Numerous genetic variants have been shown to be associated with antipsychotic response and adverse effects of schizophrenia treatment. However, the clinical application of these findings is limited. The aim of this narrative review is to summarize the most recent publications and recommendations related to the genetics of antipsychotic treatment and shed light on the clinical utility of pharmacogenetics/pharmacogenomics (PGx). We reviewed the literature on PGx studies with antipsychotic drugs (i.e., antipsychotic response and adverse effects) and commonly used commercial PGx tools for clinical practice. Publications and reviews were included with emphasis on articles published between January 2015 and April 2018. We found 44 studies focusing on antipsychotic response and 45 studies on adverse effects (e.g., antipsychotic-induced weight gain, movement disorders, hormonal abnormality, and clozapine-induced agranulocytosis/granulocytopenia), albeit with mixed results. Overall, several gene variants related to antipsychotic response and adverse effects in the treatment of patients with schizophrenia have been reported, and several commercial pharmacogenomic tests have become available. However, further well-designed investigations and replication studies in large and well-characterized samples are needed to facilitate the application of PGx findings to clinical practice.

Schizophrenia is a severe neurodevelopmental disorder with high heritability [1, 2]. Likewise, genetic factors have also been implicated in antipsychotic medication used in the treatment of schizophrenia [3, 4]. Antipsychotics are currently the main medication for treatment of schizophrenia; however, the clinical response and adverse effects vary substantially between individuals, which typically leads clinicians to “trial-and-error” procedures before optimal treatment medication is found [5].

Pharmacogenetics/pharmacogenomics (PGx) is the use of genomic data to understand drug metabolism and response, which may allow clinicians to select medications based on the genetic variability across patients [6]. Multiple studies have investigated PGx approaches in order to identify genotype-specific dosing and predict antipsychotic responses and/or adverse effects [6]. Over the past years, specific recommendations on how to apply such genetic information, including commercial tests, have been proposed for clinical practice.

This narrative review summarizes most recent publications and recommendations related to the genetics of antipsychotic treatment. Furthermore, this review aims to shed light on the clinical utility of PGx.

First, recently published articles were searched using PubMed. The following search terms were applied: (schizophreni* or schizoaffective or psychosis or psychoses or “severe mental illness”) and (antipsychotic* or neuroleptic*) and (pharmacogenetic* or genetic* or genomic* or gene or “single nucleotide polymorphism” or SNP or polymorphism). Studies were included if they (1) were peer-reviewed original articles or meta-analyses, (2) referred to PGx in the treatment of patients with schizophrenia or related psychoses (i.e., serum/plasma concentrations or dosages, treatment response, and major adverse effects including antipsychotic-induced weight gain [AIWG], movement disorders, hyperprolactinemia, and clozapine-associated neutropenia), and (3) were published in English between January 2015 and April 2018. Next, expert group recommendations and Food and Drug Administration (FDA) drug labeling were reviewed regarding PGx information for antipsychotics.

Articles Included in This Review

Ninety-two original articles and 6 systematic reviews and meta-analyses were included in this review. Among the original articles, 7 reported on serum/plasma concentrations or dosage (Table 1), 44 reported on responses to antipsychotics (Table 2), and 45 reported on antipsychotic-induced adverse effects (Table 3); some of the included articles reported on two or three topics (i.e., antipsychotic concentrations or dosage, response, and adverse effects).

Antipsychotic Serum (Plasma) Concentrations and Dosage

Cytochrome P450 (CYP) enzymes mediate the metabolism of various drugs, and for certain CYPs, the genotype affects serum (plasma) drug levels [7]. Previous studies have explored the role of pharmacokinetic (PK) genes for antipsychotic serum/plasma concentrations, particularly of the CYP genotype [8]. Four classes of CYP metabolizer profiles have been established: poor (PM), intermediate, normal, and ultrarapid metabolizers [9]. These phenotypes are based on the multiallelic nature of the CYP enzyme’s genetic construct, which reflects several polymorphisms such as single nucleotide polymorphisms (SNPs) [8]. Notably, the FDA has included warnings for higher side effects at standard dosages for CYP2D6 PM; the details are described later. Nonetheless, the relationship between antipsychotic serum/plasma concentrations and the CYP genotypes continues to be investigated.

Three studies in this review reported a potential in­fluence of polymorphisms of CYP genotypes (i.e., the CYP1A2, 2C19, 2D6, and 3A4 genes) on PK profiles (i.e., the antipsychotic serum/plasma concentration) [7, 10, 11]. One study reported a significant association between olanzapine serum concentrations and the CYP1A2*1D and *1F polymorphisms [10], while 2 other studies also reported that polymorphisms in CYP2D6, CYP1A2, and DRD3 affected antipsychotic concentrations to some extent [7, 11]. The membrane transport protein P-glycoprotein, which is encoded by the ATP-binding cassette subfamily B member 1 (ABCB1) gene, helps clear antipsychotic medications across the blood-brain barrier [12]. One study investigated the influence of the ABCB1 rs1045642 polymorphism and the ATP-binding cassette subfamily C member 1 (ABCC1) rs212090 polymorphism on clozapine and norclozapine serum levels [13]. The combination of ABCB1 and ABCC1 homozygotes was associated with increased clozapine and norclozapine serum levels, although there were no significant associations between the ABCB1 and ABCC1 SNPs and serum concentrations.

In addition, antipsychotic dosage is affected by PK and pharmacodynamic (PD) factors. Three studies that investigated the influence of genetics on antipsychotic dosage were included in the review [14‒16]. Hettige et al. [14, 17] reported a significant association between higher antipsychotic dosage and the γ-aminobutyric acid type A receptor β1 subunit (GABRB1) gene polymorphisms rs16860087 and rs4627835, although the authors were unsuccessful in reproducing the results of their previous study. However, the other 2 studies [15, 16] (using a genome-wide association approach) reported negative findings. One of the 2 studies investigated the association between antipsychotic dosage and polygenic risk scores in schizophrenia derived from significant risk loci identified by the Psychiatric Genomics Consortium (PGC2) but found no association between them [15]. Given that antipsychotic dosage can be influenced by several factors such as symptom severity and duration of illness, further research on this topic will be needed.

Antipsychotic Response

Response to antipsychotics is a complex phenotype involving genetic and clinical factors such as symptom severity and adherence level. Despite this challenging issue, various genetic variants related to PK and PD have been investigated in association with the response to antipsychotics.

Most studies included in this review focused on genetic polymorphisms, associated with response to antipsychotics, and how these polymorphisms related to clinical symptom improvement in patients with schizophrenia (e.g., a reduction in scores in the Brief Psychiatric Rating Scale [18] or the Positive and Negative Syndrome Scale [PANSS] [19]).

PK CYP450 and ABCB1 Genes

Antipsychotic drugs are primarily metabolized by CYP2D6, CYP3A4, and CYP1A2, and approximately 40, 23, and 18% of antipsychotics comprise major substrates for CYP2D6, CYP3A4, and CYP1A2, respectively [20]. Numerous studies have investigated the association between CYP variants and response to antipsychotics [8]. However, most have resulted in negative findings [8]. In the current review, 2 studies reported significant associations between antipsychotic response and the CYP3A43 rs680055 and CYP2D6 rs3892097 polymorphisms [21, 22]. Although another study found that CYP2C19 rs4986893 and CYP2D6 rs1135840 were associated with antipsychotic response, the associations did not remain significant after correction for multiple testing [22, 23].

In addition to the CYP enzymes, common polymorphisms of the P-glycoprotein-coding gene (ABCB1) have been associated with antipsychotic response. Previous studies investigating the rs1045642 (C3435T), rs2032582 (G2677T/A), and rs1128503 (C1236T) polymorphisms and their association with antipsychotic response are summarized elsewhere; the findings in this area of research are still inconsistent [12]. The current review includes 3 studies that investigated the association between polymorphisms in ABCB1 and response to antipsychotics. Of these, 1 study reported positive findings for the rs2032582 polymorphism, although the association did not remain significant after correction for multiple testing [23]. The other studies reported negative findings regarding the rs1045642 and rs2032582 polymorphisms, although a combination of a loss-of-function CYP2D6 allele and the TT genotype of ABCB1 rs2032582 was associated with poor response to antipsychotic treatment in one of those 2 studies [24, 25]. The current review includes several studies suggesting that response to antipsychotics is influenced by CYP genetic variants and ABCB1. However, this conclusion is limited by several factors such as heterogeneous ethnicity, medication, and study duration. Therefore, the association between PK gene variants and antipsychotic responses remains controversial, and further investigation is required.

Dopaminergic System Genes

Several studies investigated PD-related genes and treatment responses, particularly dopamine receptor D2 (DRD2), which plays a critical role in antipsychotic treatment. DRD2 antagonism is a common mechanism of antipsychotic action and is considered necessary for antipsychotic efficacy [26]. Accordingly, both DRD2 and D2-like receptor genes, including DRD3 and DRD4, have been extensively investigated as candidate genes that influence antipsychotic responses [27, 28]. We identified 7 studies that investigated the association between polymorphisms of the DRD2 gene and response to antipsychotics [21, 29‒34], where three polymorphisms of DRD2 (rs180498, rs2514218, and rs1079597) were significantly associated with treatment response [21, 30‒32]. In particular rs2514218, which is located 47-kb upstream of the DRD2 gene, was previously reported as one of the genome-wide significant SNPs associated with risk of schizophrenia [35]. Further, it was associated with response to antipsychotics such as clozapine and risperidone in independent samples [31].

In addition, Blasi et al. [33] reported that a combination of the DRD2 rs1076560 and serotonin 2A receptor gene (HTR2A) rs6314 polymorphisms affected the response to antipsychotic treatment. Although a previous meta-analysis reported an association of DRD2 rs1799732 (–141C Ins/Del) with antipsychotic treatment response, no statistically significant associations were found in 2 studies included in this review [29, 34]. The association of DRD3 rs6280 (Ser9Gly) with antipsychotic responses has been extensively investigated [36]. One study included in this review reported a significant association between DRD3 rs6280 (Ser9Gly) and response to quetiapine [23]. A previous meta-analysis showed a nonsignificant trend toward lower response in Ser allele carriers [27]. Although DRD3 may theoretically affect antipsychotic responses via a high affinity of antipsychotics for the D3 receptor, empirical research findings remain inconsistent [36]. In contrast, 3 studies included in this review reported no significant associations of DRD1 rs5326, rs4867798, rs4532, and rs686, DRD3 rs6280, and DRD4 rs1800955 and rs4646984 with response to antipsychotics [25, 29, 37]. Similarly, a recent meta-analysis also showed no significant association between the DRD1 rs4532 polymorphism and response to antipsychotics, including cloza­pine [38].

