Summary
Existing psychotropic medications for the treatment of mental illnesses, including antidepressants, mood stabilizers, and antipsychotics, are clinically sub-optimal. They are effective in only a subset of patients or produce partial responses, and they are often associated with debilitating side effects that discourage adherence. There is growing enthusiasm in the promise of pharmacogenetics to personalize the use of these treatments to maximize their efficacy and tolerability. However, there is still a long way to go before this promise becomes a reality. In this article, we review the progress that has been made in research towards understanding how genetic factors influence psychotropic drug responses and the challenges that lie ahead in translating the research findings into clinical practices that yield tangible benefits for patients with mental illnesses.
Keywords: Pharmacogenetics, pharmacogenomics, antidepressants, antipsychotics, mood stabilizers, genome-wide association study, efficacy, side-effects
Introduction
The primary means of treating mental illnesses is with an arsenal of psychotropic medications, including antidepressants, antipsychotics and mood stabilizers. Despite progress over the past several decades in developing new classes of such medications that are presumably safer and more effective, the ability to treat mental illnesses remains clinically sub-optimal. These medications are effective in only a subset of patients or produce partial responses, and they are often associated with debilitating side effects that discourage adherence (1).
The results from the latest and largest treatment effectiveness trials of psychotropic medications sponsored by the National Institute of Mental Health (NIMH) reinforce the notion that there is still a long way to go in the war against mental illnesses. In the recently completed Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study in which patients with non-psychotic major depression were followed for up to six years through a sequence of alternative treatment regimens, only 37% achieved remission on first line therapy with a selective serotonin reuptake inhibitor (SSRI) while another 16.3% withdrew completely from treatment due to drug intolerance (2). Even worse, in the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) in which patients with schizophrenia were treated with a menu of leading antipsychotics under conditions meant to reflect realistic clinical practice and followed for up to 18 months, over 74% eventually discontinued their study medication either due to lack of efficacy or tolerability (3). Similarly, in the Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD) trial in which patients with bipolar disorder were enrolled and provided treatments with mood stabilizers and antidepressants that followed expert consensus guidelines, up to 75% experienced symptom relapse sometime over the course of follow-up (4). These figures are sobering and suggest considerable room for improvement in psychiatric treatments.
With the advent of the genomics revolution, there has been growing excitement that pharmacogenetics can pave the way to improved treatments. The term pharmacogenetics was first coined nearly a half century ago when it was recognized that inherited variation can influence responses to medications (5). Since then, an ever growing number of pharmacogenetic traits have been studied. Earlier studies focused on variation in candidate genes or gene systems believed to influence the absorption, distribution, or clearance of drugs (i.e., pharmacokinetics) or mediate their mechanisms of actions via interactions with receptors and/or transporters and downstream second messengers (i.e., pharmacodynamics). However, the completion of the Human Genome Project and the emergence of new tools to interrogate the entire genome on an unprecedented scale have accelerated interest in studying the relevance of variation across the entire genome. These advances have spawned a new term, pharmacogenomics. Pharmacogenomics and pharmacogenetics are used interchangeably, and in both cases refer to the study of how genetic variation influences response to drug treatments in terms of efficacy (i.e., efficacy pharmacogenetics) or tolerability (i.e., safety pharmacogenetics).
The hope is that through pharmacogenetics we will be able to discover genetic profiles that can be determined by simple genetic tests and that predict how patients will respond to different psychotropic treatments before they are initiated. The benefits are obvious as it would allow physicians to tailor medications to their patients in such a way that maximizes their efficacy and tolerability, thus ushering in an era of “personalized medicine.” In addition, by elucidating the pathways by which drugs act to treat illness and provoke unwanted side effects, pharmacogenetics may inform the rational development of new treatments that are ever more safe and efficacious. Thus, the promise of pharmacogenetics in psychiatry is that it will lead to the smarter use of our existing weapons and, in turn, the development of even smarter weapons to combat mental illness.
However, there is clearly still a long way to go before the promise becomes a reality. In this article, we review the progress that has been made in research towards understanding how genetic factors influence psychotropic drug response and the challenges that lie ahead in translating the research findings into clinical practice that yield tangible benefits for patients. We discuss antidepressants, mood stabilizers, and antipsychotics in turn, and for each we review the pharmacogenetic studies that have been carried out on them, including candidate gene studies of pharmakcokinetic or pharmacodynamic factors and genome-wide studies. We then examine the few examples of pharmacogenetic biomarkers and corresponding tests that have begun to penetrate into clinical practice in psychiatry and assess their impact on patient care. Finally, we conclude with a discussion of the challenges to advancing the goals of personalized care in psychiatry.
Antidepressants
The monoamine oxidase inhibitors and tricyclics were the first antidepressants introduced back in the 1950’s. They heralded a major breakthrough in the treatment of depression, but their wider use was limited by partial efficacy and significant concerns about side effects, like sedation (6). In the late 1980’s a new class of antidepressants became available known as the selective serotonin reuptake inhibitors (SSRIs). Because of their improved efficacy and tolerability, the SSRIs quickly gained popularity and are now the most widely used antidepressants (7). They are among the first line choices for the treatment of depression (8, 9), but they are still only effective in a subset of patients (10) and are associated with certain common side effects as well, such as weight gain, insomnia and sexual dysfunction that are leading causes of non-adherence (11). More recently, a number of other new classes of antidepressants have been introduced with mixed pharmacodynamic profiles. These include serotonin-norepinephrine reuptake inhibitors (SNRIs), the dopamine-norepinephrine reuptake inihibitors (DNRIs), serotonin modulators, norepinephrine-serotonin modulators, and selective norepinephrine reuptake inhibitors (NRIs).
Overview of Pharmacogenetic Studies
At least 119 pharmacogenetic studies of candidate genes and the efficacy or tolerability of treatment with antidepressants have been reported in the literature (12–130). The overwhelming majority of these have studied SSRIs, although several have examined SNRIs or other older agents such as the monoamine oxidase inhibitors or tricyclics. Approximately 65% of the studies have been on Caucasian samples, while the remaining studies have mostly studied Asian samples, including Chinese, Japanese or Korean. The samples sizes have been generally small with a median of less than 150 subjects, and each of the studies have investigated no more than a handful of specific candidate gene polymorphisms at a time. The exceptions to this include several reports (71, 87, 90, 97, 108, 115, 123, 124, 127) from STAR*D in which pharmacogenetic studies with over 1,900 subjects who provided a DNA sample have been carried out to examine the association between a number of different candidate genes and treatment response to citalopram, including efficacy, treatment emergent suicide ideation and sexual dysfunction. In addition to these studies, three genome-wide association studies of antidepressant treatment response have recently been reported in the literature (131–133). (See Table 1 for a description of candidate genes studies from STAR*D and genome-wide association studies of treatment responses to antidepressants.)
Table 1.
Author (Year) | Drug | Gene(s) | Outcome | Sample Size | Key Finding(s) |
---|---|---|---|---|---|
Candidate gene studies from STAR*D | |||||
McMahon (2006) | Citalopram | 68 genes | Efficacy | 1,953 (split sample design) | Association with 5-HTR2A |
Hu (2007) | Citalopram | 5-HTT | Efficacy Remission Tolerance Adverse effects |
1,775 | Association with adverse effect burden |
Kraft (2007) | Citalopram | 5-HTT | Efficacy | 1,914 | No association |
Paddock (2007) | Citalopram | 68 genes | Efficacy | 1,816 (full sample) | Association with GRIK4 |
Peters (2008) | Citalopram | CYP2D6, CYP2C19, CYP3A4, CYP3A5, ABCB1 | Efficacy Tolerance |
1,953 (split sample design) | No associations |
Lekman (2008) | Citalopram | FKBP5 | Efficacy | 1,809 | Association with response |
Cabanero (2009) | Citalopram | PDE11A, PDE1A, PDE9A | Efficacy | 1,914 | No association |
Domschke (2009) | Citalopram | BDNF | Efficacy | 1,914 | No association |
Perlis (2009) | Citalopram | 68 genes | Sexual Dysfunction | 1,473 | Associations with GRIA3, GRIK2, GRIA1, GRIN3A |
Genome-wide association studies | |||||
Laje (2009) | Citalopram | Illumina Human-1 Bead Chip |
Treatment emergent suicidal ideation | 180 | Association with PAPLN |
Garriock (2009) | Citalopram | Affymetrix 500K and 5.0 | Efficacy | 1,491 | Suggestive associations with UBE3C, BMP7, RORA |
Ising (2009) | Antidepressants | Illumina Human-1 and HumanHap300 Bead Chips |
Efficacy | 339 | Association with a multifactorial SNP score |
Pharmacokinetic Studies
Multiple pharmacogenetic studies have been carried out on the relationship between genes coding for CYP450 enzymes, which are involved in the metabolism of many different xenobiotics, and antidepressant treatment responses. CYP2D6 and CYP2C19, which together with CYP2C9 metabolize virtually all SSRIs (134), have received the greatest attention.
CYP2D6 is constitutively expressed in the liver and is responsible for metabolizing approximately 25% of drugs known to be metabolized by CYP450 enzymes (135). It is the key enzyme in the metabolic pathway of many antidepressants (136). Because CYP2D6 is not inducible (137), functional genetic variation and “environmental” inhibitors of the enzyme are the only factors that can modify its activity, making it a good candidate for pharmacogenetic testing (138). Over 90 genetic variants have been identified in CYP2D6 (139). These variants have been functionally classified into four phenotypic groups based on their effects on enzyme activity: poor metabolizers (PMs), intermediate metabolizers (IMs), extensive metabolizers (EMs), and ultrarapid metabolizers (UMs). There are considerable differences in the frequencies of these classes across racial and ethnic groups (138). CYP2C19 is also polymorphic with two main phenotypic groups: EMs and the more rare PMs (138).
The contention is that CYP related PMs are at an increased risk of side effects from antidepressants, while UMs, and to a lesser extent IMs, are less likely to show positive response to treatment (138). A commercially available pharmacogenetic test has been clinically approved to test for the CYP2D6 and CYP2C19 genetic variants based on this characterization (140). However, at least 13 different studies have examined the relationship between variation in these genes and treatment response to antidepressants (12, 22, 24, 39, 40, 48, 54, 55, 59, 75, 82, 83, 108), and the results have been decidedly mixed. The largest study from STAR*D included 1,877 genotyped subjects and found no association between variation in CYP2D6 or CYP2C19 and either efficacy or tolerability (108). Moreover, a recent review of existing studies found little overall evidence of an association between these two CYP450 genes and antidepressant response, calling into question the clinical utility of testing for these variants (141). The commercially available pharmacogenetic test and its potential use in clinic practice are discussed in further detail below.
Pharmacodynamic Studies
The therapeutic action of antidepressants is thought to be mediated at least partially through their effects on monoaminergic transmission and primarily the serotonergic pathway. Consequently, genes in the serotonergic pathway have been of great interest in pharmacogenetic studies of antidepressants. 5HTT, which codes for the pre-synaptic membrane bound serotonin transporter protein that is the target of SSRIs, is by far the most widely studied of these. It has a variable length repeat polymorphism in the promoter region (5HTTLPR), in which a 44 bp long stretch of DNA is either present in the “long” form of the gene or absent in the “short” form. Experimental data suggest the long form is associated with greater expression of the gene (142), although recent findings have suggested this locus may actually be triallelic due the presence of a SNP nearby which leads to further variability in the effect on gene expression (143). Other common variants are found in the gene, including a variable number of tandem repeats in intron 2 (STin2) which has been shown to influence gene transcription (144) and has also been examined in multiple pharmacogenetic studies.
A systematic review of pharmacogenetic studies of antidepressants (145, 146) was recently reported in which the published associations between treatment response and these two well-characterized 5HTT variants were comprehensively examined. In this review, a meta-analysis of 15 studies (13–16, 21, 27, 28, 34, 37, 38, 44, 49, 73, 74, 88) showed the long allele of the promoter polymorphism was associated with better response and remission rates, while another meta-analysis of nine studies (35, 41, 46, 74, 81, 87, 100, 118, 121) indicated the long allele was also associated with lower rates of side-effects. The review of studies on the intronic variant was less clear, although a meta-analysis of seven studies (14, 30, 51, 73, 84, 110, 121) suggested an influence on efficacy, particularly among Asians. Enthusiasm for these findings are dampened by the fact that another study (90) from the STAR*D trial found no evidence of an association between any variants in 5HTT and treatment outcomes, despite having one of the largest samples to test the relationship.
The systematic review (146) also examined variants in 16 other candidate genes thought to play a role in the pharmacodynamics of antidepressants and reported on by at least two different studies. Of these, variants in four of the genes were found upon meta-analysis to be significantly associated with either efficacy or side effects. The four genes were 5HT1A, 5HT2A, TPH1, and BDNF. 5HT1A and 5HT2A code for serotonin receptors that are the targets of certain antidepressants and atypical antipsychotics. Interestingly, an association between 5HT2A and antidepressant efficacy was one of the leading pharmacogenetic findings from STAR*D (71). TPH1 codes for tryptophan hydroxylase which is the rate-limiting enzyme in the biosynthesis of serotonin. It is more commonly expressed in the periphery (147), but there is some evidence from the mouse that it is also expressed in the brain during the late developmental stages (148). BDNF is a neurotrophic factor that is involved in the development, survival and functional maintenance of neurons (149).
Genome-wide Studies
Three genome-wide association studies of antidepressant response have been published (131–133). In the first study (131) 90 Caucasians who developed treatment emergent suicidal ideation (TESI) with citalopram in STAR*D and an equal number of sex and race matched treated controls were genotyped at 109,365 SNPs on the Illumina Human-1 BeadChip. One marker was significant after correction for multiple testing in the gene PAPLN. This gene encodes a proteoglycan-like sulfated glycoprotein, but little else is known about its function and potential relevance to TESI.
The second study (132) examined efficacy responses to citalopram in 1,491 STAR*D subjects who were genotyped at 430,198 SNPs with the Affymetrix 500K and 5.0 platforms. No SNPs met criteria for genome-wide significance, but there were three with suggestive evidence in or near the genes UBE3C, BMP7, and RORA. The biological relevance of these genes to treatment response is not immediately obvious.
The most recent study (133) was carried out in the Munich Antidepressant Response Signature (MARS) project in which patients who were treated with antidepressants according to the choice of their physicians were naturalistically followed for efficacy response. A total of 339 patients were genotyped on almost 410,000 non-overlapping SNPs with the Illumina Sentrix Human-1 and HumanHap300 BeadChip arrays. A multilocus genetic variable that described the individual number of alleles of select SNPs associated with beneficial treatment outcome was constructed and then dichotomized. The dichotomized variable describing carriers with high and low number of response alleles was associated with positive outcome in the MARS sample as well as in a replication sample derived from STAR*D. This finding suggests that treatment response may in fact be multifactorial and under the control of a number of additive genetic loci instead of a limited number with large effects.
Mood Stabilizers
The leading mood stabilizers include lithium and the anticonvulsants, such as valproate, carbamazepine, and lamotrigine. Lithium has been a remarkably successful drug, but its introduction into psychiatry has had a complicated and somewhat controversial history (150). Its use in practice dates back to the mid-19th century, but it was not until the 1970’s that it was finally approved in the United States for the treatment and prophylaxis of mania. Considerable evidence has accumulated since then about the positive benefits of lithium (151), yet it lacks universal effectiveness and can provoke side effects, such as hand tremor, frequent urination, and weight gain. Despite the benefits and relatively cheap cost of lithium, its use has been steadily eclipsed over the past couple decades by the introduction of the anticonvulsants (152). The comparative safety and efficacy of lithium versus the rival anticonvulsants, however, remains a matter of debate (153).
Overview of Pharmacogenetic Studies
There is a relative dearth of pharmacogenetic studies of mood stabilizers. Some 36 candidate gene studies have been reported (154–189). Almost all of these examined lithium. With few exceptions, the studies were based on Caucasian samples with exceedingly small numbers (median = 111), and in the majority of cases they relied on retrospective characterization of response to lithium. One genome-wide association study of lithium responsiveness was recently reported (190). (See Table 2 for a description of the genome-wide association study of treatment response to mood stabilizers.)
Table 2.
Author (Year) | Drug | Gene(s) | Outcome | Sample Size | Key Finding(s) |
---|---|---|---|---|---|
Perlis (2009) | Lithium (alone or in combination with other psychotropic medications) | Affymetrix 500K | Efficacy (recurrence) | 458 | Suggestive associations with GRIA2, SDC2, ODZ4 |
Pharmacodynamic Studies
As far as we are aware, no pharmacogenetic studies have been reported on the pharmacokinetics of mood stabilizers. Published studies have instead concentrated on pharmacodynamic factors. Although the mechanisms of action of lithium and the other anticonvulsant mood stabilizers are not completely known, there are two leading hypotheses. The first involves the phosphoinositide pathway and has been referred to as the “inositol depletion hypothesis” (191). It posits that lithium, and perhaps valproate and carbamazepine as well (192), inhibit the activity of two enzymes, inositol monophosphatase (IMPA) and inositol polyphosphate 1-polyphosphatase (INPP1) which causes a reduction in the amount of free inositol available for the regeneration of phosphatidylinositol-4, 5-bisphosphate (PIP2). PIP2 is a substrate needed for the generation of important intracellular signaling molecules, inositol-1, 4, 5-triphosphate (IP3) and diacylglycerol (DAG) via activation of the enzyme G-protein-coupled phospholipase C. IP3 mediates Ca+2 release from intracellular stores and mediates a range of signaling pathways. Both Ca+2 and DAG further stimulate protein kinase C which is also a component of other signaling pathways. There have been at least seven studies (156, 157, 159, 167, 169, 179, 181) on the relationship between variation in genes coding for key enzymes in this pathway and treatment response to lithium, but the findings have been largely inconclusive.
The leading alternative hypothesis for lithium’s mechanism of action is its effects on cell survival through the inhibition of GSK3b (193). Lithium acts in the same manner as the Wnt pathway to inhibit GSK3b, leading to the translocation of β-catenin to the cell nucleus where it becomes part of complexes that regulate the transcription of genetic components involved in cell survival (194, 195). It has been shown that valproate may have similar effects (196). Motivated by these considerations, several pharmacogenetic studies of GSK3b (175, 176) have been reported, again with mixed findings.
There has also been interest in examining some of the usual suspects in psychiatric genetics including, for example, MAOA, COMT, 5HTT, TPH1, BDNF (e.g., (161, 163, 168, 170, 173, 174, 177, 179, 182, 184)). As yet, there remains no conclusive evidence that variation in any of these genes influence treatment response to mood stabilizers.
Genome-wide Studies
One genome-wide association study of lithium response has been reported (190). In this prospective study from STEP-BD, the associations between 1.4 million genotyped and imputed SNPs and the risk of mood disorder recurrence were examined among 1,177 patients with bipolar disorder, including 458 who were treated with lithium alone or in combination with other psychotropic medications. SNPs found to be associated at the threshold of p<5×10−4 were examined in a replication sample from the University College London in which 359 patients with bipolar disorder were retrospectively assessed for lithium response. These SNPs were also tested to determine if their association with recurrence was specific to treatment lithium in the STEP-BD cohort. None of the SNPs tested in the STEP-BD cohort met genome-wide significant criteria for association. However, 140 SNPs were carried forward for replication, and of these 9 were significant in the UCL sample at p<0.05. Of these, five had the same direction of effect as in the STEP-BD cohort, and three displayed associations that were specific to lithium treatment. The latter three SNPs point to associations with GRIA2, which has been found to be downregulated by chronic lithium treatment in a human neuronal cell line (197, 198); SDC2, which codes for a cell-surface proteoglycan that may play a role in dendritic spine formation in the hippocampus (197); and ODZ4, which has been implicated in brain patterning (199). These findings implicate novel candidate genes for lithium response that merit further investigation, but more generally they suggest there are few if any genes with large effects on lithium response and, instead, as with other complex traits, multiple loci may be involved.
Antipsychotics
The first generation of antipsychotics (FGAs), including the phenothiazine deriviates such as chlorpromazine, dates back to the 1950s. Their introduction was responsible for large decreases in psychiatric inpatient populations (200). However, because of side effects such as extrapyramidal symptoms (EPS) including parkinsonism, akathisia and tardive dyskinesia (TD), their appeal was limited. In the 1990s a second generation of antipsychotics (SGAs) with a more diverse mechanism of action that targeted various serotonin and dopamine receptors was developed. The first of these, clozapine, has been shown to be effective against treatment-resistant schizophrenia (201), but it is also associated with life-threatening agranulocytosis. Several other SGAs, such as risperidone, olanzapine, and quetiapine, have since been introduced. The conventional notion is that compared to FGAs these so-called “atypical antipsychotics” have better safety profiles and greater efficacy, particularly against the negative symptoms of schizophrenia (200). However, a recent meta-analysis suggested that only a few are actually more effective at reducing symptoms and have less tendency for inducing EPS (202). SGAs are also associated with notable adverse effects of their own, such as weight gain, diabetes, metabolic syndrome and sedation (203).
Overview of PGx Studies
At least 274 pharmacogenetic studies of candidate genes and antipsychotic treatment response have been published (204–476). The majority of these studies examined the use of multiple antipsychotics, although FGAs were typically considered separately from SGAs. Approximately 40% of the studies examined only one antipsychotic, with clozapine being the most widely studied, followed by olanzapine and risperidone. The outcomes of interest were split evenly between efficacy and side effects. Among the latter, TD and weight gain were mostly commonly studied, with a subset of studies focused on agranulocytosis associated with clozapine. Just over half the studies were on Caucasian samples, while the rest were mostly of Chinese, Japanese or Korean samples. As with the pharmacogenetic studies of antidepressants and mood stabilizers, the sample sizes were generally small with a median of 115, not counting four reports (398, 403, 441, 442) from the CATIE trial which each included approximately 700 patients. Five genome-wide association studies of antipsychotic treatment response have been published (477–481). (See Table 3 for a description of candidate genes studies from CATIE and genome-wide association studies of treatment responses to antipsychotics.)
Table 3.
