The influence of CYP2D6 and CYP2C19 genetic variation on diabetes mellitus risk in people taking antidepressants and antipsychotics =================================================================================================================================== * Isabelle Austin-Zimmerman * Marta Wronska * Baihan Wang * Haritz Irizar * Johan Hilge Thygesen * Anjali Bhat * Spiros Denaxas * Ghazaleh Fatemifar * Chris Finan * Jasmine Harju-Seppänen * Olga Giannakopoulou * Karoline Kuchenbaecker * Eirini Zartaloudi * Andrew McQuillin * Elvira Bramon ## Abstract **Background** CYP2D6 and CYP2C19 enzymes are essential in the metabolism of antidepressants and antipsychotics. Genetic variation in these genes may increase risk of adverse drug reactions. Antidepressants and antipsychotics have previously been associated with risk of diabetes. We examined whether individual genetic differences in CYP2D6 and CYP2C19 contribute to these effects. **Methods** We identified 31,579 individuals taking antidepressants and 2,699 taking antipsychotics within UK Biobank. Participants were classified as poor, intermediate or normal metabolisers of CYP2D6, and as poor, intermediate, normal, rapid and ultra-rapid metabolisers of CYP2C19. Risk of diabetes mellitus represented by HbA1c level was examined in relation to the metabolic phenotypes. We analysed drugs either individually (where sample size permitted) or grouped by class. **Results** CYP2D6 poor metabolisers taking paroxetine had higher Hb1Ac than normal metabolisers (mean difference: 2.29mmol/mol; p < 0.001). Among participants with diabetes who were taking venlafaxine, CYP2D6 poor metabolisers had higher HbA1c levels compared to normal metabolisers (mean differences: 10.15 mmol/mol; p < 0.001. Among participants with diabetes who were taking fluoxetine, we observe that CYP2D6 intermediate metabolisers and decreased HbA1c, compared to normal metabolisers (mean difference - 7.74mmol/mol; p=0.017). We did not observe any relationship between CYP2D6 or CYP2C19 metabolic status and HbA1c levels in participants taking antipsychotic medication. **Conclusion** Our results indicate that the impact of genetic variation in CYP2D6 differs depending on diabetes status. Although our findings support existing clinical guidelines, further research is essential to inform pharmacogenetic testing for people taking antidepressants and antipsychotics. ## Introduction The use of both antidepressant and antipsychotic medications has increased steadily in recent years. Antidepressant drugs were the third most commonly prescribed drug group in 2018, with 70.9 million prescriptions across the United Kingdom – an almost two-fold increase since 2008 (1,2). It is estimated that almost 20% of the British adult population has been prescribed an antidepressant at some stage (1–3). A similar trend is seen in the prescription of antipsychotics, with an increase from eight to 12 million prescriptions between 2008 and 2018 (2). Both antidepressant and antipsychotic medication provide essential and often lifesaving treatment for many patients. However, they are also associated with a range of common and sometimes serious adverse drug reactions including sedation, weight gain, movement disorders, and an increased risk of developing diabetes mellitus (4,5). Most first-generation antipsychotics, as well as olanzapine and clozapine, have been shown to impair glucose regulation (5–10). Other second generation (or atypical) antipsychotics such as amisulpride, ziprasidone, and aripiprazole seem less associated with this risk (5–10). Several studies have linked tricyclic antidepressants to increased diabetes risk (4,11–13). The evidence for selective serotonin reuptake inhibitors (SSRIs) is inconsistent, with some studies showing improved diabetic control and others showing the opposite (4,11). Research into serotonin-noradrenaline reuptake inhibitors (SNRIs), such as venlafaxine, has reported both a lack of influence on glycaemic control and diabetes risk (10,14–16). Some research suggests that the risk of antidepressant-induced diabetes varies substantially between similar drugs of the same class, and thus may not be a mechanism-based adverse effect, but rather an off-target effect of a single drug (17). Pharmacogenetics may help explain inter-individual differences in drug response and adverse drug reactions. Cytochrome P450 (CYP450) is a superfamily of enzymes involved in the oxidative biotransformation and clearance of the majority of prescribed drugs (18). CYP2D6 and CYP2C19 are the two CYP450 enzymes most involved in the metabolism of antidepressant and antipsychotic drugs and are both highly polymorphic (18,19). Genetic variation in these genes results in an altered enzyme activity and thus may explain some of the interindividual differences in treatment response. Typically, individuals are grouped into four to five phenotypic groups reflecting differing metabolic capabilities (19,20). Poor metabolisers lack a functional enzyme due to defective or deleted genes; intermediate metabolisers usually have one functional and one defective or deleted allele causing reduced activity of the enzyme; rapid and ultra-rapid metabolisers usually have multiple copies of a functional gene or possess variants that increase gene expression (21). Normal metabolisers (previously described as ‘extensive metabolisers’), or wild-type, are those with two fully functional copies of the gene and thus ‘normal’ enzymatic activity. The prevalence of CYP2D6 and CYP2C19 phenotypes varies across populations, but the extreme metabolisers are typically the least commonly observed: less than 10% of people are poor metabolisers, and less than 3% are ultra-rapid metabolisers, across all major populations and for both genes (22,23). Several studies have shown that poor metabolisers of CYP2D6 or CYP2C19 have higher serum levels of antidepressants and antipsychotics, compared to normal metabolisers (24–30). The Clinical Pharmacogenomics Implementation Consortium (CPIC) has developed evidence-based clinical guidelines for SSRIs and tricyclic antidepressants, recommending adjusted dosing based on CYP2D6 and CYP2C19 metabolic status (31,32). There are currently no CPIC guidelines for antipsychotics, but the Dutch Pharmacogenetics Working Group provides guidelines for aripiprazole, haloperidol, pimozide and zuclopenthixol based on CYP2D6 genotype (33). Work to incorporate similar evidence based clinical guidelines to the UK National Health Service (NHS) is ongoing (34). Thus far, research on the putative association between CYP450 metabolic phenotype and adverse drug reactions in response to antidepressants and antipsychotics has been limited by small sample sizes (34,35). Little is known about pharmacogenetic influences on the diabetes risk associated with these drugs. Therefore, this study aims to examine the association between CYP2C19 and CYP2D6 metabolic phenotypes and the risk of diabetes mellitus in UK Biobank participants taking antidepressants and antipsychotics. ## Methods ### Sample and phenotype data The UK Biobank data collection methods have been described previously in Bycroft et al (2018) and detailed study protocols are available online ([http://www.ukbiobank.ac.uk/resources/](http://www.ukbiobank.ac.uk/resources/) and [http://biobank.ctsu.ox.ac.uk/crystal/docs.cgi/](http://biobank.ctsu.ox.ac.uk/crystal/docs.cgi/)) (36,37). The study was approved by the North-West Research Ethics Committee (ref 06/MREC08/65). All