COMT Gene

In addition, catechol-O-methyltransferase (COMT), which is another dopamine pathway-related gene, has been investigated. The COMT gene is located within the 22q11 chromosomal locus, which is involved in schizophrenia susceptibility [39]. The COMT enzyme is involved in dopamine degradation [40], making the COMT gene a candidate gene for an association with antipsychotic response. In this review, 6 of 9 studies reported associations of polymorphisms of the COMT gene (including marker rs4680) with improvement in the response to treatment by antipsychotics, including clozapine [23, 41‒45]. Furthermore, a recent meta-analysis also showed that the COMT Val158Met (rs4680) polymorphism was significantly associated with response to antipsychotics [46]. These findings suggest that COMT gene variants, particularly COMT Val158Met (rs4680), are associated with antipsychotic response. However, the role of Val158Met polymorphism in antipsychotic treatment regulation is yet to be entirely elucidated [47].

Serotonergic System Genes

Most second-generation antipsychotics display a high affinity for serotonin receptors, which is considered as contributing to antipsychotic response [48]. Accordingly, several serotonin pathway-associated genes have been studied. Three studies in this review reported that the serotonin 1A receptor (HTR1A) polymorphism rs6295 was significantly associated with improvement in clinical symptoms [45, 49, 50]. Takekita et al. [49] indicated that the HTR1A polymorphism rs1364043 and the rs10042486-rs6295-rs1364043 haplotype might influence negative symptoms on the PANSS. A recent meta-analysis by Takekita et al. [51] showed that the HTR1A rs6295 polymorphism might be associated with improvement of negative symptoms but not with overall or positive symptoms. However, given the relatively small number of studies included in the analysis (n = 10) and the heterogeneity of antipsychotics, further studies using larger sample populations and homogeneous treatments are required to confirm this effect. It is thought that HTR2A contributes to the pathophysiology of hallucinations, and the receptor has been a major target among many second-generation antipsychotics [52]. Two of the 3 studies identified in this review indicated the impact of the HTR2A polymorphism rs6314 on antipsychotic treatment response modulation [21, 33, 42].

Associations of the HTR2C polymorphisms rs1328685, rs643627, rs498177, rs3813929, and rs1414334 with response to antipsychotics were also found [23, 42]. In contrast, the serotonin 7 receptor (HTR7) polymorphisms rs12412496, rs7916403, and rs1935349 did not appear to improve antipsychotic treatments [53].

Other Genes

Several other candidate genes, including glutamate-related genes, are also identified in this review (Table 2) [10, 23, 25, 34, 43, 54‒66]. For example, Stevenson et al. [67] examined the association between glutamate gene polymorphisms and treatment response to risperidone in patients with first-episode psychosis (n = 86). They found that several SNPs were associated with antipsychotic treatment response; of these, the type-7 metabotropic glutamate receptor (GRM7) polymorphism rs2069062 showed the most robust finding [67].

Similarly, GRM7 rs2133450 was significantly associated with Emsley’s positive domain derived from the PANSS in a genome-wide association study (GWAS), using a sample of patients treated with risperidone [68]. As for other genes, synaptosomal-associated protein 25 kDa (SNAP25) rs8636 and ankyrin repeat and sterile alpha motif domain-containing protein 1B (ANKS1B) rs7968606 were significantly associated with response to amisulpride [56, 69]. The brain-derived neurotrophic factor (BDNF) Val66Met (rs6265) polymorphism was not associated with response to antipsychotics in a recent meta-analysis that included 9 studies with a total of 2,461 patients treated with antipsychotics [70].

Genome-Wide Association Studies

An alternative to the candidate gene approach is to use GWAS, which is characterized as a hypothesis-free approach [71]. To our knowledge, the first GWAS investigating antipsychotic response was performed in the context of a phase III clinical trial of iloperidone, published in 2009 [72]. Subsequently, several GWAS on antipsychotic response have been conducted [73‒76]. In the current review, we found 9 studies that used this approach [67, 68, 77‒82]. The following gene polymorphisms were significantly associated with treatment response: rs72790443 in multiple EGF-like domains 10 (MEGF10); rs1471786 in solute carrier family 1 member 1 (SLC1A1); rs9291547 in protocadherin 7 (PCDH7); rs12711680 in contactin-associated protein-like 5 (CNTNAP5); rs6444970 in TRAF2 and NCK-interacting kinase (TNIK); rs2133450, rs2069062, and rs2014195 in GRM7; rs9307122 and rs1875705 in glutamate ionotropic receptor delta type subunit 2 (GRID2); rs3129996 in protein phosphatase 1 regulatory subunit 18 (PPP1R18); and rs6435681 in erb-b2 receptor tyrosine kinase 4 (ERBB4) [67, 68, 77, 81‒83]. Among these studies, the study by Yu et al. [77] has one of the largest sample sizes reported so far (n = 2,413 in the discovery cohort and n = 1,379 in the replication sample). They found five novel genome-wide significant loci associated with treatment response in samples of Han Chinese ancestry (i.e., rs72790443 in MEGF10, rs1471786 in SLC1A1, rs9291547 in PCDH7, rs12711680 in CNTNAP5, and rs6444970 in TNIK). Furthermore, three additional loci were associated with drug-specific treatment responses (rs2239063 in CANCA1C for olanzapine, rs16921385 in SLC1A1 for risperidone, and rs17022006 in CNTN4 for aripiprazole). Although several factors such as duration of illness, duration of treatment, and concomitant therapy should be considered as covariates and validation studies in other ethnic populations are needed, the genes identified are clinically relevant and the findings can provide novel insights into the underlying mechanisms of antipsychotic action [84].

In addition, Li et al. [78] reported that common genetic variants related to synaptic adhesion complexes, scaffolding, and the alternative splicing regulator were associated with treatment response to lurasidone, although their findings were not statistically significant after correction for multiple testing. Ovenden et al. [79] found tentative associations between response to treatment and the mannosidase beta (MANBA), collagen type IX alpha 2 chain (COL9A2), and nuclear factor kappa B subunit 1 (NFKB1) genes.

Whole-Exome Sequencing

Recently, the rapid progress of next-generation sequencing, also called massively parallel sequencing, has led to revolutionary changes in medical genomics, including the field of psychiatry [85]. This method enables the sequencing of the whole genomes or exomes [85]. In the first study using exome sequencing to examine the pharmacogenomics of antipsychotic response, Drögemöller et al. [86] sequenced 11 South African patients with first-episode schizophrenia to identify genetic traits related to antipsychotic response. In the current review, we found a whole-exome sequencing study by the same group [83]. They identified two novel variants that were significantly associated with antipsychotic treatment response in two independent first-episode schizophrenia cohorts: rs13025959 in myosin VIIB (MYO7B) and rs10380 in 5-methyltetrahydrofolate-homocysteine methyltransferase reductase (MTRR). Although these new findings provide valuable information regarding future pharmacogenomic antipsychotic studies, further research will be needed.

Adverse Effects of Antipsychotics

Four main categories of antipsychotic-induced adverse effects were identified: (1) metabolic dysregulation, such as AIWG and metabolic syndrome; (2) movement disorders, including extrapyramidal symptoms (EPS), tardive dyskinesia (TD), and restless legs syndrome; (3) hormonal abnormalities, such as hyperprolactinemia; and (4) clozapine-induced agranulocytosis/granulocytopenia (CIAG).

AIWG and Metabolic Syndrome

AIWG is a common side effect in patients treated with antipsychotics, coinciding with metabolic dysregulation and heritability [87]. Although the mechanisms of AIWG are not fully understood, various genes appear to contribute to symptom onset (e.g., genes related to antipsychotic metabolism, neurotransmitter systems, and neuroendocrine systems) [88].

Twenty-eight of the studies in this review investigated the association between genetic factors and AIWG and metabolic dysregulation (Table 3). Regarding genes related to antipsychotic metabolism, Czerwensky et al. [10] found no significant association between AIWG and CYP1A2*1D and *1F polymorphisms. The ABCB1 rs1045642 and ABCC1 rs212090 polymorphisms were associated with AIWG in male patients treated with clozapine, although no significant associations were found in the total sample [13].

As for neurotransmitter systems, serotonin receptors influence central pathways affecting satiety and hunger [88]. Of the serotonin receptor genes, HTR2C has been extensively studied in association with AIWG, and the −759C/T polymorphism of HTR2C has been targeted in previous studies. One study included in this review replicated an association of the −759C/T polymorphism of HTR2C with AIWG [89]. Although another study produced negative findings, the authors found a significant association between insulinemia and T allele carriers [90]. A recent meta-analysis that included papers up until the end of 2014 reported 13 SNPs from nine genes being significantly associated with AIWG, including the −759C/T polymorphism of HTR2C [91]. In contrast, the rs1414334 polymorphism of HTR2C and the studied polymorphisms of HTR3A and HTR3B were not associated with AIWG and metabolic dysregulation [92‒94]. The dopamine system also appears to be associated with AIWG and metabolic dysregulation [88].

BDNF is a neurotrophic factor that plays an important role in neurogenesis, neuronal survival, and synaptic plasticity [95]. Evidence exists to indicate that BDNF contributes to food intake and body weight control [95]. Although researchers continue to examine the BDNF Val66Met (rs6265) polymorphism and its potential association with AIWG, the results are still inconsistent [96‒98]. Two studies included in this review reported signi­ficant associations between AIWG and the BDNF Met66Met polymorphism [99, 100]. Furthermore, the recent meta-analysis mentioned above [91] also showed a significant association between the BDNF polymorphism and AIWG.

Leptin is an adipocyte hormone that acts on the hypothalamus to regulate appetite and energy expenditure, which makes it a natural candidate for genetic AIWG studies. In particular, the −2548A/G (rs7799039) polymorphism of the LEP gene has been investigated in several studies [92]. However, the association between the −2548A/G polymorphism and AIWG remains inconclusive. In the current review, 1 study reported no significant association [92].

Antipsychotic-induced lipid biosynthesis may explain the metabolic side effects of antipsychotics [101]. Le Hellard et al. [102] investigated the association between AIWG and five major genes involved in the sterol regulatory element-binding protein (SREBP) activation of fatty acids and cholesterol production (SREBF1, SREBF2, SREBP cleavage-activation protein [SCAP], insulin-induced gene 1 [INSIG1], and INSIG2). They found a significant association between three markers localized within or near INSIG2 (rs17587100, rs17047764, and rs10490624) and AIWG. In addition, nominally significant associations with AIWG were found in INSIG1 (rs13223383), SREBF2 (rs4822064), and SCAP (rs12490383). The following gene polymorphisms were significantly associated with AIWG or the metabolic syndrome in this review: rs11654081 in SREBF1; rs2267443 and rs1052717 in SREBF2; and rs12151787, rs1049626, and rs17047733 in INSIG2 [103‒105]. Taken together, these genes might enhance the risk of antipsychotic- induced metabolic dysregulation, although these mixed findings warrant further investigation.