Author (Year) | Drug | Gene(s) | Outcome | Sample Size | Key Finding(s) |
---|---|---|---|---|---|
Candidate gene studies from CATIE | |||||
Grossman (2008) | CATIE* | CYP2D6, CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP1A4, CYP3A5, CYP3A4, ABCB1, FMO3 UGT1A4 | Optimized dose Treatment stop due to side effects Tardive dyskinesia |
750 | No associations |
Campbell (2008) | CATIE* | RGS4 | Efficacy | 678 | Suggestive association |
Need (2009) | CATIE* | 118 genes | 21 phenotypes | 756 | Multiple suggestive associations |
Tsai (2009) | CATIE* | 128 genes | Tardive dyskinesia | 710 | No associations |
Genome-wide association studies | |||||
McClay (2009) | CATIE* | Affymetrix 500K | Efficacy | 738 | Significant association in 4p15 |
Alkelai (2009) | CATIE* | Affymetrix 500K plus custom 164K fill-in chip | Antipsychotic-induced parkinsonism | 397 | Suggesitve associations in EPF1, NOVA1, FIGN |
Aberg (2009) | CATIE* | Affymetrix 500K | Extra-pyramidal side effects | 738 | Significant associations in 11q24 and ZNF202 |
Lavedan (2009) | Iloperidone | Affymetrix 500K | Efficacy | 426 | Suggestive association with NPAS3 |
Volpi (2009) | Iloperidone | Affymetrix 500K | QT interval prolongation | 183 | Suggestive association with CERKL and SLCO3A1 |
In CATIE, patients were treated with up to 5 different antipsychotics: olanzapine, quetiapine, risperidone, ziprasidone, perphenazine
Pharmacokinetics
Just as they do with antidepressants, the CYP450 enzymes play a leading role in the pharmacokinetics of antipsychotics. Along with CYP2D6, CYP1A2, CYP3A4, and CYP3A5 are the key enzymes responsible for metabolizing most commonly used antipsychotics (482). A number of studies (205, 222, 232, 236, 245, 260, 262, 270, 271, 281, 292, 294, 310, 311, 324, 331, 341, 342, 348, 367, 377, 388, 389, 404, 420, 451, 459, 461, 463) have examined the association between variants in the genes coding for these enzymes and antipsychotic response. The majority of these have studied adverse effects, and in particular TD. A meta-analysis (483) of studies on TD provided evidence of an increased risk with loss of function alleles in CYP2D6. However, further analysis suggested that publication bias could not be entirely ruled out. In addition, a recent report (398) from the CATIE trial in which a number of variants across the key CYP450 genes, as well as several other Phase II and transporter genes, were examined found no strong associations with dosing, safety or efficacy of the antipsychotic treatments used in the trial.
Pharmacodynamics
Dysregulation of the dopaminergic system was among the first pathological findings observed in schizophrenia, and dopamine inhibition is a common feature of most antipsychotics, particularly the FGAs. Evidence suggests that dopamine antagonism may be required for antipsychotic activity, with PET studies showing that a certain level of blockade of dopaminergic receptors in the striatrum is needed to sustain a therapeutic effect, while excess blockade can lead to extrapyramidal side effects (484–486). There are five subtypes of dopamine receptors (D1–D5), and of these D2 and D3 are the most widely implicated in pharmacogenetic studies of antipsychotics.
Three polymorphisms in DRD2 which encodes the D2 receptor have received the greatest attention. These include the Taq1A polymorphism, which is located approximately 10 kb from the 3’ end of the gene and has no known functional effect; the −141-C Ins/Del polymorphism in the promoter region, which has been associated with lower expression of the D2 receptor in vitro (487) and higher D2 density in the striatum in vivo (488); and Ser311Cys, a relatively common coding polymorphism that has been shown to reduce signal transduction via the receptor (489). At least fourteen studies (250, 261, 265, 273, 306, 330, 345, 349, 351, 390, 392, 411, 420, 421) have examined the relationship between DRD2 polymorphisms and efficacy of both FGAs and SGAs, while twenty-one studies (243, 255, 259, 264, 274, 278, 289, 293, 295, 300, 352, 372, 375, 382, 395, 401, 421, 421, 422, 444, 452) have investigated adverse effects, including TD, weight gain and neuromalignant syndrome. In a recent meta-analysis (490) of four different genes and TD, a significant association was found with the Taq1A polymorphism in DRD2.
The DRD3 gene, which has also been extensively studied, contains a Ser9Gly polymorphism that has been shown in vitro to influence dopamine binding affinity (491). Several studies have examined the association between this polymorphism and efficacy (227, 238, 250, 284, 316, 334, 339, 400, 418, 420, 439, 446, 449) and adverse effects like TD (211, 215, 223, 228, 237, 242, 244, 252, 257, 278, 286, 375, 401, 416, 443, 452, 473, 492). A mega-analysis of combined data from several studies on 780 patients (276) suggested the Gly9 allele conferred a small, but significant, increase in risk of TD. This finding was corroborated by a later meta-analysis (492) which suggested the association was stronger in non-Asian versus Asian populations.
The serotonergic system has also been implicated in treatment responses to antipsychotics. SGAs in particular display high affinities for serotonin receptors which have been hypothesized to mediate, at least partially, their therapeutic action (493, 494). Seven distinct families of serotonin receptors have been identified (5HT1 – 5HT7) (495). Of greatest interest is the 5HT2 receptor family, especially 5-HT2A and 5-HT2C. Several polymorphisms in both 5HT2A (−1438-G/A and 102-T/C in the promoter and His425Tyr in the coding region) and 5HT2C (a VNTR, −759-T/C, and −995-G/A in the promoter and Cys23Ser in the coding region) have been investigated in multiple studies of treatment response and adverse effects (204, 206–210, 212, 214, 218, 220, 225, 226, 230, 246, 248, 249, 263, 266, 268, 269, 275, 280, 283, 285, 288, 290, 297, 308, 320, 327–329, 336, 365, 367, 370, 375, 384, 385, 387, 401, 402, 409, 413, 418, 429, 430, 443, 445, 448, 453, 456, 468, 473). Several meta-analyses of these studies have been conducted including one showing an association between the 102-T/C and His425Tyr polymorphisms in 5HT2A and treatment response to clozpaine (496), a second showing an association between the 102-T/C polymorphism in 5HT2A and TD (497), and a third showing an association between the −759C/T polymorphism in 5HT2C and weight gain (498).
A number of other genes have been investigated in relation to antipsychotic responses. These studies have been motivated by various hypotheses about the mechanisms of action of antipsychotics, such as, for example, the role of the glutamatergic system or neuronal genesis and plasticity (for a more detailed review see (482)). In addition, several pharmacogenetic studies from the CATIE trial have examined a range of candidate genes with inconclusive results (441, 442).
Genome-wide Studies
Five genome-wide association studies of antipsychotic treatment response have been reported in the literature (477–481). Three of these came from the CATIE trial in which patients with schizophrenia were randomized to treatment with either a SGA (olanzapine, quetiapine, risperidone, or ziprasidone) or a FGA (perphenazine). The first study (477) tested for genome-wide predictors of efficacy among 738 patients genotyped using the Affymetrix 500K genotyping platform supplemented with a custom 164K chip to improve genome-wide coverage. Efficacy was measured by changes over time in positive and negative symptom scores. Because the patients were allowed to switch among treatments due to lack of efficacy or tolerability, associations were examined relative to the first drug to which the patient was randomized. Only one finding, in an intergenic region on chromosome 4p15, reached the pre-specified threshold for genome-wide significance. Two other findings were close to this threshold in ANKS1B and CNTNAP5, which were found to mediate negative symptom response to olanzapine and risperidone, respectively.
The two other studies (478, 479) from CATIE were partially overlapping. The more inclusive study (479) examined symptoms of parkinsonism, akathasia, and abnormal involuntary movements among the 738 patients included in the efficacy study described above. Three findings met genome-wide significance in novel regions that have not been previously implicated in the pharmacogenetics of extrapyramidal symptoms. Two were located in an intergenic region on chromosome 11q24, and the other was in ZNF202, which is a transcriptional repressor controlling PLP1, a major component of myelin.
The remaining two genome-wide association studies (480, 481) came from a phase 3 randomized trial of iloperidone, an investigational new drug for the treatment of schizophrenia from Vanda Pharmaceuticals. The 28 day trial was double-blinded as well as placebo and ziprasidone-controlled. In the first study (480), genome-wide associations with efficacy were examined. A total of 426 patients genotyped on the Affymetrix 500K platform were included, including 218 on iloperidone, 103 on active comparator, and 103 on placebo. The outcome was change from baseline to last scheduled observation in positive and negative total symptom scores. Three complimentary analyses were carried out, and six loci were identified with consistent findings across these analyses. The single best finding was in NPAS3, a gene which circumstantial evidence has previously implicated in schizophrenia. In the second study (481), genome-wide associations with QT interval prolongation, a potentially life-threatening side effect of treatment with iloperidone and other antipsychotics, were examined. A total of 183 patients on iloperidone treatment with QT interval measurements at day 14 of the trial were included in this analysis. The top findings implicated two genes in particular, CERKL and SLC03A1, with plausible roles in this adverse effect. CERKL is thought to be part of the ceramide pathway, which regulates currents conducted by various potassium channels including the hERG channel, which, when inhibited, can prolong the QT interval. SLC03A1 plays a role in translocation of prostaglandins which may have cardioprotective effects.
Pharmacogenetics in Clinical Practice
Only one pharmacogenetics test has been approved by the Food & Drug Administration (FDA) for clinical use in psychiatry (140). This is the AmpliChip CYP450 Test marketed by Roche Molecular Systems. It uses Affymetrix microarray-based genotyping technology with more than 15,000 oligonucleotide probes to assay for 20 CYP2D6 alleles, 7 CYP2D6 duplications, and 3 CYP2C19 alleles. The test includes software with an algorithm to predict CYP2D6 and CYP2C19 phenotypes (i.e., PM, IM, EM, and UM) based on the identified alleles.
The intended use of the chip cleared by the FDA is very general and does not refer to any specific drug. Instead, it states that information about the two CYP450 genes assayed, “may be used as an aid to clinicians in determining therapeutic strategy and treatment dose for therapeutics that are metabolized” by products of these genes (499). Consequently, the FDA cleared the Roche AmpliChip without clinical studies demonstrating that it is actually beneficial for selection or dosing of any psychotropic medication, despite the fact that it has been marketed, often direct-to-consumer, for use with these medications, especially the SSRIs (500). This is consistent with the FDA’s approach to other diagnostic devices such as MRIs where it has left demonstration of clinical benefit to clinicians and payers.
The Center for Disease Control and Prevention (CDC) commissioned an independent panel to examine the analytic validity, clinical validity and clinical utility of CYP450 genotyping when prescribing SSRI antidepressants (501). These three key characteristics of a pharmacogenetics test are defined, respectively, as, the ability to a) detect different alleles accurately, b) predict clinically meaningful outcomes, and c) provide information that improves the risk/benefit ratio of clinical treatment. In their review, the independent panel determined there was strong evidence for the analytic validity of CYP450 genotyping, but only marginal evidence for its clinical validity and almost no evidence for its clinical utility (141). Thus, the independent panel concluded (501) there was, “insufficient evidence to support a recommendation for or against use of CYP450 testing in adults beginning SSRI treatment,” and further noted that, “in the absence of supporting evidence, and with consideration of other contextual issues, EGAPP discourages use of CYP450 testing for patients beginning SSRI treatment until further clinical trials are completed.”
In addition to regulating pharmacogenetic tests, the FDA also oversees the incorporation of information about relevant pharmacogenetic biomarkers into the drug labels. Biomarkers are defined as characteristics that can be objectively measured and evaluated as indicators of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic interventions (502). Genetic variants associated with therapeutic responses to a pharmacological agent are pharmarcogenetics biomarkers. The FDA collects information on pharmacogenetic biomarkers and their analytic validity, clinical validity and clinical utility from pharmaceutical companies during the drug application process (503), and then incorporates this information into approved drug labels as deemed appropriate.
In a recent survey (504) of drug labels approved between 1945–2005, it was reported that a total of 121 contained pharmacogenetic information. Of these, 69 labels referred to human genomic biomarkers, and the remainder referred to microbial genomic biomarkers. Of the labels referring to human genomic biomarkers, 43 (62%) pertained to polymorphisms in CYP450 genes, with CYP2D6 being the most common. Approximately 17% of the labels were for psychiatric drugs. In most cases the drug labels merely provided pharmacogenetic information. Only one label went further and recommended, but did not require, a specific action before making a therapeutic decision. This was for testing urea cycle enzyme deficiencies before prescribing valproic acid. Thus, while pharmacogenetic information has begun to penetrate into clinical practice, it has not yet had a meaningful impact on therapeutic decision making in psychiatry.
Challenges to Clinical Translation
Despite notable progress in research over the past decade, the promise of pharmacogenetics in psychiatry has not yet been fully realized. The biggest obstacle to translating the promise into reality is that we still do not have a clear understanding of how genetic factors influence treatment response to psychotropic medications. The studies carried out to date suggest a number of intriguing hypotheses that merit further investigation, but they do not point to any definitive associations that can be used with confidence to predict how a patient will respond to a particular treatment. The difficulty with the pharamcogenetic associations thus far reported is the lack of consistent findings. For every positive association, there are typically several negative studies that cast doubt on the finding. As a result, it is difficult to draw firm conclusions about the clinical relevance of any genes that may be implicated.
There are several reasons for the difficulty. First, treatment responses to psychotropic medications are complex phenotypes. They may, in fact, be as complex as the diseases for which they are used to treat. Psychotropic medications may act on a number of different molecular pathways to exert their therapeutic effect, and in turn they may be acted on by a number of different molecular pathways in the process of their absorption, distribution and elimination. Consequently, multiple variants in distinct and converging genetic pathways may independently and interactively contribute to a particular drug response. In addition, multiple environmental factors may further contribute to variability in the response. Demographic factors, diet, substance abuse, smoking, concomitant treatments and co-morbidities may all affect the actions of psychotropic drugs (505). For example, it has been shown that smoking induces CYP450 activity and promotes the metabolism of substrate drugs (506, 507), while conversely SSRI’s are known to inhibit CYP450 activity and may disrupt the metabolism of other concomitant medications (508). Thus, treatment responses may be the sum of a number of impinging genetic and environmental factors, making it difficult to identify any one factor in isolation and to construct more complete models of the determinants of drug response.
Second, it is particularly challenging to conduct appropriately designed pharmacogenetic studies that can illuminate the complex architecture of treatment responses. The studies carried out to date have had rather small sample sizes and short periods of follow-up, largely because it is costly and logistically challenging to ascertain and prospectively evaluate patients in such studies. Even the largest studies that have been reported are significantly underpowered to detect genes with effect sizes likely involved in treatment responses. To address this issue, efforts have been made to combine data across studies in meta or mega analyses. While this can be a useful strategy, existing studies frequently differ so considerably in design, patient populations and outcome measures that it raises serious questions about the comparability of results across studies. Finally, to complicate matters, within each study, patients often take multiple medications and have erratic patterns of adherence. As a result, the responses to any one drug during follow-up may be hopelessly obscured. Clearly, new approaches are needed to overcome these limitations in order to further advance the goals of pharmacogenetics.
Conclusions
By personalizing treatments to psychotropic medications, pharmacogenetics holds great promise to dramatically improve care in psychiatry. The genomics revolution has provided us with an unprecedented set of tools to pursue the goal of pharmacogenetics. As reviewed here, a great deal of work has begun to use these tools to unravel the complex pharmacogenetic underpinnings of treatment responses. While considerable progress has been made, much work remains to be done. It appears we are only at the end of the beginning of a long venture.
Acknowledgements
The work of Peter P. Zandi was supported by NIMH grant K01 MH072866-01. The work of Jennifer A. Judy was supported by NIMH grant T32-MH14592.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Liu-Seifert H, Adams DH, Kinon BJ. Discontinuation of treatment of schizophrenic patients is driven by poor symptom response: a pooled post-hoc analysis of four atypical antipsychotic drugs. BMC.Med. 2005;3:21. doi: 10.1186/1741-7015-3-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, Niederehe G, Thase ME, Lavori PW, Lebowitz BD, McGrath PJ, Rosenbaum JF, Sackeim HA, Kupfer DJ, Luther J, Fava M. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am.J.Psychiatry. 2006;163:1905–1917. doi: 10.1176/ajp.2006.163.11.1905. [DOI] [PubMed] [Google Scholar]
- 3.Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD, Severe J, Hsiao JK. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N.Engl.J.Med. 2005;353:1209–1223. doi: 10.1056/NEJMoa051688. [DOI] [PubMed] [Google Scholar]