participants provided written informed consent, and those who withdrew consent after providing their sample for genetic analysis were excluded from the data extraction. Data for 502,527 UK Biobank participants were considered in this study. Participants for this study were selected based on the criteria of taking one or more psychotropic drugs. Participants were asked during a verbal interview if they were taking any ‘regular prescription medication’, and to provide the name of the medication if so. Both generic and proprietary names were recorded by UK Biobank. In these instances, we reviewed the alternative names for equivalent drugs and combined them under the generic name for analysis. For additional detail, please refer to the supplementary methods section and supplementary figure 1. We identified a sample of 44,051 participants taking a drug of interest for this study. The UK Biobank measured a variety of biochemical markers in blood samples collected at the baseline visit. Glycated haemoglobin (HbA1c) was measured with the High Performance Liquid Chromatography (HPLC) method on a Bio-Rad VARIANT II Turbo analyser. The HbA1c analytical range was 15-184 mmol/mol and this measurement was recorded for over 92% of the UK Biobank cohort. Data on diabetes diagnosis (self-reported and confirmed by ICD-10 diagnosis when available), antidiabetic medications, CYP2D6 and/or CYP2C19 enzyme inhibitors and body mass index (BMI) were also downloaded. Further detail is available in the supplementary methods. We identified 49 individuals who reported taking antidiabetic medication but stated they do not have diabetes. They were excluded from the analysis due to uncertainty about their diagnosis. A total of 40,783 participants taking a psychotropic drug of interest also had HbA1c measurements available. ### Genetic data and quality control The UK Biobank conducted genome-wide genotyping for 488,377 participants. Genotyping was performed using the Affymetrix UK BiLEVE Axiom array on an initial sample of 50,000 and the Affymetrix UK Biobank Axiom® array was used on all later participants (36). These arrays include over 820,000 variants (SNPs and indel markers) and have good coverage of pharmacogenetics variants. Quality control and imputation of over 90 million variants was performed by a collaborative group led by the Wellcome Trust Centre for Human Genetics (36). Fully imputed genetic data was downloaded in March 2018. Further local post-imputation quality control was performed in each ethnic group separately to remove variants with minor allele frequency below 1% and/or Fisher information score (a measure of the imputation accuracy for each SNP) of less than 0.3. Individuals with greater than 10% missingness, excessive genetic relatedness (greater than 10 third-degree relatives based on kinship calculations as provided centrally by UK Biobank) or mismatch between reported and genetically inferred sex were removed. We included both European and non-European subjects in this analysis. A list of approximately 408,000 participants of European ancestry was provided centrally by UK Biobank, based on a combination of principal component analysis (PCA) and self-reported ethnicity data (36). Further local analysis was conducted to determine the genetic ancestry of the remaining participants: Two rounds of PCA were performed using the PC-AiR algorithm, and relatedness was estimated using PC-Relate (38–41). This resulted in the following groups: East Asian 0.5% (N=2,464), South Asian 2% (N=8,964), African 2% (N=9,233) or admixed with predominantly European origin 2.5% (N=11,251). A further 6,686 did not cluster with any main group and were excluded from analysis. One of each pair of participants with a kinship score greater than 0.083 (approximately third-degree relatives) were excluded from the analysis. This results in a total of 40,129 participants to exclude, across all ethnicities. After these quality control procedures, a total of 33,149 participants taking antidepressant and/or antipsychotic medication with HbA1c and good quality genetic data were included in the analysis. Please see supplementary figure 1 for a CONSORT diagram detailing these steps. ### Assigning CYP metabolic phenotype We extracted regions of interest for each CYP2D6 and CYP2C19, defined as being one megabase (Mb) upstream of the 5’ end of the gene and one megabase downstream of the 3’ end of the gene (see supplementary table 1). Several of the SNPs of interest in this study (i.e., those that define either CYP2D6 or CYP2C19 star alleles) are rare (MAF < 0.01) and therefore fail standard quality control protocols. For rare SNPs of interest included on the genotype panel we used Evoker v2.4 to create intensity plots and performed visual checks to determine if the data for these SNPs was reliable enough to include (42). We reviewed a total of six genotyped SNPs for CYP2C19 and five for CYP2D6. SNPs with distinct allelic clusters were included in this study. For the rare, imputed SNPs, we included only those that met a higher Fisher information score threshold of 0.6. We reviewed a total of seven imputed SNPs for CYP2C19 and five for CYP2D6. These steps enabled the inclusion of an additional four relevant SNPs for CYP2C19, and three for CYP2D6. The extraction of data and identification of rare SNPs was conducted separately for each ancestry group. Haplotypes for our sample were constructed based on extracted imputed genetic data using Beagle version 5.0 (43,44). An input map and reference panel from the 1,000 genome project were used (45). The phased data was used to construct haplotypes for all participants according to the star allele nomenclature system (20,46). We grouped individuals into CYP2C19 metabolic phenotype groups based on the activity of the individual haplotypes and resulting diplotypes (46). We grouped individuals into CYP2D6 metabolic phenotype groups according to the Gaedigk activity score method (47,48). Haplotypes containing no star-allele defining SNP variants were classified as wild-type (*1) alleles for the corresponding gene. Because not all star allele-defining SNPs were available in our genetic dataset, we expect a fraction of haplotypes to be misclassified as wild-type. Nonetheless, as the cumulative reported frequency of the missing SNPs is very low, we expect the number of misclassified haplotypes to be small.. In addition, we did not have data on CYP2D6 copy number variants (CNVs). This means we are not able to define CYP2D6 ultra-rapid metabolisers, or other whole gene deletions (e.g., CYP2D6*5). ### Statistical analysis We conducted a grouped analysis of all tricyclic antidepressants, as previous evidence suggests that they all cause an increase in HbA1c to some extent (49). We did not analyse SSRIs as a group due to variable evidence on their influence on HbA1c in the literature (15,17,49). Any antidepressants taken by over 1,800 participants were analysed independently (amitriptyline, citalopram, fluoxetine, sertraline, paroxetine, venlafaxine). Medications were grouped according to whether their primary metabolic pathway was catalysed by CYP2D6 or CYP2C19, based on the Maudsley Prescribing Guidelines and CPIC guidelines (10,31,32). Tricyclic antidepressants that are known CYP2C19 substrates are: amitriptyline, clomipramine, doxepin, imipramine and trimipramine. SSRIs that are known CYP2C19 substrates are: citalopram, escitalopram, and sertraline. Tricyclic antidepressants that are known substrates