The mitochondria play a key role in energy metabolism, and mitochondrial dysfunction contributes to metabolic disorders [106]. Thus, the mitochondrial system has become an interesting target for AIWG studies, although there have not been many studies investigating mitochondrial genetic variance in AIWG; Gonçalves et al. [107] reported an association between the mitochondrial NADH dehydrogenase (ubiquinone) Fe-S protein 1 (NDUFS1) gene and AIWG. Translocator protein 18 kDa (TSPO) is also a mitochondrial gene that may regulate weight through its effects on mitochondrial metabolism [108]. One study included in this review reported a nominal association between AIWG and the TSPO rs6971 polymorphism in two independent samples [66]. More recently, Mittal et al. [109] examined associations of variants in nuclear-encoded mitochondrial genes and mitochondrial DNA with AIWG. They identified three nuclear-encoded mitochondrial genes conferring a risk for AIWG. These findings suggest that mitochondrial genes have a significant role in AIWG, although replication studies are needed with larger samples and/or other ethnic groups.

As for other genes, Brandl et al. [110] conducted a GWAS using the data set derived from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) sample, selecting a well-characterized and homogeneous subsample of individuals of European ancestry. Although none of the SNPs were significantly associated with AIWG, there were nominal associations for rs9346455 upstream of opioid growth factor receptor-like 1 (OGFRL1) and rs1059778 in iron-sulfur cluster assembly (IBA57). Notably, rs9346455 showed a significant association with AIWG in an independent sample of Europeans patients (n = 86) [110]. Another GWAS of 534 Han Chinese patients with schizophrenia found significant associations with rs10977144 and rs10977154 in protein tyrosine phosphatase, receptor type D (PTPRD), and these results were further validated in an independent replication cohort (n = 547) [111]. Notably, a recent meta-analysis that included studies through the end of 2014 found that 13 SNPs from nine genes were significantly associated with AIWG, with the SNPs in adrenoreceptor alpha 2A (ADRA2A), DRD2, HTR2C, and melanocortin 4 receptor (MC4R) having the largest effect sizes [91]. Additional details are summarized in Table 2. The previous studies from 2010 to 2015, focusing on gene variants associated with AIWG, are summarized in a recent comprehensive review [87].

In summary, several genes and polymorphisms have been studied in relation to AIWG with some findings showing independent replications. Nevertheless, considering that AIWG is a polygenic phenotype interacting with clinical and demographic factors, further studies are required to elaborate an algorithm for clinical practice [91].

Antipsychotic-Induced Movement Disorders

Antipsychotic drugs are associated with various movement disorders. Among them, TD is a severe and often irreversible side effect, characterized by involuntary trunk, limb, and orofacial muscle movements. Several studies using genome-wide association and candidate gene approaches have been conducted to identify risk variants associated with TD development [112]. In the current review, 12 studies reported associations between variants of related genes and EPS, TD, and restless legs syndrome, with 6 of them reporting TD [113‒124]. A large number of studies have examined an association between CYP genes, including CYP2D6, and EPS/TD, and produced mixed findings [87]. One meta-analysis from 2005 suggested that loss-of-function alleles in CYP2D6 increased the risk of developing TD [125], whereas a recent study reported that CYP2D6 ultrarapid metabolizers were associated with an increased risk of TD [126]. Two studies included in this review found associations of polymorphic 1846G>A CYP2D6*4 with EPS and limb-trunk TD [116, 119]. Combined with the existing knowledge, it appears that CYP2D6 is a potential gene associated with the onset of TD [112]. In addition, CYP1A2 is involved in the metabolism of several antipsychotics, such as cloza­pine, olanzapine, and some first-generation antipsychotics [8]. Two studies included in this review reported an association between the CYP1A2*F polymorphic variant and TD [119, 122]. However, further studies using larger samples are required to confirm the association between CYP enzyme genes and TD, and to evaluate the clinical utility of these genes.

Neurotransmitter genes, including those within the dopamine, serotonin, γ-aminobutyric acid, and glutamate systems, may be linked to antipsychotic-induced movement disorders including TD [112]. For example, the Taq1A (rs1800497) polymorphism of the DRD2 gene [127], Ser9Gly (rs6280) of the DRD3 gene [128], and T102C (rs6313) of the HTR2A gene [129] have previously been investigated as risk variants for TD. Furthermore, vesicular monoamine transporter 2, which is coded by the solute carrier family 18 member A2 (SLC18A2) gene, has been implicated in TD [130, 131]. Zai et al. [131] found that the polymorphisms rs2015586, rs363390, rs363224, and rs14240 were associated with TD. Remarkably, these findings are consistent with clinical evidence demonstrating that a novel selective vesicular monoamine transporter 2 inhibitor (valbenazine) improves TD in patients with schizophrenia [132, 133]. In the current review, 1 study examined the association of 43 tag SNPs in five neurotransmitter genes with TD [118]. As a result, only rs1345423 in the glutamate receptor, ionotropic, N-methyl D-aspartate 2A (GRIN2A) gene showed a significant association with TD. A recent study focused on the influence of the neuregulin-1 (NGR1) and ERBB4 genes on TD [114], which help regulate N-methyl-D-aspartate and dopaminergic activity [134, 135]. They found a significant association between ERBB4 rs839523 and TD, although additional replication studies are needed to replicate this preliminary finding.

One study included in this review investigated an association between EPS and 202 SNPs in 31 neurotransmitter genes [117]. Four SNPs (rs9567733 [HTR2A], rs363341 [SLC18A2], rs1334802 [glutamate ionotropic receptor kainate type subunit 3 (GRIK3)], and rs1124491 [DRD2]) were significantly associated with EPS. Mas et al. [123] reported that the mTOR pathway was important for developing EPS. Using protein-protein interaction network analysis in 12 antipsychotic-naïve patients with first-episode psychosis, the same group showed that the NF-κB pathway (inflammatory response) and the mTOR pathway (lipid biosynthesis, insulin signaling, and autophagy) played a key role in EPS [121].

In summary, a large number of genes are potentially involved in antipsychotic-induced movement disorders. In the light of clinical evidence, SLC18A2 is a promising marker of TD risk. Nevertheless, further research and genetic validation studies are required to predict the risk for antipsychotic-induced movement disorders, including TD [112].

Antipsychotic-Induced Prolactin Increase

With regard to hormonal abnormality, hyperprolactinemia has been reported as a common adverse effect of antipsychotics [136]. Prolactin synthesis and secretion are inhibited by dopamine in the anterior pituitary gland through DRD2 activation [137]. Thus, an association between the DRD2 gene and hyperprolactinemia has been investigated [138]. A recent meta-analysis that included studies up until May 2015 showed that DRD2 Taq1A A1 carriers had significantly higher prolactin levels than A1 noncarriers among patients with schizophrenia, although further investigation will be needed due to several limitations, including a small sample size and heterogeneity of antipsychotics (n = 475; Hedges’ g 0.250; 95% CI: 0.068–0.433; p = 0.007) [138]. This review included 3 original studies that focused on the association between genetic variants and hyperprolactinemia (or an increase in prolactin levels). It appears that rs1341239 in the prolactin (PRL) gene, rs569959 and rs17326429 in the HTR2C gene, and rs4680 in the COMT gene are significantly associated with hyperprolactinemia [44, 139, 140]. Thus, investigating the effect of combinations of several gene variants will be required to predict the risk for hyperprolactinemia.

Clozapine-Induced Agranulocytosis/Granulocytopenia

Clozapine is the only drug with proven efficacy in treatment-resistant schizophrenia. However, it has a risk of hematological side effects such as CIAG [141]. The human leukocyte antigen (HLA) genes have been implicated in clozapine-induced agranulocytosis (CIA) [142]. The first GWAS in relation to CIA, which was conducted by the Clozapine-Induced Agranulocytosis Consortium (CIAC), demonstrated that the HLA-DQB1 126Q and HLA-B 158T alleles were significantly associated with CIAG [143]. The current review includes 2 studies that examined the genetic factor for CIAG [144, 145]. One study reported a significant association of HLA-B*59: 01 – which was not compatible with the HLA-B 158T allele detected in the previous study – with CIAG in a Japanese sample [144]. The other study, using a genome-wide association approach, HLA allele imputation, exome array, and copy number variation, reported that clozapine- associated neutropenia was associated with rs149104283 located between SLCO1B3 and SLCO1B7 (solute carrier organic anion transporter family, member 1B3 and member 1B7) through a meta-analysis using the CIAC samples [145]. HLA-DQB1 (126Q) and HLA-B (158T), implicated in a previous study [143], could not be imputed with sufficient quality and thus were investigated in the study, while no imputed classic HLA or amino acid polymorphism was associated with clozapine-induced neutropenia. As for another finding, the association of HLA-DBQ1 6672G>C (rs113332494), which had been shown to be associated with the risk for CIA in another previous study [146], was replicated in this study.

In 2007, a first commercial test kit for CIA, PGxPredict:CLOZAPINE (Clinical Data, Inc., New Haven, CT, USA), which included the 6672G/C polymorphism in HLA-DQB1 for the detection of high-risk patients carrying the C allele, was launched, with a high specificity of 98.4% but a low sensitivity of 21.5% [47]. However, this test was discontinued due to lack of general interest at that time [147]. Further research focusing on multiple genes and polymorphisms will be needed to predict CIAG.

Expert Consensus Recommendations and FDA Drug Labeling

Currently, the US FDA provides information on pharmacogenomic biomarkers in their drug labeling for nine antipsychotics (i.e., aripiprazole, aripiprazole lauroxil, brexpiprazole, clozapine, iloperidone, perphenazine, pimozide, risperidone, and thioridazine) [148]. Seven out of nine refer to the CYP2D6 PM status, where dose adjustment recommendations are provided for the following antipsychotics: aripiprazole, aripiprazole lauroxil, brexpiprazole, clozapine, iloperidone, pimozide, and thioridazine. Similarly, the Pharmacogenomics Knowledgebase (PharmGKB) website lists ten antipsychotics where caution is advised for patients who are poor CYP2D6 metabolizers [149]. Drug labels with PGx information are provided for the following ten antipsychotics: aripiprazole, aripiprazole lauroxil, brexpiprazole, clozapine, iloperidone, olanzapine, perphenazine, pimozide, risperidone, and thioridazine.

The Clinical Pharmacogenetics Implementation Consortium (CPIC) [150], which was established in 2009 as a shared project of PharmGKB and the Pharmacogenomics Research Network (PGRN) [151], also provides useful gene-drug information for clinicians. This is available on the PharmGKB website, which assigns CPIC levels to genes and drugs according to the extent to which genetic information affects drug prescription. PharmGKB also provides levels of evidence for gene-drug associations. However, recommendations for antipsychotic medications have not yet been published. In contrast, the Royal Dutch Association for the Advancement of Pharmacy – Dutch Pharmacogenetics Working Group has also provided PGx drug dosing guidelines based on CYP2D6 genotypes for six antipsychotics: aripiprazole, clozapine, haloperidol, olanzapine, risperidone, and zuclopenthixol [152]. The guidelines are available on the PharmGKB website. An overview of the current recommendations is provided in Table 4.

In summary, recommendations exist on how to use PK variants (such as CYP2D6) for some antipsychotics; however, no PD variants for antipsychotics are available for clinical practice.