- 4.Thase ME. STEP-BD and bipolar depression: what have we learned? Curr.Psychiatry Rep. 2007;9:497–503. doi: 10.1007/s11920-007-0068-9. [DOI] [PubMed] [Google Scholar]
- 5.Weinshilboum R, Wang L. Pharmacogenomics: bench to bedside. Nat.Rev.Drug Discov. 2004;3:739–748. doi: 10.1038/nrd1497. [DOI] [PubMed] [Google Scholar]
- 6.Lieberman JA., III History of the use of antidepressants in primary care. Primary Care Companion J Clin Psychiatry. 2003;5:6–10. [Google Scholar]
- 7.Preskorn SH, Stanga CY, Ross R. Selective serotonin reuptake inhibitor. 2004:242. [Google Scholar]
- 8.Anderson IM, Ferrier IN, Baldwin RC, Cowen PJ, Howard L, Lewis G, Matthews K, lister-Williams RH, Peveler RC, Scott J, Tylee A. Evidence-based guidelines for treating depressive disorders with antidepressants: a revision of the 2000 British Association for Psychopharmacology guidelines. J.Psychopharmacol. 2008;22:343–396. doi: 10.1177/0269881107088441. [DOI] [PubMed] [Google Scholar]
- 9.Crismon ML, Trivedi M, Pigott TA, Rush AJ, Hirschfeld RM, Kahn DA, DeBattista C, Nelson JC, Nierenberg AA, Sackeim HA, Thase ME. The Texas Medication Algorithm Project: report of the Texas Consensus Conference Panel on Medication Treatment of Major Depressive Disorder. J.Clin.Psychiatry. 1999;60:142–156. [PubMed] [Google Scholar]
- 10.Thase ME, Rush AJ. Treatment-resistant depression. 1995:1081–1097. [Google Scholar]
- 11.Lin EH, Von KM, Katon W, Bush T, Simon GE, Walker E, Robinson P. The role of the primary care physician in patients' adherence to antidepressant therapy. Med.Care. 1995;33:67–74. doi: 10.1097/00005650-199501000-00006. [DOI] [PubMed] [Google Scholar]
- 12.Chen S, Chou WH, Blouin RA, Mao Z, Humphries LL, Meek QC, Neill JR, Martin WL, Hays LR, Wedlund PJ. The cytochrome P450 2D6 (CYP2D6) enzyme polymorphism: screening costs and influence on clinical outcomes in psychiatry. Clin.Pharmacol.Ther. 1996;60:522–534. doi: 10.1016/S0009-9236(96)90148-4. [DOI] [PubMed] [Google Scholar]
- 13.Smeraldi E, Zanardi R, Benedetti F, Di BD, Perez J, Catalano M. Polymorphism within the promoter of the serotonin transporter gene and antidepressant efficacy of fluvoxamine. Mol.Psychiatry. 1998;3:508–511. doi: 10.1038/sj.mp.4000425. [DOI] [PubMed] [Google Scholar]
- 14.Kim DK, Lim SW, Lee S, Sohn SE, Kim S, Hahn CG, Carroll BJ. Serotonin transporter gene polymorphism and antidepressant response. Neuroreport. 2000;11:215–219. doi: 10.1097/00001756-200001170-00042. [DOI] [PubMed] [Google Scholar]
- 15.Pollock BG, Ferrell RE, Mulsant BH, Mazumdar S, Miller M, Sweet RA, Davis S, Kirshner MA, Houck PR, Stack JA, Reynolds CF, Kupfer DJ. Allelic variation in the serotonin transporter promoter affects onset of paroxetine treatment response in late-life depression. Neuropsychopharmacology. 2000;23:587–590. doi: 10.1016/S0893-133X(00)00132-9. [DOI] [PubMed] [Google Scholar]
- 16.Zanardi R, Benedetti F, Di BD, Catalano M, Smeraldi E. Efficacy of paroxetine in depression is influenced by a functional polymorphism within the promoter of the serotonin transporter gene. J.Clin.Psychopharmacol. 2000;20:105–107. doi: 10.1097/00004714-200002000-00021. [DOI] [PubMed] [Google Scholar]
- 17.Zill P, Baghai TC, Zwanzger P, Schule C, Minov C, Riedel M, Neumeier K, Rupprecht R, Bondy B. Evidence for an association between a G-protein beta3-gene variant with depression and response to antidepressant treatment. Neuroreport. 2000;11:1893–1897. doi: 10.1097/00001756-200006260-00018. [DOI] [PubMed] [Google Scholar]
- 18.Minov C, Baghai TC, Schule C, Zwanzger P, Schwarz MJ, Zill P, Rupprecht R, Bondy B. Serotonin-2A-receptor and -transporter polymorphisms: lack of association in patients with major depression. Neurosci.Lett. 2001;303:119–122. doi: 10.1016/s0304-3940(01)01704-9. [DOI] [PubMed] [Google Scholar]
- 19.Serretti A, Zanardi R, Cusin C, Rossini D, Lorenzi C, Smeraldi E. Tryptophan hydroxylase gene associated with paroxetine antidepressant activity. Eur.Neuropsychopharmacol. 2001;11:375–380. doi: 10.1016/s0924-977x(01)00113-4. [DOI] [PubMed] [Google Scholar]
- 20.Serretti A, Zanardi R, Rossini D, Cusin C, Lilli R, Smeraldi E. Influence of tryptophan hydroxylase and serotonin transporter genes on fluvoxamine antidepressant activity. Mol.Psychiatry. 2001;6:586–592. doi: 10.1038/sj.mp.4000876. [DOI] [PubMed] [Google Scholar]
- 21.Zanardi R, Serretti A, Rossini D, Franchini L, Cusin C, Lattuada E, Dotoli D, Smeraldi E. Factors affecting fluvoxamine antidepressant activity: influence of pindolol and 5-HTTLPR in delusional and nondelusional depression. Biol.Psychiatry. 2001;50:323–330. doi: 10.1016/s0006-3223(01)01118-0. [DOI] [PubMed] [Google Scholar]
- 22.Allgulander C, Nilsson B. A prospective study of 86 new patients with social anxiety disorder. Acta Psychiatr.Scand. 2001;103:447–452. doi: 10.1034/j.1600-0447.2001.00320.x. [DOI] [PubMed] [Google Scholar]
- 23.Baghai TC, Schule C, Zwanzger P, Minov C, Schwarz MJ, de JS, Rupprecht R, Bondy B. Possible influence of the insertion/deletion polymorphism in the angiotensin I-converting enzyme gene on therapeutic outcome in affective disorders. Mol.Psychiatry. 2001;6:258–259. doi: 10.1038/sj.mp.4000857. [DOI] [PubMed] [Google Scholar]
- 24.Wang JH, Liu ZQ, Wang W, Chen XP, Shu Y, He N, Zhou HH. Pharmacokinetics of sertraline in relation to genetic polymorphism of CYP2C19. Clin.Pharmacol.Ther. 2001;70:42–47. doi: 10.1067/mcp.2001.116513. [DOI] [PubMed] [Google Scholar]
- 25.Wu WH, Huo SJ, Cheng CY, Hong CJ, Tsai SJ. Association study of the 5-HT(6) receptor polymorphism (C267T) and symptomatology and antidepressant response in major depressive disorders. Neuropsychobiology. 2001;44:172–175. doi: 10.1159/000054938. [DOI] [PubMed] [Google Scholar]
- 26.Cusin C, Serretti A, Zanardi R, Lattuada E, Rossini D, Lilli R, Lorenzi C, Smeraldi E. Influence of monoamine oxidase A and serotonin receptor 2A polymorphisms in SSRI antidepressant activity. Int.J.Neuropsychopharmacol. 2002;5:27–35. doi: 10.1017/S1461145701002711. [DOI] [PubMed] [Google Scholar]
- 27.Yoshida K, Ito K, Sato K, Takahashi H, Kamata M, Higuchi H, Shimizu T, Itoh K, Inoue K, Tezuka T, Suzuki T, Ohkubo T, Sugawara K, Otani K. Influence of the serotonin transporter gene-linked polymorphic region on the antidepressant response to fluvoxamine in Japanese depressed patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2002;26:383–386. doi: 10.1016/s0278-5846(01)00287-1. [DOI] [PubMed] [Google Scholar]
- 28.Yu YW, Tsai SJ, Chen TJ, Lin CH, Hong CJ. Association study of the serotonin transporter promoter polymorphism and symptomatology and antidepressant response in major depressive disorders. Mol.Psychiatry. 2002;7:1115–1119. doi: 10.1038/sj.mp.4001141. [DOI] [PubMed] [Google Scholar]
- 29.Hong CJ, Wang YC, Tsai SJ. Association study of angiotensin I-converting enzyme polymorphism and symptomatology and antidepressant response in major depressive disorders. J.Neural Transm. 2002;109:1209–1214. doi: 10.1007/s00702-001-0686-z. [DOI] [PubMed] [Google Scholar]
- 30.Ito K, Yoshida K, Sato K, Takahashi H, Kamata M, Higuchi H, Shimizu T, Itoh K, Inoue K, Tezuka T, Suzuki T, Ohkubo T, Sugawara K, Otani K. A variable number of tandem repeats in the serotonin transporter gene does not affect the antidepressant response to fluvoxamine. Psychiatry Res. 2002;111:235–239. doi: 10.1016/s0165-1781(02)00141-5. [DOI] [PubMed] [Google Scholar]
- 31.Muller DJ, Schulze TG, Macciardi F, Ohlraun S, Gross MM, Scherk H, Neidt H, Syagailo YV, Grassle M, Nothen MM, Maier W, Lesch KP, Rietschel M. Moclobemide response in depressed patients: association study with a functional polymorphism in the monoamine oxidase A promoter. Pharmacopsychiatry. 2002;35:157–158. doi: 10.1055/s-2002-33199. [DOI] [PubMed] [Google Scholar]
- 32.Sato K, Yoshida K, Takahashi H, Ito K, Kamata M, Higuchi H, Shimizu T, Itoh K, Inoue K, Tezuka T, Suzuki T, Ohkubo T, Sugawara K, Otani K. Association between −1438G/A promoter polymorphism in the 5-HT(2A) receptor gene and fluvoxamine response in Japanese patients with major depressive disorder. Neuropsychobiology. 2002;46:136–140. doi: 10.1159/000066394. [DOI] [PubMed] [Google Scholar]
- 33.Yoshida K, Naito S, Takahashi H, Sato K, Ito K, Kamata M, Higuchi H, Shimizu T, Itoh K, Inoue K, Tezuka T, Suzuki T, Ohkubo T, Sugawara K, Otani K. Monoamine oxidase: A gene polymorphism, tryptophan hydroxylase gene polymorphism and antidepressant response to fluvoxamine in Japanese patients with major depressive disorder. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2002;26:1279–1283. doi: 10.1016/s0278-5846(02)00267-1. [DOI] [PubMed] [Google Scholar]
- 34.Rausch JL, Johnson ME, Fei YJ, Li JQ, Shendarkar N, Hobby HM, Ganapathy V, Leibach FH. Initial conditions of serotonin transporter kinetics and genotype: influence on SSRI treatment trial outcome. Biol.Psychiatry. 2002;51:723–732. doi: 10.1016/s0006-3223(01)01283-5. [DOI] [PubMed] [Google Scholar]
- 35.Takahashi H, Yoshida K, Ito K, Sato K, Kamata M, Higuchi H, Shimizu T, Ito K, Inoue K, Tezuka T, Suzuki T, Ohkubo T, Sugawara K. No association between the serotonergic polymorphisms and incidence of nausea induced by fluvoxamine treatment. Eur.Neuropsychopharmacol. 2002;12:477–481. doi: 10.1016/s0924-977x(02)00056-1. [DOI] [PubMed] [Google Scholar]
- 36.Serretti A, Lorenzi C, Cusin C, Zanardi R, Lattuada E, Rossini D, Lilli R, Pirovano A, Catalano M, Smeraldi E. SSRIs antidepressant activity is influenced by G beta 3 variants. Eur.Neuropsychopharmacol. 2003;13:117–122. doi: 10.1016/s0924-977x(02)00154-2. [DOI] [PubMed] [Google Scholar]
- 37.Arias B, Catalan R, Gasto C, Gutierrez B, Fananas L. 5-HTTLPR polymorphism of the serotonin transporter gene predicts non-remission in major depression patients treated with citalopram in a 12-weeks follow up study. J.Clin.Psychopharmacol. 2003;23:563–567. doi: 10.1097/01.jcp.0000095350.32154.73. [DOI] [PubMed] [Google Scholar]
- 38.Joyce PR, Mulder RT, Luty SE, McKenzie JM, Miller AL, Rogers GR, Kennedy MA. Age-dependent antidepressant pharmacogenomics: polymorphisms of the serotonin transporter and G protein beta3 subunit as predictors of response to fluoxetine and nortriptyline. Int.J.Neuropsychopharmacol. 2003;6:339–346. doi: 10.1017/S1461145703003663. [DOI] [PubMed] [Google Scholar]
- 39.Murphy GM, Jr, Kremer C, Rodrigues HE, Schatzberg AF. Pharmacogenetics of antidepressant medication intolerance. Am.J.Psychiatry. 2003;160:1830–1835. doi: 10.1176/appi.ajp.160.10.1830. [DOI] [PubMed] [Google Scholar]
- 40.Gerstenberg G, Aoshima T, Fukasawa T, Yoshida K, Takahashi H, Higuchi H, Murata Y, Shimoyama R, Ohkubo T, Shimizu T, Otani K. Relationship between clinical effects of fluvoxamine and the steady-state plasma concentrations of fluvoxamine and its major metabolite fluvoxamino acid in Japanese depressed patients. Psychopharmacology (Berl) 2003;167:443–448. doi: 10.1007/s00213-003-1430-1. [DOI] [PubMed] [Google Scholar]
- 41.Perlis RH, Mischoulon D, Smoller JW, Wan YJ, Lamon-Fava S, Lin KM, Rosenbaum JF, Fava M. Serotonin transporter polymorphisms and adverse effects with fluoxetine treatment. Biol.Psychiatry. 2003;54:879–883. doi: 10.1016/s0006-3223(03)00424-4. [DOI] [PubMed] [Google Scholar]
- 42.Tsai SJ, Cheng CY, Yu YW, Chen TJ, Hong CJ. Association study of a brain-derived neurotrophic-factor genetic polymorphism and major depressive disorders, symptomatology, and antidepressant response. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2003;123B:19–22. doi: 10.1002/ajmg.b.20026. [DOI] [PubMed] [Google Scholar]
- 43.Yoshida K, Naito S, Takahashi H, Sato K, Ito K, Kamata M, Higuchi H, Shimizu T, Itoh K, Inoue K, Suzuki T, Ohkubo T. Monoamine oxidase A gene polymorphism, 5-HT 2A receptor gene polymorphism and incidence of nausea induced by fluvoxamine. Neuropsychobiology. 2003;48:10–13. doi: 10.1159/000071822. [DOI] [PubMed] [Google Scholar]
- 44.Durham LK, Webb SM, Milos PM, Clary CM, Seymour AB. The serotonin transporter polymorphism, 5HTTLPR, is associated with a faster response time to sertraline in an elderly population with major depressive disorder. Psychopharmacology (Berl) 2004;174:525–529. doi: 10.1007/s00213-003-1562-3. [DOI] [PubMed] [Google Scholar]
- 45.Lee MS, Lee HY, Lee HJ, Ryu SH. Serotonin transporter promoter gene polymorphism and long-term outcome of antidepressant treatment. Psychiatr.Genet. 2004;14:111–115. doi: 10.1097/01.ypg.0000107928.32051.11. [DOI] [PubMed] [Google Scholar]
- 46.Murphy GM, Jr, Hollander SB, Rodrigues HE, Kremer C, Schatzberg AF. Effects of the serotonin transporter gene promoter polymorphism on mirtazapine and paroxetine efficacy and adverse events in geriatric major depression. Arch.Gen.Psychiatry. 2004;61:1163–1169. doi: 10.1001/archpsyc.61.11.1163. [DOI] [PubMed] [Google Scholar]
- 47.Peters EJ, Slager SL, McGrath PJ, Knowles JA, Hamilton SP. Investigation of serotonin-related genes in antidepressant response. Mol.Psychiatry. 2004;9:879–889. doi: 10.1038/sj.mp.4001502. [DOI] [PubMed] [Google Scholar]
- 48.Rau T, Wohlleben G, Wuttke H, Thuerauf N, Lunkenheimer J, Lanczik M, Eschenhagen T. CYP2D6 genotype: impact on adverse effects and nonresponse during treatment with antidepressants-a pilot study. Clin.Pharmacol.Ther. 2004;75:386–393. doi: 10.1016/j.clpt.2003.12.015. [DOI] [PubMed] [Google Scholar]
- 49.Serretti A, Cusin C, Rossini D, Artioli P, Dotoli D, Zanardi R. Further evidence of a combined effect of SERTPR and TPH on SSRIs response in mood disorders. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2004;129B:36–40. doi: 10.1002/ajmg.b.30027. [DOI] [PubMed] [Google Scholar]
- 50.Serretti A, Artioli P, Lorenzi C, Pirovano A, Tubazio V, Zanardi R. The C(−1019)G polymorphism of the 5-HT1A gene promoter and antidepressant response in mood disorders: preliminary findings. Int.J.Neuropsychopharmacol. 2004;7:453–460. doi: 10.1017/S1461145704004687. [DOI] [PubMed] [Google Scholar]
- 51.Yoshida K, Takahashi H, Higuchi H, Kamata M, Ito K, Sato K, Naito S, Shimizu T, Itoh K, Inoue K, Suzuki T, Nemeroff CB. Prediction of antidepressant response to milnacipran by norepinephrine transporter gene polymorphisms. Am.J.Psychiatry. 2004;161:1575–1580. doi: 10.1176/appi.ajp.161.9.1575. [DOI] [PubMed] [Google Scholar]
- 52.Baghai TC, Schule C, Zill P, Deiml T, Eser D, Zwanzger P, Ella R, Rupprecht R, Bondy B. The angiotensin I converting enzyme insertion/deletion polymorphism influences therapeutic outcome in major depressed women, but not in men. Neurosci.Lett. 2004;363:38–42. doi: 10.1016/j.neulet.2004.03.052. [DOI] [PubMed] [Google Scholar]
- 53.Binder EB, Salyakina D, Lichtner P, Wochnik GM, Ising M, Putz B, Papiol S, Seaman S, Lucae S, Kohli MA, Nickel T, Kunzel HE, Fuchs B, Majer M, Pfennig A, Kern N, Brunner J, Modell S, Baghai T, Deiml T, Zill P, Bondy B, Rupprecht R, Messer T, Kohnlein O, Dabitz H, Bruckl T, Muller N, Pfister H, Lieb R, Mueller JC, Lohmussaar E, Strom TM, Bettecken T, Meitinger T, Uhr M, Rein T, Holsboer F, Muller-Myhsok B. Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nat.Genet. 2004;36:1319–25. doi: 10.1038/ng1479. [DOI] [PubMed] [Google Scholar]
- 54.Grasmader K, Verwohlt PL, Rietschel M, Dragicevic A, Muller M, Hiemke C, Freymann N, Zobel A, Maier W, Rao ML. Impact of polymorphisms of cytochrome-P450 isoenzymes 2C9, 2C19 and 2D6 on plasma concentrations and clinical effects of antidepressants in a naturalistic clinical setting. Eur.J.Clin.Pharmacol. 2004;60:329–336. doi: 10.1007/s00228-004-0766-8. [DOI] [PubMed] [Google Scholar]
- 55.Kawanishi C, Lundgren S, Agren H, Bertilsson L. Increased incidence of CYP2D6 gene duplication in patients with persistent mood disorders: ultrarapid metabolism of antidepressants as a cause of nonresponse. A pilot study. Eur.J.Clin.Pharmacol. 2004;59:803–807. doi: 10.1007/s00228-003-0701-4. [DOI] [PubMed] [Google Scholar]
- 56.Lee HJ, Cha JH, Ham BJ, Han CS, Kim YK, Lee SH, Ryu SH, Kang RH, Choi MJ, Lee MS. Association between a G-protein beta 3 subunit gene polymorphism and the symptomatology and treatment responses of major depressive disorders. Pharmacogenomics.J. 2004;4:29–33. doi: 10.1038/sj.tpj.6500217. [DOI] [PubMed] [Google Scholar]
- 57.Lemonde S, Du L, Bakish D, Hrdina P, Albert PR. Association of the C(−1019)G 5-HT1A functional promoter polymorphism with antidepressant response. Int.J.Neuropsychopharmacol. 2004;7:501–506. doi: 10.1017/S1461145704004699. [DOI] [PubMed] [Google Scholar]
- 58.Licinio J, O'Kirwan F, Irizarry K, Merriman B, Thakur S, Jepson R, Lake S, Tantisira KG, Weiss ST, Wong ML. Association of a corticotropin-releasing hormone receptor 1 haplotype and antidepressant treatment response in Mexican-Americans. Mol.Psychiatry. 2004;9:1075–1082. doi: 10.1038/sj.mp.4001587. [DOI] [PubMed] [Google Scholar]
- 59.Roberts RL, Mulder RT, Joyce PR, Luty SE, Kennedy MA. No evidence of increased adverse drug reactions in cytochrome P450 CYP2D6 poor metabolizers treated with fluoxetine or nortriptyline. Hum.Psychopharmacol. 2004;19:17–23. doi: 10.1002/hup.539. [DOI] [PubMed] [Google Scholar]
- 60.Suzuki Y, Sawamura K, Someya T. The effects of a 5-hydroxytryptamine 1A receptor gene polymorphism on the clinical response to fluvoxamine in depressed patients. Pharmacogenomics.J. 2004;4:283–286. doi: 10.1038/sj.tpj.6500256. [DOI] [PubMed] [Google Scholar]
- 61.Zill P, Baghai TC, Engel R, Zwanzger P, Schule C, Eser D, Behrens S, Rupprecht R, Moller HJ, Ackenheil M, Bondy B. The dysbindin gene in major depression: an association study. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2004;129B:55–58. doi: 10.1002/ajmg.b.30064. [DOI] [PubMed] [Google Scholar]
- 62.Arias B, Catalan R, Gasto C, Gutierrez B, Fananas L. Evidence for a combined genetic effect of the 5-HT(1A) receptor and serotonin transporter genes in the clinical outcome of major depressive patients treated with citalopram. J.Psychopharmacol. 2005;19:166–172. doi: 10.1177/0269881105049037. [DOI] [PubMed] [Google Scholar]
- 63.Ham BJ, Lee MS, Lee HJ, Kang RH, Han CS, Choi MJ, Lee SH, Ryu SH. No association between the tryptophan hydroxylase gene polymorphism and major depressive disorders and antidepressant response in a Korean population. Psychiatr.Genet. 2005;15:299–301. doi: 10.1097/00041444-200512000-00014. [DOI] [PubMed] [Google Scholar]
- 64.Szegedi A, Rujescu D, Tadic A, Muller MJ, Kohnen R, Stassen HH, Dahmen N. The catechol-O-methyltransferase Val108/158Met polymorphism affects short-term treatment response to mirtazapine, but not to paroxetine in major depression. Pharmacogenomics.J. 2005;5:49–53. doi: 10.1038/sj.tpj.6500289. [DOI] [PubMed] [Google Scholar]
- 65.Choi MJ, Kang RH, Ham BJ, Jeong HY, Lee MS. Serotonin receptor 2A gene polymorphism (−1438A/G) and short-term treatment response to citalopram. Neuropsychobiology. 2005;52:155–162. doi: 10.1159/000087847. [DOI] [PubMed] [Google Scholar]
- 66.Garriock HA, Allen JJ, Delgado P, Nahaz Z, Kling MA, Carpenter L, Burke M, Burke W, Schwartz T, Marangell LB, Husain M, Erickson RP, Moreno FA. Lack of association of TPH2 exon XI polymorphisms with major depression and treatment resistance. Mol.Psychiatry. 2005;10:976–977. doi: 10.1038/sj.mp.4001712. [DOI] [PubMed] [Google Scholar]