for CYP2D6 include: amitriptyline, clomipramine, duloxetine, and doxepin. SSRIs that are known substrates for CYP2D6 are: fluoxetine, fluvoxamine, paroxetine, sertraline, as well as the SNRIs mirtazapine and venlafaxine (10,50). No single antipsychotic drug had sufficient sample size to allow for individual analysis. Therefore, we included all antipsychotic drugs known to be metabolised at least in part by CYP2D6: aripiprazole, clozapine, fluphenazine, haloperidol, olanzapine, perphenazine, pimozide, risperidone, zuclopenthixol, thioridazine. CYP2C19 does not play a significant role in the metabolism of antipsychotics (10). For each drug or drug group, we ran linear regression models with HbA1c as the outcome of interest and CYP450 metabolic phenotype and diabetes status as the main explanatory variables. All statistical models were adjusted to account for any participant taking antidiabetic treatment or taking drugs, psychotropic or otherwise, that are known inhibitors of the enzymes of interest. Additional covariates included were BMI, sex, age, and genetically determined ancestry group. We investigated the interaction of diabetes status and CYP metabolic phenotype. Where this interaction was significant (*p* < 0.05) we conducted a stratified analysis separating participants into two groups based on their diabetes status. Some of these analyses are nested (individual drug analyses overlap with drug group analyses), and, as such, we concluded that a Bonferroni correction for multiple testing would be excessively stringent (51). Therefore, we report uncorrected *p* values in all text and tables, but as recommended by Li *et al* (2012) (52), we have an adjusted significance threshold of *p* < 0.05/2 = 0.025 (threshold for a suggestive association *p* < 0.1/2 = 0.05) for the two grouped analyses, and *p* < 0.05/6 = 0.0083 (threshold for a suggestive association *p* < 0.1/6 = 0.017) for the individual drug analyses examining six specific drugs. All statistical analyses were performed using R version 3.6.0 (53–55). ## Results ### Dataset We identified 33,149 UK Biobank participants who reported taking at least one antidepressant or antipsychotic and had HbA1c and genetic data passing quality control (antidepressants N=31,579, antipsychotics N=2,699) (Table 1). Our sample included 22,632 (68.3%) females and 10,517 (31.7%) males (see Table 1). Mean age was 56.6±7.8 years, range 40 to 70 years. Full demographic data and summary statistics of our sample are shown in the Table 1 (see also supplementary Tables 5 and 6). View this table: [Table 1](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/T1) Table 1 Demographic data for study sample ### Psychotropics prescribed in the UK Biobank There were 28 different antidepressants identified in our sample (figure 1; supplementary Tables 7 and 8). Amitriptyline was the most common drug in our cohort (N=8,191). We identified 24 different antipsychotic drugs (figure 1; supplementary Table 9), with the most frequent antipsychotics being prochlorperazine (870 individuals, 30.9%), followed by olanzapine (499 individuals, 17.7%). Among UK Biobank participants taking antidepressants, 5.2% report taking more than one different antidepressant concurrently (of these, 2% report taking three or four). Of those taking antipsychotics, 4.5% report taking more than one different antipsychotic medication concurrently (of these, 7.4% report taking three or four). The co-prescription of an antidepressant with antipsychotics is very common, with 41.4% of subjects taking antipsychotics also taking at least one antidepressant. ![Figure 1](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/16/2021.07.07.21259926/F1.medium.gif) [Figure 1](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/F1) Figure 1 Frequency table of identified antipsychotics (blue bars) and antidepressants (green bars) in UK Biobank. **Other antipsychotic (N):** promazine (30), zuclopenthixol (25), perphenazine (12), pipotiazine (10), pericyazine (8), levopromazine (6), benperidol (4), pimozide (3), thioridazine (2), sertindole (1); **Other antidepressants (N)**: moclobemide (30), phenelzine (30), tranylcypromine (21), bupropion (6), mianserin (4), isocarboxazid (2). The included covariates (diabetes status, antidiabetic medications, BMI, age, sex, and ethnicity) affected HbA1c as expected. Please refer to the supplementary methods for further details. #### Antidepressants and CYP metabolic status For several of the antidepressants investigated, we consistently found that the interaction of diabetes status and CYP2D6 and CYP2C19 metabolic phenotype is statistically significant (supplementary figure 2). Where this was the case, we stratified our analyses by whether participants had diabetes or not. Among all participants (regardless of diabetes status) taking paroxetine (SSRI), we observe significantly higher HbA1c levels among CPY2D6 poor metabolisers (mean difference: 2.43mmol/mol; 95% CI [1.23,3.63]; p = 7.77×10−5) (see table 2, figure 2, and supplementary table 10). A stratified analysis of diabetic participants taking fluoxetine (SSRI) reveals a suggestive association between CYP2D6 intermediate metabolisers and lower HbA1c levels compared to normal metabolisers (mean difference = -3.74mmol/mol; 95% CI [-6.82,-0.67]; p = 0.017) (see table 3, figure 2, and supplementary table 11). In participants taking venlafaxine (SNRI), we found that, amongst people with diabetes, poor metabolisers for CYP2D6 had higher HbA1c than normal metabolisers (mean difference: 10.15mmol/mol; 95% CI [2.63,17.67]; p = 0.008) (see table 4, figure 2, and supplementary table 12). View this table: [Table 2](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/T2) Table 2 Association between CYP2D6 metabolic phenotype and HbA1c levels among participants taking fluoxetine and paroxetine. View this table: [Table 3](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/T3) Table 3 A) Association between CYP2D6 metabolic phenotype and HbA1c levels among participants taking fluoxetine; B) Stratified analysis of diabetes status among participants taking fluoxetine. View this table: [Table 4](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/T4) Table 4 A) Association between CYP2D6 metabolic phenotype and HbA1c levels among participants taking venlafaxine; B) Stratified analysis of diabetes status among participants taking venlafaxine. ![Figure 2](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/08/16/2021.07.07.21259926/F2.medium.gif) [Figure 2](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/F2) Figure 2 Violin plots showing the relationship between CYP2D6 metabolic status and HbA1c levels (mmol/mol) among subjects taking (from left to right) paroxetine, fluoxetine, venlafaxine, and all antipsychotics. Stratified analyses of citalopram, sertraline, and amitriptyline did not reveal any significant association between the relevant CYP450 metabolic status and HbA1c levels (see supplementary tables 13-18). Several tricyclic antidepressants were reported too infrequently to allow for single-drug analysis. Therefore, we grouped the remaining drugs of this class, excluding amitriptyline as its higher frequency would have heavily driven the findings. We again stratified the group based on diabetes status and found no significant associations between CYP2D6 or CYP2C19 derived metabolic groups and HbA1c (see supplementary Table 19 and 20). We did not observe any significant association between HbA1c levels and