Skip to main content
Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie logoLink to Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie
. 2014 Feb;59(2):76–88. doi: 10.1177/070674371405900203

Pharmacogenetics of Antipsychotics

Eva J Brandl 1, James L Kennedy 2, Daniel J Müller 3,
PMCID: PMC4079237  PMID: 24881126

Abstract

Objective:

During the past decades, increasing efforts have been invested in studies to unravel the influence of genetic factors on antipsychotic (AP) dosage, treatment response, and occurrence of adverse effects. These studies aimed to improve clinical care by predicting outcome of treatment with APs and thus allowing for individualized treatment strategies. We highlight most important findings obtained through both candidate gene and genome-wide association studies, including pharmacokinetic and pharmacodynamic factors.

Methods:

We reviewed studies on pharmacogenetics of AP response and adverse effects published on PubMed until early 2012. Owing to the high number of published studies, we focused our review on findings that have been replicated in independent studies or are supported by meta-analyses.

Results:

Most robust findings were reported for associations between polymorphisms of the cytochrome P450 system, the dopamine and the serotonin transmitter systems, and dosage, treatment response, and adverse effects, such as AP-induced weight gain or tardive dyskinesia. These associations were either detected for specific medications or for classes of APs.

Conclusion:

First promising and robust results show that pharmacogenetics bear promise for a widespread use in future clinical practice. This will likely be achieved by developing algorithms that will include many genetic variants. However, further investigation is warranted to replicate and validate previous findings, as well as to identify new genetic variants involved in AP response and for replication of existing findings.

Keywords: pharmacogenetic, antipsychotic, treatment response, adverse effect, dopamine, serotonin, cytochrome P450 system, candidate gene, genome-wide association


Since the first observations in the 1950s suggested genetic influences on drug response,1 pharmacogenetic studies have rapidly evolved over the past decades. Modern laboratory techniques have given insights toward the identification of genetic variants influencing drug efficacy, metabolism, and occurrence of adverse effects. This is giving pharmacogenetics an important role in contemporary psychiatric research, strengthening the notion that the widespread use of genetic tests will become available in future clinical practice.

Besides studies investigating the genetics of outcome to antidepressants and mood stabilizers2 (see also the In Review paper by Dr Serretti and colleagues3 in this issue), large efforts have been undertaken in the area of pharmacogenetics of APs. The importance of these efforts is increasing because APs are routinely used in the treatment not only of schizophrenia and related spectrum disorders but also for mood disorders and various other conditions, such as obsessive–compulsive disorder, eating disorders, or behavioural disturbances, associated with dementia. Specific APs may differ regarding alleviating various symptoms; for instance, SGAs may be more effective for the treatment of negative symptoms. APs also differ in their propensity to induce specific adverse effects; for example, SGAs may be associated with a higher risk of significant metabolic disturbances but a lower risk of TD, compared with FGAs.4 More importantly, large interindividual differences exist among patients. These differences cannot be predicted clinically but they can have serious consequences leading to repeated medication switches owing to intolerability or lack of efficacy. This issue is of particular relevance in patients with schizophrenia who need to be treated over a long period of time and whose adherence is hampered by adverse effects.5 Also, treatment resistance (that is, a lack of adequate response to medication) occurs in up to 40% of these patients.6 The possibility of meaningful prediction of individual responses and risks of adverse effects would, therefore, represent a milestone in AP pharmacotherapy.

To date, only a limited number of factors have been identified that are described to be either positively or negatively correlated with outcome of AP treatment. Among those are demographic factors (for example, family history, ethnicity, and gender), clinical factors (for example, duration of untreated psychosis and early response to APs), and environmental factors (for example, smoking habits, concomitant treatment, and diet).7 The potential influence of genetic factors is underpinned by different studies demonstrating similar treatment response and occurrence of adverse effects in monozygotic twins or first-degree relatives.812

Clinical Implications

  • Accurate prediction of individual response to AP treatment or individual risk for the occurrence of adverse effects has not yet been achieved. Large efforts have been made to find genetic factors in the hope of improving future patient care.

  • Some genetic factors have been identified. However, valid tests with substantial specificity and sensitivity measures have not yet been developed to predict drug metabolism, response to treatment, or most adverse effects.

  • Nonetheless, some results are starting to impact patient care through the development of genetic tests.

Limitations

  • Lack of independent replication in well-characterized samples limits the clinical applicability of most of these genetic findings.

  • Approaches integrating genome-wide, DNA sequencing, epigenetic and environmental factors, methods for prediction of treatment response, and occurrence of adverse effects are required to allow for a more widespread clinical use of pharmacogenetic-based algorithms.

Most studies conducted to date have used a candidate gene approach, investigating SNPs or changes in larger DNA segments of a gene considered to be involved in the mechanisms of medication response or in a given adverse effect. With further development of laboratory techniques, GWASs and next-generation sequencing are being applied to pharmacogenetic research. Although there are some inconsistencies in the results of some studies, efforts are being made to transfer some promising findings into clinical practice. Our article reviews the most robust pharmacogenetic results, including pharmacokinetic and pharmacodynamic factors, relevant to APs.

Methods

We reviewed studies on pharmacogenetics of AP response and adverse effects published on PubMed until January 2012. Owing to the large amount of available literature, a detailed summary of all studies would go beyond the scope of our review. Also, several recent reviews have focused on AP response and adverse events.7,1315 Thus we focused our review on pharmacogenetics findings that have been replicated in independent studies or are supported by meta-analyses. We also included selected studies that highlight new and promising candidate genes in relevant pathways.

Genetics and Pharmacokinetic Factors

Cytochrome Enzymes

Most psychotropics, including APs, are metabolized by cytochrome enzymes, mainly members of the CYP family, which includes CYP1A2, CYP2D6, CYP3A4, and CYP2C19. Besides environmental factors, such as induction of the CYP1A2 enzyme by smoking, genetic variation contributes to the variability in enzyme activity and drug metabolism. The genes encoding CYP enzymes can be highly polymorphic,16 with more than 80 genetic variations known in CYP2D6. For this gene, a distinction is made among extensive metabolizers, with normal enzyme activity having 2 functional alleles; PMs, with low enzyme activity caused by 2 nonfunctional alleles; IMs, with intermediate activity carrying 2 partly defective alleles or 1 nonfunctional allele; and UMs, with more than 2 functional alleles. Similarly, an activity score system was recently proposed.17 The main polymorphisms causing defective alleles in the population of European descent are CYP2D6*3, CYP2D6*4, CYP2D6*5, and CYP2D6*6.18 Genotype distribution differs considerably between ethnicities: 1% to 2% of Europeans are UMs and 5% to 10% are PMs, whereas only 1% to 2% of Asians are PMs, while 30% to 40% of the population in North Africa are UMs but only 1% are PMs.19 Metabolizer status highly influences the required dosage of substances; for example, it has been demonstrated that carriers of CYP2D6 defective allele (that is, *3, *4, *5, and *6) can have up to 80% higher serum concentrations of risperidone, compared with homozygous carriers of the wildtype CYP2D6*1.20 The *1F polymorphism in CYP1A2 seems to be related to higher inducibility of the enzyme by smoking, leading to reduced clozapine plasma levels.21 Polymorphisms in CYP2C19 and CYP3A4 have also been reported to affect clozapine plasma levels.22,23

These data suggest that CYP polymorphisms may affect treatment response. However, there are only a few studies with positive findings, most likely because plasma levels of APs often do not correlate with treatment response. For CYP1A2, the *1F polymorphism has been associated with poorer treatment response to clozapine and olanzapine.2426 One other study27 indicated a weak effect of CYP3A4 polymorphisms on risperidone response. A recent study by Bigos et al28 reported a highly significant influence of the marker rs472660 in CYP3A43, a member of the CYP3A4 enzyme family encoding the CYP family 3, subfamily A, polypeptide 43, on olanzapine clearance and response. By contrast, several studies investigating the influence of CYP2D6 polymorphisms on olanzapine response have reported no associations.2932

There are more data available indicating an impact of CYP polymorphisms on side effects: CYP2D6*3, CYP2D6*4, and CYP2D6*10, but not CYP1A2*1F, have been associated with increased AIWG.24,33,34

Today, most evidence exists for an influence of CYP polymorphisms on motor adverse effects of APs, such as TD. Although some studies reported negative findings,3538 there is growing evidence that CYP2D6 PMs are at higher risk of developing acute EPSs39,40 or TD4143 (see reviews13,44,45). Some studies have also associated CYP1A2*1F with TD,42,46 but other studies and a meta-analysis have yielded negative findings.47,48

In summary, genetic variation in CYP genes seems to exert a stronger influence on plasma levels and the occurrence of adverse effects than on medication response. As sample sizes of many studies are small and these studies have been performed using different medications and protocols, further investigation is warranted to fully assess clinical implications of these findings. Further, there is still a paucity of studies assessing the cost-effectiveness of CYP genotyping in daily practice,19,45,49 and further pharmacoeconomic investigations are needed. Despite these limitations, first steps to bring pharmacogenetics advances into clinical practice have been undertaken by offering tests for CYP2D6 and CYP2D19 usable for medication monitoring.50 For information regarding ongoing related investigations in Canada, please see the Pharmacogenetics Research Clinic website.51,52

Blood–Brain Barrier Transporters

P-gp is a member of the ATP-binding cassette transporters and is involved in the transmembrane efflux of different substrates, including toxins, peptides, and medications. P-gps are located in different organs, including the blood–brain barrier, where they pump medications back into the plasma.53 An altered transporter activity influences intracerebral drug concentration and may thereby impact on treatment response as well. Polymorphisms in the encoding gene MDR1 (synonymous with ABCB1, ATP-binding cassette, subfamily B [MDR/transporter associated with antigen processing], member 1 gene) have been investigated in several studies. Although there are negative findings,54,55 some positive findings exist for the G2677T/A and C3435T polymorphisms. Contradictory results have been reported, with studies showing better treatment response of T allele carriers of both polymorphisms,56 while others reported worse response in the T allele carriers of C3435T.57,58 Other studies found no association in the above-mentioned polymorphisms but had positive results for C1236T.59 Overall, despite ambiguous evidence for the role of MDR1 polymorphisms in treatment response, probably owing to yet undetected confounding variables (for example, ethnicity), a role of this gene is likely and awaits further clarification.

Regarding AP-induced adverse effects, there has been one study showing no association of MDR1 polymorphisms with TD,60 one study demonstrating an association with a specific haplotype and TD,61 and one study indicating an influence of MDR1 genotypes on the occurrence of EPSs,30 while another study could not find an association with EPSs.55 There are also hints of an influence of MDR1 on weight gain62 and metabolic disturbances caused by APs.63 Overall, more studies are needed to assess the role of MDR1 polymorphisms in the occurrence of adverse effects.

Genetics and Pharmacodynamic Factors of Treatment Response

The DA System

The antidopaminergic effect of FGA and SGA is thought to be their main mechanism of action.64 Therefore, genetic variation in DA receptors influencing receptor density, expression, and activity of the receptor may be an essential factor in regulation of individual treatment response.

As all APs block D2 receptors,65 polymorphisms in the DRD2 gene have been investigated most intensively. The del allele of the −141C ins/del polymorphism in the promoter region is assumed to reduce D2 density and activity. In several studies, the del allele was associated with poorer response to AP treatment, for example, see Lencz et al66 and Wu et al.67 This finding has been confirmed by a recent meta-analysis,68 despite some studies that failed to find an association with treatment response (for example, see Xing et al69). The A1 allele of TaqIA polymorphism, which is located 10 kilobases downstream DRD2, leads to reduced gene expression and was therefore also hypothesized to influence treatment response. However, previous studies have reported inconsistent findings: while some studies indicated that A17072 or A273 are associated with better treatment response, the above-mentioned meta-analysis68 did not detect any association. Also for other DRD2 polymorphisms, for example, TaqIB,74 Ser311Cys,75 or A-241G,66 a few studies have reported associations, but contradictory results and lack of replication make further investigations necessary.