- 67.Kato M, Ikenaga Y, Wakeno M, Okugawa G, Nobuhara K, Fukuda T, Fukuda K, Azuma J, Kinoshita T. Controlled clinical comparison of paroxetine and fluvoxamine considering the serotonin transporter promoter polymorphism. Int.Clin.Psychopharmacol. 2005;20:151–156. doi: 10.1097/00004850-200505000-00005. [DOI] [PubMed] [Google Scholar]
- 68.Kraft JB, Slager SL, McGrath PJ, Hamilton SP. Sequence analysis of the serotonin transporter and associations with antidepressant response. Biol.Psychiatry. 2005;58:374–381. doi: 10.1016/j.biopsych.2005.04.048. [DOI] [PubMed] [Google Scholar]
- 69.Lee SH, Lee KJ, Lee HJ, Ham BJ, Ryu SH, Lee MS. Association between the 5-HT6 receptor C267T polymorphism and response to antidepressant treatment in major depressive disorder. Psychiatry Clin.Neurosci. 2005;59:140–145. doi: 10.1111/j.1440-1819.2005.01348.x. [DOI] [PubMed] [Google Scholar]
- 70.Yu YW, Tsai SJ, Hong CJ, Chen TJ, Chen MC, Yang CW. Association study of a monoamine oxidase a gene promoter polymorphism with major depressive disorder and antidepressant response. Neuropsychopharmacology. 2005;30:1719–1723. doi: 10.1038/sj.npp.1300785. [DOI] [PubMed] [Google Scholar]
- 71.McMahon FJ, Buervenich S, Charney D, Lipsky R, Rush AJ, Wilson AF, Sorant AJ, Papanicolaou GJ, Laje G, Fava M, Trivedi MH, Wisniewski SR, Manji H. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antidepressant treatment. Am.J.Hum.Genet. 2006;78:804–814. doi: 10.1086/503820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Arias B, Serretti A, Lorenzi C, Gasto C, Catalan R, Fananas L. Analysis of COMT gene (Val 158 Met polymorphism) in the clinical response to SSRIs in depressive patients of European origin. J.Affect.Disord. 2006;90:251–256. doi: 10.1016/j.jad.2005.11.008. [DOI] [PubMed] [Google Scholar]
- 73.Hong CJ, Chen TJ, Yu YW, Tsai SJ. Response to fluoxetine and serotonin 1A receptor (C-1019G) polymorphism in Taiwan Chinese major depressive disorder. Pharmacogenomics.J. 2006;6:27–33. doi: 10.1038/sj.tpj.6500340. [DOI] [PubMed] [Google Scholar]
- 74.Kato M, Fukuda T, Wakeno M, Fukuda K, Okugawa G, Ikenaga Y, Yamashita M, Takekita Y, Nobuhara K, Azuma J, Kinoshita T. Effects of the serotonin type 2A, 3A and 3B receptor and the serotonin transporter genes on paroxetine and fluvoxamine efficacy and adverse drug reactions in depressed Japanese patients. Neuropsychobiology. 2006;53:186–195. doi: 10.1159/000094727. [DOI] [PubMed] [Google Scholar]
- 75.Shams ME, Arneth B, Hiemke C, Dragicevic A, Muller MJ, Kaiser R, Lackner K, Hartter S. CYP2D6 polymorphism and clinical effect of the antidepressant venlafaxine. J.Clin.Pharm.Ther. 2006;31:493–502. doi: 10.1111/j.1365-2710.2006.00763.x. [DOI] [PubMed] [Google Scholar]
- 76.Smeraldi E, Serretti A, Artioli P, Lorenzi C, Catalano M. Serotonin transporter gene-linked polymorphic region: possible pharmacogenetic implications of rare variants. Psychiatr.Genet. 2006;16:153–158. doi: 10.1097/01.ypg.0000218611.53064.a0. [DOI] [PubMed] [Google Scholar]
- 77.Yu YW, Tsai SJ, Liou YJ, Hong CJ, Chen TJ. Association study of two serotonin 1A receptor gene polymorphisms and fluoxetine treatment response in Chinese major depressive disorders. Eur.Neuropsychopharmacol. 2006;16:498–503. doi: 10.1016/j.euroneuro.2005.12.004. [DOI] [PubMed] [Google Scholar]
- 78.Bishop JR, Moline J, Ellingrod VL, Schultz SK, Clayton AH. Serotonin 2A −1438 G/A and G-protein Beta3 subunit C825T polymorphisms in patients with depression and SSRI-associated sexual side-effects. Neuropsychopharmacology. 2006;31:2281–2288. doi: 10.1038/sj.npp.1301090. [DOI] [PubMed] [Google Scholar]
- 79.Choi MJ, Kang RH, Lim SW, Oh KS, Lee MS. Brain-derived neurotrophic factor gene polymorphism (Val66Met) and citalopram response in major depressive disorder. Brain Res. 2006;1118:176–182. doi: 10.1016/j.brainres.2006.08.012. [DOI] [PubMed] [Google Scholar]
- 80.Ng CH, Easteal S, Tan S, Schweitzer I, Ho BK, Aziz S. Serotonin transporter polymorphisms and clinical response to sertraline across ethnicities. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2006;30:953–957. doi: 10.1016/j.pnpbp.2006.02.015. [DOI] [PubMed] [Google Scholar]
- 81.Popp J, Leucht S, Heres S, Steimer W. Serotonin transporter polymorphisms and side effects in antidepressant therapy--a pilot study. Pharmacogenomics. 2006;7:159–166. doi: 10.2217/14622416.7.2.159. [DOI] [PubMed] [Google Scholar]
- 82.Sugai T, Suzuki Y, Sawamura K, Fukui N, Inoue Y, Someya T. The effect of 5-hydroxytryptamine 3A and 3B receptor genes on nausea induced by paroxetine. Pharmacogenomics.J. 2006;6:351–356. doi: 10.1038/sj.tpj.6500382. [DOI] [PubMed] [Google Scholar]
- 83.Suzuki Y, Sawamura K, Someya T. Polymorphisms in the 5-hydroxytryptamine 2A receptor and CytochromeP4502D6 genes synergistically predict fluvoxamine-induced side effects in japanese depressed patients. Neuropsychopharmacology. 2006;31:825–831. doi: 10.1038/sj.npp.1300919. [DOI] [PubMed] [Google Scholar]
- 84.Kim H, Lim SW, Kim S, Kim JW, Chang YH, Carroll BJ, Kim DK. Monoamine transporter gene polymorphisms and antidepressant response in koreans with late-life depression. JAMA. 2006;296:1609–1618. doi: 10.1001/jama.296.13.1609. [DOI] [PubMed] [Google Scholar]
- 85.Perlis RH, Purcell S, Fava M, Fagerness J, Rush AJ, Trivedi MH, Smoller JW. Association between treatment-emergent suicidal ideation with citalopram and polymorphisms near cyclic adenosine monophosphate response element binding protein in the STAR*D study. Arch.Gen.Psychiatry. 2007;64:689–697. doi: 10.1001/archpsyc.64.6.689. [DOI] [PubMed] [Google Scholar]
- 86.Ham BJ, Lee BC, Paik JW, Kang RH, Choi MJ, Choi IG, Lee MS. Association between the tryptophan hydroxylase-1 gene A218C polymorphism and citalopram antidepressant response in a Korean population. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2007;31:104–107. doi: 10.1016/j.pnpbp.2006.08.001. [DOI] [PubMed] [Google Scholar]
- 87.Hu XZ, Rush AJ, Charney D, Wilson AF, Sorant AJ, Papanicolaou GJ, Fava M, Trivedi MH, Wisniewski SR, Laje G, Paddock S, McMahon FJ, Manji H, Lipsky RH. Association between a functional serotonin transporter promoter polymorphism and citalopram treatment in adult outpatients with major depression. Arch.Gen.Psychiatry. 2007;64:783–792. doi: 10.1001/archpsyc.64.7.783. [DOI] [PubMed] [Google Scholar]
- 88.Kirchheiner J, Nickchen K, Sasse J, Bauer M, Roots I, Brockmoller J. A 40-basepair VNTR polymorphism in the dopamine transporter (DAT1) gene and the rapid response to antidepressant treatment. Pharmacogenomics.J. 2007;7:48–55. doi: 10.1038/sj.tpj.6500398. [DOI] [PubMed] [Google Scholar]
- 89.Levin GM, Bowles TM, Ehret MJ, Langaee T, Tan JY, Johnson JA, Millard WJ. Assessment of human serotonin 1A receptor polymorphisms and SSRI responsiveness. Mol.Diagn.Ther. 2007;11:155–160. doi: 10.1007/BF03256237. [DOI] [PubMed] [Google Scholar]
- 90.Kraft JB, Peters EJ, Slager SL, Jenkins GD, Reinalda MS, McGrath PJ, Hamilton SP. Analysis of association between the serotonin transporter and antidepressant response in a large clinical sample. Biol.Psychiatry. 2007;61:734–742. doi: 10.1016/j.biopsych.2006.07.017. [DOI] [PubMed] [Google Scholar]
- 91.Kang RH, Wong ML, Choi MJ, Paik JW, Lee MS. Association study of the serotonin transporter promoter polymorphism and mirtazapine antidepressant response in major depressive disorder. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2007;31:1317–1321. doi: 10.1016/j.pnpbp.2007.05.018. [DOI] [PubMed] [Google Scholar]
- 92.Kang RH, Hahn SW, Choi MJ, Lee MS. Relationship between G-protein beta-3 subunit C825T polymorphism and mirtazapine responses in Korean patients with major depression. Neuropsychobiology. 2007;56:1–5. doi: 10.1159/000109970. [DOI] [PubMed] [Google Scholar]
- 93.Kato M, Wakeno M, Okugawa G, Fukuda T, Azuma J, Kinoshita T, Serretti A. No association of TPH1 218A/C polymorphism with treatment response and intolerance to SSRIs in Japanese patients with major depression. Neuropsychobiology. 2007;56:167–171. doi: 10.1159/000119734. [DOI] [PubMed] [Google Scholar]
- 94.Kronenberg S, Apter A, Brent D, Schirman S, Melhem N, Pick N, Gothelf D, Carmel M, Frisch A, Weizman A. Serotonin transporter polymorphism (5-HTTLPR) and citalopram effectiveness and side effects in children with depression and/or anxiety disorders. J.Child Adolesc.Psychopharmacol. 2007;17:741–750. doi: 10.1089/cap.2006.0144. [DOI] [PubMed] [Google Scholar]
- 95.Laje G, Paddock S, Manji H, Rush AJ, Wilson AF, Charney D, McMahon FJ. Genetic markers of suicidal ideation emerging during citalopram treatment of major depression. Am.J.Psychiatry. 2007;164:1530–1538. doi: 10.1176/appi.ajp.2007.06122018. [DOI] [PubMed] [Google Scholar]
- 96.Liu Z, Zhu F, Wang G, Xiao Z, Tang J, Liu W, Wang H, Liu H, Wang X, Wu Y, Cao Z, Li W. Association study of corticotropin-releasing hormone receptor1 gene polymorphisms and antidepressant response in major depressive disorders. Neurosci.Lett. 2007;414:155–158. doi: 10.1016/j.neulet.2006.12.013. [DOI] [PubMed] [Google Scholar]
- 97.Paddock S, Laje G, Charney D, Rush AJ, Wilson AF, Sorant AJ, Lipsky R, Wisniewski SR, Manji H, McMahon FJ. Association of GRIK4 with outcome of antidepressant treatment in the STAR*D cohort. Am.J.Psychiatry. 2007;164:1181–1188. doi: 10.1176/appi.ajp.2007.06111790. [DOI] [PubMed] [Google Scholar]
- 98.Pae CU, Serretti A, Mandelli L, De RD, Patkar AA, Jun TY, Kim JJ, Lee CU, Lee SJ, Lee C, Paik IH. Dysbindin associated with selective serotonin reuptake inhibitor antidepressant efficacy. Pharmacogenet.Genomics. 2007;17:69–75. doi: 10.1097/01.fpc.0000236330.03681.6d. [DOI] [PubMed] [Google Scholar]
- 99.Papiol S, Arias B, Gasto C, Gutierrez B, Catalan R, Fananas L. Genetic variability at HPA axis in major depression and clinical response to antidepressant treatment. J.Affect.Disord. 2007;104:83–90. doi: 10.1016/j.jad.2007.02.017. [DOI] [PubMed] [Google Scholar]
- 100.Smits K, Smits L, Peeters F, Schouten J, Janssen R, Smeets H, van OJ, Prins M. Serotonin transporter polymorphisms and the occurrence of adverse events during treatment with selective serotonin reuptake inhibitors. Int.Clin.Psychopharmacol. 2007;22:137–143. doi: 10.1097/YIC.0b013e328014822a. [DOI] [PubMed] [Google Scholar]
- 101.Tadic A, Muller MJ, Rujescu D, Kohnen R, Stassen HH, Dahmen N, Szegedi A. The MAOA T941G polymorphism and short-term treatment response to mirtazapine and paroxetine in major depression. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:325–331. doi: 10.1002/ajmg.b.30462. [DOI] [PubMed] [Google Scholar]
- 102.Tsai SJ, Hong CJ, Chen TJ, Yu YW. Lack of supporting evidence for a genetic association of the FKBP5 polymorphism and response to antidepressant treatment. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:1097–1098. doi: 10.1002/ajmg.b.30246. [DOI] [PubMed] [Google Scholar]
- 103.Wilkie MJ, Smith D, Reid IC, Day RK, Matthews K, Wolf CR, Blackwood D, Smith GK, Wolf CR, Blackwood D, Smith G. A splice site polymorphism in the G-protein beta subunit influences antidepressant efficacy in depression. Pharmacogenet.Genomics. 2007;17:207–215. doi: 10.1097/FPC.0b013e32801a3be6. [DOI] [PubMed] [Google Scholar]
- 104.Yoshida K, Higuchi H, Kamata M, Takahashi H, Inoue K, Suzuki T, Itoh K, Ozaki N. The G196A polymorphism of the brain-derived neurotrophic factor gene and the antidepressant effect of milnacipran and fluvoxamine. J.Psychopharmacol. 2007;21:650–656. doi: 10.1177/0269881106072192. [DOI] [PubMed] [Google Scholar]
- 105.Benedetti F, Barbini B, Bernasconi A, Fulgosi MC, Campori E, Colombo C, Dallaspezia S, Lorenzi C, Pontiggia A, Radaelli D, Smeraldi E. Lithium overcomes the influence of 5-HTTLPR gene polymorphism on antidepressant response to sleep deprivation. J.Clin.Psychopharmacol. 2008;28:249–251. doi: 10.1097/JCP.0b013e318167461e. [DOI] [PubMed] [Google Scholar]
- 106.Lotrich FE, Pollock BG, Kirshner M, Ferrell RF, Reynolds IiiCF. Serotonin transporter genotype interacts with paroxetine plasma levels to influence depression treatment response in geriatric patients. J.Psychiatry Neurosci. 2008;33:123–130. [PMC free article] [PubMed] [Google Scholar]
- 107.Uhr M, Tontsch A, Namendorf C, Ripke S, Lucae S, Ising M, Dose T, Ebinger M, Rosenhagen M, Kohli M, Kloiber S, Salyakina D, Bettecken T, Specht M, Putz B, Binder EB, Muller-Myhsok B, Holsboer F. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203–209. doi: 10.1016/j.neuron.2007.11.017. [DOI] [PubMed] [Google Scholar]
- 108.Peters EJ, Slager SL, Kraft JB, Jenkins GD, Reinalda MS, McGrath PJ, Hamilton SP. Pharmacokinetic genes do not influence response or tolerance to citalopram in the STAR*D sample. PLoS.One. 2008;3:e1872. doi: 10.1371/journal.pone.0001872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Baune BT, Hohoff C, Roehrs T, Deckert J, Arolt V, Domschke K. Serotonin receptor 1A-1019C/G variant: impact on antidepressant pharmacoresponse in melancholic depression? Neurosci.Lett. 2008;436:111–115. doi: 10.1016/j.neulet.2008.03.001. [DOI] [PubMed] [Google Scholar]
- 110.Bozina N, Peles AM, Sagud M, Bilusic H, Jakovljevic M. Association study of paroxetine therapeutic response with SERT gene polymorphisms in patients with major depressive disorder. World J.Biol.Psychiatry. 2008;9:190–197. doi: 10.1080/15622970701308397. [DOI] [PubMed] [Google Scholar]
- 111.Gex-Fabry M, Eap CB, Oneda B, Gervasoni N, Aubry JM, Bondolfi G, Bertschy G. CYP2D6 and ABCB1 genetic variability: influence on paroxetine plasma level and therapeutic response. Ther.Drug Monit. 2008;30:474–482. doi: 10.1097/FTD.0b013e31817d6f5d. [DOI] [PubMed] [Google Scholar]
- 112.Gratacos M, Soria V, Urretavizcaya M, Gonzalez JR, Crespo JM, Bayes M, de CR, Menchon JM, Vallejo J, Estivill X. A brain-derived neurotrophic factor (BDNF) haplotype is associated with antidepressant treatment outcome in mood disorders. Pharmacogenomics.J. 2008;8:101–112. doi: 10.1038/sj.tpj.6500460. [DOI] [PubMed] [Google Scholar]
- 113.Kato M, Fukuda T, Serretti A, Wakeno M, Okugawa G, Ikenaga Y, Hosoi Y, Takekita Y, Mandelli L, Azuma J, Kinoshita T. ABCB1 (MDR1) gene polymorphisms are associated with the clinical response to paroxetine in patients with major depressive disorder. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:398–404. doi: 10.1016/j.pnpbp.2007.09.003. [DOI] [PubMed] [Google Scholar]
- 114.Kato M, Wakeno M, Okugawa G, Fukuda T, Takekita Y, Hosoi Y, Azuma J, Kinoshita T, Serretti A. Antidepressant response and intolerance to SSRI is not influenced by Gprotein beta3 subunit gene C825T polymorphism in Japanese major depressive patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:1041–1044. doi: 10.1016/j.pnpbp.2008.01.019. [DOI] [PubMed] [Google Scholar]
- 115.Lekman M, Laje G, Charney D, Rush AJ, Wilson AF, Sorant AJ, Lipsky R, Wisniewski SR, Manji H, McMahon FJ, Paddock S. The FKBP5-gene in depression and treatment response--an association study in the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Cohort. Biol.Psychiatry. 2008;63:1103–1110. doi: 10.1016/j.biopsych.2007.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mihaljevic PA, Bozina N, Sagud M, Rojnic KM, Lovric M. MDR1 gene polymorphism: therapeutic response to paroxetine among patients with major depression. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:1439–1444. doi: 10.1016/j.pnpbp.2008.03.018. [DOI] [PubMed] [Google Scholar]
- 117.Smits KM, Smits LJ, Peeters FP, Schouten JS, Janssen RG, Smeets HJ, van OJ, Prins MH. The influence of 5-HTTLPR and STin2 polymorphisms in the serotonin transporter gene on treatment effect of selective serotonin reuptake inhibitors in depressive patients. Psychiatr.Genet. 2008;18:184–190. doi: 10.1097/YPG.0b013e3283050aca. [DOI] [PubMed] [Google Scholar]
- 118.Tanaka M, Kobayashi D, Murakami Y, Ozaki N, Suzuki T, Iwata N, Haraguchi K, Ieiri I, Kinukawa N, Hosoi M, Ohtani H, Sawada Y, Mine K. Genetic polymorphisms in the 5-hydroxytryptamine type 3B receptor gene and paroxetine-induced nausea. Int.J.Neuropsychopharmacol. 2008;11:261–267. doi: 10.1017/S1461145707007985. [DOI] [PubMed] [Google Scholar]
- 119.Yoshida K, Higuchi H, Takahashi H, Kamata M, Sato K, Inoue K, Suzuki T, Itoh K, Ozaki N. Influence of the tyrosine hydroxylase val81met polymorphism and catechol-O-methyltransferase val158met polymorphism on the antidepressant effect of milnacipran. Hum.Psychopharmacol. 2008;23:121–128. doi: 10.1002/hup.907. [DOI] [PubMed] [Google Scholar]
- 120.Bishop JR, Ellingrod VL, Akroush M, Moline J. The association of serotonin transporter genotypes and selective serotonin reuptake inhibitor (SSRI)-associated sexual side effects: possible relationship to oral contraceptives. Hum.Psychopharmacol. 2009;24:207–215. doi: 10.1002/hup.1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wilkie MJ, Smith G, Day RK, Matthews K, Smith D, Blackwood D, Reid IC, Wolf CR. Polymorphisms in the SLC6A4 and HTR2A genes influence treatment outcome following antidepressant therapy. Pharmacogenomics.J. 2009;9:61–70. doi: 10.1038/sj.tpj.6500491. [DOI] [PubMed] [Google Scholar]
- 122.Arias B, Serretti A, Mandelli L, Gasto C, Catalan R, Ronchi DD, Fananas L. Dysbindin gene (DTNBP1) in major depression: association with clinical response to selective serotonin reuptake inhibitors. Pharmacogenet.Genomics. 2009;19:121–128. doi: 10.1097/FPC.0b013e32831ebb4b. [DOI] [PubMed] [Google Scholar]
- 123.Cabanero M, Laje G, Detera-Wadleigh S, McMahon FJ. Association study of phosphodiesterase genes in the Sequenced Treatment Alternatives to Relieve Depression sample. Pharmacogenet.Genomics. 2009;19:235–238. doi: 10.1097/FPC.0b013e328320a3e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Domschke K, Lawford B, Laje G, Berger K, Young R, Morris P, Deckert J, Arolt V, McMahon FJ, Baune BT. Brain-derived neurotrophic factor (BDNF) gene: no major impact on antidepressant treatment response. Int.J.Neuropsychopharmacol. 2009:1–9. doi: 10.1017/S1461145709000030. [DOI] [PubMed] [Google Scholar]
- 125.Kang R, Chang H, Wong M, Choi M, Park J, Lee H, Jung I, Joe S, Kim L, Kim S, Kim Y, Han C, Ham B, Lee H, Ko Y, Lee M, Lee M. Brain-derived neurotrophic factor gene polymorphisms and mirtazapine responses in Koreans with major depression. J.Psychopharmacol. 2009 doi: 10.1177/0269881109105457. [DOI] [PubMed] [Google Scholar]
- 126.Kato M, Fukuda T, Wakeno M, Okugawa G, Takekita Y, Watanabe S, Yamashita M, Hosoi Y, Azuma J, Kinoshita T, Serretti A. Effect of 5-HT1A gene polymorphisms on antidepressant response in major depressive disorder. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2009;150B:115–123. doi: 10.1002/ajmg.b.30783. [DOI] [PubMed] [Google Scholar]
- 127.Mrazek DA, Rush AJ, Biernacka JM, O'Kane DJ, Cunningham JM, Wieben ED, Schaid DJ, Drews MS, Courson VL, Snyder KA, Black JL, III, Weinshilboum RM. SLC6A4 variation and citalopram response. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2009;150B:341–351. doi: 10.1002/ajmg.b.30816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Perlis RH, Fijal B, Adams DH, Sutton VK, Trivedi MH, Houston JP. Variation in catechol-O-methyltransferase is associated with duloxetine response in a clinical trial for major depressive disorder. Biol.Psychiatry. 2009;65:785–791. doi: 10.1016/j.biopsych.2008.10.002. [DOI] [PubMed] [Google Scholar]