CYP2C19 metabolic status in individuals taking antidepressants. In addition, we find that participants taking drugs that act as CYP2C19 inhibitors, regardless of CYP2C19 metabolic status, experience higher levels of HbA1c. Citalopram: mean difference: 0.36mmol/mol, 95% CI [0.07,0.65]; p = 0.016); Amitriptyline: mean difference: 0.37mmol/mol; 95% CI [0.09,0.64]; p = 0.009; Tricyclics: mean difference = 0.39mmol/mol; 95% CI [0.13,0.66]; p = 0.004). We did not see this relationship with sertraline (see supplementary Tables 13, 15, 17, 19). ### Antipsychotics and CYP metabolic status We find no evidence that the metabolic phenotypes of CYP2D6 influence HbA1c levels amongst 2,699 people taking antipsychotic medications. Similarly, taking a CYP2D6 inhibitor drug was not significantly associated with HbA1c levels amongst people taking antipsychotic medication. See table 5, figure 2 and supplementary Table 21. View this table: [Table 5](http://medrxiv.org/content/early/2021/08/16/2021.07.07.21259926/T5) Table 5 Association between CYP2D6 metabolic phenotype and HbA1c levels in participants taking antipsychotics ## Discussion Non-normal metabolic phenotypes of CYP2D6 and CYP2C19 have been linked to QT prolongation (56,57), weight gain (58–61), hormonal changes among patients taking psychotropic medication, as well as increased risk of extrapyramidal adverse reactions to antipsychotics (62). However, recent studies and meta-analyses have yielded inconclusive or negative findings and the clinical significance of CYP450 metabolic phenotypes is still in question (30,63). Several studies agree that long-term antidepressant treatment increases risk of developing diabetes (4,64–66), but the extent to which this specific adverse drug reaction is impacted by genetics is unknown. To our knowledge, this study is the first to explore if variation in the CYP2D6 and CYP2C19 genes influences HbA1c levels in individuals taking antidepressants and antipsychotics. Most previous studies of CYP450 metabolic status and adverse drug reactions are limited by small sample sizes and low representation of the less common poor or ultra-rapid metabolisers (30,56–66). This study represents one of the largest available samples of individuals taking antidepressants and antipsychotics and includes a much higher number of extreme CYP450 metabolisers than seen in previous publications (N=9,878 non-wild-type CYP2D6 metabolisers and N=21,273 non-wild-type CYP2C19 metabolisers). We find a significant association between CYP2D6 poor metabolisers and higher levels of HbA1c among all participants taking paroxetine with an average increase of 2.3mmol/mol, a substantial effect. The Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines recommend using lower doses of paroxetine for poor metabolisers of CYP2D6 (32). Thus, our findings are consistent with existing pharmacokinetic evidence and provide further support for the CPIC guidelines. Of interest, some research found that prolonged use of paroxetine was associated with phenocopying, an environmentally induced conversion of normal metabolisers to poor metabolisers (67–69). We observe a significant interaction between diabetic status and non-wild-type CYP status for participants taking amitriptyline, fluoxetine, citalopram, sertraline, and venlafaxine. We conducted stratified analyses of these drugs and found suggestive evidence that, in diabetic participants taking venlafaxine, CYP2D6 poor and intermediate metabolisers have higher HbA1c levels. Like paroxetine, venlafaxine has been previously associated with an increased risk of diabetes (4,15,70). Our study finds that diabetic CYP2D6 poor metabolisers treated with venlafaxine have on average 10.15 mmol/mol higher HbA1c levels than diabetic normal metabolisers. Though this is a suggestive association only with a comparatively small sample size, it is consistent with the guidelines published by the Dutch Pharmacogenetics Working Group which suggest that CYP2D6 poor metabolisers should be treated with an alternative antidepressants or have their venlafaxine dose reduced (33). In addition, a stratified analysis reveals suggestive evidence that diabetic CYP2D6 intermediate metabolisers taking fluoxetine have lower HbA1c levels compared to diabetic CYP2D6 normal metabolisers. Although this is contrary to our initial hypothesis, there is some evidence to suggest that fluoxetine can lower HbA1c levels in diabetic patients, despite increasing risk of type 2 diabetes in non-diabetic patients (71–73). Our findings add support to this theory, suggesting that decreased CYP2D6 metabolism may in fact be somewhat beneficial for patients with diabetes who take fluoxetine. Contrary to our hypotheses, we did not find evidence of associations between CYP2D6 or CYP2C19 metabolic status and HbA1c in people treated with amitriptyline and other tricyclics. Although CPIC guidelines exist for CYP2C19 and CYP2D6 poor metabolisers taking tricyclic antidepressants, they state that suggested dose alterations or treatment changes are optional based on the limited strength of existing evidence (31). Our analyses of tricyclics antidepressants and amitriptyline alone were adequately powered with over 400 poor metabolisers of each gene, making it one of the largest samples of abnormal CYP metabolisers available. However, the metabolic pathway of amitriptyline (and other tertiary amine tricyclic antidepressants) involves two steps: the first step is catalysed by CYP2C19 and produces an active metabolite (nortriptyline). The second step is the metabolism of nortriptyline to an inactive metabolite, via CYP2D6 (74,75). This is not adequately accounted for in this study, and thus future studies investigating the synergistic action of CYP2D6 and CYP2C19 on amitriptyline metabolism are required. In addition, we did not find associations between CYP2D6 variation and HbA1c amongst people taking antipsychotics, nor did we observe an impact of CYP2D6 inhibitors. Given the total sample size of 2,699, we undertook a combined analysis including all antipsychotics, which have various levels of influence on glucose regulation and diabetes risk. Although this sample is the largest available with 135 CYP2D6 poor metabolisers overall, statistical power remains limited given the heterogeneity of the sample. Analysis in a larger sample would allow for the separate analysis of individual drugs and should yield more conclusive results. This limitation also applies to the less common antidepressants in our sample, which were included in grouped analyses only. Given that UK Biobank is a population study, utilizing existing data from large patient-based biobanks such as the Million Veteran Program could be a valuable continuation of this work (76). Biobanks from countries with more historically isolated populations, such as Finngen, may contain a higher proportion of some rare SNPs that are necessary to define additional CYP450 star alleles. As well as being impacted by genetic variation, CYP2D6 and CYP2C19 enzyme activity is susceptible to inhibition by other compounds. We observed that taking CYP2C19 inhibitors (of which proton pump inhibitors were the most common in our sample) led to higher HbA1c levels in people taking tricyclic antidepressants, amitriptyline, and citalopram. Thus, based on our data, there is substantial potential for drug interactions and drug-drug-CYP2C19 interactions. These should be investigated further and considered for inclusion in future clinical guidelines. We