In DRD3, the Gly9 variant of the Ser9Gly polymorphism, changes D3 receptor density. Thus this impact of this variant on AP treatment response has been studied extensively, with at least 18 published studies.13 While some studies have indicated better response of the Ser allele, a meta-analysis has reported a nonsignificant trend toward lower response in carriers of Ser allele.76 Because clozapine also binds to D4 receptors, a 48-bp variable number tandem repeat in the DRD4 gene has been investigated in several studies. However, findings have been contradictory,77,78 and many studies have yielded negative results (for example, see Rietschel et al79 and Ikeda et al80); the role of the polymorphism in treatment response is not fully resolved, to date. Polymorphisms of the DRD181 and DRD580 genes have not yet been investigated intensively and their clinical relevance needs to be demonstrated in future studies.

The 5-HT System

Because of the higher affinity of SGAs to 5-HT receptors, compared with FGAs, genes in the serotonergic transmitter system are interesting candidates for investigation of AP treatment response. Several studies have focused on polymorphisms in HTR2A, the gene encoding 5-HT2A receptors. The functional −1438A/G SNP in the promoter region of the gene and the SNP 102T/C are in high linkage disequlibrium with each other82 and −1438A/G has been demonstrated to influence promoter activity.83 Association studies have repeatedly indicated poorer treatment response in G allele carriers of the −1438A/G SNP,84,85 although there are negative findings71 and a study showing better response in G allele carriers.86 Regarding the 102T/C marker, results are less consistent with some studies (see Anttila et al87) and an early meta-analysis focusing on clozapine demonstrating better response in T allele carriers,88 while later studies investigating other APs found no association80 or association of the C allele with better symptom improvement.89 Another polymorphism repeatedly investigated in HTR2A is His452Tyr (histidine and Tyr at the 452nd amino acid): the Tyr allele seems to be associated with poorer treatment response,88 but this has not been observed in all studies.90

In HTR2C, encoding 5-HT2C receptors, the C/C genotype of the −759C/T SNP, which has been shown to lead to reduced transcription,91 was described to be associated with improvement of negative symptoms in one study.92 Also, association of the Ser allele of Cys23Ser has been reported once,93 but all other studies90,94 have failed to replicate this finding, indicating no major role of the SNP in treatment response. In the gene encoding solute carrier family 6 (neurotransmitter transporter), member 4 (SLC6A4, synonymous with 5-HTT) 5-HT transporters, which terminate 5-HT action by transporting it back into the presynaptic neuron, an association of the short allele of the 44-bp ins/del polymorphism serotonin-transporter-linked polymorphic region (commonly referred to as 5-HTTLPR) with poorer AP response has consistently been demonstrated.9597

Further association with AP treatment response has been described for the G allele of −1019C/G SNP in 5-HT receptor 1A, G protein-coupled (HTR1A), encoding 5-HT1A receptors,98,99 for polymorphisms in the genes encoding subtypes 3A, B, and E of 5-HT3 receptors100,101 and for the 267C/T SNP in the gene encoding 5-HT6 receptors.102,103 However, this last finding has not been confirmed in all studies.80

Other Systems

Other candidate genes that may be involved in regulation of individual treatment response to APs have not been studied as extensively as the above-mentioned genes. However, several studies have suggested an association between response and several genetic variants. For example, the COMT gene encodes catechol-O-methyltransferase, which is involved in the degradation of monoamines. The Val(108/158)Met polymorphism of this gene influences activity of the enzyme, which is less active in Met/Met carriers. The Met allele has been repeatedly associated with better improvement of cognitive function104,105 during AP treatment, compared with the Val allele. Other studies106 have found no association or reported an association of the Val allele with better response.107 Therefore, the role of the Val(108/158)Met SNP in the regulation of treatment response is not yet fully understood and deserves further study.

Further, studies reported associations in GNB3, encoding the β-subunit of G-protein, with the T allele of the −825C/T polymorphism leading to worse treatment response,108,109 and in the genes encoding brain-derived neurotrophic factor,110,111 GFRA,112 oxytocin,113 and TNF114; however, those genes need to be studies more extensively.

In summary, candidate gene studies have delivered some promising results, consisting mainly of associations between genes involved in the DA system and overall treatment response or between genes involved in the serotonergic system and improvement of negative symptoms (Table 1). Therefore, findings suggest a clinical impact of those genetic variants. However, given the relatively small sample sizes and inconsistent results, further studies with larger sample sizes and more homogenous studies are needed before these findings can be incorporated in clinical algorithms.

Table 1.

Polymorphisms associated with AP treatment response supported by a meta-analysis or by 3 or more studies reporting the same allele association

Gene Polymorphism Main association finding Study
CYP1A2 *1F *1F/*1F genotype associated with lower plasma concentrations and lower response Laika et al,24 Ozdemir et al,25 Eap et al26
DRD2 −141C ins/del Del allele is associated with poorer response Lencz et al,66 Wu et al,67 Zhang et al68
TaqIA A1 allele associated with better response in some studies; but a meta-analysis showed negative results Suzuki et al,70 Schafer et al,71 Yamanouchi et al,72 Zhang et al68
DRD3 Ser9Gly Meta-analysis with trend for lower response in Ser-allele carriers Hwang et al76
HTR2A −1438G/A G allele was associated with poorer response Arranz et al,84 Ellingrod et al,85 Chen et al177
102T/C T allele was associated with better response in a meta-analysis Arranz et al88
His452Tyr Tyr allele was reported to be associated with poor response Arranz et al,88 Arranz et al,178 Masellis et al179
5-HTT 5-HTTLPR Short-allele was demonstrated to be associated with poorer treatment response Dolzan et al,95 Arranz et al,96 Vazquez-Bourgon et al97
COMT Val(108/158)Met Better improvement of cognitive function was associated with Met allele Bertolino et al,104 Woodward et al,105 Bertolino et al180

Genome-Wide Association Studies

During the past few years, the development of technologies has facilitated the investigation of polymorphisms in the whole genome through GWASs. Studies investigating the CATIE sample5 have reported the association of an intergenetic SNP on chromosome 4p15 with response to APs. Also, borderline significant results have been reported for SNPs in the ankyrin repeat and sterile alpha motif domain containing 1B (ANKS1B) and contactin associated protein-like 5 (CNTNAP5) genes115 as well as association of 6 SNPs in or near to the ets homologous factor (EHF), solute carrier family 26 (anion exchanger), member 9 (SLC26A9), DRD2, G protein-coupled receptor 137B (GPR137B), carbohydrate (N-acetylgalactosamine 4-0) sulfotransferase 8 (CHST8), and interleukin 1, alpha (IL1A) genes.116 In other samples, ATPase, Ca++ transporting, plasma membrane 2 (ATP2B2), heparan sulfate (glucosamine) 3-O-sulfotransferase 2 (HS3ST2), unc-5 homolog C (C elegans) (UNC5C), BCL2-associated athanogene 3 (BAG3), phosphodiesterase 7B (PDE7B), phosphoribosylaminoimidazole carboxylase, phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS), prostaglandin F2 receptor inhibitor (PTGFRN), nuclear receptor subfamily 3, group C, member 2 (NR3C2), zinc finger and BTB domain containing 20 (ZBTB20), ST6 beta-galactosamide alpha-2,6-sialyltranferase 2 (ST6GAL2), phosphatidylinositol-4-phosphate 5-kinase, type I, beta (PIP5K1B), ephrin receptor type A6 (EPHA6), potassium voltage-gated channel, sub-family H (eag-related), member 5 (KCNH5), and adherens junctions associated protein 1 (AJAP1) have been reported to be associated with response to risperidone.117 Similarly, SNPs within neuronal PAS domain protein 3 (NPAS3), XK, Kell blood group complex subunit-related family, member 4 (XKR4), tenascin R (TNR), glutamate receptor, ionotropic, AMPA 4 (GRIA4), GFRA2, and the nudix (nucleoside diphosphate linked moiety X)-type motif 9 pseudogene 1 (NUDT9P1) located in the 5-HT receptor 7, adenylate cyclase-coupled (HTR7) gene have been reported to be associated with response to iloperidone.118 In summary, hypothesis-free GWASs have yielded numerous interesting findings that warrant further investigation.

Genetics and Pharmacodynamic Factors of Adverse Effects

Antipsychotic-Induced Weight Gain

AIWG is observed in up to 30% of patients treated with SGAs. Owing to the higher morbidity and mortality associated with obesity and metabolic syndrome, as well as the social stigmatization and noncompliance that arises from weight gain (extensively reviewed elsewhere14,119), this serious adverse effect needs special consideration in clinical practice. However, there is a large variation in the propensity of various APs to cause AIWG,120 with olanzapine and clozapine causing the most extensive amount of weight gain. Interindividual variability is also influenced by clinical factors, such as sex, baseline weight, and age. However, there are no reliable clinical predictors available in clinical practice. As results of twin studies indicate a genetic influence on AIWG,11,12,121 numerous candidate studies have been performed during the past few years. Most consistent findings exist for an association between polymorphisms of the HTR2C gene and AIWG. The functional polymorphism −759C/T122 has repeatedly been shown to influence AIWG, with the C allele being overrepresented in patients with higher AIWG (for example, see Reynolds et al,123 Miller et al,124 and Opgen-Rhein et al,125 including 2 meta-analyses, see De Luca et al126 and Sicard et al127). Although negative findings have been reported,128,129 with one study indicating the opposite allele as a risk variant,130 to date, the role of this polymorphism is supported by the most robust studies. Other HTR2C polymorphisms have been shown to be associated with AIWG as well, including the −995G/A, −1165A/G,125 and Cys23Ser polymorphisms as part of a high risk haplotype.127

Leptin plays a major role in energy homoeostasis. The −2548A/G polymorphism in the encoding gene leptin (LEP) has been shown to impact on AIWG, with the G allele being the risk allele in some but not in all studies.131,132 Nonetheless, negative findings (for example, see Opgen-Rhein et al125; for review, see Lett et al14 and Lee and Bishop133) hamper the assessment of the clinical impact of this polymorphism. Other replicated associations include Ddell, Mnll, and Tail polymorphisms in synaptosomal-associated protein, 25kDa (SNAP25),134,135 and in the gene encoding alpha-2-adrenergic receptor, adrenoceptor alpha 2A (ADRA2A), with the G/G genotype of −1291C/G being at higher risk for AIWG.136,137 Similarly, the T allele carriers of the −825C/T polymorphism in the gene encoding β3-subunit of G-protein receptors, that is, GNB3, are at higher risk for AIWG.129,138 Other reported associations, such as polymorphisms in insulin-induced gene 2 (INSIG2),139 have failed to be replicated in other samples.125,140 Recent studies indicate an influence of DRD2 polymorphisms,141,142 cholecystokinin B receptors,143 and adiponectin.144 A systematic analysis of the DA D1–D5 receptors and the association with weight gain reported a significant finding for the 957C/T polymorphism and 2 weak signals for DRD3 polymorphisms.142 Most recent data indicate a strong effect in the region of MC4R, the gene encoding the melanocortin 4 receptor, on development of obesity.145 A GWAS in a cohort of drug-naive adolescents showed significant association of an SNP near this gene with weight gain induced by several APs, which was replicated independently by our group.146 In a different study, our group has also shown a significant association of another SNP in the promoter region of MC4R with AIWG.147

A recent GWAS found significant associations of AIWG with SNPs in the Meis homeobox 2 (MEIS2), protein kinase, cyclic adenosine monophosphate-dependent, regulatory, type II, beta (PRKAR2B), G protein-coupled receptor 98 (GPR98), formin homology 2 domain containing 3 (FHOD3), ring finger protein 144A (RNF144A), astrotactin 2 (ASTN2), sex determining region Y-box 5 (SOX5), and activating transcription factor 7 interacting protein 2 (ATF7IP2) genes148 but these findings have not yet been replicated in other studies. Thus further investigation remains necessary. Table 2 presents a summary of the most important genetic findings regarding AIWG. Overall, except for the polymorphism −759C/T of HTR2C, with most robust findings obtained in a large number of studies, genetic findings regarding AIWG are not yet applicable in clinical practice.

