism of amitriptyline and nortriptyline in the Faroese population. Br J Clin Pharmacol. 2008;65(1):134-138. doi: 10.1111/j.1365-2125.2007.02969.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Steimer W, Zöpf K, von Amelunxen S, et al. Allele-specific change of concentration and functional gene dose for the prediction of steady-state serum concentrations of amitriptyline and nortriptyline in CYP2C19 and CYP2D6 extensive and intermediate metabolizers. Clin Chem. 2004;50(9):1623-1633. doi: 10.1373/clinchem.2003.030825 [DOI] [PubMed] [Google Scholar]
- 105.Jiang ZP, Shu Y, Chen XP, et al. The role of CYP2C19 in amitriptyline N-demethylation in Chinese subjects. Eur J Clin Pharmacol. 2002;58(2):109-113. doi: 10.1007/s00228-002-0445-6 [DOI] [PubMed] [Google Scholar]
- 106.Hayashi Y, Watanabe T, Aoki A, et al. Factors affecting steady-state plasma concentrations of enantiomeric mirtazapine and its desmethylated metabolites in Japanese psychiatric patients. Pharmacopsychiatry. 2015;48(7):279-285. doi: 10.1055/s-0035-1565069 [DOI] [PubMed] [Google Scholar]
- 107.Charlier C, Broly F, Lhermitte M, Pinto E, Ansseau M, Plomteux G. Polymorphisms in the CYP 2D6 gene: association with plasma concentrations of fluoxetine and paroxetine. Ther Drug Monit. 2003;25(6):738-742. doi: 10.1097/00007691-200312000-00014 [DOI] [PubMed] [Google Scholar]
- 108.Ververs FF, Voorbij HA, Zwarts P, et al. Effect of cytochrome P450 2D6 genotype on maternal paroxetine plasma concentrations during pregnancy. Clin Pharmacokinet. 2009;48(10):677-683. doi: 10.2165/11318050-000000000-00000 [DOI] [PubMed] [Google Scholar]
- 109.Dalén P, Dahl ML, Bernal Ruiz ML, Nordin J, Bertilsson L. 10-Hydroxylation of nortriptyline in white persons with 0, 1, 2, 3, and 13 functional CYP2D6 genes. Clin Pharmacol Ther. 1998;63(4):444-452. doi: 10.1016/S0009-9236(98)90040-6 [DOI] [PubMed] [Google Scholar]
- 110.Yasui-Furukori N, Takahata T, Nakagami T, et al. Different inhibitory effect of fluvoxamine on omeprazole metabolism between CYP2C19 genotypes. Br J Clin Pharmacol. 2004;57(4):487-494. doi: 10.1111/j.1365-2125.2003.02047.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Scordo MG, Spina E, Dahl ML, Gatti G, Perucca E. Influence of CYP2C9, 2C19 and 2D6 genetic polymorphisms on the steady-state plasma concentrations of the enantiomers of fluoxetine and norfluoxetine. Basic Clin Pharmacol Toxicol. 2005;97(5):296-301. doi: 10.1111/j.1742-7843.2005.pto_194.x [DOI] [PubMed] [Google Scholar]
- 112.Eap CB, Bondolfi G, Zullino D, et al. Concentrations of the enantiomers of fluoxetine and norfluoxetine after multiple doses of fluoxetine in cytochrome P4502D6 poor and extensive metabolizers. J Clin Psychopharmacol. 2001;21(3):330-334. doi: 10.1097/00004714-200106000-00013 [DOI] [PubMed] [Google Scholar]
- 113.Liu ZQ, Cheng ZN, Huang SL, et al. Effect of the CYP2C19 oxidation polymorphism on fluoxetine metabolism in Chinese healthy subjects. Br J Clin Pharmacol. 2001;52(1):96-99. doi: 10.1046/j.0306-5251.2001.01402.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bakken GV, Molden E, Hermann M. Impact of genetic variability in CYP2D6, CYP3A5, and ABCB1 on serum concentrations of quetiapine and N-desalkylquetiapine in psychiatric patients. Ther Drug Monit. 2015;37(2):256-261. doi: 10.1097/FTD.0000000000000135 [DOI] [PubMed] [Google Scholar]
- 115.Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA Group . Preferred Reporting Items for Systematic Reviews and Meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097. doi: 10.1371/journal.pmed.1000097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Koller D, Belmonte C, Lubomirov R, et al. Effects of aripiprazole on pupillometric parameters related to pharmacokinetics and pharmacogenetics after single oral administration to healthy subjects. J Psychopharmacol. 2018;32(11):1212-1222. doi: 10.1177/0269881118798605 [DOI] [PubMed] [Google Scholar]