did not find evidence that taking CYP2D6 inhibitors affected HbA1c levels in people taking antipsychotics or antidepressants. This enzyme inhibition could, however, still be important for other psychotropic adverse effects such as QT prolongation. It is also worth noting that CYP2C19 inhibitor drugs were taken by approximately a quarter of the subjects included in these analyses. That may have decreased the power to detect a significant association between the (genetically-determined) CYP2C19 metabolic status and HbA1c levels, because if the inhibitory impact of a drug was strong enough it would reduce or eliminate the impact of the genetic variation; i.e. an CYP2C19 normal metaboliser taking an inhibitor may have the same enzymatic activity and as CYP2C19 poor metaboliser taking an inhibitor. Future analyses with larger sample sizes should investigate this interaction further. A clear limitation of this study is the reliance on certain self-reported data (including diabetes diagnosis). In addition, we have used only the baseline, cross-sectional UK Biobank data and therefore lack detail on treatment dose and duration. Most adverse drug reactions to antidepressants and antipsychotics are dose-dependent, and thus further analysis including this data is warranted. Besides, diabetes is a complex disease with many genetic and environmental risk factors. Although the SNP-based heritability of diabetes is estimated to be less than 20%, the inclusion of polygenic risk scores for diabetes may improve analyses of pharmacogenetic associations by capturing background genetic disease risk (77). A genome-wide gene-environment interaction study may also highlight other genes of potential interest. Finally, although we included participants of all ethnicities in this analysis, UK Biobank is predominantly European. There is a great deal of variation in the frequency of functional variants within the CYP450 genes across different populations (22,78), as well as in the risk of diabetes. The field of pharmacogenetics would be greatly benefitted by further study in more diverse samples. Although both arrays used by UK Biobank have relatively good coverage of CYP2C19 and CYP2D6, several SNPs that define known star alleles were neither genotyped nor imputed or did not otherwise meet the criteria for inclusion as described in the methods. Therefore, we expect a small number of individuals to be misclassified as normal metabolisers. However, we anticipate this number to be small given the low minor allele frequency of the missing variants. We were unable to include CYP2D6 ultra-rapid metabolisers in this study, as copy number and other structural variants were not defined. CYP2D6 ultra-rapid metabolisers are the least common phenotypic group across all populations, with a frequency of less than 2% in European, South Asian, East Asian and Admixed European groups, and approximately 3-6% in African ancestry groups (22,78). CYP2D6 ultra-rapid metabolisers therefore represent a very small minority in our sample, and they have been combined with the normal metabolisers group by default. We estimate this to have a small effect on our results as we would expect ultra-rapid metabolisers to be less susceptible to adverse drug reactions, though it will be important to consider this group in future studies of treatment failure. The availability of whole genome sequencing data will improve the accuracy with which highly polymorphic pharmacogenes like CYP2D6 can be characterised, whilst still capturing the important splicing or non-coding variants that may be missed with exome sequencing data (79). Overall, our findings are broadly consistent with existing guidelines for antidepressants and point towards the necessity to include more antidepressants and antipsychotics in pharmacogenetic clinical trials and experimental medicine studies. These results also suggest that there is a need for randomised double-blinded clinical trials to further explore genetic testing as a guide to antidepressant/antipsychotic treatment. Indeed, studies show that pharmacogenetic testing is practical (80), accurately predicts the outcomes of antidepressant treatments (81) and improves outcomes (82,83). It has also been demonstrated that it can reduce the total cost of antipsychotic treatment by 28% (84). Findings from this study need to be followed up with further longitudinal testing, with a focus on singular antidepressants and antipsychotics, more adverse drug reactions, and in more diverse populations. ## Supporting information Supplementary material [[supplements/259926_file02.pdf]](pending:yes) ## Data Availability Data used in this paper is available to all UK Biobank approved researchers. ## Acknowledgements The authors thank all UK Biobank participants. This research has been conducted using the UK Biobank under application ID 20737 (PI: Andrew McQuillin, Co-I: Elvira Bramon). The UK Biobank study was approved by the North-West Research Ethics Committee (ref 06/MREC08/65) in accordance with the Declaration of Helsinki. This study was supported by a Medical Research Council doctoral studentships awarded to Isabelle Austin-Zimmerman, Anjali Bhat, and Jasmine Harju-Seppänen. Baihan Wang was supported by the China Scholarship Council-University College London Joint Research Scholarship. Haritz Irizar has received funding from the European Union’s Horizon 220 research and innovation programme under the Marie Sklodowska-Curie grant agreement no 747429 and is currently supported by a grant from the National Institute of Allergy and Infectious Diseases, National Institutes of Health. Gazaleh Fatemifar is funded by the American Heart Association Institutional Data Fellowship Program (AHA Award 17IF3389000). Elvira Bramon has recieved the following grant funding that supported this work: National Institute of Health Research UK (NIHR200756); Mental Health Research UK John Grace QC Scholarship 2018; BMA Margaret Temple Fellowship 2016; Medical Research Council New Investigator and Centenary Awards (G0901310, G1100583), MRC (G1100583); NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust and University College London. ## Footnotes * * Joint first authorship * Correcting author list and adding additional figure (fig2). * Received July 7, 2021. * Revision received August 16, 2021. * Accepted August 16, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## References 1. 1.Taylor S, Annand F, Burkinshaw P, Greaves F, Knight J, Perkins C, et al. Dependence and withdrawal associated with some prescribed medicines: an evidence review. 2019 p. 152. 2. 2.Prescription Cost Analysis - England, 2018 [PAS] [Internet]. NHS Digital; 2018 [cited 2020 Aug 24]. Available from: Available from: [https://digital.nhs.uk/data-and-information/publications/statistical/prescription-cost-analysis/2018](https://digital.nhs.uk/data-and-information/publications/statistical/prescription-cost-analysis/2018) 3. 3.Iacobucci G. NHS prescribed record number of antidepressants last year. BMJ. 2019 Mar 29;364:|1508. 4. 4.Andersohn F, Schade R, Suissa S, Garbe E. Long-Term Use of Antidepressants for Depressive Disorders and the Risk of Diabetes Mellitus. Am J Psychiatry. 2009 May 1;166(5):591–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2008.08071065&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19339356&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265662500014&link_type=ISI) 5. 5.Holt RIG. Association Between Antipsychotic Medication Use and Diabetes. Curr Diab Rep. 2019 Sep 2;19(10):96. 