Since the FDA first approved a PGx testing platform in 2004 (i.e., that of Roche Molecular Systems Inc. and Affymetrix Inc. [Pleasanton, CA, USA] for CYP2D6 and CYP2C19), several pharmacogenomic tests have become commercially available [153]. Several randomized clinical trials have previously evaluated the effectiveness of PGx testing for antidepressants and compared it with that for standard treatments (i.e., treatment as usual), and promising results regarding the clinical utility of PGx testing have been observed [154‒156]. However, the precise clinical validity and utility of PGx tests regarding antipsychotics remain to be validated in randomized clinical trials. Therefore, randomized clinical trials investigating the effectiveness of pharmacogenomic tests for antipsychotics in clinical practice are required.

In this study, we reviewed recent developments regarding PGx in the treatment of schizophrenia. Numerous new studies have been published in the past years on antipsychotic serum/plasma concentrations, response to treatment, and adverse effects. While some studies could replicate previous findings, others could not, but many new associations have been reported which warrant further validation. Overall, the strongest associations are being noted of CYP2D6 metabolizer status and outcome with antipsychotic medications. The influence of CYP2D6 on antipsychotic dosage and the occurrence of side effects has also been integrated in recommendations provided by various expert groups and agencies such as the FDA. Not surprisingly, genetic tests are now being commercially offered across the world and have recently been reviewed for their clinical use [153]. In addition, some promising studies have detected associations between “older” candidate genes such as COMT and treatment response.

However, newer findings need to be considered as preliminary until further replication studies will have been conducted. Importantly, there is still a large heterogeneity across studies, possibly explaining some of the inconsistent findings. Sriretnakumar et al. [141] proposed five factors in addressing mixed findings: (1) heterogeneity of the study design, (2) adherence to treatment, (3) individual study-wide limitations, (4) population stratification, and (5) additional mechanisms beyond polymorphic DNA sequence effects. Although these authors particularly focused on the field of clozapine pharmacogenetics in patients with schizophrenia, their suggestions may be valid for the overall field of PGx regarding the response to antipsychotics and adverse effects in patients with schizophrenia.

In addition to the traditional candidate gene approach, new methodologies including GWAS, whole-exome sequencing, and gene-gene interaction have been adopted in PGx research [157]. These studies have produced novel findings and keep the promise to develop PGx research, although further replication studies are needed in other ethnic populations. Moreover, collaborations with pooled samples using robust biostatistical strategies are strongly encouraged, similar to the efforts made by the PGC in the broader field of psychiatric genetics or by the Pharmacogenomics of Antipsychot ic-Induced Weight Gain Consortium [158] and the CRESTAR group for clozapine response [159] in psychiatric pharmacogenetics.

In conclusion, PGx has the potential for optimizing antipsychotic treatment through the prediction of clinical outcomes without the need for conventional “trial-and-error” approaches. However, further well-designed investigations (such as trials to create algorithms for use in clinical practice and replication studies in large, well-characterized samples) are needed.

D.J.M. is supported by the Canadian Institutes of Health Research (CIHR Operating Grant MOP 142192), the National Institutes of Health (R01MH085801), and the Centre for Addiction and Mental Health Foundation (Joanne Murphy Professorship).

The authors declare no conflict of interest.

K.Y. wrote the first draft of the manuscript, and both authors conducted the literature search and contributed to and approved the final manuscript.