- 129.Perlis RH, Laje G, Smoller JW, Fava M, Rush AJ, McMahon FJ. Genetic and clinical predictors of sexual dysfunction in citalopram-treated depressed patients. Neuropsychopharmacology. 2009;34:1819–1828. doi: 10.1038/npp.2009.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Secher A, Bukh J, Bock C, Koefoed P, Rasmussen HB, Werge T, Kessing LV, Mellerup E. Antidepressive-drug-induced bodyweight gain is associated with polymorphisms in genes coding for COMT and TPH1. Int.Clin.Psychopharmacol. 2009;24:199–203. doi: 10.1097/YIC.0b013e32832d6be2. [DOI] [PubMed] [Google Scholar]
- 131.Laje G, Allen AS, Akula N, Manji H, John RA, McMahon FJ. Genome-wide association study of suicidal ideation emerging during citalopram treatment of depressed outpatients. Pharmacogenet.Genomics. 2009;19:666–674. doi: 10.1097/FPC.0b013e32832e4bcd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Garriock HA, Kraft JB, Shyn SI, Peters EJ, Yokoyama JS, Jenkins GD, Reinalda MS, Slager SL, McGrath PJ, Hamilton SP. A Genome-wide Association Study of Citalopram Response in Major Depressive Disorder. Biol.Psychiatry. 2009 doi: 10.1016/j.biopsych.2009.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ising M, Lucae S, Binder EB, Bettecken T, Uhr M, Ripke S, Kohli MA, Hennings JM, Horstmann S, Kloiber S, Menke A, Bondy B, Rupprecht R, Domschke K, Baune BT, Arolt V, Rush AJ, Holsboer F, Muller-Myhsok B. A genome-wide association study points to multiple loci that predict antidepressant drug treatment outcome in depression. Arch Gen.Psychiatry. 2009;66:966–975. doi: 10.1001/archgenpsychiatry.2009.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Brosen K. Some aspects of genetic polymorphism in the biotransformation of antidepressants. Therapie. 2004;59:5–12. doi: 10.2515/therapie:2004003. [DOI] [PubMed] [Google Scholar]
- 135.Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics.J. 2005;5:6–13. doi: 10.1038/sj.tpj.6500285. [DOI] [PubMed] [Google Scholar]
- 136.Bertilsson L, Dahl ML, Dalen P, Al-Shurbaji A. Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs. Br.J.Clin.Pharmacol. 2002;53:111–122. doi: 10.1046/j.0306-5251.2001.01548.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Madan A, Graham RA, Carroll KM, Mudra DR, Burton LA, Krueger LA, Downey AD, Czerwinski M, Forster J, Ribadeneira MD, Gan LS, LeCluyse EL, Zech K, Robertson P, Jr, Koch P, Antonian L, Wagner G, Yu L, Parkinson A. Effects of prototypical microsomal enzyme inducers on cytochrome P450 expression in cultured human hepatocytes. Drug Metab Dispos. 2003;31:421–431. doi: 10.1124/dmd.31.4.421. [DOI] [PubMed] [Google Scholar]
- 138.de LJ, Armstrong SC, Cozza KL. Clinical guidelines for psychiatrists for the use of pharmacogenetic testing for CYP450 2D6 and CYP450 2C19. Psychosomatics. 2006;47:75–85. doi: 10.1176/appi.psy.47.1.75. [DOI] [PubMed] [Google Scholar]
- 139.Nebert DW, Dieter MZ. The evolution of drug metabolism. Pharmacology. 2000;61:124–135. doi: 10.1159/000028393. [DOI] [PubMed] [Google Scholar]
- 140.de LJ. AmpliChip CYP450 test: personalized medicine has arrived in psychiatry. Expert.Rev.Mol.Diagn. 2006;6:277–286. doi: 10.1586/14737159.6.3.277. [DOI] [PubMed] [Google Scholar]
- 141.Thakur M, Grossman I, McCrory DC, Orlando LA, Steffens DC, Cline KE, Gray RN, Farmer J, DeJesus G, O'Brien C, Samsa G, Goldstein DB, Matchar DB. Review of evidence for genetic testing for CYP450 polymorphisms in management of patients with nonpsychotic depression with selective serotonin reuptake inhibitors. Genet.Med. 2007;9:826–835. doi: 10.1097/gim.0b013e31815bf98f. [DOI] [PubMed] [Google Scholar]
- 142.Heils A, Teufel A, Petri S, Stober G, Riederer P, Bengel D, Lesch KP. Allelic variation of human serotonin transporter gene expression. J.Neurochem. 1996;66:2621–2624. doi: 10.1046/j.1471-4159.1996.66062621.x. [DOI] [PubMed] [Google Scholar]
- 143.Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, Xu K, Arnold PD, Richter MA, Kennedy JL, Murphy DL, Goldman D. Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am.J.Hum.Genet. 2006;78:815–826. doi: 10.1086/503850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Hranilovic D, Stefulj J, Schwab S, Borrmann-Hassenbach M, Albus M, Jernej B, Wildenauer D. Serotonin transporter promoter and intron 2 polymorphisms: relationship between allelic variants and gene expression. Biol.Psychiatry. 2004;55:1090–1094. doi: 10.1016/j.biopsych.2004.01.029. [DOI] [PubMed] [Google Scholar]
- 145.Serretti A, Kato M, De RD, Kinoshita T. Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with selective serotonin reuptake inhibitor efficacy in depressed patients. Mol.Psychiatry. 2007;12:247–257. doi: 10.1038/sj.mp.4001926. [DOI] [PubMed] [Google Scholar]
- 146.Kato M, Serretti A. Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol.Psychiatry. 2008 doi: 10.1038/mp.2008.116. [DOI] [PubMed] [Google Scholar]
- 147.Walther DJ, Peter JU, Bashammakh S, Hortnagl H, Voits M, Fink H, Bader M. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science. 2003;299:76. doi: 10.1126/science.1078197. [DOI] [PubMed] [Google Scholar]
- 148.Nakamura K, Sugawara Y, Sawabe K, Ohashi A, Tsurui H, Xiu Y, Ohtsuji M, Lin QS, Nishimura H, Hasegawa H, Hirose S. Late developmental stage-specific role of tryptophan hydroxylase 1 in brain serotonin levels. J.Neurosci. 2006;26:530–534. doi: 10.1523/JNEUROSCI.1835-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Binder DK, Scharfman HE. Brain-derived neurotrophic factor. Growth Factors. 2004;22:123–131. doi: 10.1080/08977190410001723308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Shorter E. The history of lithium therapy. Bipolar.Disord. 2009;11 Suppl 2:4–9. doi: 10.1111/j.1399-5618.2009.00706.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Grof P, Muller-Oerlinghausen B. A critical appraisal of lithium's efficacy and effectiveness: the last 60 years. Bipolar.Disord. 2009;11 Suppl 2:10–19. doi: 10.1111/j.1399-5618.2009.00707.x. [DOI] [PubMed] [Google Scholar]
- 152.Young AH, Hammond JM. Lithium in mood disorders: increasing evidence base, declining use? Br.J.Psychiatry. 2007;191:474–476. doi: 10.1192/bjp.bp.107.043133. [DOI] [PubMed] [Google Scholar]
- 153.Coryell W. Maintenance treatment in bipolar disorder: a reassessment of lithium as the first choice. Bipolar.Disord. 2009;11 Suppl 2:77–83. doi: 10.1111/j.1399-5618.2009.00712.x. [DOI] [PubMed] [Google Scholar]
- 154.Cavazzoni P, Alda M, Turecki G, Rouleau G, Grof E, Martin R, Duffy A, Grof P. Lithium-responsive affective disorders: no association with the tyrosine hydroxylase gene. Psychiatry Res. 1996;64:91–96. doi: 10.1016/0165-1781(96)02888-0. [DOI] [PubMed] [Google Scholar]
- 155.Turecki G, Alda M, Grof P, Martin R, Cavazzoni PA, Duffy A, Maciel P, Rouleau GA. No association between chromosome-18 markers and lithium-responsive affective disorders. Psychiatry Res. 1996;63:17–23. doi: 10.1016/0165-1781(96)02864-8. [DOI] [PubMed] [Google Scholar]
- 156.Steen VM, Gulbrandsen AK, Eiken HG, Berle JO. Lack of genetic variation in the coding region of the myo-inositol monophosphatase gene in lithium-treated patients with manic depressive illness. Pharmacogenetics. 1996;6:113–116. doi: 10.1097/00008571-199602000-00010. [DOI] [PubMed] [Google Scholar]
- 157.Turecki G, Grof P, Cavazzoni P, Duffy A, Grof E, Ahrens B, Berghofer A, Muller-Oerlinghausen B, Dvorakova M, Libigerova E, Vojtechovsky M, Zvolsky P, Joober R, Nilsson A, Prochazka H, Licht RW, Rasmussen NA, Schou M, Vestergaard P, Holzinger A, Schumann C, Thau K, Rouleau GA, Alda M. Evidence for a role of phospholipase C-gamma1 in the pathogenesis of bipolar disorder. Mol.Psychiatry. 1998;3:534–538. doi: 10.1038/sj.mp.4000447. [DOI] [PubMed] [Google Scholar]
- 158.Alda M, Grof P, Grof E. MN blood groups and bipolar disorder: evidence of genotypic association and Hardy-Weinberg disequilibrium. Biol.Psychiatry. 1998;44:361–363. doi: 10.1016/s0006-3223(98)00013-4. [DOI] [PubMed] [Google Scholar]
- 159.Steen VM, Lovlie R, Osher Y, Belmaker RH, Berle JO, Gulbrandsen AK. The polymorphic inositol polyphosphate 1-phosphatase gene as a candidate for pharmacogenetic prediction of lithium-responsive manic-depressive illness. Pharmacogenetics. 1998;8:259–268. [PubMed] [Google Scholar]
- 160.Serretti A, Lilli R, Lorenzi C, Franchini L, Smeraldi E. Dopamine receptor D3 gene and response to lithium prophylaxis in mood disorders. Int.J.Neuropsychopharmacol. 1998;1:125–129. doi: 10.1017/S1461145798001230. [DOI] [PubMed] [Google Scholar]
- 161.Turecki G, Grof P, Cavazzoni P, Duffy A, Grof E, Ahrens B, Berghofer A, Muller-Oerlinghausen B, Dvorakova M, Libigerova E, Vojtechovsky M, Zvolsky P, Joober R, Nilsson A, Prochazka H, Licht RW, Rasmussen NA, Schou M, Vestergaard P, Holzinger A, Schumann C, Thau K, Rouleau GA, Alda M. MAOA: association and linkage studies with lithium responsive bipolar disorder. Psychiatr.Genet. 1999;9:13–16. [PubMed] [Google Scholar]
- 162.Serretti A, Lilli R, Lorenzi C, Franchini L, Di BD, Catalano M, Smeraldi E. Dopamine receptor D2 and D4 genes, GABA(A) alpha-1 subunit genes and response to lithium prophylaxis in mood disorders. Psychiatry Res. 1999;87:7–19. doi: 10.1016/s0165-1781(99)00056-6. [DOI] [PubMed] [Google Scholar]
- 163.Serretti A, Lilli R, Lorenzi C, Gasperini M, Smeraldi E. Tryptophan hydroxylase gene and response to lithium prophylaxis in mood disorders. Tryptophan hydroxylase gene and response to lithium prophylaxis in mood disorders. J.Psychiatr.Res. 1999;33:371–377. doi: 10.1016/s0022-3956(99)00013-8. [DOI] [PubMed] [Google Scholar]
- 164.Duffy A, Turecki G, Grof P, Cavazzoni P, Grof E, Joober R, Ahrens B, Berghofer A, Muller-Oerlinghausen B, Dvorakova M, Libigerova E, Vojtechovsky M, Zvolsky P, Nilsson A, Licht RW, Rasmussen NA, Schou M, Vestergaard P, Holzinger A, Schumann C, Thau K, Robertson C, Rouleau GA, Alda M. Association and linkage studies of candidate genes involved in GABAergic neurotransmission in lithium-responsive bipolar disorder. J.Psychiatry Neurosci. 2000;25:353–358. [PMC free article] [PubMed] [Google Scholar]
- 165.Alda M, Turecki G, Grof P, Cavazzoni P, Duffy A, Grof E, Ahrens B, Berghofer A, Muller-Oerlinghausen B, Dvorakova M, Libigerova E, Vojtechovsky M, Zvolsky P, Joober R, Nilsson A, Prochazka H, Licht RW, Rasmussen NA, Schou M, Vestergaard P, Holzinger A, Schumann C, Thau K, Rouleau GA. Association and linkage studies of CRH and PENK genes in bipolar disorder: a collaborative IGSLI study. Am.J.Med.Genet. 2000;96:178–181. doi: 10.1002/(sici)1096-8628(20000403)96:2<178::aid-ajmg11>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 166.Serretti A, Lorenzi C, Lilli R, Smeraldi E. Serotonin receptor 2A, 2C, 1A genes and response to lithium prophylaxis in mood disorders. J.Psychiatr.Res. 2000;34:89–98. doi: 10.1016/s0022-3956(00)00004-2. [DOI] [PubMed] [Google Scholar]
- 167.Ftouhi-Paquin N, Alda M, Grof P, Chretien N, Rouleau G, Turecki G. Identification of three polymorphisms in the translated region of PLC-gamma1 and their investigation in lithium responsive bipolar disorder. Am.J.Med.Genet. 2001;105:301–305. doi: 10.1002/ajmg.1326. [DOI] [PubMed] [Google Scholar]
- 168.Serretti A, Lilli R, Mandelli L, Lorenzi C, Smeraldi E. Serotonin transporter gene associated with lithium prophylaxis in mood disorders. Pharmacogenomics.J. 2001;1:71–77. doi: 10.1038/sj.tpj.6500006. [DOI] [PubMed] [Google Scholar]
- 169.Lovlie R, Berle JO, Stordal E, Steen VM. The phospholipase C-gamma1 gene (PLCG1) and lithium-responsive bipolar disorder: re-examination of an intronic dinucleotide repeat polymorphism. Psychiatr.Genet. 2001;11:41–43. doi: 10.1097/00041444-200103000-00008. [DOI] [PubMed] [Google Scholar]
- 170.Serretti A, Lorenzi C, Lilli R, Mandelli L, Pirovano A, Smeraldi E. Pharmacogenetics of lithium prophylaxis in mood disorders: analysis of COMT, MAO-A, and Gbeta3 variants. Am.J.Med.Genet. 2002;114:370–379. doi: 10.1002/ajmg.10357. [DOI] [PubMed] [Google Scholar]
- 171.Washizuka S, Ikeda A, Kato N, Kato T. Possible relationship between mitochondrial DNA polymorphisms and lithium response in bipolar disorder. Int.J.Neuropsychopharmacol. 2003;6:421–424. doi: 10.1017/S1461145703003778. [DOI] [PubMed] [Google Scholar]
- 172.Dmitrzak-Weglarz M, Rybakowski JK, Suwalska A, Slopien A, Czerski PM, Leszczynska-Rodziewicz A, Hauser J. Association studies of 5-HT2A and 5-HT2C serotonin receptor gene polymorphisms with prophylactic lithium response in bipolar patients. Pharmacol.Rep. 2005;57:761–765. [PubMed] [Google Scholar]
- 173.Rybakowski JK, Suwalska A, Skibinska M, Szczepankiewicz A, Leszczynska-Rodziewicz A, Permoda A, Czerski PM, Hauser J. Prophylactic lithium response and polymorphism of the brain-derived neurotrophic factor gene. Pharmacopsychiatry. 2005;38:166–170. doi: 10.1055/s-2005-871239. [DOI] [PubMed] [Google Scholar]
- 174.Rybakowski JK, Suwalska A, Czerski PM, Dmitrzak-Weglarz M, Leszczynska-Rodziewicz A, Hauser J. Prophylactic effect of lithium in bipolar affective illness may be related to serotonin transporter genotype. Pharmacol.Rep. 2005;57:124–127. [PubMed] [Google Scholar]
- 175.Benedetti F, Serretti A, Pontiggia A, Bernasconi A, Lorenzi C, Colombo C, Smeraldi E. Long-term response to lithium salts in bipolar illness is influenced by the glycogen synthase kinase 3-beta −50 T/C SNP. Neurosci.Lett. 2005;376:51–55. doi: 10.1016/j.neulet.2004.11.022. [DOI] [PubMed] [Google Scholar]
- 176.Szczepankiewicz A, Rybakowski JK, Suwalska A, Skibinska M, Leszczynska-Rodziewicz A, Dmitrzak-Weglarz M, Czerski PM, Hauser J. Association study of the glycogen synthase kinase-3beta gene polymorphism with prophylactic lithium response in bipolar patients. World J.Biol.Psychiatry. 2006;7:158–161. doi: 10.1080/15622970600554711. [DOI] [PubMed] [Google Scholar]
- 177.Masui T, Hashimoto R, Kusumi I, Suzuki K, Tanaka T, Nakagawa S, Suzuki T, Iwata N, Ozaki N, Kato T, Kunugi H, Koyama T. Lithium response and Val66Met polymorphism of the brain-derived neurotrophic factor gene in Japanese patients with bipolar disorder. Psychiatr.Genet. 2006;16:49–50. doi: 10.1097/01.ypg.0000180680.72922.57. [DOI] [PubMed] [Google Scholar]
- 178.Masui T, Hashimoto R, Kusumi I, Suzuki K, Tanaka T, Nakagawa S, Kunugi H, Koyama T. A possible association between the −116C/G single nucleotide polymorphism of the XBP1 gene and lithium prophylaxis in bipolar disorder. Int.J.Neuropsychopharmacol. 2006;9:83–88. doi: 10.1017/S1461145705005523. [DOI] [PubMed] [Google Scholar]
- 179.Michelon L, Meira-Lima I, Cordeiro Q, Miguita K, Breen G, Collier D, Vallada H. Association study of the INPP1, 5HTT, BDNF, AP-2beta and GSK-3beta GENE variants and restrospectively scored response to lithium prophylaxis in bipolar disorder. Neurosci.Lett. 2006;403:288–293. doi: 10.1016/j.neulet.2006.05.001. [DOI] [PubMed] [Google Scholar]
- 180.Mamdani F, Sequeira A, Alda M, Grof P, Rouleau G, Turecki G. No association between the PREP gene and lithium responsive bipolar disorder. BMC.Psychiatry. 2007;7:9. doi: 10.1186/1471-244X-7-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Bremer T, Diamond C, McKinney R, Shehktman T, Barrett TB, Herold C, Kelsoe JR. The pharmacogenetics of lithium response depends upon clinical co-morbidity. Mol.Diagn.Ther. 2007;11:161–170. doi: 10.1007/BF03256238. [DOI] [PubMed] [Google Scholar]
- 182.Rybakowski JK, Suwalska A, Skibinska M, Dmitrzak-Weglarz M, Leszczynska-Rodziewicz A, Hauser J. Response to lithium prophylaxis: interaction between serotonin transporter and BDNF genes. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:820–823. doi: 10.1002/ajmg.b.30420. [DOI] [PubMed] [Google Scholar]
- 183.Mamdani F, Alda M, Grof P, Young LT, Rouleau G, Turecki G. Lithium response and genetic variation in the CREB family of genes. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2008;147B:500–504. doi: 10.1002/ajmg.b.30617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Dmitrzak-Weglarz M, Rybakowski JK, Suwalska A, Skibinska M, Leszczynska-Rodziewicz A, Szczepankiewicz A, Hauser J. Association studies of the BDNF and the NTRK2 gene polymorphisms with prophylactic lithium response in bipolar patients. Pharmacogenomics. 2008;9:1595–1603. doi: 10.2217/14622416.9.11.1595. [DOI] [PubMed] [Google Scholar]
- 185.Masui T, Hashimoto R, Kusumi I, Suzuki K, Tanaka T, Nakagawa S, Suzuki T, Iwata N, Ozaki N, Kato T, Takeda M, Kunugi H, Koyama T. A possible association between missense polymorphism of the breakpoint cluster region gene and lithium prophylaxis in bipolar disorder. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:204–208. doi: 10.1016/j.pnpbp.2007.08.010. [DOI] [PubMed] [Google Scholar]
- 186.Kim B, Kim CY, Lee MJ, Joo YH. Preliminary evidence on the association between XBP1-116C/G polymorphism and response to prophylactic treatment with valproate in bipolar disorders. Psychiatry Res. 2009;168:209–212. doi: 10.1016/j.psychres.2008.05.010. [DOI] [PubMed] [Google Scholar]
- 187.Szczepankiewicz A, Skibinska M, Suwalska A, Hauser J, Rybakowski JK. No association of three GRIN2B polymorphisms with lithium response in bipolar patients. Pharmacol.Rep. 2009;61:448–452. doi: 10.1016/s1734-1140(09)70085-4. [DOI] [PubMed] [Google Scholar]
- 188.Szczepankiewicz A, Skibinska M, Suwalska A, Hauser J, Rybakowski JK. The association study of three FYN polymorphisms with prophylactic lithium response in bipolar patients. Hum.Psychopharmacol. 2009;24:287–291. doi: 10.1002/hup.1018. [DOI] [PubMed] [Google Scholar]
- 189.Rybakowski JK, Dmitrzak-Weglarz M, Suwalska A, Leszczynska-Rodziewicz A, Hauser J. Dopamine D1 receptor gene polymorphism is associated with prophylactic lithium response in bipolar disorder. Pharmacopsychiatry. 2009;42:20–22. doi: 10.1055/s-0028-1085441. [DOI] [PubMed] [Google Scholar]
- 190.Perlis RH, Smoller JW, Ferreira MA, McQuillin A, Bass N, Lawrence J, Sachs GS, Nimgaonkar V, Scolnick EM, Gurling H, Sklar P, Purcell S. A genome-wide association study of response to lithium for prevention of recurrence in bipolar disorder. Am.J.Psychiatry. 2009;166:718–725. doi: 10.1176/appi.ajp.2009.08111633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Berridge MJ, Downes CP, Hanley MR. Neural and developmental actions of lithium: a unifying hypothesis. Cell. 1989;59:411–419. doi: 10.1016/0092-8674(89)90026-3. [DOI] [PubMed] [Google Scholar]
- 192.Harwood AJ. Lithium and bipolar mood disorder: the inositol-depletion hypothesis revisited. Mol.Psychiatry. 2005;10:117–126. doi: 10.1038/sj.mp.4001618. [DOI] [PubMed] [Google Scholar]
- 193.Klein PS, Melton DA. A molecular mechanism for the effect of lithium on development. Proc.Natl.Acad.Sci.U.S.A. 1996;93:8455–8459. doi: 10.1073/pnas.93.16.8455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Williams RS, Harwood AJ. Lithium therapy and signal transduction. Trends Pharmacol.Sci. 2000;21:61–64. doi: 10.1016/s0165-6147(99)01428-5. [DOI] [PubMed] [Google Scholar]
- 195.Jope RS, Bijur GN. Mood stabilizers, glycogen synthase kinase-3beta and cell survival. Mol.Psychiatry. 2002;7 Suppl 1:S35–S45. doi: 10.1038/sj.mp.4001017. [DOI] [PubMed] [Google Scholar]
- 196.Chen G, Huang LD, Jiang YM, Manji HK. The mood-stabilizing agent valproate inhibits the activity of glycogen synthase kinase-3. J.Neurochem. 1999;72:1327–1330. doi: 10.1046/j.1471-4159.2000.0721327.x. [DOI] [PubMed] [Google Scholar]