6. 6.Association AD. Consensus Development Conference on Antipsychotic Drugs and Obesity and Diabetes. Diabetes Care. 2004 Feb 1;27(2):596–601. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czo4OiIyNy8yLzU5NiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA4LzE2LzIwMjEuMDcuMDcuMjEyNTk5MjYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 7. 7.BMJ Publishing Group Ltd RC of P and BPS. Stable monotherapy with clozapine or olanzapine increases the incidence of diabetes mellitus in people with schizophrenia. Evid Based Ment Health. 2005 Feb 1;8(1):24–24. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15671515&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 8. 8.Leslie DL, Rosenheck RA. Incidence of newly diagnosed diabetes attributable to atypical antipsychotic medications. Am J Psychiatry. 2004 Sep;161(9):1709–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.161.9.1709&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15337666&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223800600029&link_type=ISI) 9. 9.Schwenkreis P, Assion H-J. Atypical antipsychotics and diabetes mellitus. World J Biol Psychiatry Off J World Fed Soc Biol Psychiatry. 2004 Apr;5(2):73–82. 10. 10.Taylor D, Barnes T, Young A. The Maudsley Prescribing Guidelines in Psychiatry, 13th Edition. Wiley-Blackwell; 2018. 11. 11.Kivimäki M, Hamer M, Batty GD, Geddes JR, Tabak AG, Pentti J, et al. Antidepressant medication use, weight gain, and risk of type 2 diabetes: a population-based study. Diabetes Care. 2010 Dec;33(12):2611–6. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czoxMDoiMzMvMTIvMjYxMSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA4LzE2LzIwMjEuMDcuMDcuMjEyNTk5MjYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 12. 12.Lustman PJ, Griffith LS, Clouse RE, Freedland KE, Eisen SA, Rubin EH, et al. Effects of Nortriptyline on Depression and Glycemic Control in Diabetes: Results of a Double-blind, Placebo-controlled Trial. Psychosom Med. 1997 Jun;59(3):241–50. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoicHN5Y2htZWQiO3M6NToicmVzaWQiO3M6ODoiNTkvMy8yNDEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wOC8xNi8yMDIxLjA3LjA3LjIxMjU5OTI2LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 13. 13.Mumoli N, Cocciolo M, Vitale J, Mantellassi M, Sabatini S, Gambaccini L, et al. Diabetes mellitus associated with clomipramine treatment: a retrospective analysis. Acta Diabetol. 2014 Feb;51(1):167–8. 14. 14.Burcu M, Zito JM, Safer DJ, Magder LS, dosReis S, Shaya FT, et al. Association of Antidepressant Medications With Incident Type 2 Diabetes Among Medicaid-Insured Youths. JAMA Pediatr. 2017 Dec 1;171(12):1200–7. 15. 15.Gagnon J, Lussier M-T, MacGibbon B, Daskalopoulou SS, Bartlett G. The Impact of Antidepressant Therapy on Glycemic Control in Canadian Primary Care Patients With Diabetes Mellitus. Front Nutr. 2018;5:47. 16. 16.Hall JA, Wang F, Oakes TMM, Utterback BG, Crucitti A, Acharya N. Safety and tolerability of duloxetine in the acute management of diabetic peripheral neuropathic pain: analysis of pooled data from three placebo-controlled clinical trials. Expert Opin Drug Saf. 2010 Jul 1;9(4):525–37. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20465525&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 17. 17.Barnard K, Peveler RC, Holt RIG. Antidepressant medication as a risk factor for type 2 diabetes and impaired glucose regulation: systematic review. Diabetes Care. 2013 Oct;36(10):3337–45. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czoxMDoiMzYvMTAvMzMzNyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA4LzE2LzIwMjEuMDcuMDcuMjEyNTk5MjYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 18. 18.Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013 Apr;138(1):103–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pharmthera.2012.12.007&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23333322&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 19. 19.Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J. 2005;5(1):6–13. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.tpj.6500285&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15492763&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000227168500002&link_type=ISI) 20. 20.Sim SC, Ingelman-Sundberg M. The Human Cytochrome P450 (CYP) Allele Nomenclature website: a peer-reviewed database of CYP variants and their associated effects. Hum Genomics. 2010; 21. 21.Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C. Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther. 2007 Dec;116(3):496–526. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pharmthera.2007.09.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18001838&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251812000010&link_type=ISI) 22. 22.Gaedigk A, Sangkuhl K, Whirl-Carrillo M, Klein T, Leeder JS. Prediction of CYP2D6 phenotype from genotype across world populations. Genet Med Off J Am Coll Med Genet. 2017 Jan;19(1):69–76. 23. 23.Martis S, Peter I, Hulot J-S, Kornreich R, Desnick RJ, Scott SA. Multi-ethnic distribution of clinically relevant CYP2C genotypes and haplotypes. Pharmacogenomics J. 2013 Aug;13(4):369–77. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/tpj.2012.10&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22491019&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 24. 24.Grasmäder K, Verwohlt PL, Rietschel M, Dragicevic A, Müller M, Hiemke C, et al. Impact of polymorphisms of cytochrome-P450 isoenzymes 2C9, 2C19 and 2D6 on plasma concentrations and clinical effects of antidepressants in a naturalistic clinical setting. Eur J Clin Pharmacol. 2004 Jul;60(5):329–36. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15168101&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 25. 25.Chang M, Tybring G, Dahl M-L, Lindh JD. Impact of cytochrome P450 2C19 polymorphisms on citalopram/escitalopram exposure: a systematic review and meta-analysis. Clin Pharmacokinet. 2014 Sep;53(9):801–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s40262-014-0162-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25154506&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 26. 26.Huezo-Diaz P, Perroud N, Spencer EP, Smith R, Sim S, Virding S, et al. CYP2C19 genotype predicts steady state escitalopram concentration in GENDEP. J Psychopharmacol Oxf Engl. 2012 Mar;26(3):398–407. 27. 27.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 Jul;71(7):835–41. 28. 28.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 Dec;64(12):1181–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00228-008-0533-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18677622&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 29. 29.Schenk PW, van Vliet M, Mathot R a. A, van Gelder T, Vulto AG, van Fessem M a. C, et al. The CYP2C19*17 genotype is associated with lower imipramine plasma concentrations in a large group of depressed patients. Pharmacogenomics J. 2010 Jun;10(3):219–25. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/tpj.2009.50&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19884907&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000277941300006&link_type=ISI) 30. 30.Milosavljevic F, Bukvic N, Pavlovic Z, Miljevic C, Pešic V, Molden E, et al. Association of CYP2C19 and CYP2D6 Poor and Intermediate Metabolizer Status With Antidepressant and Antipsychotic Exposure: A Systematic Review and Meta-analysis. JAMA Psychiatry. 