Table 2.

Polymorphisms associated with AIWS or EPSs supported by a meta-analysis or by 3 or more studies reporting the same allele association

Gene Polymorphism Main association finding Study
CYP2D6 metabolizer status PMs were at higher risk for EPSs in several studies Kobylecki et al,41 Fu et al,42 Patsopoulos et al43
HTR2C −759C/T Two meta-analyzes reported C allele to be associated with larger AIWG Sicard et al,127 De Luca et al181
LEP −2548A/G G allele was associated with higher AIWG in most studies; others showed A allele to be associated Ellingrod et al,131 Zhang et al,132 Templeman et al182; Zhang et al183
GNB3 −825C/T T allele was associated with higher AIWG in several studies, and with a nonsignificant trend in a meta-analysis Ujike et al,129 Wang et al,138 Souza et al184
DRD2 TaqIA A2 allele was associated with higher risk for TD in 2 meta-analyzes Bakker et al,48 Zai et al154
DRD3 Ser9Gly Gly allele was associated with higher risk for TD in several studies de Leon et al,60 Steen et al,155 Al Hadithy et al,156 Lerer et al,157 Bakker et al158
HTR2A T102C C allele was associated with TD in a meta-analysis Bakker et al,48 Lerer et al164
COMT Val(108/158)Met Met allele was protective against TD in a meta-analysis Bakker et al48
MnSOD Ala-9Val Val allele was protective against TD in a meta-analysis Bakker et al48

Ala = alanine

Agranulocytosis

Agranulocytosis is a serious adverse effect occurring with a wide range of psychotropic medications. While clozapine is the most effective medication for treatment-resistant schizophrenia,15 it is associated with an incidence of up to 2% of agranulocytosis. Hypothesized mechanisms of CIA include immune-mediated response against neutrophils, enhanced release or destruction of neutrophils, and direct toxicity against bone marrow stromal cells.149 Association studies investigating genetic factors of CIA have mainly focused on genes building the HLA system. Positive findings exist for the HLA-B38 marker, which is overrepresented in patients affected by CIA as well as a haplotype consisting of HLA-B38, DR4, and DQw3.150 Other HLA-antigens associated with CIA include HLA-DRB5*0201 and HLA-Cw-7 and haplotypes with HLA-Cw-B and HLA-DRB5-DRB4.151 In addition, other genes for non-HLA components of the MHC have been investigated. For both TNF and heat shock protein replicated positive findings have been reported.15 There are also associations for genes that are not part of the MHC, for example, myeloperoxidase (MPO) and nicotinamide adenine dinucleotide phosphate-oxidase gene polymorphisms.152,153 First steps toward a clinical use have been made by the temporary release of a commercial test-kit in 2007, that is, PGxPredict:CLOZAPINE (Clinical Data, Inc, New Haven, CT), which included the 6672G/C polymorphism in MHC, class II, DQ beta 1 (HLA-DQB1) for detection of high-risk patients carrying the C allele, with high specificity of 98.4% but a low of sensitivity of 21.5%.

TD and EPSs

TD is a serious adverse effect that occurs in roughly 25% of all AP-treated patients. FGAs bear a much higher risk for the development of TD than SGAs. Clinical risk factors include advanced age, dose and duration of medication, and sex.45 Familial occurrence points to a genetic influence on TD.10 Candidate genes for TD have been extensively investigated during the past 2 decades, and there are numerous studies supporting the hypothesis of a genetic implication. Besides positive findings for pharmacokinetic factors (see above), there is growing evidence for pharmacodynamic factors having an impact on TD.

In DRD2, an increased risk for the development of TD has been described for carriers of the A2 allele of the TaqIA polymorphism in 2 meta-analyzes,48,154 although more studies have delivered negative findings than positive ones.13 Most robust findings exist for the Ser9Gly polymorphism in DRD3, with the Gly allele being associated with higher risk of TD in different ethnicities (for examples, see de Leon et al,60 Steen et al,155 Al Hadithy,156 Lerer et al,157 and Bakker et al158). Nonetheless, results are not unambiguous as negative results also exist,38,159,160 and one study indicated the opposite allele as risk variant.161 DRD4 has not been studied as extensively as DRD2 and DRD3. Nonetheless, there have been positive findings for DRD4 polymorphisms,162,163 which need further replication. Several studies have reported associations for polymorphisms in serotonergic genes. A meta-analysis found an association of the C allele in the 102T/C polymorphism of HTR2A with TD.164 Nonetheless, there are numerous negative findings for this SNP as well (for example, see Wilffert et al165). Studies also reported positive results with the −1438G/A166 in HTR2A and the Cys23Ser polymorphism in HTR2C (for example, see Al Hadithy et al156). As there are many studies with negative findings,38,167 further studies of 5-HT genes remain necessary to evaluate their role in the development of TD. Other significant findings in meta-analyses exist for the Val(108/158)Met polymorphism in COMT48 (with the Met allele being protective against TD) and for manganese superoxide dismutase (MnSOD)44 (with the Val allele of alanine (Ala-9Val) polymorphism being protective against TD48). There are single reports of positive results for a few other genes, including different COMT polymorphisms,168 glycogen synthase kinase 3 beta (GSK3B), linked to dopaminergic signalling,169 and nitric oxide synthase 3 (endothelial cell) (NOS3).170

There has also been reports of an influence of regulator of G-protein signalling 2, 24kDa (RGS2) polymorphisms on EPSs.171,172 A GWAS analyzing the CATIE sample5,173 reported associations in the gene encoding zinc finger protein 202, (ZNF202), in GLI family zinc finger 2 (GLI2), a gene encoding a transcription factor involved in embryonal development of the dopaminergic system,174 and statistical trends for other genes.175

In summary, several candidate genes have been found to be involved in TD, and the most robust findings were obtained in dopaminergic genes. Table 2 summarizes the most important genetic findings. As TD and EPSs are mainly mediated by the influence of APs on the DA system, those findings are hardly surprising. However, to date, there is no definite evidence on the genetics of TD, and further research is needed to develop predictive tests that can be used in clinical practice.

Summary and Outlook

During the past 20 years, pharmacogenetic studies have identified many genetic variants implicated in serum levels, response to APs, and occurrence of adverse effects. While some of these results have been replicated in independent samples, and therefore are likely to represent true associations, negative studies have also been published, albeit, often in smaller and likely underpowered studies. Currently, most promising findings involve associations between DA receptor polymorphisms and response, or HTR2C SNPs and AIWG. There are several possible explanations for these inconsistencies: differences in study design, small effect sizes of most SNPs, small sample sizes, incomplete coverage of most genes, lack of control of environmental and clinical confounders, or varying definition of outcome parameters. Conflicting results may also be due to an insufficient incorporation of gene–gene and gene–environment interactions. Therefore, new approaches, beyond candidate gene studies and GWASs are needed: DNA-sequencing, gene expression studies, novel bioinformatic approaches, animal studies, epigenetic approaches, and large and prospectively assessed samples will illuminate underlying genetic mechanisms.

Although widespread clinical use of pharmacogenetics will likely take several more years of investigation, some findings are starting to be clinically applied. These include the assessment of drug metabolizer status and inclusion of other pharmacodynamics genes with promising findings showing superiority to treatment as usual.176

Those tests are still relatively expensive and need to be improved by adding more gene variants. They also need to be implemented in ways that allow their use in clinical routine (that is, rapid processing and convenient costs), and physicians need to learn how to use their results in their practice. However, the clinical availability of these few tests suggests that pharmacogenetics is becoming a reality in patient care and will help to individualize and substantially improve treatment in the near future.

Acknowledgments

Dr Müller has received a Canadian Institutes for Health Research (CIHR) operating grant; a Canadian Foundation for Innovation (CFI) grant; the National Alliance for Research on Schizophrenia and Depression (commonly referred to as NARSAD) Independent Investigator Award; the Early Researcher Award from the Ontario Ministry of Economic Development and Innovation (MEDI); the CIHR Michael Smith New Investigator Salary Prize for Research in Schizophrenia; and a New Investigator Fellowship form the Ontario Mental Health Foundation (OMHF).

Dr Kennedy has also received a CIHR operating grant; and OMHF, CFI, and MEDI grants.

Dr Brandl and Dr Müller have no conflicts of interest to declare. Dr Kennedy has been a consultant to GlaxoSmithKline, Sanofi-Aventis, and Dinippon-Sumitomo.

Abbreviations

5-HT

5-hydroxytryptamine (serotonin)