1.
McGuffin P, Owen MJ, Farmer AE: Genetic basis of schizophrenia. Lancet 1995; 346: 678–682.
2.
McGrath J, Saha S, Chant D, Welham J: Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol Rev 2008; 30: 67–76.
3.
Arranz MJ, de Leon J: Pharmacogenetics and pharmacogenomics of schizophrenia: a review of last decade of research. Mol Psychiatry 2007; 12: 707–747.
4.
Müller DJ, Schulze TG, Knapp M, Held T, Krauss H, Weber T, Ahle G, Maroldt A, Alfter D, Maier W, Nöthen MM, Rietschel M: Familial occurrence of tardive dyskinesia. Acta Psychiatr Scand 2001; 104: 375–379.
5.
Pouget JG, Shams TA, Tiwari AK, Müller DJ: Pharmacogenetics and outcome with antipsychotic drugs. Dialogues Clin Neurosci 2014; 16: 555–566.
6.
Malhotra AK, Zhang J-P, Lencz T: Pharmacogenetics in psychiatry: translating research into clinical practice. Mol Psychiatry 2012; 17: 760–769.
7.
van der Weide K, van der Weide J: The Influence of the CYP3A4*22 polymorphism and CYP2D6 polymorphisms on serum concentrations of aripiprazole, haloperidol, pimozide, and risperidone in psychiatric patients. J Clin Psychopharmacol 2015; 35: 228–236.
8.
Ravyn D, Ravyn V, Lowney R, Nasrallah HA: CYP450 pharmacogenetic treatment strategies for antipsychotics: a review of the evidence. Schizophr Res 2013; 149: 1–14.
9.
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; 83: 234–242.
10.
Czerwensky F, Leucht S, Steimer W: CYP1A2*1D and *1F polymorphisms have a significant impact on olanzapine serum concentrations. Ther Drug Monit 2015; 37: 152–160.
11.
Cabaleiro T, López-Rodríguez R, Román M, Ochoa D, Novalbos J, Borobia A, Carcas A, Abad-Santos F: Pharmacogenetics of quetia­pine in healthy volunteers: association with pharmacokinetics, pharmacodynamics, and adverse effects. Int Clin Psychopharmacol 2015; 30: 82–88.
12.
Moons T, de Roo M, Claes S, Dom G: Relationship between P-glycoprotein and second-generation antipsychotics. Pharmacogenomics 2011; 12: 1193–1211.
13.
Piatkov I, Caetano D, Assur Y, Lau SL, Jones T, Boyages SC, McLean M: ABCB1 and ABCC1 single-nucleotide polymorphisms in patients treated with clozapine. Pharmgenomics Pers Med 2017; 10: 235–242.
14.
Hettige NC, de Moraes GH, Kennedy JL, De Luca V: Candidate gene analysis of pharmacodynamic targets for antipsychotic dosage. Pharmacogenomics 2016; 17: 199–208.
15.
Hettige NC, Cole CB, Khalid S, De Luca V: Polygenic risk score prediction of antipsychotic dosage in schizophrenia. Schizophr Res 2016; 170: 265–270.
16.
Koga AT, Strauss J, Zai C, Remington G, De Luca V: Genome-wide association analysis to predict optimal antipsychotic dosage in schizophrenia: a pilot study. J Neural Transm (Vienna) 2016; 123: 329–338.
17.
Hettige NC, Zai C, Hazra M, Borlido C, Kennedy JL, Strauss J, Le Foll B, Wong A, Remington G, De Luca V: Use of candidate gene markers to guide antipsychotic dosage adjustment. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54: 315–320.
18.
Beller SA, Overall JE: The Brief Psychiatric Rating Scale (BPRS) in geropsychiatric research: II. Representative profile patterns. J Gerontol 1984; 39: 194–200.
19.
Kay SR, Fiszbein A, Opler LA: The Positive and Negative Syndrome Scale (PANSS) for schizophrenia. Schizophr Bull 1987; 13: 261–276.
20.
Cacabelos R, Hashimoto R, Takeda M: Pharmacogenomics of antipsychotics efficacy for schizophrenia. Psychiatry Clin Neurosci 2011; 65: 3–19.
21.
Kaur G, Gupta D, Chavan BS, Sinhmar V, Prasad R, Tripathi A, Garg PD, Gupta R, Khurana H, Gautam S, Margoob MA, Aneja J: Identification of genetic correlates of response to risperidone: findings of a multicentric schizophrenia study from India. Asian J Psychiatr 2017; 29: 174–182.
22.
Brandl EJ, Chowdhury NI, Tiwari AK, Lett TAP, Meltzer HY, Kennedy JL, Müller DJ: Genetic variation in CYP3A43 is associated with response to antipsychotic medication. J Neural Transm (Vienna) 2015; 122: 29–34.
23.
Xu Q, Wu X, Li M, Huang H, Minica C, Yi Z, Wang G, Shen L, Xing Q, Shi Y, He L, Qin S: Association studies of genomic variants with treatment response to risperidone, clozapine, quetiapine and chlorpromazine in the Chinese Han population. Pharmacogenomics J 2016; 16: 357–365.
24.
Papazisis G, Goulas A, Sarrigiannidis A, Bargiota S, Antoniadis D, Raikos N, Basgiouraki E, Bozikas VP, Garyfallos G: ABCB1 and CYP2D6 polymorphisms and treatment response of psychotic patients in a naturalistic setting. Hum Psychopharmacol 2018; 33: e2644.
25.
Pai P, Arathil P, Kotambail A, Nair R, Gupta M, Moily NS, Kukreti R, Jain S, Rai PS, Gopinath PM, Sharma PS, Satyamoorthy K: Association of GRIN1, ABCB1, and DRD4 genes and response to antipsychotic drug treatment in schizophrenia. Psychiatr Genet 2015; 25: 135–136.
26.
Kapur S, Seeman P: Does fast dissociation from the dopamine D2 receptor explain the action of atypical antipsychotics? A new hypothesis. Am J Psychiatry 2001; 158: 360–369.
27.
Hwang R, Zai C, Tiwari A, Müller DJ, Arranz MJ, Morris AG, McKenna PJ, Munro J, Potkin SG, Lieberman JA, Meltzer HY, Kennedy JL: 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: 200–218.
28.
Hwang R, Tiwari AK, Zai CC, Felsky D, Remington E, Wallace T, Tong RP, Souza RP, Oh G, Potkin SG, Lieberman JA, Meltzer HY, Kennedy JL: Dopamine D4 and D5 receptor gene variant effects on clozapine response in schizophrenia: replication and exploration. Prog Neuropsychopharmacol Biol Psychiatry 2012; 37: 62–75.
29.
Terzić T, Kastelic M, Dolžan V, Plesničar BK: Genetic polymorphisms in dopaminergic system and treatment-resistant schizophrenia. Psychiatr Danub 2016; 28: 127–131.
30.
Huang E, Maciukiewicz M, Zai CC, Tiwari AK, Li J, Potkin SG, Liberman JA, Meltzer HY, Müller DJ, Kennedy JL: Preliminary evidence for association of genome-wide significant DRD2 schizophrenia risk variant with clozapine response. Pharmacogenomics 2016; 17: 103–109.
31.
Zhang JP, Robinson DG, Gallego JA, John M, Yu J, Addington J, Tohen M, Kane JM, Malhotra AK, Lencz T: Association of a schizophrenia risk variant at the DRD2 locus with antipsychotic treatment response in first-episode psychosis. Schizophr Bull 2015; 41: 1248–1255.
32.
Kang SG, Na KS, Lee HJ, Chee IS, Lee K, Lee J: DRD2 genotypic and haplotype variation is associated with improvements in negative symptoms after 6 weeks’ amisulpride treatment. J Clin Psychopharmacol 2015; 35: 158–162.
33.
Blasi G, Selvaggi P, Fazio L, Antonucci LA, Taurisano P, Masellis R, Romano R, Mancini M, Zhang F, Caforio G, Popolizio T, Apud J, Weinberger DR, Bertolino A: Variation in dopamine D2 and serotonin 5-HT2A receptor genes is associated with working memory processing and response to treatment with antipsychotics. Neuropsychopharmacology 2015; 40: 1600–1608.
34.
Bishop JR, Reilly JL, Harris MSH, Patel SR, Kittles R, Badner JA, Prasad KM, Nimgaonkar VL, Keshavan MS, Sweeney JA: Pharmacogenetic associations of the type-3 metabotropic glutamate receptor (GRM3) gene with working memory and clinical symptom response to antipsychotics in first-episode schizophrenia. Psychopharmacology (Berl) 2015; 232: 145–154.
35.
Schizophrenia Working Group of the Psychiatric Genomics Consortium: Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014; 511: 421–427.
36.
Zhang JP, Malhotra AK: Pharmacogenetics and antipsychotics: therapeutic efficacy and side effects prediction. Expert Opin Drug Metab Toxicol 2011; 7: 9–37.
37.
Huo R, Wei Z, Xiong Y, Jiang J, Liu Y, Yan Y, Shi J, Li W, Cui D, Xing Q, He L, Qin S: Association of dopamine receptor D1 (DRD1) polymorphisms with risperidone treatment response in Chinese schizophrenia patients. Neurosci Lett 2015; 584: 178–183.
38.
de Matos LPR, Santana CVN, Souza RP: Meta-analysis of dopamine receptor D1 rs4532 polymorphism and susceptibility to antipsychotic treatment response. Psychiatry Res 2015; 229: 586–588.
39.
Williams NM, Owen MJ: Genetic abnormalities of chromosome 22 and the development of psychosis. Curr Psychiatry Rep 2004; 6: 176–182.
40.
Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, Weinshilboum RM: Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 1996; 6: 243–250.
41.
Sun Z, Zhang Z, Mao P, Ma Y, Li W, Li J, Yang X, Ling S, Tang Y: Association between COMT gene polymorphisms, clinical symptoms, and cognitive functions in Han Chinese patients with schizophrenia. Psychiatr Genet 2018; 28: 47–54.
42.
Calabrò M, Porcelli S, Crisafulli C, Wang S-M, Lee S-J, Han C, Patkar AA, Masand PS, Albani D, Raimondi I, Forloni G, Bin S, Cristalli C, Mantovani V, Pae CU, Serretti A: Genetic variants within molecular targets of antipsychotic treatment: effects on treatment response, schizophrenia risk, and psychopathological features. J Mol Neurosci 2018; 64: 62–74.
43.
Shi Y, Li M, Song C, Xu Q, Huo R, Shen L, Xing Q, Cui D, Li W, Zhao J, He L, Qin S: Combined study of genetic and epigenetic biomarker risperidone treatment efficacy in Chinese Han schizophrenia patients. Transl Psychiatry 2017; 7:e1170.
44.
Chen CY, Yeh YW, Kuo SC, Ho PS, Liang CS, Yen CH, Lu RB, Huang SY: Catechol-O-methyltransferase gene variants may associate with negative symptom response and plasma concentrations of prolactin in schizophrenia after amisulpride treatment. Psycho neuroendocrinology 2016; 65: 67–75.
45.
Bosia M, Lorenzi C, Pirovano A, Guglielmino C, Cocchi F, Spangaro M, Bramanti P, Sme­raldi E, Cavallaro R: COMT Val158Met and 5-HT1A-R –1019 C/G polymorphisms: effects on the negative symptom response to clozapine. Pharmacogenomics 2015; 16: 35–44.
46.
Huang E, Zai CC, Lisoway A, Maciukiewicz M, Felsky D, Tiwari AK, Bishop JR, Ikeda M, Molero P, Ortuno F, Porcelli S, Samochowiec J, Mierzejewski P, Gao S, Crespo-Facorro B, Pelayo-Terán JM, Kaur H, Kukreti R, Meltzer HY Lieberman JA, Potkin SG, Müller DJ, Kennedy JL: Catechol-O-methyltransferase Val158Met polymorphism and clinical response to antipsychotic treatment in schizophrenia and schizo-affective disorder patients: a meta-analysis. Int J Neuropsychopharmacol 2016; 19:pyv132.
47.
Brandl EJ, Kennedy JL, Müller DJ: Pharmacogenetics of antipsychotics. Can J Psychiatry 2014; 59: 76–88.
48.
Tang H, McGowan OO, Reynolds GP: Polymorphisms of serotonin neurotransmission and their effects on antipsychotic drug action. Pharmacogenomics 2014; 15: 1599–1609.
49.