2021 Mar 1;78(3):270–80. 31. 31.Hicks JK, Sangkuhl K, Swen JJ, Ellingrod VL, Müller DJ, Shimoda K, et al. Clinical pharmacogenetics implementation consortium guideline (CPIC) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin Pharmacol Ther. 2016;102(1):37–44. 32. 32.Hicks JK, Bishop JR, Sangkuhl K, Muller DJ, Ji Y, Leckband SG, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake inhibitors. Clin Pharmacol Ther. 2015;98(2):127–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/cpt.147&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25974703&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 33. 33.Dutch Pharmacogenetic Working Group Guidelines November 2018. 2018. 34. 34.Turner RM, Newman WG, Bramon E, McNamee CJ, Wong WL, Misbah S, et al. Pharmacogenomics in the UK National Health Service: opportunities and challenges. Pharmacogenomics. 2020 Nov;21(17):1237–46. 35. 35.van Westrhenen R, Aitchison KJ, Ingelman-Sundberg M, Jukić MM. Pharmacogenomics of Antidepressant and Antipsychotic Treatment: How Far Have We Got and Where Are We Going? Front Psychiatry. 2020;11:94. 36. 36.Bycroft C, Freeman C, Petkova D, Band G, Elliott LT, Sharp K, et al. The UK Biobank resource with deep phenotyping and genomic data. Nature. 2018;562:203–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-018-0579-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30305743&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 37. 37.Sudlow C, Gallacher J, Allen N, Beral V, Burton P, Danesh J, et al. UK Biobank: An Open Access Resource for Identifying the Causes of a Wide Range of Complex Diseases of Middle and Old Age. PLoS Med. 2015;12(3). 38. 38.Conomos MP, Reiner AP, Weir BS, Thornton TA. Model-free Estimation of Recent Genetic Relatedness. Am J Hum Genet. 2016;98(1):127–48. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2015.11.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26748516&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 39. 39.Conomos MP, Miller MB, Thornton TA. Robust inference of population structure for ancestry prediction and correction of stratification in the presence of relatedness. Genet Epidemiol. 2015;39(4):276–93. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/gepi.21896&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25810074&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 40. 40.Manichaikul A, Mychaleckyj JC, Rich SS, Daly K, Sale M, Chen W-M. Robust relationship inference in genome-wide association studies. Bioinforma Oxf Engl. 2010 Nov 15;26(22):2867–73. 41. 41.Gogarten SM, Bhangale T, Conomos MP, Laurie CA, McHugh CP, Painter I, et al. GWASTools: an R/Bioconductor package for quality control and analysis of genome-wide association studies. Bioinformatics. 2012 Dec 1;28(24):3329–31. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bts610&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23052040&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 42. 42.Morris JA, Randall JC, Maller JB, Barrett JC. Evoker: A visualization tool for genotype intensity data. Bioinformatics. 2010; 43. 43.Browning B, Zhou Y, Browning S. A one-penny imputed genome from next generation reference panels. Am J Hum Genet. 2016;103(3):338–48. 44. 44.Browning SR, Browning BL. Rapid and accurate haplotype phasing and missing-data inference for whole-genome association studies by use of localized haplotype clustering. Am J Hum Genet. 2007 Nov;81(5):1084–97. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/521987&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17924348&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250480900018&link_type=ISI) 45. 45.Delaneau O, Marchini JL, McVeanh GA, Donnelly P, Lunter G, Marchini JL, et al. Integrating sequence and array data to create an improved 1000 Genomes Project haplotype reference panel. Nat Commun. 2014;5(3934). 46. 46.(Gaedigk et al. 2018, CPT 103:399; Gaedigk et al. 2019 C 105:29). Pharmacogene Variation Consortium (PharmVar) [Internet]. Available from: [www.PharmVar.org](http://www.PharmVar.org) 47. 47.Gaedigk A, Simon SD, Pearce RE, Bradford LD, Kennedy MJ, Leeder JS. The CYP2D6 activity score: Translating genotype information into a qualitative measure of phenotype. Clin Pharmacol Ther. 2008; 48. 48.Gaedigk A, Dinh JC, Jeong H, Prasad B, Leeder JS. Ten years’ experience with the CYP2D6 activity score: A perspective on future investigations to improve clinical predictions for precision therapeutics. Vol. 8, Journal of Personalized Medicine. 2018. 15 p. 49. 49.Roopan S, Larsen ER. Use of antidepressants in patients with depression and comorbid diabetes mellitus: a systematic review. Acta Neuropsychiatr. 2017 Jun;29(3):127–39. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 50. 50.1. Lyubimov AV Stingl J, Oesterheld J, Turpeinen M. Metabolism of psychotropic drugs. In: Lyubimov AV, editor. Encyclopedia of Drug Metabolism and Interaction. First edit. John Wiley & Sons; 2012. 51. 51.Perneger TV. What’s wrong with Bonferroni adjustments. BMJ. 1998;316(7139):1236–8. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjEzOiIzMTYvNzEzOS8xMjM2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDgvMTYvMjAyMS4wNy4wNy4yMTI1OTkyNi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 52. 52.Li MX, Yeung JMY, Cherny SS, Sham PC. Evaluating the effective numbers of independent tests and significant p-value thresholds in commercial genotyping arrays and public imputation reference datasets. Hum Genet. 2012;131(5):747–56. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00439-011-1118-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22143225&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 53. 53.R Core Team. R: A Language and Environment for Statistical Computing [Internet]. Vienna, Austria: R Foundation for Statistical Computing; 2019. Available from: [https://www.R-project.org/](https://www.R-project.org/) 54. 54.Wickham H. ggplot2: Elegant Graphics for Data Analysis [Internet]. Media. Springer-Verlag New York; 2016. Available from: [http://ggplot2.org](http://ggplot2.org) 55. 55. Matt Dowle, Srinivasan A. data.table: Extension of ‘data.frame [Internet]. 2019. Available from: [https://CRAN.R-project.org/package=data.table](https://CRAN.R-project.org/package=data.table) 56. 56.Kumar Y, Kung S, Shinozaki G. CYP2C19 variation, not citalopram dose nor serum level, is associated with QTc prolongation. J Psychopharmacol Oxf Engl. 2014 Dec;28(12):1143–8. 57. 57.Tay JKX, Tan CH, Chong SA, Tan EC. Functional polymorphisms of the cytochrome P450 1A2 (CYP1A2) gene and prolonged QTc interval in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2007; 58. 58.Lane H-Y, Liu Y-C, Huang C-L, Chang Y-C, Wu P-L, Lu C-T, et al. Risperidone-related weight gain: genetic and nongenetic predictors. J Clin Psychopharmacol. 2006 Apr;26(2):128–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/01.jcp.0000203196.65710.2b&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16633140&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000236559600004&link_type=ISI) 59. 59.Zhang JP, Lencz T, Zhang RX, Nitta M, Maayan L, John M, et al. Pharmacogenetic Associations of Antipsychotic Drug-Related Weight Gain: A Systematic Review and Meta-analysis. Schizophr Bull. 2016; 60. 60.Correia CT, Almeida JP, Santos PE, Sequeira AF, Marques CE, Miguel TS, et al. Pharmacogenetics of risperidone therapy in autism: association analysis of eight candidate genes with drug efficacy and adverse drug reactions. Pharmacogenomics J. 2010 Oct;10(5):418–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/tpj.2009.63&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19997080&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000282619700005&link_type=ISI) 61. 