AIWG

AP-induced weight gain

AP

antipsychotic

ATP

adenosine triphosphate

bp

base pair

CATIE

Clinical Antipsychotic Trials of Intervention Effectiveness

CIA

clozapine-induced agranulocytosis

COMT

catechol-O-methyltransferase

CYP

cytochrome P450

CYP1A2

CYP, family 1, subfamily A, polypeptide 2

CYP2C19

CYP, family 2, subfamily C, polypeptide 19

CYP2D6

CYP, family 2, subfamily D, polypeptide 6

CYP3A4

CYP, family 3, subfamily A, polypeptide 4

Cys

cysteine

DA

dopamine

del

deletion

DNA

deoxyribonucleic acid

DRB5

MHC, class II, DR beta 5

DRD

DA receptor D

EPS

extra pyramidal symptom

FGA

first-generation AP

GFRA

glial cell derived neurotrophic factor family receptor alpha

Gly

glycine

GNB3

guanine nucleotide binding protein (G protein), beta polypeptide 3

GWAS

genome-wide association study

HLA

human leukocyte antigen

HTR2A

5-HT receptor 2A, G protein-coupled

HTR2C

5-HT receptor 2C, G protein-coupled

IM

intermediate metabolizer

ins

insertion

MC4R

melanocortin 4 receptor

MDR1

multi-drug resistance gene 1

Met

methionine

MHC

major histocompatibility complex

P-gp

permeability glycoprotein

PM

poor metabolizer

rs

reference SNP

SLC

solute carrier

Ser

serine

SGA

second-generation AP

SNP

single-nucleotide polymorphism

TD

tardive dyskinesia

TNF

tumour necrosis factor

Tyr

tyrosine

UM

ultra-rapid metabolizer

Val

valine

References

  • 1.Gardiner SJ, Begg EJ. Pharmacogenetics, drug-metabolizing enzymes, and clinical practice. Pharmacol Rev. 2006;58(3):521–590. doi: 10.1124/pr.58.3.6. [DOI] [PubMed] [Google Scholar]
  • 2.Kato M, Serretti A. Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol Psychiatry. 2010;15(5):473–500. doi: 10.1038/mp.2008.116. [DOI] [PubMed] [Google Scholar]
  • 3.Fabbri C, Porcelli S, Serretti A. From pharmacogenetics to pharmacogenomics: the way toward the personalization of antidepressant treatment. Can J Psychiatry. 2014;59(2):62–75. doi: 10.1177/070674371405900202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tandon R, Belmaker RH, Gattaz WF, et al. World Psychiatric Association Pharmacopsychiatry Section statement on comparative effectiveness of antipsychotics in the treatment of schizophrenia. Schizophr Res. 2008;100(1–3):20–38. doi: 10.1016/j.schres.2007.11.033. [DOI] [PubMed] [Google Scholar]
  • 5.Lieberman JA, Stroup TS, McEvoy JP, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353(12):1209–1223. doi: 10.1056/NEJMoa051688. [DOI] [PubMed] [Google Scholar]
  • 6.Weiss EL, Longhurst JG, Bowers MB, Jr, et al. Olanzapine for treatment-refractory psychosis in patients responsive to, but intolerant of, clozapine. J Clin Psychopharmacol. 1999;19(4):378–380. doi: 10.1097/00004714-199908000-00017. [DOI] [PubMed] [Google Scholar]
  • 7.Arranz MJ, Kapur S. Pharmacogenetics in psychiatry: are we ready for widespread clinical use? Schizophr Bull. 2008;34(6):1130–1144. doi: 10.1093/schbul/sbn114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Vojvoda D, Grimmell K, Sernyak M, et al. Monozygotic twins concordant for response to clozapine. Lancet. 1996;347(8993):61. doi: 10.1016/s0140-6736(96)91594-9. [DOI] [PubMed] [Google Scholar]
  • 9.Mata I, Madoz V, Arranz MJ, et al. Olanzapine: concordant response in monozygotic twins with schizophrenia. Br J Psychiatry. 2001;178(1):86. doi: 10.1192/bjp.178.1.86. [DOI] [PubMed] [Google Scholar]
  • 10.Müller DJ, Schulze TG, Knapp M, et al. Familial occurrence of tardive dyskinesia. Acta Psychiatr Scand. 2001;104(5):375–379. doi: 10.1034/j.1600-0447.2001.00401.x. [DOI] [PubMed] [Google Scholar]
  • 11.Theisen FM, Cichon S, Linden A, et al. Clozapine and weight gain. Am J Psychiatry. 2001;158(5):816. doi: 10.1176/appi.ajp.158.5.816. [DOI] [PubMed] [Google Scholar]
  • 12.Wehmeier PM, Gebhardt S, Schmidtke J, et al. Clozapine: weight gain in a pair of monozygotic twins concordant for schizophrenia and mild mental retardation. Psychiatry Res. 2005;133(2–3):273–276. doi: 10.1016/j.psychres.2004.02.018. [DOI] [PubMed] [Google Scholar]
  • 13.Zhang JP, Malhotra AK. Pharmacogenetics and antipsychotics: therapeutic efficacy and side effects prediction. Expert Opin Drug Metab Toxicol. 2011;7(1):9–37. doi: 10.1517/17425255.2011.532787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lett TA, Wallace TJ, Chowdhury NI, et al. Pharmacogenetics of antipsychotic-induced weight gain: review and clinical implications. Mol Psychiatry. 2012;17(3):242–266. doi: 10.1038/mp.2011.109. [DOI] [PubMed] [Google Scholar]
  • 15.Chowdhury NI, Remington G, Kennedy JL. Genetics of antipsychotic-induced side effects and agranulocytosis. Curr Psychiatry Rep. 2011;13(2):156–165. doi: 10.1007/s11920-011-0185-3. [DOI] [PubMed] [Google Scholar]
  • 16.Bertilsson L, Dahl ML, Dalen P, et al. Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs. Br J Clin Pharmacol. 2002;53(2):111–122. doi: 10.1046/j.0306-5251.2001.01548.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gaedigk A, Simon SD, Pearce RE, et al. The CYP2D6 activity score: translating genotype information into a qualitative measure of phenotype. Clin Pharmacol Ther. 2008;83(2):234–242. doi: 10.1038/sj.clpt.6100406. [DOI] [PubMed] [Google Scholar]
  • 18.Arranz MJ, de Leon J. Pharmacogenetics and pharmacogenomics of schizophrenia: a review of last decade of research. Mol Psychiatry. 2007;12(8):707–747. doi: 10.1038/sj.mp.4002009. [DOI] [PubMed] [Google Scholar]
  • 19.Vetti HH, Molven A, Eliassen AK, et al. Is pharmacogenetic CYP2D6 testing useful? Tidsskr Nor Laegeforen. 2010;130(22):2224–2228. doi: 10.4045/tidsskr.09.1445. English, Norwegian. [DOI] [PubMed] [Google Scholar]
  • 20.Hendset M, Molden E, Refsum H, et al. 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]
  • 21.van der Weide J, Steijns LS, van Weelden MJ. The effect of smoking and cytochrome P450 CYP1A2 genetic polymorphism on clozapine clearance and dose requirement. Pharmacogenetics. 2003;13(3):169–172. doi: 10.1097/00008571-200303000-00006. [DOI] [PubMed] [Google Scholar]
  • 22.García-Martín E, Martínez C, Pizarro RM, et al. CYP3A4 variant alleles in white individuals with low CYP3A4 enzyme activity. Clin Pharmacol Ther. 2002;71(3):196–204. doi: 10.1067/mcp.2002.121371. [DOI] [PubMed] [Google Scholar]
  • 23.Olesen OV, Linnet K. Contributions of five human cytochrome P450 isoforms to the N-demethylation of clozapine in vitro at low and high concentrations. J Clin Pharmacol. 2001;41(8):823–832. doi: 10.1177/00912700122010717. [DOI] [PubMed] [Google Scholar]
  • 24.Laika B, Leucht S, Heres S, et al. Pharmacogenetics and olanzapine treatment: CYP1A2*1F and serotonergic polymorphisms influence therapeutic outcome. Pharmacogenomics J. 2010;10(1):20–29. doi: 10.1038/tpj.2009.32. [DOI] [PubMed] [Google Scholar]
  • 25.Ozdemir V, Kalow W, Okey AB, et al. Treatment-resistance to clozapine in association with ultrarapid CYP1A2 activity and the C—>A polymorphism in intron 1 of the CYP1A2 gene: effect of grapefruit juice and low-dose fluvoxamine. J Clin Psychopharmacol. 2001;21(6):603–607. doi: 10.1097/00004714-200112000-00011. [DOI] [PubMed] [Google Scholar]
  • 26.Eap CB, Bender S, Jaquenoud Sirot E, et al. Nonresponse to clozapine and ultrarapid CYP1A2 activity: clinical data and analysis of CYP1A2 gene. J Clin Psychopharmacol. 2004;24(2):214–219. doi: 10.1097/01.jcp.0000116646.91923.2f. [DOI] [PubMed] [Google Scholar]
  • 27.Du J, Zhang A, Wang L, et al. Relationship between response to risperidone, plasma concentrations of risperidone and CYP3A4 polymorphisms in schizophrenia patients. J Psychopharmacol. 2010;24(7):1115–1120. doi: 10.1177/0269881109104932. [DOI] [PubMed] [Google Scholar]
  • 28.Bigos KL, Bies RR, Pollock BG, et al. Genetic variation in CYP3A43 explains racial difference in olanzapine clearance. Mol Psychiatry. 2011;16(6):620–625. doi: 10.1038/mp.2011.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Arranz MJ, Dawson E, Shaikh S, et al. Cytochrome P4502D6 genotype does not determine response to clozapine. Br J Clin Pharmacol. 1995;39(4):417–420. doi: 10.1111/j.1365-2125.1995.tb04471.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jovanović N, Božina N, Lovrić M, et al. The role of CYP2D6 and ABCB1 pharmacogenetics in drug-naive 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]
  • 31.Kakihara S, Yoshimura R, Shinkai K, et al. Prediction of response to risperidone treatment with respect to plasma concencentrations of risperidone, catecholamine metabolites, and polymorphism of cytochrome P450 2D6. Int Clin Psychopharmacol. 2005;20(2):71–78. doi: 10.1097/00004850-200503000-00002. [DOI] [PubMed] [Google Scholar]
  • 32.Riedel M, Schwarz MJ, Strassnig M, et al. Risperidone plasma levels, clinical response and side-effects. Eur Arch Psychiatry Clin Neurosci. 2005;255(4):261–268. doi: 10.1007/s00406-004-0556-4. [DOI] [PubMed] [Google Scholar]
  • 33.Ellingrod VL, Miller D, Schultz SK, et al. CYP2D6 polymorphisms and atypical antipsychotic weight gain. Psychiatr Genet. 2002;12(1):55–58. doi: 10.1097/00041444-200203000-00008. [DOI] [PubMed] [Google Scholar]
  • 34.Lane HY, Liu YC, Huang CL, et al. Risperidone-related weight gain: genetic and nongenetic predictors. J Clin Psychopharmacol. 2006;26(2):128–134. doi: 10.1097/01.jcp.0000203196.65710.2b. [DOI] [PubMed] [Google Scholar]
  • 35.Lohmann PL, Bagli M, Krauss H, et al. CYP2D6 polymorphism and tardive dyskinesia in schizophrenic patients. Pharmacopsychiatry. 2003;36(2):73–78. doi: 10.1055/s-2003-39048. [DOI] [PubMed] [Google Scholar]
  • 36.Plesnicar BK, Zalar B, Breskvar K, et al. The influence of the CYP2D6 polymorphism on psychopathological and extrapyramidal symptoms in the patients on long-term antipsychotic treatment. J Psychopharmacol. 2006;20(6):829–833. doi: 10.1177/0269881106062894. [DOI] [PubMed] [Google Scholar]
  • 37.Tiwari AK, Deshpande SN, Rao AR, et al. Genetic susceptibility to tardive dyskinesia in chronic schizophrenia subjects: III. Lack of association of CYP3A4 and CYP2D6 gene polymorphisms. Schizophr Res. 2005;75(1):21–26. doi: 10.1016/j.schres.2004.12.011. [DOI] [PubMed] [Google Scholar]