- 117.Zeier Z, Carpenter LL, Kalin NH, et al. Clinical implementation of pharmacogenetic decision support tools for antidepressant drug prescribing. Am J Psychiatry. 2018;175(9):873-886. doi: 10.1176/appi.ajp.2018.17111282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Stingl JC, Brockmöller J, Viviani R. Genetic variability of drug-metabolizing enzymes: the dual impact on psychiatric therapy and regulation of brain function. Mol Psychiatry. 2013;18(3):273-287. doi: 10.1038/mp.2012.42 [DOI] [PubMed] [Google Scholar]
- 119.Bousman CA, Hopwood M. Commercial pharmacogenetic-based decision-support tools in psychiatry. Lancet Psychiatry. 2016;3(6):585-590. doi: 10.1016/S2215-0366(16)00017-1 [DOI] [PubMed] [Google Scholar]
- 120.US Food and Drug Administration. Risperidal drug label. Published June 2009. Accessed June 20, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/020272s056,020588s044,021346s033,021444s03lbl.pdf
- 121.US Food and Drug Administration. Abilify drug label. Published August 2016. Accessed June 30, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021436s041,021713s032,021729s024,021866s026lbl.pdf
- 122.US Food and Drug Administration. Lexapro drug label. Published January 2017. Accessed June 30, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021323s047lbl.pdf
- 123.US Food and Drug Administration. Zoloft drug label. Published December 2016. Accessed June 30, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/019839S74S86S87_20990S35S44S45lbl.pdf
- 124.US Food and Drug Administration. Paxil drug label. Published December 2012. Accessed June 30, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020031s067,020710s031.pdf
- 125.US Food and Drug Administration. Pamelor drug label. Published May 2007. Accessed June 30, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/018013s58lbl.pdf
- 126.McCutcheon R, Beck K, D’Ambrosio E, et al. Antipsychotic plasma levels in the assessment of poor treatment response in schizophrenia. Acta Psychiatr Scand. 2018;137(1):39-46. doi: 10.1111/acps.12825 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Veselinović T, Scharpenberg M, Heinze M, et al. ; NeSSy Study Group . Dopamine D2 receptor occupancy estimated from plasma concentrations of four different antipsychotics and the subjective experience of physical and mental well-being in schizophrenia: results from the randomized NeSSy Trial. J Clin Psychopharmacol. 2019;39(6):550-560. doi: 10.1097/JCP.0000000000001131 [DOI] [PubMed] [Google Scholar]
- 128.Pérez V, Salavert A, Espadaler J, et al. ; AB-GEN Collaborative Group . Efficacy of prospective pharmacogenetic testing in the treatment of major depressive disorder: results of a randomized, double-blind clinical trial. BMC Psychiatry. 2017;17(1):250. doi: 10.1186/s12888-017-1412-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bradley P, Shiekh M, Mehra V, et al. Improved efficacy with targeted pharmacogenetic-guided treatment of patients with depression and anxiety: a randomized clinical trial demonstrating clinical utility. J Psychiatr Res. 2018;96:100-107. doi: 10.1016/j.jpsychires.2017.09.024 [DOI] [PubMed] [Google Scholar]
- 130.Greden JF, Parikh SV, Rothschild AJ, et al. Impact of pharmacogenomics on clinical outcomes in major depressive disorder in the GUIDED trial: a large, patient- and rater-blinded, randomized, controlled study. J Psychiatr Res. 2019;111:59-67. doi: 10.1016/j.jpsychires.2019.01.003 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.