- 197.Bajjalieh SM, Peterson K, Linial M, Scheller RH. Brain contains two forms of synaptic vesicle protein 2. Proc.Natl.Acad.Sci.U.S.A. 1993;90:2150–2154. doi: 10.1073/pnas.90.6.2150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Seelan RS, Khalyfa A, Lakshmanan J, Casanova MF, Parthasarathy RN. Deciphering the lithium transcriptome: microarray profiling of lithium-modulated gene expression in human neuronal cells. Neuroscience. 2008;151:1184–1197. doi: 10.1016/j.neuroscience.2007.10.045. [DOI] [PubMed] [Google Scholar]
- 199.Ben-Zur T, Feige E, Motro B, Wides R. The mammalian Odz gene family: homologs of a Drosophila pair-rule gene with expression implying distinct yet overlapping developmental roles. Dev.Biol. 2000;217:107–120. doi: 10.1006/dbio.1999.9532. [DOI] [PubMed] [Google Scholar]
- 200.Shen WW. A history of antipsychotic drug development. Compr.Psychiatry. 1999;40:407–414. doi: 10.1016/s0010-440x(99)90082-2. [DOI] [PubMed] [Google Scholar]
- 201.Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment-resistant schizophrenic. A double-blind comparison with chlorpromazine. Arch Gen.Psychiatry. 1988;45:789–796. doi: 10.1001/archpsyc.1988.01800330013001. [DOI] [PubMed] [Google Scholar]
- 202.Leucht S, Corves C, Arbter D, Engel RR, Li C, Davis JM. Second-generation versus firstgeneration antipsychotic drugs for schizophrenia: a meta-analysis. Lancet. 2009;373:31–41. doi: 10.1016/S0140-6736(08)61764-X. [DOI] [PubMed] [Google Scholar]
- 203.Sharif Z. Side effects as influencers of treatment outcome. J.Clin.Psychiatry. 2008;69 Suppl 3:38–43. [PubMed] [Google Scholar]
- 204.Arranz M, Collier D, Sodhi M, Ball D, Roberts G, Price J, Sham P, Kerwin R. Association between clozapine response and allelic variation in 5-HT2A receptor gene. Lancet. 1995;346:281–282. doi: 10.1016/s0140-6736(95)92168-0. [DOI] [PubMed] [Google Scholar]
- 205.Arranz MJ, Dawson E, Shaikh S, Sham P, Sharma T, Aitchison K, Crocq MA, Gill M, Kerwin R, Collier DA. Cytochrome P4502D6 genotype does not determine response to clozapine. Br.J.Clin.Pharmacol. 1995;39:417–420. doi: 10.1111/j.1365-2125.1995.tb04471.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Masellis M, Paterson AD, Badri F, Lieberman JA, Meltzer HY, Cavazzoni P, Kennedy JL. Genetic variation of 5-HT2A receptor and response to clozapine. Lancet. 1995;346:1108. doi: 10.1016/s0140-6736(95)91785-3. [DOI] [PubMed] [Google Scholar]
- 207.Nothen MM, Rietschel M, Erdmann J, Oberlander H, Moller HJ, Nober D, Propping P. Genetic variation of the 5-HT2A receptor and response to clozapine. Lancet. 1995;346:908–909. doi: 10.1016/s0140-6736(95)92756-5. [DOI] [PubMed] [Google Scholar]
- 208.Sodhi MS, Arranz MJ, Curtis D, Ball DM, Sham P, Roberts GW, Price J, Collier DA, Kerwin RW. Association between clozapine response and allelic variation in the 5-HT2C receptor gene. Neuroreport. 1995;7:169–172. [PubMed] [Google Scholar]
- 209.Malhotra AK, Goldman D, Ozaki N, Breier A, Buchanan R, Pickar D. Lack of association between polymorphisms in the 5-HT2A receptor gene and the antipsychotic response to clozapine. Am.J.Psychiatry. 1996;153:1092–1094. doi: 10.1176/ajp.153.8.1092. [DOI] [PubMed] [Google Scholar]
- 210.Arranz MJ, Collier DA, Munro J, Sham P, Kirov G, Sodhi M, Roberts G, Price J, Kerwin RW. Analysis of a structural polymorphism in the 5-HT2A receptor and clinical response to clozapine. Neurosci.Lett. 1996;217:177–178. [PubMed] [Google Scholar]
- 211.Gaitonde EJ, Morris A, Sivagnanasundaram S, McKenna PJ, Hunt DM, Mollon JD. Assessment of association of D3 dopamine receptor MscI polymorphism with schizophrenia: analysis of symptom ratings, family history, age at onset, and movement disorders. Am.J.Med.Genet. 1996;67:455–458. doi: 10.1002/(SICI)1096-8628(19960920)67:5<455::AID-AJMG3>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 212.Jonsson E, Nothen MM, Bunzel R, Propping P, Sedvall G. 5HT 2a receptor T102C polymorphism and schizophrenia. Lancet. 1996;347:1831. [PubMed] [Google Scholar]
- 213.Rietschel M, Naber D, Oberlander H, Holzbach R, Fimmers R, Eggermann K, Moller HJ, Propping P, Nothen MM. Efficacy and side-effects of clozapine: testing for association with allelic variation in the dopamine D4 receptor gene. Neuropsychopharmacology. 1996;15:491–496. doi: 10.1016/S0893-133X(96)00090-5. [DOI] [PubMed] [Google Scholar]
- 214.Rietschel M, Naber D, Fimmers R, Moller HJ, Propping P, Nothen MM. Efficacy and side-effects of clozapine not associated with variation in the 5-HT2C receptor. Neuroreport. 1997;8:1999–2003. doi: 10.1097/00001756-199705260-00040. [DOI] [PubMed] [Google Scholar]
- 215.Steen VM, Lovlie R, MacEwan T, McCreadie RG. Dopamine D3-receptor gene variant and susceptibility to tardive dyskinesia in schizophrenic patients. Mol.Psychiatry. 1997;2:139–145. doi: 10.1038/sj.mp.4000249. [DOI] [PubMed] [Google Scholar]
- 216.Turbay D, Lieberman J, Alper CA, Delgado JC, Corzo D, Yunis JJ, Yunis EJ. Tumor necrosis factor constellation polymorphism and clozapine-induced agranulocytosis in two different ethnic groups. Blood. 1997;89:4167–4174. [PubMed] [Google Scholar]
- 217.Amar A, Segman RH, Shtrussberg S, Sherman L, Safirman C, Lerer B, Brautbar C. An association between clozapine-induced agranulocytosis in schizophrenics and HLADQB1*0201. Int.J.Neuropsychopharmacol. 1998;1:41–44. doi: 10.1017/S1461145798001023. [DOI] [PubMed] [Google Scholar]
- 218.Arranz MJ, Munro J, Owen MJ, Spurlock G, Sham PC, Zhao J, Kirov G, Collier DA, Kerwin RW. Evidence for association between polymorphisms in the promoter and coding regions of the 5-HT2A receptor gene and response to clozapine. Mol.Psychiatry. 1998;3:61–66. doi: 10.1038/sj.mp.4000348. [DOI] [PubMed] [Google Scholar]
- 219.Hwu HG, Hong CJ, Lee YL, Lee PC, Lee SF. Dopamine D4 receptor gene polymorphisms and neuroleptic response in schizophrenia. Biol.Psychiatry. 1998;44:483–487. doi: 10.1016/s0006-3223(98)00134-6. [DOI] [PubMed] [Google Scholar]
- 220.Masellis M, Basile V, Meltzer HY, Lieberman JA, Sevy S, Macciardi FM, Cola P, Howard A, Badri F, Nothen MM, Kalow W, Kennedy JL. Serotonin subtype 2 receptor genes and clinical response to clozapine in schizophrenia patients. Neuropsychopharmacology. 1998;19:123–132. doi: 10.1016/S0893-133X(98)00007-4. [DOI] [PubMed] [Google Scholar]
- 221.Valevski A, Klein T, Gazit E, Meged S, Stein D, Elizur A, Narinsky ER, Kutzuk D, Weizman A. HLA-B38 and clozapine-induced agranulocytosis in Israeli Jewish schizophrenic patients. Eur.J.Immunogenet. 1998;25:11–13. doi: 10.1046/j.1365-2370.1998.00091.x. [DOI] [PubMed] [Google Scholar]
- 222.Aitchison KJ, Munro J, Wright P, Smith S, Makoff AJ, Sachse C, Sham PC, Murray RM, Collier DA, Kerwin RW. Failure to respond to treatment with typical antipsychotics is not associated with CYP2D6 ultrarapid hydroxylation. Br.J.Clin.Pharmacol. 1999;48:388–394. doi: 10.1046/j.1365-2125.1999.00006.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Basile VS, Masellis M, Badri F, Paterson AD, Meltzer HY, Lieberman JA, Potkin SG, Macciardi F, Kennedy JL. Association of the MscI polymorphism of the dopamine D3 receptor gene with tardive dyskinesia in schizophrenia. Neuropsychopharmacology. 1999;21:17–27. doi: 10.1016/S0893-133X(98)00114-6. [DOI] [PubMed] [Google Scholar]
- 224.Cohen BM, Ennulat DJ, Centorrino F, Matthysse S, Konieczna H, Chu HM, Cherkerzian S. Polymorphisms of the dopamine D4 receptor and response to antipsychotic drugs. Psychopharmacology (Berl) 1999;141:6–10. doi: 10.1007/s002130050799. [DOI] [PubMed] [Google Scholar]
- 225.Joober R, Benkelfat C, Brisebois K, Toulouse A, Turecki G, Lal S, Bloom D, Labelle A, Lalonde P, Fortin D, Alda M, Palmour R, Rouleau GA. T102C polymorphism in the 5HT2A gene and schizophrenia: relation to phenotype and drug response variability. J.Psychiatry Neurosci. 1999;24:141–146. [PMC free article] [PubMed] [Google Scholar]
- 226.Lin CH, Tsai SJ, Yu YW, Song HL, Tu PC, Sim CB, Hsu CP, Yang KH, Hong CJ. No evidence for association of serotonin-2A receptor variant (102T/C) with schizophrenia or clozapine response in a Chinese population. Neuroreport. 1999;10:57–60. doi: 10.1097/00001756-199901180-00011. [DOI] [PubMed] [Google Scholar]
- 227.Scharfetter J, Chaudhry HR, Hornik K, Fuchs K, Sieghart W, Kasper S, Aschauer HN. Dopamine D3 receptor gene polymorphism and response to clozapine in schizophrenic Pakastani patients. Eur.Neuropsychopharmacol. 1999;10:17–20. doi: 10.1016/s0924-977x(99)00044-9. [DOI] [PubMed] [Google Scholar]
- 228.Segman R, Neeman T, Heresco-Levy U, Finkel B, Karagichev L, Schlafman M, Dorevitch A, Yakir A, Lerner A, Shelevoy A, Lerer B. Genotypic association between the dopamine D3 receptor and tardive dyskinesia in chronic schizophrenia. Mol.Psychiatry. 1999;4:247–253. doi: 10.1038/sj.mp.4000511. [DOI] [PubMed] [Google Scholar]
- 229.Yu YW, Tsai SJ, Lin CH, Hsu CP, Yang KH, Hong CJ. Serotonin-6 receptor variant (C267T) and clinical response to clozapine. Neuroreport. 1999;10:1231–1233. doi: 10.1097/00001756-199904260-00014. [DOI] [PubMed] [Google Scholar]
- 230.Arranz MJ, Munro J, Birkett J, Bolonna A, Mancama D, Sodhi M, Lesch KP, Meyer JF, Sham P, Collier DA, Murray RM, Kerwin RW. Pharmacogenetic prediction of clozapine response. Lancet. 2000;355:1615–1616. doi: 10.1016/s0140-6736(00)02221-2. [DOI] [PubMed] [Google Scholar]
- 231.Arranz MJ, Bolonna AA, Munro J, Curtis CJ, Collier DA, Kerwin RW. The serotonin transporter and clozapine response. Mol.Psychiatry. 2000;5:124–125. doi: 10.1038/sj.mp.4000652. [DOI] [PubMed] [Google Scholar]
- 232.Basile VS, Ozdemir V, Masellis M, Walker ML, Meltzer HY, Lieberman JA, Potkin SG, Alva G, Kalow W, Macciardi FM, Kennedy JL. A functional polymorphism of the cytochrome P450 1A2 (CYP1A2) gene: association with tardive dyskinesia in schizophrenia. Mol.Psychiatry. 2000;5:410–417. doi: 10.1038/sj.mp.4000736. [DOI] [PubMed] [Google Scholar]
- 233.Birkett JT, Arranz MJ, Munro J, Osbourn S, Kerwin RW, Collier DA. Association analysis of the 5-HT5A gene in depression, psychosis and antipsychotic response. Neuroreport. 2000;11:2017–2020. doi: 10.1097/00001756-200006260-00042. [DOI] [PubMed] [Google Scholar]
- 234.Bolonna AA, Arranz MJ, Munro J, Osborne S, Petouni M, Martinez M, Kerwin RW. No influence of adrenergic receptor polymorphisms on schizophrenia and antipsychotic response. Neurosci.Lett. 2000;280:65–68. doi: 10.1016/s0304-3940(99)01000-9. [DOI] [PubMed] [Google Scholar]
- 235.Chong SA, Tan EC, Tan CH, Mahendren R, Tay AH, Chua HC. Tardive dyskinesia is not associated with the serotonin gene polymorphism (5-HTTLPR) in Chinese. Am.J.Med.Genet. 2000;96:712–715. doi: 10.1002/1096-8628(20001204)96:6<712::aid-ajmg2>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 236.Dettling M, Sachse C, Muller-Oerlinghausen B, Roots I, Brockmoller J, Rolfs A, Cascorbi I. Clozapine-induced agranulocytosis and hereditary polymorphisms of clozapine metabolizing enzymes: no association with myeloperoxidase and cytochrome P4502D6. Pharmacopsychiatry. 2000;33:218–220. doi: 10.1055/s-2000-8359. [DOI] [PubMed] [Google Scholar]
- 237.Eichhammer P, Albus M, Borrmann-Hassenbach M, Schoeler A, Putzhammer A, Frick U, Klein HE, Rohrmeier T. Association of dopamine D3-receptor gene variants with neuroleptic induced akathisia in schizophrenic patients: a generalization of Steen's study on DRD3 and tardive dyskinesia. Am.J.Med.Genet. 2000;96:187–191. doi: 10.1002/(sici)1096-8628(20000403)96:2<187::aid-ajmg13>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 238.Joober R, Toulouse A, Benkelfat C, Lal S, Bloom D, Labelle A, Lalonde P, Turecki G, Rouleau GA. DRD3 and DAT1 genes in schizophrenia: an association study. J.Psychiatr.Res. 2000;34:285–291. doi: 10.1016/s0022-3956(00)00018-2. [DOI] [PubMed] [Google Scholar]
- 239.Joober R, Benkelfat C, Lal S, Bloom D, Labelle A, Lalonde P, Turecki G, Rozen R, Rouleau GA. Association between the methylenetetrahydrofolate reductase 677C-->T missense mutation and schizophrenia. Mol.Psychiatry. 2000;5:323–326. doi: 10.1038/sj.mp.4000724. [DOI] [PubMed] [Google Scholar]
- 240.Kaiser R, Konneker M, Henneken M, Dettling M, Muller-Oerlinghausen B, Roots I, Brockmoller J. Dopamine D4 receptor 48-bp repeat polymorphism: no association with response to antipsychotic treatment, but association with catatonic schizophrenia. Mol.Psychiatry. 2000;5:418–424. doi: 10.1038/sj.mp.4000729. [DOI] [PubMed] [Google Scholar]
- 241.Krebs MO, Guillin O, Bourdell MC, Schwartz JC, Olie JP, Poirier MF, Sokoloff P. Brain derived neurotrophic factor (BDNF) gene variants association with age at onset and therapeutic response in schizophrenia. Mol.Psychiatry. 2000;5:558–562. doi: 10.1038/sj.mp.4000749. [DOI] [PubMed] [Google Scholar]
- 242.Lovlie R, Daly AK, Blennerhassett R, Ferrier N, Steen VM. Homozygosity for the Gly-9 variant of the dopamine D3 receptor and risk for tardive dyskinesia in schizophrenic patients. Int.J.Neuropsychopharmacol. 2000;3:61–65. doi: 10.1017/S1461145700001796. [DOI] [PubMed] [Google Scholar]
- 243.Mihara K, Suzuki A, Kondo T, Nagashima U, Ono S, Otani K, Kaneko S. No relationship between Taq1 a polymorphism of dopamine D(2) receptor gene and extrapyramidal adverse effects of selective dopamine D(2) antagonists, bromperidol, and nemonapride in schizophrenia: a preliminary study. Am.J.Med.Genet. 2000;96:422–424. doi: 10.1002/1096-8628(20000612)96:3<422::aid-ajmg35>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
- 244.Rietschel M, Krauss H, Muller DJ, Schulze TG, Knapp M, Marwinski K, Maroldt AO, Paus S, Grunhage F, Propping P, Maier W, Held T, Nothen MM. Dopamine D3 receptor variant and tardive dyskinesia. Eur.Arch.Psychiatry Clin.Neurosci. 2000;250:31–35. doi: 10.1007/pl00007536. [DOI] [PubMed] [Google Scholar]
- 245.Scordo MG, Spina E, Romeo P, Dahl ML, Bertilsson L, Johansson I, Sjoqvist F. CYP2D6 genotype and antipsychotic-induced extrapyramidal side effects in schizophrenic patients. Eur.J.Clin.Pharmacol. 2000;56:679–683. doi: 10.1007/s002280000222. [DOI] [PubMed] [Google Scholar]
- 246.Segman RH, Heresco-Levy U, Finkel B, Inbar R, Neeman T, Schlafman M, Dorevitch A, Yakir A, Lerner A, Goltser T, Shelevoy A, Lerer B. Association between the serotonin 2C receptor gene and tardive dyskinesia in chronic schizophrenia: additive contribution of 5-HT2Cser and DRD3gly alleles to susceptibility. Psychopharmacology (Berl) 2000;152:408–413. doi: 10.1007/s002130000521. [DOI] [PubMed] [Google Scholar]
- 247.Tsai SJ, Hong CJ, Yu YW, Lin CH, Song HL, Lai HC, Yang KH. Association study of a functional serotonin transporter gene polymorphism with schizophrenia, psychopathology and clozapine response. Schizophr.Res. 2000;44:177–181. doi: 10.1016/s0920-9964(99)00170-x. [DOI] [PubMed] [Google Scholar]
- 248.Basile VS, Ozdemir V, Masellis M, Meltzer HY, Lieberman JA, Potkin SG, Macciardi FM, Petronis A, Kennedy JL. Lack of association between serotonin-2A receptor gene (HTR2A) polymorphisms and tardive dyskinesia in schizophrenia. Mol.Psychiatry. 2001;6:230–234. doi: 10.1038/sj.mp.4000847. [DOI] [PubMed] [Google Scholar]
- 249.Basile VS, Masellis M, McIntyre RS, Meltzer HY, Lieberman JA, Kennedy JL. Genetic dissection of atypical antipsychotic-induced weight gain: novel preliminary data on the pharmacogenetic puzzle. J.Clin.Psychiatry. 2001;62 Suppl 23:45–66. [PubMed] [Google Scholar]
- 250.Dahmen N, Muller MJ, Germeyer S, Rujescu D, Anghelescu I, Hiemke C, Wetzel H. Genetic polymorphisms of the dopamine D2 and D3 receptor and neuroleptic drug effects in schizophrenic patients. Schizophr.Res. 2001;49:223–225. doi: 10.1016/s0920-9964(99)00147-4. [DOI] [PubMed] [Google Scholar]
- 251.Dettling M, Cascorbi I, Roots I, Mueller-Oerlinghausen B. Genetic determinants of clozapine-induced agranulocytosis: recent results of HLA subtyping in a non-jewish caucasian sample. Arch.Gen.Psychiatry. 2001;58:93–94. doi: 10.1001/archpsyc.58.1.93. [DOI] [PubMed] [Google Scholar]
- 252.Garcia-Barcelo MM, Lam LC, Ungvari GS, Lam VK, Tang WK. Dopamine D3 receptor gene and tardive dyskinesia in Chinese schizophrenic patients. J.Neural Transm. 2001;108:671–677. doi: 10.1007/s007020170044. [DOI] [PubMed] [Google Scholar]
- 253.Hong CJ, Lin CH, Yu YW, Yang KH, Tsai SJ. Genetic variants of the serotonin system and weight change during clozapine treatment. Pharmacogenetics. 2001;11:265–268. doi: 10.1097/00008571-200104000-00010. [DOI] [PubMed] [Google Scholar]
- 254.Hong CJ, Yu YW, Lin CH, Cheng CY, Tsai SJ. Association analysis for NMDA receptor subunit 2B (GRIN2B) genetic variants and psychopathology and clozapine response in schizophrenia. Psychiatr.Genet. 2001;11:219–222. doi: 10.1097/00041444-200112000-00007. [DOI] [PubMed] [Google Scholar]
- 255.Hori H, Ohmori O, Shinkai T, Kojima H, Nakamura J. Association between three functional polymorphisms of dopamine D2 receptor gene and tardive dyskinesia in schizophrenia. Am.J.Med.Genet. 2001;105:774–778. doi: 10.1002/ajmg.10045. [DOI] [PubMed] [Google Scholar]
- 256.Kaiser R, Tremblay PB, Schmider J, Henneken M, Dettling M, Muller-Oerlinghausen B, Uebelhack R, Roots I, Brockmoller J. Serotonin transporter polymorphisms: no association with response to antipsychotic treatment, but associations with the schizoparanoid and residual subtypes of schizophrenia. Mol.Psychiatry. 2001;6:179–185. doi: 10.1038/sj.mp.4000821. [DOI] [PubMed] [Google Scholar]
- 257.Liao DL, Yeh YC, Chen HM, Chen H, Hong CJ, Tsai SJ. Association between the Ser9Gly polymorphism of the dopamine D3 receptor gene and tardive dyskinesia in Chinese schizophrenic patients. Neuropsychobiology. 2001;44:95–98. doi: 10.1159/000054924. [DOI] [PubMed] [Google Scholar]
- 258.Masellis M, Basile VS, Meltzer HY, Lieberman JA, Sevy S, Goldman DA, Hamblin MW, Macciardi FM, Kennedy JL. Lack of association between the T-->C 267 serotonin 5-HT6 receptor gene (HTR6) polymorphism and prediction of response to clozapine in schizophrenia. Schizophr.Res. 2001;47:49–58. doi: 10.1016/s0920-9964(00)00016-5. [DOI] [PubMed] [Google Scholar]
- 259.Mihara K, Kondo T, Suzuki A, Yasui N, Ono S, Otani K, Kaneko S. No relationship between--141C Ins/Del polymorphism in the promoter region of dopamine D2 receptor and extrapyramidal adverse effects of selective dopamine D2 antagonists in schizophrenic patients: a preliminary study. Psychiatry Res. 2001;101:33–38. doi: 10.1016/s0165-1781(00)00247-x. [DOI] [PubMed] [Google Scholar]
- 260.Roh HK, Kim CE, Chung WG, Park CS, Svensson JO, Bertilsson L. Risperidone metabolism in relation to CYP2D6*10 allele in Korean schizophrenic patients. Eur.J.Clin.Pharmacol. 2001;57:671–675. doi: 10.1007/s002280100372. [DOI] [PubMed] [Google Scholar]
- 261.Schafer M, Rujescu D, Giegling I, Guntermann A, Erfurth A, Bondy B, Moller HJ. Association of short-term response to haloperidol treatment with a polymorphism in the dopamine D(2) receptor gene. Am.J.Psychiatry. 2001;158:802–804. doi: 10.1176/appi.ajp.158.5.802. [DOI] [PubMed] [Google Scholar]
- 262.Schulze TG, Schumacher J, Muller DJ, Krauss H, Alfter D, Maroldt A, Ahle G, Maroldt AO, Fernandez A, Weber T, Held T, Propping P, Maier W, Nothen MM, Rietschel M. Lack of association between a functional polymorphism of the cytochrome P450 1A2 (CYP1A2) gene and tardive dyskinesia in schizophrenia. Am.J.Med.Genet. 2001;105:498–501. doi: 10.1002/ajmg.1472. [DOI] [PubMed] [Google Scholar]
- 263.Segman RH, Heresco-Levy U, Finkel B, Goltser T, Shalem R, Schlafman M, Dorevitch A, Yakir A, Greenberg D, Lerner A, Lerer B. Association between the serotonin 2A receptor gene and tardive dyskinesia in chronic schizophrenia. Mol.Psychiatry. 2001;6:225–229. doi: 10.1038/sj.mp.4000842. [DOI] [PubMed] [Google Scholar]