  • 38.Tsai HT, Caroff SN, Miller DD, et al. A candidate gene study of tardive dyskinesia in the CATIE schizophrenia trial. Am J Med Genet B Neuropsychiatr Genet. 2010;153B(1):336–340. doi: 10.1002/ajmg.b.30981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Crescenti A, Mas S, Gassó P, et al. Cyp2d6*3, *4, *5 and *6 polymorphisms and antipsychotic-induced extrapyramidal side-effects in patients receiving antipsychotic therapy. Clin Exp Pharmacol Physiol. 2008;35(7):807–811. doi: 10.1111/j.1440-1681.2008.04918.x. [DOI] [PubMed] [Google Scholar]
  • 40.Laika B, Leucht S, Heres S, et al. Intermediate metabolizer: increased side effects in psychoactive drug therapy. The key to cost-effectiveness of pretreatment CYP2D6 screening? Pharmacogenomics J. 2009;9(6):395–403. doi: 10.1038/tpj.2009.23. [DOI] [PubMed] [Google Scholar]
  • 41.Kobylecki CJ, Jakobsen KD, Hansen T, et al. CYP2D6 genotype predicts antipsychotic side effects in schizophrenia inpatients: a retrospective matched case–control study. Neuropsychobiology. 2009;59(4):222–226. doi: 10.1159/000223734. [DOI] [PubMed] [Google Scholar]
  • 42.Fu Y, Fan CH, Deng HH, et al. Association of CYP2D6 and CYP1A2 gene polymorphism with tardive dyskinesia in Chinese schizophrenic patients. Acta Pharmacol Sin. 2006;27(3):328–332. doi: 10.1111/j.1745-7254.2006.00279.x. [DOI] [PubMed] [Google Scholar]
  • 43.Patsopoulos NA, Ntzani EE, Zintzaras E, et al. CYP2D6 polymorphisms and the risk of tardive dyskinesia in schizophrenia: a meta-analysis. Pharmacogenet Genomics. 2005;15(3):151–158. doi: 10.1097/01213011-200503000-00003. [DOI] [PubMed] [Google Scholar]
  • 44.Müller DJ, Shinkai T, De Luca V, et al. Clinical implications of pharmacogenomics for tardive dyskinesia. Pharmacogenomics J. 2004;4(2):77–87. doi: 10.1038/sj.tpj.6500233. [DOI] [PubMed] [Google Scholar]
  • 45.Fleeman N, McLeod C, Bagust A, et al. The clinical effectiveness and cost-effectiveness of testing for cytochrome P450 polymorphisms in patients with schizophrenia treated with antipsychotics: a systematic review and economic evaluation. Health Technol Assess. 2010;14(3):1–157. iii. doi: 10.3310/hta14030. [DOI] [PubMed] [Google Scholar]
  • 46.Basile VS, Ozdemir V, Masellis M, et al. A functional polymorphism of the cytochrome P450 1A2 (CYP1A2) gene: association with tardive dyskinesia in schizophrenia. Mol Psychiatry. 2000;5(4):410–417. doi: 10.1038/sj.mp.4000736. [DOI] [PubMed] [Google Scholar]
  • 47.Tiwari AK, Deshpande SN, Rao AR, et al. Genetic susceptibility to tardive dyskinesia in chronic schizophrenia subjects: I. Association of CYP1A2 gene polymorphism. Pharmacogenomics J. 2005;5(1):60–69. doi: 10.1038/sj.tpj.6500282. [DOI] [PubMed] [Google Scholar]
  • 48.Bakker PR, van Harten PN, van Os J. Antipsychotic-induced tardive dyskinesia and polymorphic variations in COMT, DRD2, CYP1A2 and MnSOD genes: a meta-analysis of pharmacogenetic interactions. Mol Psychiatry. 2008;13(5):544–556. doi: 10.1038/sj.mp.4002142. [DOI] [PubMed] [Google Scholar]
  • 49.Rodriguez-Antona C, Gurwitz D, de Leon J, et al. CYP2D6 genotyping for psychiatric patients treated with risperidone: considerations for cost-effectiveness studies. Pharmacogenomics. 2009;10(4):685–699. doi: 10.2217/pgs.09.15. [DOI] [PubMed] [Google Scholar]
  • 50.de Leon J, Susce MT, Murray-Carmichael E. The AmpliChip CYP450 genotyping test: integrating a new clinical tool. Mol Diagn Ther. 2006;10(3):135–151. doi: 10.1007/BF03256453. [DOI] [PubMed] [Google Scholar]
  • 51.Pharmacogenetics Research Clinic, Centre for Addiction and Mental Health (CAMH) Pharmacogenetics [Internet] Toronto (ON): CAMH; 2013. [date cited unknown]. Available from: http://www.pharmacogenetics.ca. [Google Scholar]
  • 52.Müller DJ, Kekin I, Kao AC, et al. Towards the implementation of CYP2D6 and CYP2C19 genotypes in clinical practice: update and report from a pharmacogenetic service clinic. Int Rev Psychiatry. 2013;25(5):554–571. doi: 10.3109/09540261.2013.838944. [DOI] [PubMed] [Google Scholar]
  • 53.Nnadi CU, Malhotra AK. Individualizing antipsychotic drug therapy in schizophrenia: the promise of pharmacogenetics. Curr Psychiatry Rep. 2007;9(4):313–318. doi: 10.1007/s11920-007-0038-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Takao T, Tachikawa H, Kawanishi Y, et al. Association of treatment-resistant schizophrenia with the G2677A/T and C3435T polymorphisms in the ATP-binding cassette subfamily B member 1 gene. Psychiatr Genet. 2006;16(2):47–48. doi: 10.1097/01.ypg.0000194441.04684.db. [DOI] [PubMed] [Google Scholar]
  • 55.Kastelic M, Koprivsek J, Plesnicar BK, et al. MDR1 gene polymorphisms and response to acute risperidone treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(2):387–392. doi: 10.1016/j.pnpbp.2010.01.005. [DOI] [PubMed] [Google Scholar]
  • 56.Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42(2):89–97. doi: 10.1016/j.jpsychires.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 57.Nikisch G, Baumann P, Oneda B, et al. Cytochrome P450 and ABCB1 genetics: association with quetiapine and norquetiapine plasma and cerebrospinal fluid concentrations and with clinical response in patients suffering from schizophrenia. A pilot study. J Psychopharmacol. 2011;25(7):896–907. doi: 10.1177/0269881110389208. [DOI] [PubMed] [Google Scholar]
  • 58.Yasui-Furukori N, Saito M, Nakagami T, et al. Association between multidrug resistance 1 (MDR1) gene polymorphisms and therapeutic response to bromperidol in schizophrenic patients: a preliminary study. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(2):286–291. doi: 10.1016/j.pnpbp.2005.06.019. [DOI] [PubMed] [Google Scholar]
  • 59.Xing Q, Gao R, Li H, et al. Polymorphisms of the ABCB1 gene are associated with the therapeutic response to risperidone in Chinese schizophrenia patients. Pharmacogenomics. 2006;7(7):987–993. doi: 10.2217/14622416.7.7.987. [DOI] [PubMed] [Google Scholar]
  • 60.de Leon J, Susce MT, Pan RM, et al. Polymorphic variations in GSTM1, GSTT1, PgP, CYP2D6, CYP3A5, and dopamine D2 and D3 receptors and their association with tardive dyskinesia in severe mental illness. J Clin Psychopharmacol. 2005;25(5):448–456. doi: 10.1097/01.jcp.0000177546.34799.af. [DOI] [PubMed] [Google Scholar]
  • 61.De Luca V, Souza RP, Viggiano E, et al. MDR1 gene in tardive dyskinesia scale scores: comparison of strategies for quantitative trait haplotype analysis. Schizophr Res. 2009;110(1–3):200–201. doi: 10.1016/j.schres.2009.01.021. [DOI] [PubMed] [Google Scholar]
  • 62.Kuzman MR, Medved V, Bozina N, et al. The influence of 5-HT(2C) and MDR1 genetic polymorphisms on antipsychotic-induced weight gain in female schizophrenic patients. Psychiatry Res. 2008;160(3):308–315. doi: 10.1016/j.psychres.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 63.Kuzman MR, Medved V, Bozina N, et al. Association study of MDR1 and 5-HT2C genetic polymorphisms and antipsychotic-induced metabolic disturbances in female patients with schizophrenia. Pharmacogenomics J. 2011;11(1):35–44. doi: 10.1038/tpj.2010.7. [DOI] [PubMed] [Google Scholar]
  • 64.Seeman P, Lee T. Antipsychotic drugs: direct correlation between clinical potency and presynaptic action on dopamine neurons. Science. 1975;188(4194):1217–1219. doi: 10.1126/science.1145194. [DOI] [PubMed] [Google Scholar]
  • 65.Kapur S, Mamo D. Half a century of antipsychotics and still a central role for dopamine D2 receptors. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(7):1081–1090. doi: 10.1016/j.pnpbp.2003.09.004. [DOI] [PubMed] [Google Scholar]
  • 66.Lencz T, Robinson DG, Xu K, et al. DRD2 promoter region variation as a predictor of sustained response to antipsychotic medication in first-episode schizophrenia patients. Am J Psychiatry. 2006;163(3):529–531. doi: 10.1176/appi.ajp.163.3.529. [DOI] [PubMed] [Google Scholar]
  • 67.Wu S, Xing Q, Gao R, et al. Response to chlorpromazine treatment may be associated with polymorphisms of the DRD2 gene in Chinese schizophrenic patients. Neurosci Lett. 2005;376(1):1–4. doi: 10.1016/j.neulet.2004.11.014. [DOI] [PubMed] [Google Scholar]
  • 68.Zhang JP, Lencz T, Malhotra AK. D2 receptor genetic variation and clinical response to antipsychotic drug treatment: a meta-analysis. Am J Psychiatry. 2010;167(7):763–772. doi: 10.1176/appi.ajp.2009.09040598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Xing Q, Qian X, Li H, et al. The relationship between the therapeutic response to risperidone and the dopamine D2 receptor polymorphism in Chinese schizophrenia patients. Int J Neuropsychopharmacol. 2007;10(5):631–637. doi: 10.1017/S146114570600719X. [DOI] [PubMed] [Google Scholar]
  • 70.Suzuki A, Mihara K, Kondo T, et al. The relationship between dopamine D2 receptor polymorphism at the Taq1 A locus and therapeutic response to nemonapride, a selective dopamine antagonist, in schizophrenic patients. Pharmacogenetics. 2000;10(4):335–341. doi: 10.1097/00008571-200006000-00007. [DOI] [PubMed] [Google Scholar]
  • 71.Schafer M, Rujescu D, Giegling I, et al. Association of short-term response to haloperidol treatment with a polymorphism in the dopamine D(2) receptor gene. Am J Psychiatry. 2001;158(5):802–804. doi: 10.1176/appi.ajp.158.5.802. [DOI] [PubMed] [Google Scholar]
  • 72.Yamanouchi Y, Iwata N, Suzuki T, et al. Effect of DRD2, 5-HT2A, and COMT genes on antipsychotic response to risperidone. Pharmacogenomics J. 2003;3(6):356–361. doi: 10.1038/sj.tpj.6500211. [DOI] [PubMed] [Google Scholar]
  • 73.Dahmen N, Müller MJ, Germeyer S, et al. Genetic polymorphisms of the dopamine D2 and D3 receptor and neuroleptic drug effects in schizophrenic patients. Schizophr Res. 2001;49(1–2):223–225. doi: 10.1016/s0920-9964(99)00147-4. [DOI] [PubMed] [Google Scholar]