61.Sukasem C, Hongkaew Y, Ngamsamut N, Puangpetch A, Vanwong N, Chamnanphon M, et al. Impact of Pharmacogenetic Markers of CYP2D6 and DRD2 on Prolactin Response in Risperidone-Treated Thai Children and Adolescents With Autism Spectrum Disorders. J Clin Psychopharmacol. 2016;36(2):1. 62. 62.Fleeman N, Dundar Y, Dickson R, Jorgensen A, Pushpakom S, McLeod C, et al. Cytochrome P450 testing for prescribing antipsychotics in adults with schizophrenia: systematic review and meta-analyses. Pharmacogenomics J. 2011; 63. 63.Calafato MS, Austin-Zimmerman I, Thygesen JH, Sairam M, Metastasio A, Marston L, et al. The effect of CYP2D6 variation on antipsychotic-induced hyperprolactinaemia: a systematic review and meta-analysis. Pharmacogenomics J. 2020 Oct;20(5):629–37. 64. 64.Chávez-Castillo M, Ortega Á, Nava M, Fuenmayor J, Lameda V, Velasco M, et al. Metabolic risk in depression and treatment with selective serotonin reuptake inhibitors: are the metabolic syndrome and an increase in cardiovascular risk unavoidable? Vessel Plus [Internet]. 2018 Apr 18 [cited 2021 Mar 17];2. Available from: [https://vpjournal.net/article/view/2486](https://vpjournal.net/article/view/2486) 65. 65.Pan A, Sun Q, Okereke OI, Rexrode KM, Rubin RR, Lucas M, et al. Use of antidepressant medication and risk of type 2 diabetes: results from three cohorts of US adults. Diabetologia. 2012 Jan;55(1):63–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00125-011-2268-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21811871&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000297739000012&link_type=ISI) 66. 66.Yoon JM, Cho E-G, Lee H-K, Park SM. Antidepressant use and diabetes mellitus risk: a meta-analysis. Korean J Fam Med. 2013 Jul;34(4):228–40. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4082/kjfm.2013.34.4.228&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23904952&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 67. 67.Sindrup SH, Brøsen K, Gram LF, Hallas J, Skjelbo E, Allen A, et al. The relationship between paroxetine and the sparteine oxidation polymorphism. Clin Pharmacol Ther. 1992 Mar;51(3):278–87. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/clpt.1992.23&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1531950&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1992HK42900007&link_type=ISI) 68. 68.Solai LK, Pollock BG, Mulsant BH, Frye RF, Miller MD, Sweet RA, et al. Effect of nortriptyline and paroxetine on CYP2D6 activity in depressed elderly patients. J Clin Psychopharmacol. 2002 Oct;22(5):481–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00004714-200210000-00007&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12352271&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 69. 69.Zourková A, Hadasová E. Paroxetine-induced conversion of cytochrome P450 2D6 phenotype and occurence of adverse effects. Gen Physiol Biophys. 2003 Mar;22(1):103–13. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12870705&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000183539200009&link_type=ISI) 70. 70.Khoza S, Barner JC, Bohman TM, Rascati K, Lawson K, Wilson JP. Use of antidepressant agents and the risk of type 2 diabetes. Eur J Clin Pharmacol. 2012 Sep;68(9):1295–302. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00228-011-1168-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22120432&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 71. 71.Biagetti B, Corcoy R. Hypoglycemia associated with fluoxetine treatment in a patient with type 1 diabetes. World J Clin Cases. 2013 Aug 16;1(5):169–71. 72. 72.Rebai R, Jasmin L, Boudah A. The antidepressant effect of melatonin and fluoxetine in diabetic rats is associated with a reduction of the oxidative stress in the prefrontal and hippocampal cortices. Brain Res Bull. 2017 Sep 1;134:142–50. 73. 73.Baumeister H, Hutter N, Bengel J. Psychological and pharmacological interventions for depression in patients with diabetes mellitus and depression. Cochrane Database Syst Rev [Internet]. 2012 [cited 2021 Mar 26];(12). Available from: [https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD008381.pub2/full](https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD008381.pub2/full) 74. 74.Baumann P, Jonzier-Perey M, Koeb L, Küpfer A, Tinguely D, Schöpf J. Amitriptyline pharmacokinetics and clinical response: II. Metabolic polymorphism assessed by hydroxylation of debrisoquine and mephenytoin. Int Clin Psychopharmacol. 1986 Apr;1(2):102–12. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=3571939&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1986F845500002&link_type=ISI) 75. 75.1. Pratt VM, 2. Scott SA, 3. Pirmohamed M, 4. Esquivel B, 5. Kane MS, 6. Kattman BL, et al., editors Dean L. Amitriptyline Therapy and CYP2D6 and CYP2C19 Genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kane MS, Kattman BL, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012 [cited 2021 Jun 4]. Available from: [http://www.ncbi.nlm.nih.gov/books/NBK425165/](http://www.ncbi.nlm.nih.gov/books/NBK425165/) 76. 76.Gaziano JM, Concato J, Brophy M, Fiore L, Pyarajan S, Breeling J, et al. Million Veteran Program: A mega-biobank to study genetic influences on health and disease. J Clin Epidemiol. 2016;70:214–23. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jclinepi.2015.09.016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26441289&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 77. 77.Xue A, Wu Y, Zhu Z, Zhang F, Kemper KE, Zheng Z, et al. Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat Commun. 2018 Jul 27;9(1):2941. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-018-04951-w&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30054458&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F08%2F16%2F2021.07.07.21259926.atom) 78. 78.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; 79. 79.McInnes G, Lavertu A, Sangkuhl K, Klein TE, Whirl-Carrillo M, Altman RB. Pharmacogenetics at Scale: An Analysis of the UK Biobank. Clin Pharmacol Ther. 2020 Nov 25; 80. 80.Müller DJ, Kekin I, Kao ACC, Brandl EJ. Towards the implementation of CYP2D6 and CYP2C19 genotypes in clinical practice: update and report from a pharmacogenetic service clinic. Int Rev Psychiatry Abingdon Engl. 2013 Oct;25(5):554–71. 81. 81.Altar CA, Carhart JM, Allen JD, Hall-Flavin DK, Dechairo BM, Winner JG. Clinical validity: Combinatorial pharmacogenomics predicts antidepressant responses and healthcare utilizations better than single gene phenotypes. Pharmacogenomics J. 2015 Oct;15(5):443–51. 82. 82.Arranz MJ, Gonzalez-Rodriguez A, Perez-Blanco J, Penadés R, Gutierrez B, Ibañez L, et al. A pharmacogenetic intervention for the improvement of the safety profile of antipsychotic treatments. Transl Psychiatry. 2019 Jul 25;9(1):1–8. 83. 83.Greden JF, Parikh SV, Rothschild AJ, Thase ME, Dunlop BW, DeBattista C, 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 Apr;111:59–67. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpsychires.2019.01.003&link_type=DOI) 84. 84.Herbild L, Andersen SE, Werge T, Rasmussen HB, Jürgens G. Does pharmacogenetic testing for CYP450 2D6 and 2C19 among patients with diagnoses within the schizophrenic spectrum reduce treatment costs? Basic Clin Pharmacol Toxicol. 2013;113(4):266–72.