- 264.Suzuki A, Kondo T, Otani K, Mihara K, Yasui-Furukori N, Sano A, Koshiro K, Kaneko S. Association of the TaqI A polymorphism of the dopamine D(2) receptor gene with predisposition to neuroleptic malignant syndrome. Am.J.Psychiatry. 2001;158:1714–1716. doi: 10.1176/appi.ajp.158.10.1714. [DOI] [PubMed] [Google Scholar]
- 265.Suzuki A, Kondo T, Mihara K, Yasui-Furukori N, Ishida M, Furukori H, Kaneko S, Inoue Y, Otani K. The −141C Ins/Del polymorphism in the dopamine D2 receptor gene promoter region is associated with anxiolytic and antidepressive effects during treatment with dopamine antagonists in schizophrenic patients. Pharmacogenetics. 2001;11:545–550. doi: 10.1097/00008571-200108000-00009. [DOI] [PubMed] [Google Scholar]
- 266.Tan EC, Chong SA, Mahendran R, Dong F, Tan CH. Susceptibility to neuroleptic-induced tardive dyskinesia and the T102C polymorphism in the serotonin type 2A receptor. Biol.Psychiatry. 2001;50:144–147. doi: 10.1016/s0006-3223(01)01076-9. [DOI] [PubMed] [Google Scholar]
- 267.Tsai SJ, Wang YC, Yu Younger WY, Lin CH, Yang KH, Hong CJ. Association analysis of polymorphism in the promoter region of the alpha2a-adrenoceptor gene with schizophrenia and clozapine response. Schizophr.Res. 2001;49:53–58. doi: 10.1016/s0920-9964(00)00127-4. [DOI] [PubMed] [Google Scholar]
- 268.Yu YW, Tsai SJ, Yang KH, Lin CH, Chen MC, Hong CJ. Evidence for an association between polymorphism in the serotonin-2A receptor variant (102T/C) and increment of N100 amplitude in schizophrenics treated with clozapine. Neuropsychobiology. 2001;43:79–82. doi: 10.1159/000054871. [DOI] [PubMed] [Google Scholar]
- 269.Basile VS, Masellis M, De LV, Meltzer HY, Kennedy JL. 759C/T genetic variation of 5HT(2C) receptor and clozapine-induced weight gain. Lancet. 2002;360:1790–1791. doi: 10.1016/s0140-6736(02)11706-5. [DOI] [PubMed] [Google Scholar]
- 270.Brockmoller J, Kirchheiner J, Schmider J, Walter S, Sachse C, Muller-Oerlinghausen B, Roots I. The impact of the CYP2D6 polymorphism on haloperidol pharmacokinetics and on the outcome of haloperidol treatment. Clin.Pharmacol.Ther. 2002;72:438–452. doi: 10.1067/mcp.2002.127494. [DOI] [PubMed] [Google Scholar]
- 271.Ellingrod VL, Miller D, Schultz SK, Wehring H, Arndt S. CYP2D6 polymorphisms and atypical antipsychotic weight gain. Psychiatr.Genet. 2002;12:55–58. doi: 10.1097/00041444-200203000-00008. [DOI] [PubMed] [Google Scholar]
- 272.Gutierrez B, Arranz MJ, Huezo-Diaz P, Dempster D, Matthiasson P, Travis M, Munro J, Osborne S, Kerwin RW. Novel mutations in 5-HT3A and 5-HT3B receptor genes not associated with clozapine response. Schizophr.Res. 2002;58:93–97. doi: 10.1016/s0920-9964(02)00205-0. [DOI] [PubMed] [Google Scholar]
- 273.Himei A, Koh J, Sakai J, Inada Y, Akabame K, Yoneda H. The influence on the schizophrenic symptoms by the DRD2Ser/Cys311 and −141C Ins/Del polymorphisms. Psychiatry Clin.Neurosci. 2002;56:97–102. doi: 10.1046/j.1440-1819.2002.00935.x. [DOI] [PubMed] [Google Scholar]
- 274.Kaiser R, Tremblay PB, Klufmoller F, Roots I, Brockmoller J. Relationship between adverse effects of antipsychotic treatment and dopamine D(2) receptor polymorphisms in patients with schizophrenia. Mol.Psychiatry. 2002;7:695–705. doi: 10.1038/sj.mp.4001054. [DOI] [PubMed] [Google Scholar]
- 275.Lane HY, Chang YC, Chiu CC, Chen ML, Hsieh MH, Chang WH. Association of risperidone treatment response with a polymorphism in the 5-HT(2A) receptor gene. Am.J.Psychiatry. 2002;159:1593–1595. doi: 10.1176/appi.ajp.159.9.1593. [DOI] [PubMed] [Google Scholar]
- 276.Lerer B, Segman RH, Fangerau H, Daly AK, Basile VS, Cavallaro R, Aschauer HN, McCreadie RG, Ohlraun S, Ferrier N, Masellis M, Verga M, Scharfetter J, Rietschel M, Lovlie R, Levy UH, Meltzer HY, Kennedy JL, Steen VM, Macciardi F. Pharmacogenetics of tardive dyskinesia: combined analysis of 780 patients supports association with dopamine D3 receptor gene Ser9Gly polymorphism. Neuropsychopharmacology. 2002;27:105–119. doi: 10.1016/S0893-133X(02)00293-2. [DOI] [PubMed] [Google Scholar]
- 277.Mancama D, Arranz MJ, Munro J, Osborne S, Makoff A, Collier D, Kerwin R. Investigation of promoter variants of the histamine 1 and 2 receptors in schizophrenia and clozapine response. Neurosci.Lett. 2002;333:207–211. doi: 10.1016/s0304-3940(02)00178-7. [DOI] [PubMed] [Google Scholar]
- 278.Mihara K, Kondo T, Higuchi H, Takahashi H, Yoshida K, Shimizu T, Kaneko S. Tardive dystonia and genetic polymorphisms of cytochrome P4502D6 and dopamine D2 and D3 receptors: a preliminary finding. Am.J.Med.Genet. 2002;114:693–695. doi: 10.1002/ajmg.10602. [DOI] [PubMed] [Google Scholar]
- 279.Ohmori O, Shinkai T, Hori H, Nakamura J. Genetic association analysis of 5-HT(6) receptor gene polymorphism (267C/T) with tardive dyskinesia. Psychiatry Res. 2002;110:97–102. doi: 10.1016/s0165-1781(02)00095-1. [DOI] [PubMed] [Google Scholar]
- 280.Reynolds GP, Zhang ZJ, Zhang XB. Association of antipsychotic drug-induced weight gain with a 5-HT2C receptor gene polymorphism. Lancet. 2002;359:2086–2087. doi: 10.1016/S0140-6736(02)08913-4. [DOI] [PubMed] [Google Scholar]
- 281.Schillevoort I, de BA, van der Weide J, Steijns LS, Roos RA, Jansen PA, Leufkens HG. Antipsychotic-induced extrapyramidal syndromes and cytochrome P450 2D6 genotype: a case-control study. Pharmacogenetics. 2002;12:235–240. doi: 10.1097/00008571-200204000-00008. [DOI] [PubMed] [Google Scholar]
- 282.Segman RH, Heresco-Levy U, Yakir A, Goltser T, Strous R, Greenberg DA, Lerer B. Interactive effect of cytochrome P450 17alpha-hydroxylase and dopamine D3 receptor gene polymorphisms on abnormal involuntary movements in chronic schizophrenia. Biol.Psychiatry. 2002;51:261–263. doi: 10.1016/s0006-3223(01)01302-6. [DOI] [PubMed] [Google Scholar]
- 283.Segman RH, Lerer B. Age and the relationship of dopamine D3, serotonin 2C and serotonin 2A receptor genes to abnormal involuntary movements in chronic schizophrenia. Mol.Psychiatry. 2002;7:137–139. doi: 10.1038/sj.mp.4000960. [DOI] [PubMed] [Google Scholar]
- 284.Staddon S, Arranz MJ, Mancama D, Mata I, Kerwin RW. Clinical applications of pharmacogenetics in psychiatry. Psychopharmacology (Berl) 2002;162:18–23. doi: 10.1007/s00213-002-1084-4. [DOI] [PubMed] [Google Scholar]
- 285.Tsai SJ, Hong CJ, Yu YW, Lin CH. −759C/T genetic variation of 5HT(2C) receptor and clozapine-induced weight gain. Lancet. 2002;360:1790. doi: 10.1016/S0140-6736(02)11705-3. [DOI] [PubMed] [Google Scholar]
- 286.Woo SI, Kim JW, Rha E, Han SH, Hahn KH, Park CS, Sohn JW. Association of the Ser9Gly polymorphism in the dopamine D3 receptor gene with tardive dyskinesia in Korean schizophrenics. Psychiatry Clin.Neurosci. 2002;56:469–474. doi: 10.1046/j.1440-1819.2002.01038.x. [DOI] [PubMed] [Google Scholar]
- 287.Zhang Z, Zhang X, Hou G, Sha W, Reynolds GP. The increased activity of plasma manganese superoxide dismutase in tardive dyskinesia is unrelated to the Ala-9Val polymorphism. J.Psychiatr.Res. 2002;36:317–324. doi: 10.1016/s0022-3956(02)00007-9. [DOI] [PubMed] [Google Scholar]
- 288.Zhang ZJ, Zhang XB, Sha WW, Zhang XB, Reynolds GP. Association of a polymorphism in the promoter region of the serotonin 5-HT2C receptor gene with tardive dyskinesia in patients with schizophrenia. Mol.Psychiatry. 2002;7:670–671. doi: 10.1038/sj.mp.4001052. [DOI] [PubMed] [Google Scholar]
- 289.Chong SA, Tan EC, Tan CH, Mythily, Chan YH. Polymorphisms of dopamine receptors and tardive dyskinesia among Chinese patients with schizophrenia. Am.J.Med.Genet.BNeuropsychiatr.Genet. 2003;116B:51–54. doi: 10.1002/ajmg.b.10004. [DOI] [PubMed] [Google Scholar]
- 290.Herken H, Erdal ME, Boke O, Savas HA. Tardive dyskinesia is not associated with the polymorphisms of 5-HT2A receptor gene, serotonin transporter gene and catechol-omethyltransferase gene. Eur.Psychiatry. 2003;18:77–81. doi: 10.1016/s0924-9338(03)00005-1. [DOI] [PubMed] [Google Scholar]
- 291.Hong CJ, Yu YW, Lin CH, Tsai SJ. An association study of a brain-derived neurotrophic factor Val66Met polymorphism and clozapine response of schizophrenic patients. Neurosci.Lett. 2003;349:206–208. doi: 10.1016/s0304-3940(03)00828-0. [DOI] [PubMed] [Google Scholar]
- 292.Inada T, Senoo H, Iijima Y, Yamauchi T, Yagi G. Cytochrome P450 II D6 gene polymorphisms and the neuroleptic-induced extrapyramidal symptoms in Japanese schizophrenic patients. Psychiatr.Genet. 2003;13:163–168. doi: 10.1097/00041444-200309000-00005. [DOI] [PubMed] [Google Scholar]
- 293.Kishida I, Kawanishi C, Furuno T, Matsumura T, Hasegawa H, Sugiyama N, Suzuki K, Yamada Y, Kosaka K. Lack of association in Japanese patients between neuroleptic malignant syndrome and the TaqI A polymorphism of the dopamine D2 receptor gene. Psychiatr.Genet. 2003;13:55–57. doi: 10.1097/00041444-200303000-00010. [DOI] [PubMed] [Google Scholar]
- 294.Lohmann PL, Bagli M, Krauss H, Muller DJ, Schulze TG, Fangerau H, Ludwig M, Barkow K, Held T, Heun R, Maier W, Rietschel M, Rao ML. CYP2D6 polymorphism and tardive dyskinesia in schizophrenic patients. Pharmacopsychiatry. 2003;36:73–78. doi: 10.1055/s-2003-39048. [DOI] [PubMed] [Google Scholar]
- 295.Mihara K, Kondo T, Suzuki A, Yasui-Furukori N, Ono S, Sano A, Koshiro K, Otani K, Kaneko S. Relationship between functional dopamine D2 and D3 receptors gene polymorphisms and neuroleptic malignant syndrome. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2003;117B:57–60. doi: 10.1002/ajmg.b.10025. [DOI] [PubMed] [Google Scholar]
- 296.Ostrousky O, Meged S, Loewenthal R, Valevski A, Weizman A, Carp H, Gazit E. NQO2 gene is associated with clozapine-induced agranulocytosis. Tissue Antigens. 2003;62:483–491. doi: 10.1046/j.1399-0039.2003.00133.x. [DOI] [PubMed] [Google Scholar]
- 297.Reynolds GP, Zhang Z, Zhang X. Polymorphism of the promoter region of the serotonin 5-HT(2C) receptor gene and clozapine-induced weight gain. Am.J.Psychiatry. 2003;160:677–679. doi: 10.1176/appi.ajp.160.4.677. [DOI] [PubMed] [Google Scholar]
- 298.Tsai SJ, Hong CJ, Yu YW, Lin CH, Liu LL. No association of tumor necrosis factor alpha gene polymorphisms with schizophrenia or response to clozapine. Schizophr.Res. 2003;65:27–32. doi: 10.1016/s0920-9964(02)00531-5. [DOI] [PubMed] [Google Scholar]
- 299.Zalsman G, Frisch A, Lev-Ran S, Martin A, Michaelovsky E, Bensason D, Gothelf D, Nahshoni E, Tyano S, Weizman A. DRD4 exon III polymorphism and response to risperidone in Israeli adolescents with schizophrenia: a pilot pharmacogenetic study. Eur.Neuropsychopharmacol. 2003;13:183–185. doi: 10.1016/s0924-977x(03)00006-3. [DOI] [PubMed] [Google Scholar]
- 300.Zhang ZJ, Yao ZJ, Zhang XB, Chen JF, Sun J, Yao H, Hou G, Zhang XB. No association of antipsychotic agent-induced weight gain with a DA receptor gene polymorphism and therapeutic response. Acta Pharmacol.Sin. 2003;24:235–240. [PubMed] [Google Scholar]
- 301.Anttila S, Illi A, Kampman O, Mattila KM, Lehtimaki T, Leinonen E. Interaction between NOTCH4 and catechol-O-methyltransferase genotypes in schizophrenia patients with poor response to typical neuroleptics. Pharmacogenetics. 2004;14:303–307. doi: 10.1097/00008571-200405000-00005. [DOI] [PubMed] [Google Scholar]
- 302.Bertolino A, Caforio G, Blasi G, De CM, Latorre V, Petruzzella V, Altamura M, Nappi G, Papa S, Callicott JH, Mattay VS, Bellomo A, Scarabino T, Weinberger DR, Nardini M. Interaction of COMT (Val(108/158)Met) genotype and olanzapine treatment on prefrontal cortical function in patients with schizophrenia. Am.J.Psychiatry. 2004;161:1798–1805. doi: 10.1176/ajp.161.10.1798. [DOI] [PubMed] [Google Scholar]
- 303.Huezo-Diaz P, Arranz MJ, Munro J, Osborne S, Makoff A, Kerwin RW, Austin J, O'Donovan M. An association study of the neurotensin receptor gene with schizophrenia and clozapine response. Schizophr.Res. 2004;66:193–195. doi: 10.1016/S0920-9964(03)00128-2. [DOI] [PubMed] [Google Scholar]
- 304.Kampman O, Anttila S, Illi A, Saarela M, Rontu R, Mattila KM, Leinonen E, Lehtimaki T. Neuregulin genotype and medication response in Finnish patients with schizophrenia. Neuroreport. 2004;15:2517–2520. doi: 10.1097/00001756-200411150-00017. [DOI] [PubMed] [Google Scholar]
- 305.Kishida I, Kawanishi C, Furuno T, Kato D, Ishigami T, Kosaka K. Association in Japanese patients between neuroleptic malignant syndrome and functional polymorphisms of the dopamine D(2) receptor gene. Mol.Psychiatry. 2004;9:293–298. doi: 10.1038/sj.mp.4001422. [DOI] [PubMed] [Google Scholar]
- 306.Lane HY, Lee CC, Chang YC, Lu CT, Huang CH, Chang WH. 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;7:461–470. doi: 10.1017/S1461145704004389. [DOI] [PubMed] [Google Scholar]
- 307.Lane HY, Lin CC, Huang CH, Chang YC, Hsu SK, Chang WH. Risperidone response and 5-HT6 receptor gene variance: genetic association analysis with adjustment for nongenetic confounders. Schizophr.Res. 2004;67:63–70. doi: 10.1016/j.schres.2003.08.006. [DOI] [PubMed] [Google Scholar]
- 308.Lattuada E, Cavallaro R, Serretti A, Lorenzi C, Smeraldi E. Tardive dyskinesia and DRD2, DRD3, DRD4, 5-HT2A variants in schizophrenia: an association study with repeated assessment. Int.J.Neuropsychopharmacol. 2004;7:489–493. doi: 10.1017/S1461145704004614. [DOI] [PubMed] [Google Scholar]
- 309.Liou YJ, Liao DL, Chen JY, Wang YC, Lin CC, Bai YM, Yu SC, Lin MW, Lai IC. 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;5:243–251. doi: 10.1385/NMM:5:3:243. [DOI] [PubMed] [Google Scholar]
- 310.Liou YJ, Wang YC, Bai YM, Lin CC, Yu SC, Liao DL, Lin MW, Chen JY, Lai IC. Cytochrome P-450 2D6*10 C188T polymorphism is associated with antipsychoticinduced persistent tardive dyskinesia in Chinese schizophrenic patients. Neuropsychobiology. 2004;49:167–173. doi: 10.1159/000077360. [DOI] [PubMed] [Google Scholar]
- 311.Matsumoto C, Ohmori O, Shinkai T, Hori H, Nakamura J. Genetic association analysis of functional polymorphisms in the cytochrome P450 1A2 (CYP1A2) gene with tardive dyskinesia in Japanese patients with schizophrenia. Psychiatr.Genet. 2004;14:209–213. doi: 10.1097/00041444-200412000-00008. [DOI] [PubMed] [Google Scholar]
- 312.Matsumoto C, Shinkai T, Hori H, Ohmori O, Nakamura J. Polymorphisms of dopamine degradation enzyme (COMT and MAO) genes and tardive dyskinesia in patients with schizophrenia. Psychiatry Res. 2004;127:1–7. doi: 10.1016/j.psychres.2004.03.011. [DOI] [PubMed] [Google Scholar]
- 313.Mosyagin I, Dettling M, Roots I, Mueller-Oerlinghausen B, Cascorbi I. Impact of myeloperoxidase and NADPH-oxidase polymorphisms in drug-induced agranulocytosis. J.Clin.Psychopharmacol. 2004;24:613–617. doi: 10.1097/01.jcp.0000144891.52858.a6. [DOI] [PubMed] [Google Scholar]
- 314.Pae CU, Yu HS, Kim JJ, Lee CU, Lee SJ, Jun TY, Lee C, Paik IH. Quinone oxidoreductase (NQO1) gene polymorphism (609C/T) may be associated with tardive dyskinesia, but not with the development of schizophrenia. Int.J.Neuropsychopharmacol. 2004;7:495–500. doi: 10.1017/S1461145704004419. [DOI] [PubMed] [Google Scholar]
- 315.Shinkai T, Ohmori O, Matsumoto C, Hori H, Kennedy JL, Nakamura J. Genetic association analysis of neuronal nitric oxide synthase gene polymorphism with tardive dyskinesia. Neuromolecular.Med. 2004;5:163–170. doi: 10.1385/NMM:5:2:163. [DOI] [PubMed] [Google Scholar]
- 316.Szekeres G, Keri S, Juhasz A, Rimanoczy A, Szendi I, Czimmer C, Janka Z. Role of dopamine D3 receptor (DRD3) and dopamine transporter (DAT) polymorphism in cognitive dysfunctions and therapeutic response to atypical antipsychotics in patients with schizophrenia. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2004;124B:1–5. doi: 10.1002/ajmg.b.20045. [DOI] [PubMed] [Google Scholar]
- 317.Tsai SJ, Yu YW, Lin CH, Wang YC, Chen JY, Hong CJ. Association study of adrenergic beta3 receptor (Trp64Arg) and G-protein beta3 subunit gene (C825T) polymorphisms and weight change during clozapine treatment. Neuropsychobiology. 2004;50:37–40. doi: 10.1159/000077939. [DOI] [PubMed] [Google Scholar]
- 318.Wang YC, Liou YJ, Liao DL, Bai YM, Lin CC, Yu SC, Chen JY. Association analysis of a neural nitric oxide synthase gene polymorphism and antipsychotics-induced tardive dyskinesia in Chinese schizophrenic patients. J.Neural Transm. 2004;111:623–629. doi: 10.1007/s00702-004-0118-y. [DOI] [PubMed] [Google Scholar]
- 319.Weickert TW, Goldberg TE, Mishara A, Apud JA, Kolachana BS, Egan MF, Weinberger DR. Catechol-O-methyltransferase val108/158met genotype predicts working memory response to antipsychotic medications. Biol.Psychiatry. 2004;56:677–682. doi: 10.1016/j.biopsych.2004.08.012. [DOI] [PubMed] [Google Scholar]
- 320.Templeman LA, Reynolds GP, Arranz B, San L. 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;15:195–200. doi: 10.1097/01213011-200504000-00002. [DOI] [PubMed] [Google Scholar]
- 321.Theisen FM, Gebhardt S, Haberhausen M, Heinzel-Gutenbrunner M, Wehmeier PM, Krieg JC, Kuhnau W, Schmidtke J, Remschmidt H, Hebebrand J. Clozapine-induced weight gain: a study in monozygotic twins and same-sex sib pairs. Psychiatr.Genet. 2005;15:285–289. doi: 10.1097/00041444-200512000-00011. [DOI] [PubMed] [Google Scholar]
- 322.Anttila S, Illi A, Kampman O, Mattila KM, Lehtimaki T, Leinonen E. Lack of association between two polymorphisms of brain-derived neurotrophic factor and response to typical neuroleptics. J.Neural Transm. 2005;112:885–890. doi: 10.1007/s00702-004-0233-9. [DOI] [PubMed] [Google Scholar]
- 323.Bishop JR, Ellingrod VL, Moline J, Miller D. Association between the polymorphic GRM3 gene and negative symptom improvement during olanzapine treatment. Schizophr.Res. 2005;77:253–260. doi: 10.1016/j.schres.2005.04.001. [DOI] [PubMed] [Google Scholar]
- 324.de LJ, Susce MT, Pan RM, Fairchild M, Koch WH, Wedlund PJ. The CYP2D6 poor metabolizer phenotype may be associated with risperidone adverse drug reactions and discontinuation. J.Clin.Psychiatry. 2005;66:15–27. doi: 10.4088/jcp.v66n0103. [DOI] [PubMed] [Google Scholar]
- 325.de LJ, Susce MT, Pan RM, Koch WH, Wedlund PJ. 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;25:448–456. doi: 10.1097/01.jcp.0000177546.34799.af. [DOI] [PubMed] [Google Scholar]
- 326.De LV, Vincent JB, Muller DJ, Hwang R, Shinkai T, Volavka J, Czobor P, Sheitman BB, Lindenmayer JP, Citrome L, McEvoy JP, Lieberman JA, Kennedy JL. Identification of a naturally occurring 21 bp deletion in alpha 2c noradrenergic receptor gene and cognitive correlates to antipsychotic treatment. Pharmacol.Res. 2005;51:381–384. doi: 10.1016/j.phrs.2004.10.013. [DOI] [PubMed] [Google Scholar]
- 327.Deshpande SN, Varma PG, Semwal P, Rao AR, Bhatia T, Nimgaonkar VL, Lerer B, Thelma BK., II Serotonin receptor gene polymorphisms and their association with tardive dyskinesia among schizophrenia patients from North India. Psychiatr.Genet. 2005;15:157–158. doi: 10.1097/00041444-200509000-00002. [DOI] [PubMed] [Google Scholar]
- 328.Ellingrod VL, Perry PJ, Ringold JC, Lund BC, Bever-Stille K, Fleming F, Holman TL, Miller D. Weight gain associated with the −759C/T polymorphism of the 5HT2C receptor and olanzapine. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2005;134B:76–78. doi: 10.1002/ajmg.b.20169. [DOI] [PubMed] [Google Scholar]
- 329.Hamdani N, Bonniere M, Ades J, Hamon M, Boni C, Gorwood P. Negative symptoms of schizophrenia could explain discrepant data on the association between the 5-HT2A receptor gene and response to antipsychotics. Neurosci.Lett. 2005;377:69–74. doi: 10.1016/j.neulet.2004.11.070. [DOI] [PubMed] [Google Scholar]