  • 74.Hwang R, Shinkai T, De Luca V, et al. Association study of 12 polymorphisms spanning the dopamine D(2) receptor gene and clozapine treatment response in two treatment refractory/intolerant populations. Psychopharmacology (Berl) 2005;181(1):179–187. doi: 10.1007/s00213-005-2223-5. [DOI] [PubMed] [Google Scholar]
  • 75.Lane HY, Lee CC, Chang YC, et al. Effects of dopamine D2 receptor Ser311Cys polymorphism and clinical factors on risperidone efficacy for positive and negative symptoms and social function. Int J Neuropsychopharmacol. 2004;7(4):461–470. doi: 10.1017/S1461145704004389. [DOI] [PubMed] [Google Scholar]
  • 76.Hwang R, Zai C, Tiwari A, et al. Effect of dopamine D3 receptor gene polymorphisms and clozapine treatment response: exploratory analysis of nine polymorphisms and meta-analysis of the Ser9Gly variant. Pharmacogenomics J. 2010;10(3):200–218. doi: 10.1038/tpj.2009.65. [DOI] [PubMed] [Google Scholar]
  • 77.Hwu HG, Hong CJ, Lee YL, et al. Dopamine D4 receptor gene polymorphisms and neuroleptic response in schizophrenia. Biol Psychiatry. 1998;44(6):483–487. doi: 10.1016/s0006-3223(98)00134-6. [DOI] [PubMed] [Google Scholar]
  • 78.Zhao AL, Zhao JP, Zhang YH, et al. Dopamine D4 receptor gene exon III polymorphism and interindividual variation in response to clozapine. Int J Neurosci. 2005;115(11):1539–1547. doi: 10.1080/00207450590957863. [DOI] [PubMed] [Google Scholar]
  • 79.Rietschel M, Naber D, Oberlander H, et al. Efficacy and side-effects of clozapine: testing for association with allelic variation in the dopamine D4 receptor gene. Neuropsychopharmacology. 1996;15(5):491–496. doi: 10.1016/S0893-133X(96)00090-5. [DOI] [PubMed] [Google Scholar]
  • 80.Ikeda M, Yamanouchi Y, Kinoshita Y, et al. Variants of dopamine and serotonin candidate genes as predictors of response to risperidone treatment in first-episode schizophrenia. Pharmacogenomics. 2008;9(10):1437–1443. doi: 10.2217/14622416.9.10.1437. [DOI] [PubMed] [Google Scholar]
  • 81.Thomas P, Srivastava V, Singh A, et al. Correlates of response to olanzapine in a North Indian Schizophrenia sample. Psychiatry Res. 2008;161(3):275–283. doi: 10.1016/j.psychres.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 82.Spurlock G, Heils A, Holmans P, et al. A family based association study of T102C polymorphism in 5HT2A and schizophrenia plus identification of new polymorphisms in the promoter. Mol Psychiatry. 1998;3(1):42–49. doi: 10.1038/sj.mp.4000342. [DOI] [PubMed] [Google Scholar]
  • 83.Parsons MJ, D’Souza UM, Arranz MJ, et al. The −1438A/G polymorphism in the 5-hydroxytryptamine type 2A receptor gene affects promoter activity. Biol Psychiatry. 2004;56(6):406–410. doi: 10.1016/j.biopsych.2004.06.020. [DOI] [PubMed] [Google Scholar]
  • 84.Arranz MJ, Munro J, Owen MJ, et al. Evidence for association between polymorphisms in the promoter and coding regions of the 5-HT2A receptor gene and response to clozapine. Mol Psychiatry. 1998;3(1):61–66. doi: 10.1038/sj.mp.4000348. [DOI] [PubMed] [Google Scholar]
  • 85.Ellingrod VL, Lund BC, Miller D, et al. 5-HT2A receptor promoter polymorphism, −1438G/A and negative symptom response to olanzapine in schizophrenia. Psychopharmacol Bull. 2003;37(2):109–112. [PubMed] [Google Scholar]
  • 86.Benmessaoud D, Hamdani N, Boni C, et al. Excess of transmission of the G allele of the −1438A/G polymorphism of the 5-HT2A receptor gene in patients with schizophrenia responsive to antipsychotics. BMC Psychiatry. 2008;8:40. doi: 10.1186/1471-244X-8-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Anttila S, Kampman O, Illi A, et al. Association between 5-HT2A, TPH1 and GNB3 genotypes and response to typical neuroleptics: a serotonergic approach. BMC Psychiatry. 2007;7:22. doi: 10.1186/1471-244X-7-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Arranz MJ, Munro J, Sham P, et al. Meta-analysis of studies on genetic variation in 5-HT2A receptors and clozapine response. Schizophr Res. 1998;32(2):93–99. doi: 10.1016/s0920-9964(98)00032-2. [DOI] [PubMed] [Google Scholar]
  • 89.Kim B, Choi EY, Kim CY, et al. Could HTR2A T102C and DRD3 Ser9Gly predict clinical improvement in patients with acutely exacerbated schizophrenia? Results from treatment responses to risperidone in a naturalistic setting. Hum Psychopharmacol. 2008;23(1):61–67. doi: 10.1002/hup.897. [DOI] [PubMed] [Google Scholar]
  • 90.Ellingrod VL, Perry PJ, Lund BC, et al. 5HT2A and 5HT2C receptor polymorphisms and predicting clinical response to olanzapine in schizophrenia. J Clin Psychopharmacol. 2002;22(6):622–624. doi: 10.1097/00004714-200212000-00014. [DOI] [PubMed] [Google Scholar]
  • 91.Buckland PR, Hoogendoorn B, Guy CA, et al. Low gene expression conferred by association of an allele of the 5-HT2C receptor gene with antipsychotic-induced weight gain. Am J Psychiatry. 2005;162(3):613–615. doi: 10.1176/appi.ajp.162.3.613. [DOI] [PubMed] [Google Scholar]
  • 92.Reynolds GP, Yao Z, Zhang X, et al. Pharmacogenetics of treatment in first-episode schizophrenia: D3 and 5-HT2C receptor polymorphisms separately associate with positive and negative symptom response. Eur Neuropsychopharmacol. 2005;15(2):143–151. doi: 10.1016/j.euroneuro.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 93.Sodhi MS, Arranz MJ, Curtis D, et al. Association between clozapine response and allelic variation in the 5-HT2C receptor gene. Neuroreport. 1995;7(1):169–172. [PubMed] [Google Scholar]
  • 94.Rietschel M, Naber D, Fimmers R, et al. Efficacy and side-effects of clozapine not associated with variation in the 5-HT2C receptor. Neuroreport. 1997;8(8):1999–2003. doi: 10.1097/00001756-199705260-00040. [DOI] [PubMed] [Google Scholar]
  • 95.Dolzan V, Serretti A, Mandelli L, et al. Acute antipyschotic efficacy and side effects in schizophrenia: association with serotonin transporter promoter genotypes. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(6):1562–1566. doi: 10.1016/j.pnpbp.2008.05.022. [DOI] [PubMed] [Google Scholar]
  • 96.Arranz MJ, Munro J, Birkett J, et al. Pharmacogenetic prediction of clozapine response. Lancet. 2000;355(9215):1615–1616. doi: 10.1016/s0140-6736(00)02221-2. [DOI] [PubMed] [Google Scholar]
  • 97.Vazquez-Bourgon J, Arranz MJ, Mata I, et al. Serotonin transporter polymorphisms and early response to antipsychotic treatment in first episode of psychosis. Psychiatry Res. 2010;175(3):189–194. doi: 10.1016/j.psychres.2008.12.011. [DOI] [PubMed] [Google Scholar]
  • 98.Reynolds GP, Arranz B, Templeman LA, et al. Effect of 5-HT1A receptor gene polymorphism on negative and depressive symptom response to antipsychotic treatment of drug-naive psychotic patients. Am J Psychiatry. 2006;163(10):1826–1829. doi: 10.1176/ajp.2006.163.10.1826. [DOI] [PubMed] [Google Scholar]
  • 99.Mössner R, Schuhmacher A, Kühn KU, et al. Functional serotonin 1A receptor variant influences treatment response to atypical antipsychotics in schizophrenia. Pharmacogenet Genomics. 2009;19(1):91–94. doi: 10.1097/FPC.0b013e328311a917. [DOI] [PubMed] [Google Scholar]
  • 100.Schuhmacher A, Mössner R, Quednow BB, et al. Influence of 5-HT3 receptor subunit genes HTR3A, HTR3B, HTR3C, HTR3D and HTR3E on treatment response to antipsychotics in schizophrenia. Pharmacogenet Genomics. 2009;19(11):843–851. doi: 10.1097/FPC.0b013e3283313296. [DOI] [PubMed] [Google Scholar]
  • 101.Souza RP, de Luca V, Meltzer HY, et al. Influence of serotonin 3A and 3B receptor genes on clozapine treatment response in schizophrenia. Pharmacogenet Genomics. 2010;20(4):274–276. doi: 10.1097/FPC.0b013e328337ce3e. [DOI] [PubMed] [Google Scholar]
  • 102.Yu YW, Tsai SJ, Lin CH, et al. Serotonin-6 receptor variant (C267T) and clinical response to clozapine. Neuroreport. 1999;10(6):1231–1233. doi: 10.1097/00001756-199904260-00014. [DOI] [PubMed] [Google Scholar]
  • 103.Lane HY, Lin CC, Huang CH, et al. Risperidone response and 5-HT6 receptor gene variance: genetic association analysis with adjustment for nongenetic confounders. Schizophr Res. 2004;67(1):63–70. doi: 10.1016/j.schres.2003.08.006. [DOI] [PubMed] [Google Scholar]
  • 104.Bertolino A, Caforio G, Blasi G, et al. Interaction of COMT (Val(108/158)Met) genotype and olanzapine treatment on prefrontal cortical function in patients with schizophrenia. Am J Psychiatry. 2004;161(10):1798–1805. doi: 10.1176/ajp.161.10.1798. [DOI] [PubMed] [Google Scholar]
  • 105.Woodward ND, Jayathilake K, Meltzer HY. COMT val108/158met genotype, cognitive function, and cognitive improvement with clozapine in schizophrenia. Schizophr Res. 2007;90(1–3):86–96. doi: 10.1016/j.schres.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 106.Pelayo-Teran JM, Perez-Iglesias R, Vazquez-Bourgon J, et al. Catechol-O-methyltransferase Val158Met polymorphism and negative symptoms after acute antipsychotic treatment in first-episode non-affective psychosis. Psychiatry Res. 2011;185(1–2):286–289. doi: 10.1016/j.psychres.2010.06.006. [DOI] [PubMed] [Google Scholar]
  • 107.Illi A, Kampman O, Anttila S, et al. Interaction between angiotensin-converting enzyme and catechol-O-methyltransferase genotypes in schizophrenics with poor response to conventional neuroleptics. Eur Neuropsychopharmacol. 2003;13(3):147–151. doi: 10.1016/s0924-977x(02)00176-1. [DOI] [PubMed] [Google Scholar]
  • 108.Müller DJ, De Luca V, Sicard T, et al. Suggestive association between the C825T polymorphism of the G-protein beta3 subunit gene (GNB3) and clinical improvement with antipsychotics in schizophrenia. Eur Neuropsychopharmacol. 2005;15(5):525–531. doi: 10.1016/j.euroneuro.2005.02.001. [DOI] [PubMed] [Google Scholar]
  • 109.Kohlrausch FB, Salatino-Oliveira A, Gama CS, et al. G-protein gene 825C>T polymorphism is associated with response to clozapine in Brazilian schizophrenics. Pharmacogenomics. 2008;9(10):1429–1436. doi: 10.2217/14622416.9.10.1429. [DOI] [PubMed] [Google Scholar]
  • 110.Hong CJ, Liu HC, Liu TY, et al. Brain-derived neurotrophic factor (BDNF) Val66Met polymorphisms in Parkinson’s disease and age of onset. Neurosci Lett. 2003;353(1):75–77. doi: 10.1016/j.neulet.2003.09.009. [DOI] [PubMed] [Google Scholar]