Takekita Y, Fabbri C, Kato M, Nonen S, Sakai S, Sunada N, Koshikawa Y, Wakeno M, Okugawa G, Kinoshita T, Serretti A: HTR1A gene polymorphisms and 5-HT1A receptor partial agonist antipsychotics efficacy in schizophrenia. J Clin Psychopharmacol 2015; 35: 220–227.
50.
Terzić T, Kastelic M, Dolžan V, Plesničar BK: Influence of 5-HT1A and 5-HTTLPR genetic variants on the schizophrenia symptoms and occurrence of treatment-resistant schizophrenia. Neuropsychiatr Dis Treat 2015; 11: 453–459.
51.
Takekita Y, Fabbri C, Kato M, Koshikawa Y, Tajika A, Kinoshita T, Serretti A: HTR1A polymorphisms and clinical efficacy of antipsychotic drug treatment in schizophrenia: a meta-analysis. Int J Neuropsychopharmacol 2016; 19:pyv125.
52.
Aghajanian GK, Marek GJ: Serotonin and hallucinogens. Neuropsychopharmacology 1999; 21(suppl): 16S–23S.
53.
Takekita Y, Fabbri C, Kato M, Nonen S, Sakai S, Sunada N, Koshikawa Y, Wakeno M, Okugawa G, Kinoshita T, Serretti A: Serotonin 7 receptor variants are not associated with response to second-generation antipsychotics in Japanese schizophrenia patients. Neuropsychobiology 2015; 72: 118–125.
54.
McGregor N, Thompson N, O’Connell KS, Emsley R, van der Merwe L, Warnich L: Modification of the association between antipsychotic treatment response and childhood adversity by MMP9 gene variants in a first-episode schizophrenia cohort. Psychiatry Res 2018; 262: 141–148.
55.
Calabrò M, Porcelli S, Crisafulli C, Wang SM, Lee SJ, Han C, Patkar AA, Masand PS, Albani D, Raimondi I, Forloni G, Bin S, Mattiaccio A, Mantovani V, Jun TY, Pae CU, Serretti A: Genetic variants within key nodes of the cascade of antipsychotic mechanisms: effects on antipsychotic response and schizophrenia psychopathology in a naturalistic treatment setting in two independent Korean and Italian samples. Adv Ther 2017; 34: 1482–1497.
56.
Kang SG, Chee IS, Chang HS, Na KS, Lee K, Lee J: Polymorphism of the SNAP25 gene is associated with symptom improvement in schizophrenic patients treated with amisulpride. Neurosci Lett 2017; 661: 46–50.
57.
Taylor DL, Tiwari AK, Lieberman JA, Potkin SG, Meltzer HY, Knight J, Remington G, Müller DJ, Kennedy JL: Pharmacogenetic analysis of functional glutamate system gene variants and clinical response to clozapine. Mol Neuropsychiatry 2017; 2: 185–197.
58.
Labad J, Martorell L, Huerta-Ramos E, Cobo J, Vilella E, Rubio-Abadal E, Garcia-Pares G, Creus M, Núñez C, Ortega L, Miquel E; RALOPSYCAT Group, Usall J: Pharmacogenetic study of the effects of raloxifene on negative symptoms of postmenopausal women with schizophrenia: a double-blind, randomized, placebo-controlled trial. Eur Neuropsychopharmacol 2016; 26: 1683–1689.
59.
Brandl EJ, Lett TA, Chowdhury NI, Tiwari AK, Bakanidze G, Meltzer HY, Potkin SG, Lieberman JA, Kennedy JL, Müller DJ: The role of the ITIH3 rs2535629 variant in antipsychotic response. Schizophr Res 2016; 176: 131–135.
60.
Porcelli S, Balzarro B, Lee SJ, Han C, Patkar AA, Pae CU, Serretti A: PDE7B, NMBR and EPM2A variants and schizophrenia: a case-control and pharmacogenetics study. Neuropsychobiology 2016; 73: 160–168.
61.
Taylor DL, Tiwari AK, Lieberman JA, Potkin SG, Meltzer HY, Knight J, Remington G, Müller DJ, Kennedy JL: Genetic association analysis of N-methyl-D-aspartate receptor subunit gene GRIN2B and clinical response to cloza­pine. Hum Psychopharmacol 2016; 31: 121–134.
62.
Jajodia A, Kaur H, Kumari K, Kanojia N, Gupta M, Baghel R, Sood M, Jain S, Chadda RK, Kukreti R: Evaluation of genetic association of neurodevelopment and neuroimmunological genes with antipsychotic treatment response in schizophrenia in Indian populations. Mol Genet Genomic Med 2016; 4: 18–27.
63.
Lee SY, Chen SL, Chang YH, Chen PS, Huang SY, Tzeng NS, Wang LJ, Lee IH, Wang TY, Chen KC, Yang YK, Hong JS, Lu RB: ALDH2 polymorphism, associated with attenuating negative symptoms in patients with schizophrenia treated with add-on dextromethorphan. J Psychiatr Res 2015; 69: 50–56.
64.
Porcelli S, Lee SJ, Han C, Patkar AA, Serretti A, Pae CU: CACNA1C gene and schizophrenia: a case-control and pharmacogenetic study. Psychiatr Genet 2015; 25: 163–167.
65.
Porcelli S, Pae CU, Han C, Lee SJ, Patkar AA, Masand PS, Balzarro B, Alberti S, De Ronchi D, Serretti A: The influence of AHI1 variants on the diagnosis and treatment outcome in schizophrenia. Int J Mol Sci 2015; 16: 2517–2529.
66.
Pouget JG, Gonçalves VF, Nurmi EL, P Laughlin C, Mallya KS, McCracken JT, Aman MG, McDougle CJ, Scahill L, Misener VL, Tiwari AK, Zai CC, Brandl EJ, Felsky D, Leung AQ, Lieberman JA, Meltzer HY, Potkin SG, Niedling C, Steimer W, Leucht S, Knight J, Müller DJ, Kennedy JL: Investigation of TSPO variants in schizophrenia and antipsychotic treatment outcomes. Pharmacogenomics 2015; 16: 5–22.
67.
Stevenson JM, Reilly JL, Harris MS, Patel SR, Weiden PJ, Prasad KM, Badner JA, Nimgaonkar VL, Keshavan MS, Sweeney JA, Bishop JR: Antipsychotic pharmacogenomics in first episode psychosis: a role for glutamate genes. 2016; 6:e739.
68.
Sacchetti E, Magri C, Minelli A, Valsecchi P, Traversa M, Calza S, Vita A, Gennarelli M: The GRM7 gene, early response to risperidone, and schizophrenia: a genome-wide association study and a confirmatory pharmacogenetic analysis. Pharmacogenomics J 2017; 17: 146–154.
69.
Kang SG, Chee IS, Lee K, Lee J: rs7968606 polymorphism of ANKS1B is associated with improvement in the PANSS general score of schizophrenia caused by amisulpride. Hum Psychopharmacol 2017; 32:e2562.
70.
Cargnin S, Massarotti A, Terrazzino S: BDNF Val66Met and clinical response to antipsychotic drugs: a systematic review and meta-analysis. Eur Psychiatry 2016; 33: 45–53.
71.
Pearson TA, Manolio TA: How to interpret a genome-wide association study. JAMA 2008; 299: 1335–1344.
72.
Lavedan C, Licamele L, Volpi S, Hamilton J, Heaton C, Mack K, Lannan R, Thompson A, Wolfgang CD, Polymeropoulos MH: 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: 804–819.
73.
Ikeda M, Tomita Y, Mouri A, Koga M, Okochi T, Yoshimura R, Yamanouchi Y, Kinoshita Y, Hashimoto R, Williams HJ, Takeda M, Nakamura J, Nabeshima T, Owen MJ, O’Donovan MC, Honda H, Arinami T, Ozaki N, Iwata N: 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: 263–269.
74.
McClay JL, Adkins DE, Åberg K, Stroup S, Perkins DO, Vladimirov VI, Lieberman JA, Sullivan PF, van den Oord EJ: Genome-wide pharmacogenomic analysis of response to treatment with antipsychotics. Mol Psychiatry 2011; 16: 76–85.
75.
Clark SL, Souza RP, Adkins DE, Åberg K, Bukszár J, McClay JL, Sullivan PF, van den Oord EJ: Genome-wide association study of patient-rated and clinician-rated global impression of severity during antipsychotic treatment. Pharmacogenet Genomics 2013; 23: 69–77.
76.
Drago A, Giegling I, Schäfer M, Hartmann AM, Konte B, Friedl M, Serretti A, Rujescu D: Genome-wide association study supports the role of the immunological system and of the neurodevelopmental processes in response to haloperidol treatment. Pharmacogenet Genomics 2014; 24: 314–319.
77.
Yu H, Yan H, Wang L, Li J, Tan L, Deng W, Chen Q, Yang G, Zhang F, Lu T, Yang J, Li K, Lv L, Tan Q, Zhang H, Xiao X, Li M, Ma X, Yang F, Li L, Wang C, Li T, Zhang D, Yue W; Chinese Antipsychotics Pharmacogenomics Consortium: Five novel loci associated with antipsychotic treatment response in patients with schizophrenia: a genome-wide association study. Lancet Psychiatry 2018; 5: 327–338.
78.
Li J, Yoshikawa A, Brennan MD, Ramsey TL, Meltzer HY: Genetic predictors of antipsychotic response to lurasidone identified in a genome wide association study and by schizophrenia risk genes. Schizophr Res 2018; 192: 194–204.
79.
Ovenden ES, Drögemöller BI, van der Merwe L, Chiliza B, Asmal L, Emsley RA, Warnich L: Fine-mapping of antipsychotic response genome-wide association studies reveals novel regulatory mechanisms. Pharmacogenomics 2017; 18: 105–120.
80.
Li Q, Wineinger NE, Fu DJ, Libiger O, Alphs L, Savitz A, Gopal S, Cohen N, Schork NJ: Genome-wide association study of paliperidone efficacy. Pharmacogenet Genomics 2017; 27: 7–18.
81.
Le Clerc S, Taing L, Fond G, Meary A, Llorca P-M, Blanc O, Beaune P, Rajagopal K, Jamain S, Tamouza R, Zagury JF, Leboyer M: A double amino-acid change in the HLA-A peptide-binding groove is associated with response to psychotropic treatment in patients with schizophrenia. 2015; 5:e608.
82.
Wang D, Fu DJ, Wu X, Shapiro A, Favis R, Savitz A, Chung H, Alphs L, Gopal S, Haas M, Cohen N, Li Q: Large-scale candidate gene study to identify genetic risk factors predictive of paliperidone treatment response in patients with schizophrenia. Pharmacogenet Genomics 2015; 25: 173–185.
83.
Drögemöller BI, Emsley R, Chiliza B, van der Merwe L, Wright GE, Daya M, Hoal E, Malhotra AK, Lencz T, Robinson DG, Zhang JP, Asmal L, Niehaus DJ, Warnich L: The identification of novel genetic variants associated with antipsychotic treatment response outcomes in first-episode schizophrenia patients. Pharmacogenet Genomics 2016; 26: 235–242.
84.
Ikeda M, Mushiroda T: The genetics of antipsychotic response in schizophrenia. Lancet Psychiatry 2018; 5: 291–292.
85.
Kato T: Whole genome/exome sequencing in mood and psychotic disorders. Psychiatry Clin Neurosci 2015; 69: 65–76.
86.
Drögemöller BI, Niehaus DJH, Chiliza B, van der Merwe L, Asmal L, Malhotra AK, Wright GE, Emsley R, Warnich L: Patterns of variation influencing antipsychotic treatment outcomes in South African first-episode schizophrenia patients. Pharmacogenomics 2014; 15: 189–199.
87.
MacNeil RR, Müller DJ: Genetics of common antipsychotic-induced adverse effects. Mol Neuropsychiatry 2016; 2: 61–78.
88.
Lett TA, Wallace TJ, Chowdhury NI, Tiwari AK, Kennedy JL, Müller DJ: Pharmacogenetics of antipsychotic-induced weight gain: review and clinical implications. Mol Psychiatry 2012; 17: 242–266.
89.
Daray FM, Rodante D, Carosella LG, Silva ME, Martínez M, Fernández Busch MV, Faccone DF, Rothlin RP, Maffía PC: –759C>T polymorphism of the HTR2C gene is associated with second generation antipsychotic-induced weight gain in female patients with schizophrenia. Pharmacopsychiatry 2017; 50: 14–18.
90.
Grădinaru RC, Andreescu NI, Nussbaum LA, Farcaş SS, Dumitraşcu V, Suciu L, Puiu M: –759C/T polymorphism of the HTR2C gene is not correlated with atypical antipsychotics-induced weight gain, among Romanian psychotic patients. Rom J Morphol Embryol 2016; 57: 1343–1349.
91.