  • 111.Xu M, Li S, Xing Q, et al. Genetic variants in the BDNF gene and therapeutic response to risperidone in schizophrenia patients: a pharmacogenetic study. Eur J Hum Genet. 2010;18(6):707–712. doi: 10.1038/ejhg.2009.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Souza RP, Romano-Silva MA, Lieberman JA, et al. Genetic association of the GDNF alpha-receptor genes with schizophrenia and clozapine response. J Psychiatr Res. 2010;44(11):700–706. doi: 10.1016/j.jpsychires.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 113.Souza RP, Ismail P, Meltzer HY, et al. Variants in the oxytocin gene and risk for schizophrenia. Schizophr Res. 2010;121(1–3):279–280. doi: 10.1016/j.schres.2010.04.019. [DOI] [PubMed] [Google Scholar]
  • 114.Zai G, Müller DJ, Volavka J, et al. Family and case–control association study of the tumor necrosis factor-alpha (TNF-alpha) gene with schizophrenia and response to antipsychotic medication. Psychopharmacology (Berl) 2006;188(2):171–182. doi: 10.1007/s00213-006-0482-4. [DOI] [PubMed] [Google Scholar]
  • 115.McClay JL, Adkins DE, Aberg K, et al. Genome-wide pharmacogenomic analysis of response to treatment with antipsychotics. Mol Psychiatry. 2011;16(1):76–85. doi: 10.1038/mp.2009.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.McClay JL, Adkins DE, Aberg K, et al. Genome-wide pharmacogenomic study of neurocognition as an indicator of antipsychotic treatment response in schizophrenia. Neuropsychopharmacology. 2011;36(3):616–626. doi: 10.1038/npp.2010.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ikeda M, Tomita Y, Mouri A, et al. Identification of novel candidate genes for treatment response to risperidone and susceptibility for schizophrenia: integrated analysis among pharmacogenomics, mouse expression, and genetic case–control association approaches. Biol Psychiatry. 2010;67(3):263–269. doi: 10.1016/j.biopsych.2009.08.030. [DOI] [PubMed] [Google Scholar]
  • 118.Lavedan C, Licamele L, Volpi S, et al. Association of the NPAS3 gene and five other loci with response to the antipsychotic iloperidone identified in a whole genome association study. Mol Psychiatry. 2009;14(8):804–819. doi: 10.1038/mp.2008.56. [DOI] [PubMed] [Google Scholar]
  • 119.Müller DJ, Kennedy JL. Genetics of antipsychotic treatment emergent weight gain in schizophrenia. Pharmacogenomics. 2006;7(6):863–887. doi: 10.2217/14622416.7.6.863. [DOI] [PubMed] [Google Scholar]
  • 120.Allison DB, Mentore JL, Heo M, et al. Antipsychotic-induced weight gain: a comprehensive research synthesis. Am J Psychiatry. 1999;156(11):1686–1696. doi: 10.1176/ajp.156.11.1686. [DOI] [PubMed] [Google Scholar]
  • 121.Gebhardt S, Theisen FM, Haberhausen M, et al. Body weight gain induced by atypical antipsychotics: an extension of the monozygotic twin and sib pair study. J Clin Pharm Ther. 2010;35(2):207–211. doi: 10.1111/j.1365-2710.2009.01084.x. [DOI] [PubMed] [Google Scholar]
  • 122.Hill MJ, Reynolds GP. Functional consequences of two HTR2C polymorphisms associated with antipsychotic-induced weight gain. Pharmacogenomics. 2011;12(5):727–734. doi: 10.2217/pgs.11.16. [DOI] [PubMed] [Google Scholar]
  • 123.Reynolds GP, Zhang ZJ, Zhang XB. Association of antipsychotic drug-induced weight gain with a 5-HT2C receptor gene polymorphism. Lancet. 2002;359(9323):2086–2087. doi: 10.1016/S0140-6736(02)08913-4. [DOI] [PubMed] [Google Scholar]
  • 124.Miller DD, Ellingrod VL, Holman TL, et al. Clozapine-induced weight gain associated with the 5HT2C receptor −759C/T polymorphism. Am J Med Genet B Neuropsychiatr Genet. 2005;133B(1):97–100. doi: 10.1002/ajmg.b.30115. [DOI] [PubMed] [Google Scholar]
  • 125.Opgen-Rhein C, Brandl EJ, Müller DJ, et al. Association of HTR2C, but not LEP or INSIG2, genes with antipsychotic-induced weight gain in a German sample. Pharmacogenomics. 2010;11(6):773–780. doi: 10.2217/pgs.10.50. [DOI] [PubMed] [Google Scholar]
  • 126.De Luca V, Mueller DJ, de Bartolomeis A, et al. Association of the HTR2C gene and antipsychotic induced weight gain: a meta-analysis. Int J Neuropsychopharmacol. 2007;10(5):697–704. doi: 10.1017/S1461145707007547. [DOI] [PubMed] [Google Scholar]
  • 127.Sicard MN, Zai CC, Tiwari AK, et al. Polymorphisms of the HTR2C gene and antipsychotic-induced weight gain: an update and meta-analysis. Pharmacogenomics. 2010;11(11):1561–1571. doi: 10.2217/pgs.10.123. [DOI] [PubMed] [Google Scholar]
  • 128.Tsai SJ, Hong CJ, Yu YW, et al. −759C/T genetic variation of 5HT(2C) receptor and clozapine-induced weight gain. Lancet. 2002;360(9347):1790. doi: 10.1016/S0140-6736(02)11705-3. [DOI] [PubMed] [Google Scholar]
  • 129.Ujike H, Nomura A, Morita Y, et al. Multiple genetic factors in olanzapine-induced weight gain in schizophrenia patients: a cohort study. J Clin Psychiatry. 2008;69(9):1416–1422. doi: 10.4088/jcp.v69n0909. [DOI] [PubMed] [Google Scholar]
  • 130.Basile VS, Masellis M, De Luca V, et al. 759C/T genetic variation of 5HT(2C) receptor and clozapine-induced weight gain. Lancet. 2002;360(9347):1790–1791. doi: 10.1016/s0140-6736(02)11706-5. [DOI] [PubMed] [Google Scholar]
  • 131.Ellingrod VL, Bishop JR, Moline J, et al. Leptin and leptin receptor gene polymorphisms and increases in body mass index (BMI) from olanzapine treatment in persons with schizophrenia. Psychopharmacol Bull. 2007;40(1):57–62. [PubMed] [Google Scholar]
  • 132.Zhang XY, Tan YL, Zhou DF, et al. Association of clozapine-induced weight gain with a polymorphism in the leptin promoter region in patients with chronic schizophrenia in a Chinese population. J Clin Psychopharmacol. 2007;27(3):246–451. doi: 10.1097/jcp.0b013e3180582412. [DOI] [PubMed] [Google Scholar]
  • 133.Lee AK, Bishop JR. Pharmacogenetics of leptin in antipsychotic-associated weight gain and obesity-related complications. Pharmacogenomics. 2011;12(7):999–1016. doi: 10.2217/pgs.11.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Müller DJ, Klempan TA, De Luca V, et al. The SNAP-25 gene may be associated with clinical response and weight gain in antipsychotic treatment of schizophrenia. Neurosci Lett. 2005;379(2):81–89. doi: 10.1016/j.neulet.2004.12.037. [DOI] [PubMed] [Google Scholar]
  • 135.Musil R, Spellmann I, Riedel M, et al. SNAP-25 gene polymorphisms and weight gain in schizophrenic patients. J Psychiatr Res. 2008;42(12):963–970. doi: 10.1016/j.jpsychires.2007.11.003. [DOI] [PubMed] [Google Scholar]
  • 136.Park YM, Chung YC, Lee SH, et al. Weight gain associated with the alpha2a-adrenergic receptor −1,291 C/G polymorphism and olanzapine treatment. Am J Med Genet B Neuropsychiatr Genet. 2006;141B(4):394–397. doi: 10.1002/ajmg.b.30311. [DOI] [PubMed] [Google Scholar]
  • 137.Wang YC, Bai YM, Chen JY, et al. Polymorphism of the adrenergic receptor alpha 2a −1291C>G genetic variation and clozapine-induced weight gain. J Neural Transm. 2005;112(11):1463–1468. doi: 10.1007/s00702-005-0291-7. [DOI] [PubMed] [Google Scholar]
  • 138.Wang YC, Bai YM, Chen JY, et al. C825T polymorphism in the human G protein beta3 subunit gene is associated with long-term clozapine treatment-induced body weight change in the Chinese population. Pharmacogenet Genomics. 2005;15(10):743–748. doi: 10.1097/01.fpc.0000175600.26893.fa. [DOI] [PubMed] [Google Scholar]
  • 139.Le Hellard S, Theisen FM, Haberhausen M, et al. Association between the insulin-induced gene 2 (INSIG2) and weight gain in a German sample of antipsychotic-treated schizophrenic patients: perturbation of SREBP-controlled lipogenesis in drug-related metabolic adverse effects? Mol Psychiatry. 2009;14(3):308–317. doi: 10.1038/sj.mp.4002133. [DOI] [PubMed] [Google Scholar]
  • 140.Tiwari AK, Zai CC, Meltzer HY, et al. Association study of polymorphisms in insulin induced gene 2 (INSIG2) with antipsychotic-induced weight gain in European and African-American schizophrenia patients. Hum Psychopharmacol. 2010;25(3):253–259. doi: 10.1002/hup.1111. [DOI] [PubMed] [Google Scholar]
  • 141.Hong CJ, Liou YJ, Bai YM, et al. Dopamine receptor D2 gene is associated with weight gain in schizophrenic patients under long-term atypical antipsychotic treatment. Pharmacogenet Genomics. 2010;20(6):359–366. doi: 10.1097/FPC.0b013e3283397d06. [DOI] [PubMed] [Google Scholar]
  • 142.Müller DJ, Zai CC, Sicard M, et al. Systematic analysis of dopamine receptor genes (DRD1-DRD5) in antipsychotic-induced weight gain. Pharmacogenomics J. 2012;12(2):156–164. doi: 10.1038/tpj.2010.65. [DOI] [PubMed] [Google Scholar]
  • 143.Tiwari AK, Rodgers JB, Sicard M, et al. Association study of polymorphisms in cholecystokinin gene and its receptors with antipsychotic induced weight gain in schizophrenia patients. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34(8):1484–1490. doi: 10.1016/j.pnpbp.2010.08.009. [DOI] [PubMed] [Google Scholar]
  • 144.Jassim G, Ferno J, Theisen FM, et al. Association study of energy homeostasis genes and antipsychotic-induced weight gain in patients with schizophrenia. Pharmacopsychiatry. 2011;44(1):15–20. doi: 10.1055/s-0030-1263174. [DOI] [PubMed] [Google Scholar]
  • 145.Beckers S, Zegers D, de Freitas F, et al. Association study of MC4R with complex obesity and replication of the rs17782313 association signal. Mol Genet Metab. 2011;103(1):71–75. doi: 10.1016/j.ymgme.2011.01.007. [DOI] [PubMed] [Google Scholar]
  • 146.Malhotra AK, Correll CU, Chowdhury NI, et al. Association between common variants near the melanocortin 4 receptor gene and severe antipsychotic drug-induced weight gain. Arch Gen Psychiatry. 2012;69(9):904–912. doi: 10.1001/archgenpsychiatry.2012.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Chowdhury NI, Tiwari AK, Souza RP, et al. Genetic association study between antipsychotic-induced weight gain and the melanocortin-4 receptor gene. Pharmacogenomics J. 2013;13(3):272–279. doi: 10.1038/tpj.2011.66. [DOI] [PubMed] [Google Scholar]