Zhang JP, Lencz T, Zhang RX, Nitta M, Maa­yan L, John M, Robinson DG, Fleischhacker WW, Kahn RS, Ophoff RA, Kane JM, Malhotra AK, Correll CU: Pharmacogenetic associations of antipsychotic drug-related weight gain: a systematic review and meta-analysis. Schizophr Bull 2016; 42: 1418–1437.
92.
Klemettilä J-P, Kampman O, Seppälä N, Viikki M, Hämäläinen M, Moilanen E, Mononen N, Lehtimäki T, Leinonen E: Association study of the HTR2C, leptin and adiponectin genes and serum marker analyses in clozapine treated long-term patients with schizophrenia. Eur Psychiatry 2015; 30: 296–302.
93.
Rico-Gomis JM, Palazón-Bru A, Triano-García I, Mahecha-García LF, García-Monsalve A, Navarro-Ruiz A, Villagordo-Peñalver B, Jiménez-Abril J, Martínez-Hortelano A, Gil-Guillén VF: Association between the HTR2C rs1414334 C/G gene polymorphism and the development of the metabolic syndrome in patients treated with atypical antipsychotics. PeerJ 2016; 4:e2163.
94.
Zai CC, Tiwari AK, Chowdhury NI, Brandl EJ, Shaikh SA, Freeman N, Lieberman JA, Meltzer HY, Kennedy JL, Müller DJ: Association study of serotonin 3 receptor subunit gene variants in antipsychotic-induced weight gain. Neuropsychobiology 2016; 74: 169–175.
95.
Lebrun B, Bariohay B, Moyse E, Jean A: Brain-derived neurotrophic factor (BDNF) and food intake regulation: a minireview. Auton Neurosci 2006; 126–127: 30–38.
96.
Zhang XY, Zhou DF, Wu GY, Cao LY, Tan YL, Haile CN, Li J, Lu L, Kosten TA, Kosten TR: BDNF levels and genotype are associated with antipsychotic-induced weight gain in patients with chronic schizophrenia. Neuropsychopharmacology 2008; 33: 2200–2205.
97.
Tsai A, Liou YJ, Hong CJ, Wu CL, Tsai SJ, Bai YM: Association study of brain-derived neurotrophic factor gene polymorphisms and body weight change in schizophrenic patients under long-term atypical antipsychotic treatment. Neuromolecular Med 2011; 13: 328–333.
98.
Zai GC, Zai CC, Chowdhury NI, Tiwari AK, Souza RP, Lieberman JA, Meltzer HY, Potkin SG, Müller DJ, Kennedy JL: The role of brain-derived neurotrophic factor (BDNF) gene variants in antipsychotic response and antipsychotic-induced weight gain. Prog Neuropsychopharmacol Biol Psychiatry 2012; 39: 96–101.
99.
Bonaccorso S, Sodhi M, Li J, Bobo WV, Chen Y, Tumuklu M, Theleritis C, Jayathilake K, Meltzer HY: The brain-derived neurotrophic factor (BDNF) Val66Met polymorphism is associated with increased body mass index and insulin resistance measures in bipolar disorder and schizophrenia. Bipolar Disord 2015; 17: 528–535.
100.
Fang H, Zhen YF, Liu XY, Xu G, Soares JC, Zhao J, Zhang XY: Association of the BDNF Val66Met polymorphism with BMI in chronic schizophrenic patients and healthy controls. Int Clin Psychopharmacol 2016; 31: 353–357.
101.
Ferno J, Skrede S, Vik-Mo AO, Jassim G, Le Hellard S, Steen VM: Lipogenic effects of psychotropic drugs: focus on the SREBP system. Front Biosci (Landmark Ed) 2011; 16: 49–60.
102.
Le Hellard S, Theisen FM, Haberhausen M, Raeder MB, Fernø J, Gebhardt S, Hinney A, Remschmidt H, Krieg JC, Mehler-Wex C, Nöthen MM, Hebebrand J, Steen VM: 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: 308–317.
103.
Yang L, Chen J, Liu D, Yu S, Cong E, Li Y, Wu H, Yue Y, Zuo S, Wang Y, Liang S, Shi Y, Shi S, Xu Y: Association between SREBF2 gene polymorphisms and metabolic syndrome in clozapine-treated patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56: 136–141.
104.
Yang L, Chen J, Li Y, Wang Y, Liang S, Shi Y, Shi S, Xu Y: Association between SCAP and SREBF1 gene polymorphisms and metabolic syndrome in schizophrenia patients treated with atypical antipsychotics. World J Biol Psychiatry 2016; 17: 467–474.
105.
Koskinen S, Kampman O, Solismaa A, Lyytikäinen L-P, Seppälä N, Viikki M, Hä­mäläinen M, Moilanen E, Mononen N, Lehtimäki T, Leinonen E: INSIG2 polymorphism and weight gain, dyslipidemia and serum adiponectin in Finnish patients with schizophrenia treated with clozapine. Pharmacogenomics 2016; 17: 1987–1997.
106.
Bournat JC, Brown CW: Mitochondrial dysfunction in obesity. Curr Opin Endocrinol Diabetes Obes 2010; 17: 446–452.
107.
Gonçalves VF, Zai CC, Tiwari AK, Brandl EJ, Derkach A, Meltzer HY, Lieberman JA, Müller DJ, Sun L, Kennedy JL: A hypothesis-driven association study of 28 nuclear-encoded mitochondrial genes with antipsychotic-induced weight gain in schizophrenia. Neuropsychopharmacology 2014; 39: 1347–1354.
108.
Giannaccini G, Betti L, Palego L, Pirone A, Schmid L, Lanza M, Fabbrini L, Pelosini C, Maffei M, Santini F, Pinchera A, Lucacchini A: Serotonin transporter (SERT) and translocator protein (TSPO) expression in the obese ob/ob mouse. BMC Neurosci 2011; 12: 18.
109.
Mittal K, Gonçalves VF, Harripaul R, Cuperfain AB, Rollins B, Tiwari AK, Zai CC, Maciukiewicz M, Müller DJ, Vawter MP, Kennedy JL: A comprehensive analysis of mitochondrial genes variants and their association with antipsychotic-induced weight gain. Schizophr Res 2017; 187: 67–73.
110.
Brandl EJ, Tiwari AK, Zai CC, Nurmi EL, Chowdhury NI, Arenovich T, Sanches M, Gonçalves VF, Shen JJ, Lieberman JA, Meltzer HY, Kennedy JL, Müller DJ: Genome-wide association study on antipsychotic- induced weight gain in the CATIE sample. Pharmacogenomics J 2016; 16: 352–356.
111.
Yu H, Wang L, Lv L, Ma C, Du B, Lu T, Jin C, Yan H, Yang Y, Li W, Ruan Y, Zhang H, Zhang H, Mi W, Mowry B, Ma W, Li K, Zhang D, Yue W: Genome-wide association study suggested the PTPRD polymorphisms were associated with weight gain effects of atypical antipsychotic medications. Schizophr Bull 2016; 42: 814–823.
112.
Lanning RK, Zai CC, Müller DJ: Pharmacogenetics of tardive dyskinesia: an updated review of the literature. Pharmacogenomics 2016; 17: 1339–1351.
113.
Kang SG, Lee YJ, Park YM, Kim L, Lee HJ: Haplotype association of the MAP2K5 gene with antipsychotics-induced symptoms of restless legs syndrome among patients with schizophrenia. Psychiatry Investig 2018; 15: 84–89.
114.
Zai CC, Tiwari AK, Chowdhury NI, Yilmaz Z, De Luca V, Müller DJ, Potkin SG, Lieberman JA, Meltzer HY, Voineskos AN, Remington G, Kennedy JL: Genetic study of neuregulin 1 and receptor tyrosine-protein kinase erbB-4 in tardive dyskinesia. World J Biol Psychiatry 2017, Epub ahead of print.
115.
Lanning R, Lett TA, Tiwari AK, Brandl EJ, De Luca V, Voineskos AN, Potkin SG, Lieberman JA, Meltzer HY, Müller DJ, Remington G, Kennedy JL, Zai CC: Association study between the neurexin-1 gene and tardive dyskinesia. Hum Psychopharmacol 2017; 32:e2568.
116.
Sychev DA, Burashnikova IS, Kazakov RE: 1846G>A polymorphism of CYP2D6 gene and extrapyramidal side effects during antipsychotic therapy among Russians and Tatars: a pilot study. Drug Metab Pers Ther 2016; 31: 205–212.
117.
Mas S, Gassó P, Lafuente A, Bioque M, Lobo A, Gonzàlez-Pinto A, Olmeda MS, Corripio I, Llerena A, Cabrera B, Saiz-Ruiz J, Bernardo M; PEPs GROUP: Pharmacogenetic study of antipsychotic induced acute extrapyramidal symptoms in a first episode psychosis cohort: role of dopamine, serotonin and glutamate candidate genes. Pharmacogenomics J 2016; 16: 439–445.
118.
Ivanova SA, Loonen AJ, Bakker PR, Freidin MB, Ter Woerds NJ, Al Hadithy AF, Semke AV, Fedorenko OY, Brouwers JR, Bokhan NA, van Os J, van Harten PN, Wilffert B: Likelihood of mechanistic roles for dopaminergic, serotonergic and glutamatergic receptors in tardive dyskinesia: a comparison of genetic variants in two independent patient populations. SAGE Open Med 2016; 4: 2050312116643673.
119.
Ivanova SA, Filipenko ML, Vyalova NM, Voronina EN, Pozhidaev IV, Osmanova DZ, Ivanov MV, Fedorenko OY, Semke AV, Bokhan NA: CYP1A2 and CYP2D6 gene polymorphisms in schizophrenic patients with neuroleptic drug-induced side effects. Bull Exp Biol Med 2016; 160: 687–690.
120.
Kang SG, Lee HJ, Lee SH, Kim L: MEIS1, a promising candidate gene, is not associated with the core symptoms of antipsychotic-induced restless legs syndrome in Korean schizophrenia patients. Psychiatry Investig 2015; 12: 263–267.
121.
Mas S, Gassó P, Parellada E, Bernardo M, Lafuente A: Network analysis of gene expression in peripheral blood identifies mTOR and NF-κB pathways involved in antipsychotic-induced extrapyramidal symptoms. Pharmacogenomics J 2015; 15: 452–460.
122.
Ivanova SA, Toshchakova VA, Filipenko ML, Fedorenko OY, Boyarko EG, Boiko AS, Semke AV, Bokhan NA, Aftanas LI, Loonen AJ: Cytochrome P450 1A2 co-determines neuroleptic load and may diminish tardive dyskinesia by increased inducibility. World J Biol Psychiatry 2015; 16: 200–205.
123.
Mas S, Gassó P, Ritter MA, Malagelada C, Bernardo M, Lafuente A: Pharmacogenetic predictor of extrapyramidal symptoms induced by antipsychotics: multilocus interaction in the mTOR pathway. Eur Neuropsychopharmacol 2015; 25: 51–59.
124.
Hui L, Han M, Yin GZ, Zhang Y, Huang XF, Qian ZK, Gu WG, Gu XC, Zhu XM, Soares JC, Ning Y, Zheng Y, Du XD, Zhang XY: Association between DBH 19bp insertion/deletion polymorphism and cognition in schizophrenia with and without tardive dyskinesia. Schizophr Res 2017; 182: 104–109.
125.
Patsopoulos NA, Ntzani EE, Zintzaras E, Ioannidis JP: CYP2D6 polymorphisms and the risk of tardive dyskinesia in schizophrenia: a meta-analysis. Pharmacogenet Genomics 2005; 15: 151–158.
126.
Koola MM, Tsapakis EM, Wright P, Smith S, Kerwin Rip RW, Nugent KL, Aitchison KJ: Association of tardive dyskinesia with variation in CYP2D6: is there a role for active metabolites? J Psychopharmacol 2014; 28: 665–670.
127.
Zai CC, De Luca V, Hwang RW, Voineskos A, Müller DJ, Remington G, Kennedy JL: Meta-analysis of two dopamine D2 receptor gene polymorphisms with tardive dyskinesia in schizophrenia patients. Mol Psychiatry 2007; 12: 794–795.
128.
Tsai HT, North KE, West SL, Poole C: The DRD3 rs6280 polymorphism and prevalence of tardive dyskinesia: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2010; 153B: 57–66.
129.
Lerer B, Segman RH, Tan E-C, Basile VS, Cavallaro R, Aschauer HN, Strous R, Chong SA, Heresco-Levy U, Verga M, Scharfetter J, Meltzer HY, Kennedy JL, Macciardi F: Combined analysis of 635 patients confirms an age-related association of the serotonin 2A receptor gene with tardive dyskinesia and specificity for the non-orofacial subtype. Int J Neuropsychopharmacol 2005; 8: 411–425.
130.
Tsai HT, Caroff SN, Miller DD, McEvoy J, Lieberman JA, North KE, Stroup TS, Sullivan PF: A candidate gene study of tardive dyskinesia in the CATIE schizophrenia trial. Am J Med Genet B Neuropsychiatr Genet 2010; 153B: 336–340.