  • 148.Adkins DE, Aberg K, McClay JL, et al. Genomewide pharmacogenomic study of metabolic side effects to antipsychotic drugs. Mol Psychiatry. 2011;16(3):321–332. doi: 10.1038/mp.2010.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Nooijen PM, Carvalho F, Flanagan RJ. Haematological toxicity of clozapine and some other drugs used in psychiatry. Hum Psychopharmacol. 2011;26(2):112–119. doi: 10.1002/hup.1181. [DOI] [PubMed] [Google Scholar]
  • 150.Lieberman JA, Yunis J, Egea E, et al. HLA-B38, DR4, DQw3 and clozapine-induced agranulocytosis in Jewish patients with schizophrenia. Arch Gen Psychiatry. 1990;47(10):945–948. doi: 10.1001/archpsyc.1990.01810220061007. [DOI] [PubMed] [Google Scholar]
  • 151.Dettling M, Cascorbi I, Opgen-Rhein C, et al. Clozapine-induced agranulocytosis in schizophrenic Caucasians: confirming clues for associations with human leukocyte class I and II antigens. Pharmacogenomics J. 2007;7(5):325–332. doi: 10.1038/sj.tpj.6500423. [DOI] [PubMed] [Google Scholar]
  • 152.Mosyagin I, Dettling M, Roots I, et al. Impact of myeloperoxidase and NADPH-oxidase polymorphisms in drug-induced agranulocytosis. J Clin Psychopharmacol. 2004;24(6):613–617. doi: 10.1097/01.jcp.0000144891.52858.a6. [DOI] [PubMed] [Google Scholar]
  • 153.Opgen-Rhein C, Dettling M. Clozapine-induced agranulocytosis and its genetic determinants. Pharmacogenomics. 2008;9(8):1101–1111. doi: 10.2217/14622416.9.8.1101. [DOI] [PubMed] [Google Scholar]
  • 154.Zai CC, De Luca V, Hwang RW, et al. Meta-analysis of two dopamine D2 receptor gene polymorphisms with tardive dyskinesia in schizophrenia patients. Mol Psychiatry. 2007;12(9):794–795. doi: 10.1038/sj.mp.4002023. [DOI] [PubMed] [Google Scholar]
  • 155.Steen VM, Lovlie R, MacEwan T, et al. Dopamine D3-receptor gene variant and susceptibility to tardive dyskinesia in schizophrenic patients. Mol Psychiatry. 1997;2(2):139–145. doi: 10.1038/sj.mp.4000249. [DOI] [PubMed] [Google Scholar]
  • 156.Al Hadithy AF, Ivanova SA, Pechlivanoglou P, et al. Tardive dyskinesia and DRD3, HTR2A and HTR2C gene polymorphisms in Russian psychiatric inpatients from Siberia. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(3):475–481. doi: 10.1016/j.pnpbp.2009.01.010. [DOI] [PubMed] [Google Scholar]
  • 157.Lerer B, Segman RH, Fangerau H, et al. Pharmacogenetics of tardive dyskinesia: combined analysis of 780 patients supports association with dopamine D3 receptor gene Ser9Gly polymorphism. Neuropsychopharmacology. 2002;27(1):105–119. doi: 10.1016/S0893-133X(02)00293-2. [DOI] [PubMed] [Google Scholar]
  • 158.Bakker PR, van Harten PN, van Os J. Antipsychotic-induced tardive dyskinesia and the Ser9Gly polymorphism in the DRD3 gene: a meta analysis. Schizophr Res. 2006;83(2–3):185–192. doi: 10.1016/j.schres.2006.01.010. [DOI] [PubMed] [Google Scholar]
  • 159.Tsai HT, North KE, West SL, et al. The DRD3 rs6280 polymorphism and prevalence of tardive dyskinesia: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet. 2010;153B(1):57–66. doi: 10.1002/ajmg.b.30946. [DOI] [PubMed] [Google Scholar]
  • 160.Liou YJ, Liao DL, Chen JY, et al. Association analysis of the dopamine D3 receptor gene ser9gly and brain-derived neurotrophic factor gene val66met polymorphisms with antipsychotic-induced persistent tardive dyskinesia and clinical expression in Chinese schizophrenic patients. Neuromolecular Med. 2004;5(3):243–251. doi: 10.1385/NMM:5:3:243. [DOI] [PubMed] [Google Scholar]
  • 161.Chong SA, Tan EC, Tan CH, et al. Polymorphisms of dopamine receptors and tardive dyskinesia among Chinese patients with schizophrenia. Am J Med Genet B Neuropsychiatr Genet. 2003;116B(1):51–54. doi: 10.1002/ajmg.b.10004. [DOI] [PubMed] [Google Scholar]
  • 162.Lattuada E, Cavallaro R, Serretti A, et al. Tardive dyskinesia and DRD2, DRD3, DRD4, 5-HT2A variants in schizophrenia: an association study with repeated assessment. Int J Neuropsychopharmacol. 2004;7(4):489–493. doi: 10.1017/S1461145704004614. [DOI] [PubMed] [Google Scholar]
  • 163.Zai CC, Tiwari AK, Basile V, et al. Association study of tardive dyskinesia and five DRD4 polymorphisms in schizophrenia patients. Pharmacogenomics J. 2009;9(3):168–174. doi: 10.1038/tpj.2009.2. [DOI] [PubMed] [Google Scholar]
  • 164.Lerer B, Segman RH, Tan EC, et al. Combined analysis of 635 patients confirms an age-related association of the serotonin 2A receptor gene with tardive dyskinesia and specificity for the nonorofacial subtype. Int J Neuropsychopharmacol. 2005;8(3):411–425. doi: 10.1017/S1461145705005389. [DOI] [PubMed] [Google Scholar]
  • 165.Wilffert B, Al Hadithy AF, Sing VJ, et al. The role of dopamine D3, 5-HT2A and 5-HT2C receptor variants as pharmacogenetic determinants in tardive dyskinesia in African-Caribbean patients under chronic antipsychotic treatment: Curacao extrapyramidal syndromes study IX. J Psychopharmacol. 2009;23(6):652–659. doi: 10.1177/0269881108091594. [DOI] [PubMed] [Google Scholar]
  • 166.Segman RH, Heresco-Levy U, Finkel B, et al. Association between the serotonin 2A receptor gene and tardive dyskinesia in chronic schizophrenia. Mol Psychiatry. 2001;6(2):225–229. doi: 10.1038/sj.mp.4000842. [DOI] [PubMed] [Google Scholar]
  • 167.Basile VS, Ozdemir V, Masellis M, et al. Lack of association between serotonin-2A receptor gene (HTR2A) polymorphisms and tardive dyskinesia in schizophrenia. Mol Psychiatry. 2001;6(2):230–234. doi: 10.1038/sj.mp.4000847. [DOI] [PubMed] [Google Scholar]
  • 168.Zai CC, Tiwari AK, Müller DJ, et al. The catechol-O-methyltransferase gene in tardive dyskinesia. World J Biol Psychiatry. 2010;11(6):803–812. doi: 10.3109/15622975.2010.486043. [DOI] [PubMed] [Google Scholar]
  • 169.Souza RP, Remington G, Chowdhury NI, et al. Association study of the GSK-3B gene with tardive dyskinesia in European Caucasians. Eur Neuropsychopharmacol. 2010;20(10):688–694. doi: 10.1016/j.euroneuro.2010.05.002. [DOI] [PubMed] [Google Scholar]
  • 170.Liou YJ, Lai IC, Lin MW, et al. Haplotype analysis of endothelial nitric oxide synthase (NOS3) genetic variants and tardive dyskinesia in patients with schizophrenia. Pharmacogenet Genomics. 2006;16(3):151–157. doi: 10.1097/01.fpc.0000184958.05775.66. [DOI] [PubMed] [Google Scholar]
  • 171.Greenbaum L, Strous RD, Kanyas K, et al. Association of the RGS2 gene with extrapyramidal symptoms induced by treatment with antipsychotic medication. Pharmacogenet Genomics. 2007;17(7):519–528. doi: 10.1097/FPC.0b013e32800ffbb4. [DOI] [PubMed] [Google Scholar]
  • 172.Higa M, Ohnuma T, Maeshima H, et al. Association analysis between functional polymorphism of the rs4606 SNP in the RGS2 gene and antipsychotic-induced Parkinsonism in Japanese patients with schizophrenia: results from the Juntendo University Schizophrenia Projects (JUSP) Neurosci Lett. 2010;469(1):55–59. doi: 10.1016/j.neulet.2009.11.043. [DOI] [PubMed] [Google Scholar]
  • 173.Aberg K, Adkins DE, Liu Y, et al. Genome-wide association study of antipsychotic-induced QTc interval prolongation. Pharmacogenomics J. 2012;12(2):165–172. doi: 10.1038/tpj.2010.76. Epub 2010 Oct 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Greenbaum L, Alkelai A, Rigbi A, et al. Evidence for association of the GLI2 gene with tardive dyskinesia in patients with chronic schizophrenia. Mov Disord. 2010;25(16):2809–2817. doi: 10.1002/mds.23377. [DOI] [PubMed] [Google Scholar]
  • 175.Alkelai A, Greenbaum L, Rigbi A, et al. Genome-wide association study of antipsychotic-induced parkinsonism severity among schizophrenia patients. Psychopharmacology (Berl) 2009;206(3):491–499. doi: 10.1007/s00213-009-1627-z. [DOI] [PubMed] [Google Scholar]
  • 176.Hall-Flavin DK, Winner JG, Allen JD, et al. Using a pharmacogenomic algorithm to guide the treatment of depression. Transl Psychiatry. 2012;2:e172. doi: 10.1038/tp.2012.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Chen SF, Shen YC, Chen CH. HTR2A A-1438G/T102C polymorphisms predict negative symptoms performance upon aripiprazole treatment in schizophrenic patients. Psychopharmacology (Berl) 2009;205(2):285–292. doi: 10.1007/s00213-009-1538-z. [DOI] [PubMed] [Google Scholar]
  • 178.Arranz MJ, Collier DA, Munro J, et al. Analysis of a structural polymorphism in the 5-HT2A receptor and clinical response to clozapine. Neurosci Lett. 1996;217(2–3):177–178. [PubMed] [Google Scholar]
  • 179.Masellis M, Basile V, Meltzer HY, et al. Serotonin subtype 2 receptor genes and clinical response to clozapine in schizophrenia patients. Neuropsychopharmacology. 1998;19(2):123–132. doi: 10.1016/S0893-133X(98)00007-4. [DOI] [PubMed] [Google Scholar]
  • 180.Bertolino A, Caforio G, Blasi G, et al. COMT Val158Met polymorphism predicts negative symptoms response to treatment with olanzapine in schizophrenia. Schizophr Res. 2007;95(1–3):253–255. doi: 10.1016/j.schres.2007.06.014. [DOI] [PubMed] [Google Scholar]
  • 181.De Luca V, Müller DJ, Hwang R, et al. HTR2C haplotypes and antipsychotics-induced weight gain: X-linked multimarker analysis. Hum Psychopharmacol. 2007;22(7):463–467. doi: 10.1002/hup.868. [DOI] [PubMed] [Google Scholar]
  • 182.Templeman LA, Reynolds GP, Arranz B, et al. Polymorphisms of the 5-HT2C receptor and leptin genes are associated with antipsychotic drug-induced weight gain in Caucasian subjects with a first-episode psychosis. Pharmacogenet Genomics. 2005;15(4):195–200. doi: 10.1097/01213011-200504000-00002. [DOI] [PubMed] [Google Scholar]
  • 183.Zhang ZJ, Yao ZJ, Mou XD, et al. [Association of −2548G/A functional polymorphism in the promoter region of leptin gene with antipsychotic agent-induced weight gain] Zhonghua Yi Xue Za Zhi. 2003;83(24):2119–2123. Chinese. [PubMed] [Google Scholar]
  • 184.Souza RP, De Luca V, Muscettola G, et al. Association of antipsychotic induced weight gain and body mass index with GNB3 gene: a meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(8):1848–1853. doi: 10.1016/j.pnpbp.2008.08.014. [DOI] [PubMed] [Google Scholar]

Articles from Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie are provided here courtesy of SAGE Publications

RESOURCES