131.
Zai CC, Tiwari AK, Mazzoco M, De Luca V, Müller DJ, Shaikh SA, Lohoff FW, Freeman N, Voineskos AN, Potkin SG, Lieberman JA, Meltzer HY, Remington G, Kennedy JL: Association study of the vesicular monoamine transporter gene SLC18A2 with tardive dyskinesia. J Psychiatr Res 2013; 47: 1760–1765.
132.
Factor SA, Remington G, Comella CL, Correll CU, Burke J, Jimenez R, Liang GS, O’Brien CF: The effects of valbenazine in participants with tardive dyskinesia: results of the 1-year KINECT 3 extension study. J Clin Psychiatry 2017; 78: 1344–1350.
133.
Hauser RA, Factor SA, Marder SR, Knese­vich MA, Ramirez PM, Jimenez R, Burke J, Liang GS, O’Brien CF: KINECT 3: a phase 3 randomized, double-blind, placebo-controlled trial of valbenazine for tardive dyskinesia. Am J Psychiatry 2017; 174: 476–484.
134.
Hahn CG, Wang HY, Cho DS, Talbot K, Gur RE, Berrettini WH, Bakshi K, Kamins J, Borgmann-Winter KE, Siegel SJ, Gallop RJ, Arnold SE: Altered neuregulin 1-erbB4 signaling contributes to NMDA receptor hypofunction in schizophrenia. Nat Med 2006; 12: 824–828.
135.
Kato T, Abe Y, Sotoyama H, Kakita A, Ko­minami R, Hirokawa S, Ozaki M, Takahashi H, Nawa H: Transient exposure of neonatal mice to neuregulin-1 results in hyperdopaminergic states in adulthood: implication in neurodevelopmental hypothesis for schizophrenia. Mol Psychiatry 2011; 16: 307–320.
136.
Peuskens J, Pani L, Detraux J, De Hert M: The effects of novel and newly approved antipsychotics on serum prolactin levels: a comprehensive review. CNS Drugs 2014; 28: 421–453.
137.
Fitzgerald P, Dinan TG: Prolactin and dopamine: what is the connection? A review article. J Psychopharmacol 2008; 22: 12–19.
138.
Miura I, Zhang JP, Hagi K, Lencz T, Kane JM, Yabe H, Malhotra AK, Correll CU: Variants in the DRD2 locus and antipsychotic-related prolactin levels: a meta-analysis. Psychoneuroendocrinology 2016; 72: 1–10.
139.
Ivanova SA, Osmanova DZ, Boiko AS, Pozhidaev IV, Freidin MB, Fedorenko OY, Semke AV, Bokhan NA, Kornetova EG, Rakhmazova LD, Wilffert B, Loonen AJ: Prolactin gene polymorphism (–1149 G/T) is associated with hyperprolactinemia in patients with schizophrenia treated with antipsychotics. Schizophr Res 2017; 182: 110–114.
140.
Ivanova SA, Osmanova DZ, Freidin MB, Fedorenko OY, Boiko AS, Pozhidaev IV, Semke AV, Bokhan NA, Agarkov AA, Wilf­fert B: Identification of 5-hydroxytryptamine receptor gene polymorphisms modulating hyperprolactinaemia in antipsychotic drug-treated patients with schizophrenia. World J Biol Psychiatry 2017; 18: 239–246.
141.
Sriretnakumar V, Huang E, Müller DJ: Pharmacogenetics of clozapine treatment response and side-effects in schizophrenia: an update. Expert Opin Drug Metab Toxicol 2015; 11: 1709–1731.
142.
Dettling M, Cascorbi I, Opgen-Rhein C, Schaub R: Clozapine-induced agranulocytosis in schizophrenic Caucasians: confirming clues for associations with human leukocyte class I and II antigens. Pharmacogenomics J 2007; 7: 325–332.
143.
Goldstein JI, Jarskog LF, Hilliard C, Alfirevic A, Duncan L, Fourches D, Huang H, Lek M, Neale BM, Ripke S, Shianna K, Szatkiewicz JP, Tropsha A, van den Oord EJ, Cascorbi I, Dettling M, Gazit E, Goff DC, Holden AL, Kelly DL, Malhotra AK, Nielsen J, Pirmohamed M, Rujescu D, Werge T, Levy DL, Josiassen RC, Kennedy JL, Lieberman JA, Daly MJ, Sullivan PF: Clozapine-induced agranulocytosis is associated with rare HLA-DQB1 and HLA-B alleles. Nat Commun 2014; 5: 4757.
144.
Saito T, Ikeda M, Mushiroda T, Ozeki T, Kondo K, Shimasaki A, Kawase K, Hashimoto S, Yamamori H, Yasuda Y, Fujimoto M, Ohi K, Takeda M, Kamatani Y, Numata S, Ohmori T, Ueno S, Makinodan M, Nishihata Y, Kubota M, Kimura T, Kanahara N, Hashimoto N, Fujita K, Nemoto K, Fukao T, Suwa T, Noda T, Yada Y, Takaki M, Kida N, Otsuru T, Murakami M, Takahashi A, Kubo M, Hashimoto R, Iwata N: Pharmacogenomic study of clozapine-induced agranulocytosis/granulocytopenia in a Japanese population. Biol Psychiatry 2016; 80: 636–642.
145.
Legge SE, Hamshere ML, Ripke S, Pardinas AF, Goldstein JI, Rees E, Richards AL, Leonenko G, Jorskog LF; Clozapine-Induced Agranulocytosis Consortium, Chambert KD, Collier DA, Genovese G, Giegling I, Holmans P, Jonasdottir A, Kirov G, McCarroll SA, MacCabe JH, Mantripragada K, Moran JL, Neale BM, Stefansson H, Rujescu D, Daly MJ, Sullivan PF, Owen MJ, O’Donovan MC, Walters JTR: Genome-wide common and rare variant analysis provides novel insights into clozapine-associated neutropenia. Mol Psychiatry 2017; 22: 1502–1508.
146.
Athanasiou MC, Dettling M, Cascorbi I, Mosyagin I, Salisbury BA, Pierz KA, Zou W, Whalen H, Malhotra AK, Lencz T, Gerson SL, Kane JM, Reed CR: Candidate gene analysis identifies a polymorphism in HLA-DQB1 associated with clozapine-induced agranulocytosis. J Clin Psychiatry 2011; 72: 458–463.
147.
Changasi AH, Shams TA, Pouget JG, Müller DJ: Genetics of antipsychotic drug outcome and implications for the clinician: into the limelight. Transl Dev Psychiatry 2014; 2: 24663.
148.
FDA: Table of Pharmacogenomic Biomarkers in Drug Labeling. https://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm (accessed April 10, 2018).
149.
PharmGKB. https://www.pharmgkb.org (accessed April 10, 2018).
150.
CPIC: Genes-Drugs. https://cpicpgx.org/genes-drugs/ (accessed April 10, 2018).
151.
PGRN. http://www.pgrn.org (accessed April 10, 2018).
152.
DPWG: Dosing Guidelines. https://www.pharmgkb.org/view/dosing-guidelines.do?source=DPWG# (accessed April 10, 2018).
153.
Bousman CA, Hopwood M: Commercial pharmacogenetic-based decision-support tools in psychiatry. Lancet Psychiatry 2016; 3: 585–590.
154.
Winner JG, Carhart JM, Altar CA, Allen JD, Dechairo BM: A prospective, randomized, double-blind study assessing the clinical impact of integrated pharmacogenomic testing for major depressive disorder. Discov Med 2013; 16: 219–227.
155.
Pérez V, Salavert A, Espadaler J, Tuson M, Saiz-Ruiz J, Sáez-Navarro C, Bobes J, Baca-García E, Vieta E, Olivares JM, Rodriguez-Jimenez R, Villagrán JM, Gascón J, Cañete-Crespillo J, Solé M, Saiz PA, Ibáñez Á, de Diego-Adeliño J; AB-GEN Collaborative Group, Menchón JM: Efficacy of prospective pharmacogenetic testing in the treatment of major depressive disorder: results of a randomized, double-blind clinical trial. BMC Psychiatry 2017; 17: 250.
156.
Bradley P, Shiekh M, Mehra V, Vrbicky K, Layle S, Olson MC, Maciel A, Cullors A, Graces JA, Lukowiak AA: Improved efficacy with targeted pharmacogenetic-guided treatment of patients with depression and anxiety: a randomized clinical trial demonstrating clinical utility. J Psychiatr Res 2018; 96: 100–107.
157.
Zhang JP, Malhotra AK: Recent progress in pharmacogenomics of antipsychotic drug response. Curr Psychiatry Rep 2018; 20: 24.
158.
PGRN: Pharmacogenomics of antipsychotic-induced weight gain and metabolic syndrome. http://www.pgrn.org/aiwg.html (accessed April 10, 2018).
159.
Spencer BW, Prainsack B, Rujescu D, Giegling I, Collier DA, Gaughran F, MacCabe JH, Barr CL, Sigurdsson E, Stovring H, Malhotra AK, Curran SR; CRESTAR Consortium: Opening Pandora’s box in the UK: a hypothetical pharmacogenetic test for clozapine. Pharmacogenomics 2013; 14: 1907–1914.
160.
Li S, Xu C, Tian Y, Wang X, Jiang R, Zhang M, Wang L, Yang G, Gao Y, Song C, He Y, Zhang Y, Li J, Li WD: TOX and ADIPOQ gene polymorphisms are associated with antipsychotic-induced weight gain in Han Chinese. Sci Rep 2017; 7: 45203.
161.
Klemettilä JP, Kampman O, Solismaa A, Lyytikäinen LP, Seppälä N, Viikki M, Hämäläinen M, Moilanen E, Mononen N, Lehtimäki T, Leinonen E: Association study of arcuate nucleus neuropeptide Y neuron receptor gene variation and serum NPY levels in clozapine treated patients with schizophrenia. Eur Psychiatry 2017; 40: 13–19.
162.
Zhang C, Zhang Y, Cai J, Chen M, Song L: Complement 3 and metabolic syndrome induced by clozapine: a cross-sectional study and retrospective cohort analysis. Pharmacogenomics J 2017; 17: 92–97.
163.
Tiwari AK, Zhang D, Pouget JG, Zai CC, Chowdhury NI, Brandl EJ, Qin L, Freeman N, Lieberman JA, Meltzer HY, Kennedy JL, Müller DJ: Impact of histamine receptors H1 and H3 polymorphisms on antipsychotic-induced weight gain. World J Biol Psychiatry 2016, Epub ahead of print.
164.
Ryu S, Huh IS, Cho EY, Cho Y, Park T, Yoon SC, Joo YH, Hong KS: Association study of 60 candidate genes with antipsychotic-induced weight gain in schizophrenia patients. Pharmacopsychiatry 2016; 49: 51–56.
165.
Tiwari AK, Brandl EJ, Zai CC, Gonçalves VF, Chowdhury NI, Freeman N, Lieberman JA, Meltzer HY, Kennedy JL, Müller DJ: Association of orexin receptor polymorphisms with antipsychotic-induced weight gain. World J Biol Psychiatry 2016; 17: 221–229.
166.
Wang F, Mi W, Ma W, Ma C, Yang Y, Zhang H, Du B, Li K, Liu C, Wang L, Lu T, Zhang H, Lv L, Zhang D, Yue W: A pharmaco­genomic study revealed an association between SLC6A4 and risperidone-induced weight gain in Chinese Han population. Pharmacogenomics 2015; 16: 1943–1949.
167.
Chen PY, Lu ML, Huang MC, Kao CF, Kuo PH, Chiu CC, Lin SK, Chen CH: The relationships of obesity-related genetic variants with metabolic profiles and response to metformin in clozapine-treated patients with schizophrenia. J Clin Psychopharmacol 2015; 35: 574–578.
168.
Dong L, Yan H, Huang X, Hu X, Yang Y, Ma C, Du B, Lu T, Jin C, Wang L, Yu H, Dong Z, Li W, Ruan Y, Zhang H, Zhang H, Mi W, Ma W, Li K, Lv L, Zhang D, Yue W: A2BP1 gene polymorphisms association with olanzapine-induced weight gain. Pharmacol Res 2015; 99: 155–161.
169.
Fonseka TM, Tiwari AK, Gonçalves VF, Lieberman JA, Meltzer HY, Goldstein BI, Kennedy JL, Kennedy SH, Müller DJ: The role of genetic variation across IL-1β, IL-2, IL-6, and BDNF in antipsychotic-induced weight gain. World J Biol Psychiatry 2015; 16: 45–56.
170.
Zai CC, Tiwari AK, Chowdhury NI, Brandl EJ, Shaikh SA, Freeman N, Lieberman JA, Meltzer HY, Müller DJ, Kennedy JL: Association study of GABAA α2 receptor subunit gene variants in antipsychotic-associated weight gain. J Clin Psychopharmacol 2015; 35: 7–12.
171.
Ono S, Suzuki Y, Fukui N, Sawamura K, Sugai T, Watanabe J, Tsuneyama N, Someya T: GIPR gene polymorphism and weight gain in patients with schizophrenia treated with olanzapine. J Neuropsychiatry Clin Neurosci 2015; 27: 162–164.