- 330.Hwang R, Shinkai T, De LV, Muller DJ, Ni X, Macciardi F, Potkin S, Lieberman JA, Meltzer HY, Kennedy JL. 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;181:179–187. doi: 10.1007/s00213-005-2223-5. [DOI] [PubMed] [Google Scholar]
- 331.Kakihara S, Yoshimura R, Shinkai K, Matsumoto C, Goto M, Kaji K, Yamada Y, Ueda N, Ohmori O, Nakamura J. 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;20:71–78. doi: 10.1097/00004850-200503000-00002. [DOI] [PubMed] [Google Scholar]
- 332.Kim JY, Jung IK, Han C, Cho SH, Kim L, Kim SH, Lee BH, Lee HJ, Kim YK. Antipsychotics and dopamine transporter gene polymorphisms in delirium patients. Psychiatry Clin.Neurosci. 2005;59:183–188. doi: 10.1111/j.1440-1819.2005.01355.x. [DOI] [PubMed] [Google Scholar]
- 333.Lai IC, Wang YC, Lin CC, Bai YM, Liao DL, Yu SC, Lin CY, Chen JY, Liou YJ. Negative association between catechol-O-methyltransferase (COMT) gene Val158Met polymorphism and persistent tardive dyskinesia in schizophrenia. J.Neural Transm. 2005;112:1107–1113. doi: 10.1007/s00702-004-0252-6. [DOI] [PubMed] [Google Scholar]
- 334.Lane HY, Hsu SK, Liu YC, Chang YC, Huang CH, Chang WH. Dopamine D3 receptor Ser9Gly polymorphism and risperidone response. J.Clin.Psychopharmacol. 2005;25:6–11. doi: 10.1097/01.jcp.0000150226.84371.76. [DOI] [PubMed] [Google Scholar]
- 335.Liou YJ, Wang YC, Lin CC, Bai YM, Lai IC, Liao DL, Chen JY. Association analysis of NAD(P)Hratioquinone oxidoreductase (NQO1) Pro187Ser genetic polymorphism and tardive dyskinesia in patients with schizophrenia in Taiwan. Int.J.Neuropsychopharmacol. 2005;8:483–486. doi: 10.1017/S1461145705005262. [DOI] [PubMed] [Google Scholar]
- 336.Miller DD, Ellingrod VL, Holman TL, Buckley PF, Arndt S. Clozapine-induced weight gain associated with the 5HT2C receptor −759C/T polymorphism. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2005;133B:97–100. doi: 10.1002/ajmg.b.30115. [DOI] [PubMed] [Google Scholar]
- 337.Muller DJ, Klempan TA, De LV, Sicard T, Volavka J, Czobor P, Sheitman BB, Lindenmayer JP, Citrome L, McEvoy JP, Lieberman JA, Honer WG, Kennedy JL. The SNAP-25 gene may be associated with clinical response and weight gain in antipsychotic treatment of schizophrenia. Neurosci.Lett. 2005;379:81–89. doi: 10.1016/j.neulet.2004.12.037. [DOI] [PubMed] [Google Scholar]
- 338.Muller DJ, De LV, Sicard T, King N, Hwang R, Volavka J, Czobor P, Sheitman BB, Lindenmayer JP, Citrome L, McEvoy JP, Lieberman JA, Meltzer HY, Kennedy JL. Suggestive association between the C825T polymorphism of the G-protein beta3 subunit gene (GNB3) and clinical improvement with antipsychotics in schizophrenia. Eur.Neuropsychopharmacol. 2005;15:525–531. doi: 10.1016/j.euroneuro.2005.02.001. [DOI] [PubMed] [Google Scholar]
- 339.Reynolds GP, Yao Z, Zhang X, Sun J, Zhang Z. Pharmacogenetics of treatment in first-episode schizophrenia: D3 and 5-HT2C receptor polymorphisms separately associate with positive and negative symptom response. Eur.Neuropsychopharmacol. 2005;15:143–151. doi: 10.1016/j.euroneuro.2004.07.001. [DOI] [PubMed] [Google Scholar]
- 340.Shinkai T, De LV, Hwang R, Matsumoto C, Hori H, Ohmori O, Remington G, Meltzer HY, Lieberman JA, Potkin SG, Nakamura J, Kennedy JL. Association study between a functional glutathione S-transferase (GSTP1) gene polymorphism (Ile105Val) and tardive dyskinesia. Neurosci.Lett. 2005;388:116–120. doi: 10.1016/j.neulet.2005.06.038. [DOI] [PubMed] [Google Scholar]
- 341.Tiwari AK, Deshpande SN, Rao AR, Bhatia T, Mukit SR, Shriharsh V, Lerer B, Nimagaonkar VL, Thelma BK. Genetic susceptibility to tardive dyskinesia in chronic schizophrenia subjects: I. Association of CYP1A2 gene polymorphism. Pharmacogenomics.J. 2005;5:60–69. doi: 10.1038/sj.tpj.6500282. [DOI] [PubMed] [Google Scholar]
- 342.Tiwari AK, Deshpande SN, Rao AR, Bhatia T, Lerer B, Nimgaonkar VL, Thelma BK. Genetic susceptibility to tardive dyskinesia in chronic schizophrenia subjects: III. Lack of association of CYP3A4 and CYP2D6 gene polymorphisms. Schizophr.Res. 2005;75:21–26. doi: 10.1016/j.schres.2004.12.011. [DOI] [PubMed] [Google Scholar]
- 343.Wang YC, Bai YM, Chen JY, Lin CC, Lai IC, Liou YJ. Polymorphism of the adrenergic receptor alpha 2a −1291C>G genetic variation and clozapine-induced weight gain. J.Neural Transm. 2005;112:1463–1468. doi: 10.1007/s00702-005-0291-7. [DOI] [PubMed] [Google Scholar]
- 344.Wang YC, Bai YM, Chen JY, Lin CC, Lai IC, Liou YJ. 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;15:743–748. doi: 10.1097/01.fpc.0000175600.26893.fa. [DOI] [PubMed] [Google Scholar]
- 345.Wu S, Xing Q, Gao R, Li X, Gu N, Feng G, He L. Response to chlorpromazine treatment may be associated with polymorphisms of the DRD2 gene in Chinese schizophrenic patients. Neurosci.Lett. 2005;376:1–4. doi: 10.1016/j.neulet.2004.11.014. [DOI] [PubMed] [Google Scholar]
- 346.Zhao AL, Zhao JP, Zhang YH, Xue ZM, Chen JD, Chen XG. Dopamine D4 receptor gene exon III polymorphism and interindividual variation in response to clozapine. Int.J.Neurosci. 2005;115:1539–1547. doi: 10.1080/00207450590957863. [DOI] [PubMed] [Google Scholar]
- 347.Bishop JR, Ellingrod VL, Moline J, Miller D. Pilot study of the G-protein beta3 subunit gene (C825T) polymorphism and clinical response to olanzapine or olanzapine-related weight gain in persons with schizophrenia. Med.Sci.Monit. 2006;12:BR47–BR50. [PubMed] [Google Scholar]
- 348.Fu Y, Fan CH, Deng HH, Hu SH, Lv DP, Li LH, Wang JJ, Lu XQ. Association of CYP2D6 and CYP1A2 gene polymorphism with tardive dyskinesia in Chinese schizophrenic patients. Acta Pharmacol.Sin. 2006;27:328–332. doi: 10.1111/j.1745-7254.2006.00279.x. [DOI] [PubMed] [Google Scholar]
- 349.Hwang R, Shinkai T, Deluca V, Macciardi F, Potkin S, Meltzer HY, Kennedy JL. Dopamine D2 receptor gene variants and quantitative measures of positive and negative symptom response following clozapine treatment. Eur.Neuropsychopharmacol. 2006;16:248–259. doi: 10.1016/j.euroneuro.2005.09.004. [DOI] [PubMed] [Google Scholar]
- 350.Kampman O, Illi A, Hanninen K, Katila H, Anttila S, Rontu R, Mattila KM, Leinonen E, Lehtimaki T. RGS4 genotype is not associated with antipsychotic medication response in schizophrenia. J.Neural Transm. 2006;113:1563–1568. doi: 10.1007/s00702-006-0445-2. [DOI] [PubMed] [Google Scholar]
- 351.Lencz T, Robinson DG, Xu K, Ekholm J, Sevy S, Gunduz-Bruce H, Woerner MG, Kane JM, Goldman D, Malhotra AK. DRD2 promoter region variation as a predictor of sustained response to antipsychotic medication in first-episode schizophrenia patients. Am.J.Psychiatry. 2006;163:529–531. doi: 10.1176/appi.ajp.163.3.529. [DOI] [PubMed] [Google Scholar]
- 352.Liou YJ, Lai IC, Liao DL, Chen JY, Lin CC, Lin CY, Chen CM, Bai YM, Chen TT, Wang YC. The human dopamine receptor D2 (DRD2) gene is associated with tardive dyskinesia in patients with schizophrenia. Schizophr.Res. 2006;86:323–325. doi: 10.1016/j.schres.2006.04.008. [DOI] [PubMed] [Google Scholar]
- 353.Liou YJ, Lai IC, Lin MW, Bai YM, Lin CC, Liao DL, Chen JY, Lin CY, Wang YC. Haplotype analysis of endothelial nitric oxide synthase (NOS3) genetic variants and tardive dyskinesia in patients with schizophrenia. Pharmacogenet.Genomics. 2006;16:151–157. doi: 10.1097/01.fpc.0000184958.05775.66. [DOI] [PubMed] [Google Scholar]
- 354.Mata I, Crespo-Facorro B, Perez-Iglesias R, Carrasco-Marin E, Arranz MJ, Pelayo-Teran JM, Leyva-Cobian F, Vazquez-Barquero JL. Association between the interleukin-1 receptor antagonist gene and negative symptom improvement during antipsychotic treatment. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2006;141B:939–943. doi: 10.1002/ajmg.b.30405. [DOI] [PubMed] [Google Scholar]
- 355.Park YM, Chung YC, Lee SH, Lee KJ, Kim H, Byun YC, Lim SW, Paik JW, Lee HJ. 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:394–397. doi: 10.1002/ajmg.b.30311. [DOI] [PubMed] [Google Scholar]
- 356.Reynolds GP, Arranz B, Templeman LA, Fertuzinhos S, San L. 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;163:1826–1829. doi: 10.1176/ajp.2006.163.10.1826. [DOI] [PubMed] [Google Scholar]
- 357.Richardson MA, Chao HM, Read LL, Clelland JD, Suckow RF. Investigation of the phenylalanine hydroxylase gene and tardive dyskinesia. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2006;141B:195–197. doi: 10.1002/ajmg.b.30281. [DOI] [PubMed] [Google Scholar]
- 358.Shinkai T, Muller DJ, De LV, Shaikh S, Matsumoto C, Hwang R, King N, Trakalo J, Potapova N, Zai G, Hori H, Ohmori O, Meltzer HY, Nakamura J, Kennedy JL. Genetic association analysis of the glutathione peroxidase (GPX1) gene polymorphism (Pro197Leu) with tardive dyskinesia. Psychiatry Res. 2006;141:123–128. doi: 10.1016/j.psychres.2004.06.023. [DOI] [PubMed] [Google Scholar]
- 359.Srivastava V, Varma PG, Prasad S, Semwal P, Nimgaonkar VL, Lerer B, Deshpande SN, BK T. Genetic susceptibility to tardive dyskinesia among schizophrenia subjects: IV. Role of dopaminergic pathway gene polymorphisms. Pharmacogenet.Genomics. 2006;16:111–117. doi: 10.1097/01.fpc.0000184957.98150.0f. [DOI] [PubMed] [Google Scholar]
- 360.Takao T, Tachikawa H, Kawanishi Y, Katano T, Sen B, Homma M, Kohda Y, Mizukami K, Asada T. 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;16:47–48. doi: 10.1097/01.ypg.0000194441.04684.db. [DOI] [PubMed] [Google Scholar]
- 361.Yasui-Furukori N, Saito M, Nakagami T, Kaneda A, Tateishi T, Kaneko S. Association between multidrug resistance 1 (MDR1) gene polymorphisms and therapeutic response to bromperidol in schizophrenic patients: a preliminary study. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2006;30:286–291. doi: 10.1016/j.pnpbp.2005.06.019. [DOI] [PubMed] [Google Scholar]
- 362.Zai G, Muller DJ, Volavka J, Czobor P, Lieberman JA, Meltzer HY, Kennedy JL. 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;188:171–182. doi: 10.1007/s00213-006-0482-4. [DOI] [PubMed] [Google Scholar]
- 363.Strous RD, Greenbaum L, Kanyas K, Merbl Y, Horowitz A, Karni O, Viglin D, Olender T, Deshpande SN, Lancet D, Ben-Asher E, Lerer B. Association of the dopamine receptor interacting protein gene, NEF3, with early response to antipsychotic medication. Int.J.Neuropsychopharmacol. 2007;10:321–333. doi: 10.1017/S1461145706006651. [DOI] [PubMed] [Google Scholar]
- 364.Woodward ND, Jayathilake K, Meltzer HY. COMT val108/158met genotype, cognitive function, and cognitive improvement with clozapine in schizophrenia. Schizophr.Res. 2007;90:86–96. doi: 10.1016/j.schres.2006.10.002. [DOI] [PubMed] [Google Scholar]
- 365.Anttila S, Kampman O, Illi A, Rontu R, Lehtimaki T, Leinonen E. 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]
- 366.Bertolino A, Caforio G, Blasi G, Rampino A, Nardini M, Weinberger DR, Dallapiccola B, Sinibaldi L, Douzgou S. COMT Val158Met polymorphism predicts negative symptoms response to treatment with olanzapine in schizophrenia. Schizophr.Res. 2007;95:253–255. doi: 10.1016/j.schres.2007.06.014. [DOI] [PubMed] [Google Scholar]
- 367.Boke O, Gunes S, Kara N, Aker S, Sahin AR, Basar Y, Bagci H. Association of serotonin 2A receptor and lack of association of CYP1A2 gene polymorphism with tardive dyskinesia in a Turkish population. DNA Cell Biol. 2007;26:527–531. doi: 10.1089/dna.2007.0605. [DOI] [PubMed] [Google Scholar]
- 368.Bozina N, Medved V, Kuzman MR, Sain I, Sertic J. Association study of olanzapine-induced weight gain and therapeutic response with SERT gene polymorphisms in female schizophrenic patients. J.Psychopharmacol. 2007;21:728–734. doi: 10.1177/0269881106072750. [DOI] [PubMed] [Google Scholar]
- 369.Chagnon YC, Bureau A, Gendron D, Bouchard RH, Merette C, Roy MA, Maziade M. Possible association of the pro-melanin-concentrating hormone gene with a greater body mass index as a side effect of the antipsychotic olanzapine. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:1063–1069. doi: 10.1002/ajmg.b.30554. [DOI] [PubMed] [Google Scholar]
- 370.De LV, Muller DJ, Hwang R, Lieberman JA, Volavka J, Meltzer HY, Kennedy JL. HTR2C haplotypes and antipsychotics-induced weight gain: X-linked multimarker analysis. Hum.Psychopharmacol. 2007;22:463–467. doi: 10.1002/hup.868. [DOI] [PubMed] [Google Scholar]
- 371.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. doi: 10.1038/sj.tpj.6500423. [DOI] [PubMed] [Google Scholar]
- 372.Dolzan V, Plesnicar BK, Serretti A, Mandelli L, Zalar B, Koprivsek J, Breskvar K. Polymorphisms in dopamine receptor DRD1 and DRD2 genes and psychopathological and extrapyramidal symptoms in patients on long-term antipsychotic treatment. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:809–815. doi: 10.1002/ajmg.b.30544. [DOI] [PubMed] [Google Scholar]
- 373.Ellingrod VL, Bishop JR, Moline J, Lin YC, Miller dD. Leptin and leptin receptor gene polymorphisms and increases in body mass index (BMI) from olanzapine treatment in persons with schizophrenia. Psychopharmacol.Bull. 2007;40:57–62. [PubMed] [Google Scholar]
- 374.Greenbaum L, Strous RD, Kanyas K, Merbl Y, Horowitz A, Karni O, Katz E, Kotler M, Olender T, Deshpande SN, Lancet D, Ben-Asher E, Lerer B. Association of the RGS2 gene with extrapyramidal symptoms induced by treatment with antipsychotic medication. Pharmacogenet.Genomics. 2007;17:519–528. doi: 10.1097/FPC.0b013e32800ffbb4. [DOI] [PubMed] [Google Scholar]
- 375.Gunes A, Scordo MG, Jaanson P, Dahl ML. Serotonin and dopamine receptor gene polymorphisms and the risk of extrapyramidal side effects in perphenazine-treated schizophrenic patients. Psychopharmacology (Berl) 2007;190:479–484. doi: 10.1007/s00213-006-0622-x. [DOI] [PubMed] [Google Scholar]
- 376.Hwang R, Shinkai T, De LV, Ni X, Potkin SG, Lieberman JA, Meltzer HY, Kennedy JL. Association study of four dopamine D1 receptor gene polymorphisms and clozapine treatment response. J.Psychopharmacol. 2007;21:718–727. doi: 10.1177/0269881106072341. [DOI] [PubMed] [Google Scholar]
- 377.Kato D, Kawanishi C, Kishida I, Furuno T, Suzuki K, Onishi H, Hirayasu Y. Effects of CYP2D6 polymorphisms on neuroleptic malignant syndrome. Eur.J.Clin.Pharmacol. 2007;63:991–996. doi: 10.1007/s00228-007-0355-8. [DOI] [PubMed] [Google Scholar]
- 378.Lee HJ, Kang SG, Paik JW, Lee MS, Cho BH, Park YM, Kim W, Choi JE, Jung IK, Kim L, Lee MS. No evidence for an association between G protein beta3 subunit gene C825T polymorphism and tardive dyskinesia in schizophrenia. Hum.Psychopharmacol. 2007;22:501–504. doi: 10.1002/hup.875. [DOI] [PubMed] [Google Scholar]
- 379.Lee HJ, Kang SG, Choi JE, Paik JW, Kim YK, Kim SH, Lee MS, Joe SH, Jung IK, Kim L. No association between dopamine D4 receptor gene −521 C/T polymorphism and tardive dyskinesia in schizophrenia. Neuropsychobiology. 2007;55:47–51. doi: 10.1159/000103576. [DOI] [PubMed] [Google Scholar]
- 380.Liou YJ, Wang YC, Chen JY, Bai YM, Lin CC, Liao DL, Chen TT, Chen ML, Mo GH, Lai IC. Association analysis of polymorphisms in the N-methyl-D-aspartate (NMDA) receptor subunit 2B (GRIN2B) gene and tardive dyskinesia in schizophrenia. Psychiatry Res. 2007;153:271–275. doi: 10.1016/j.psychres.2006.08.007. [DOI] [PubMed] [Google Scholar]
- 381.Mancama D, Mata I, Kerwin RW, Arranz MJ. Choline acetyltransferase variants and their influence in schizophrenia and olanzapine response. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2007;144B:849–853. doi: 10.1002/ajmg.b.30468. [DOI] [PubMed] [Google Scholar]
- 382.Mo GH, Lai IC, Wang YC, Chen JY, Lin CY, Chen TT, Chen ML, Liou YJ, Liao DL, Bai YM, Lin CC. Support for an association of the C939T polymorphism in the human DRD2 gene with tardive dyskinesia in schizophrenia. Schizophr.Res. 2007;97:302–304. doi: 10.1016/j.schres.2007.06.026. [DOI] [PubMed] [Google Scholar]
- 383.Molero P, Ortuno F, Zalacain M, Patino-Garcia A. Clinical involvement of catechol-O-methyltransferase polymorphisms in schizophrenia spectrum disorders: influence on the severity of psychotic symptoms and on the response to neuroleptic treatment. Pharmacogenomics.J. 2007;7:418–426. doi: 10.1038/sj.tpj.6500441. [DOI] [PubMed] [Google Scholar]
- 384.Mulder H, Franke B, van der-Beek van der AA, Arends J, Wilmink FW, Scheffer H, Egberts AC. The association between HTR2C gene polymorphisms and the metabolic syndrome in patients with schizophrenia. J.Clin.Psychopharmacol. 2007;27:338–343. doi: 10.1097/JCP.0b013e3180a76dc0. [DOI] [PubMed] [Google Scholar]
- 385.Mulder H, Franke B, van der-Beek van der AA, Arends J, Wilmink FW, Egberts AC, Scheffer H. The association between HTR2C polymorphisms and obesity in psychiatric patients using antipsychotics: a cross-sectional study. Pharmacogenomics.J. 2007;7:318–324. doi: 10.1038/sj.tpj.6500422. [DOI] [PubMed] [Google Scholar]
- 386.Pae CU, Kim TS, Patkar AA, Kim JJ, Lee CU, Lee SJ, Jun TY, Lee C, Paik IH. Manganese superoxide dismutase (MnSOD: Ala-9Val) gene polymorphism may not be associated with schizophrenia and tardive dyskinesia. Psychiatry Res. 2007;153:77–81. doi: 10.1016/j.psychres.2006.04.011. [DOI] [PubMed] [Google Scholar]
- 387.Ryu S, Cho EY, Park T, Oh S, Jang WS, Kim SK, Lee D, Hong KS. -759 C/T polymorphism of 5-HT2C receptor gene and early phase weight gain associated with antipsychotic drug treatment. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2007;31:673–677. doi: 10.1016/j.pnpbp.2006.12.021. [DOI] [PubMed] [Google Scholar]
- 388.Tay JK, Tan CH, Chong SA, Tan EC. Functional polymorphisms of the cytochrome P450 1A2 (CYP1A2) gene and prolonged QTc interval in schizophrenia. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2007;31:1297–1302. doi: 10.1016/j.pnpbp.2007.05.013. [DOI] [PubMed] [Google Scholar]
- 389.Tiwari AK, Deshpande SN, Lerer B, Nimgaonkar VL, Thelma BK. Genetic susceptibility to Tardive Dyskinesia in chronic schizophrenia subjects: V. Association of CYP1A2 1545 C>T polymorphism. Pharmacogenomics.J. 2007;7:305–311. doi: 10.1038/sj.tpj.6500415. [DOI] [PubMed] [Google Scholar]
- 390.Vijayan NN, Bhaskaran S, Koshy LV, Natarajan C, Srinivas L, Nair CM, Allencherry PM, Banerjee M. Association of dopamine receptor polymorphisms with schizophrenia and antipsychotic response in a South Indian population. Behav.Brain Funct. 2007;3:34. doi: 10.1186/1744-9081-3-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391.Wang L, Yu L, He G, Zhang J, Zhang AP, Du J, Tang RQ, Zhao XZ, Ma J, Xuan JK, Xiao Y, Gu NF, Feng GY, Xu MQ, Xing QH, He L. Response of risperidone treatment may be associated with polymorphisms of HTT gene in Chinese schizophrenia patients. Neurosci.Lett. 2007;414:1–4. doi: 10.1016/j.neulet.2006.09.014. [DOI] [PubMed] [Google Scholar]
- 392.Xing Q, Qian X, Li H, Wong S, Wu S, Feng G, Duan S, Xu M, Gao R, Qin W, Gao J, Meng J, He L. The relationship between the therapeutic response to risperidone and the dopamine D2 receptor polymorphism in Chinese schizophrenia patients. Int.J.Neuropsychopharmacol. 2007;10:631–637. doi: 10.1017/S146114570600719X. [DOI] [PubMed] [Google Scholar]
- 393.Xu MQ, Xing QH, Zheng YL, Li S, Gao JJ, He G, Guo TW, Feng GY, Xu F, He L. Association of AKT1 gene polymorphisms with risk of schizophrenia and with response to antipsychotics in the Chinese population. J.Clin.Psychiatry. 2007;68:1358–1367. doi: 10.4088/jcp.v68n0906. [DOI] [PubMed] [Google Scholar]
- 394.Yue W, Kang G, Zhang Y, Qu M, Tang F, Han Y, Ruan Y, Lu T, Zhang J, Zhang D. Association of DAOA polymorphisms with schizophrenia and clinical symptoms or therapeutic effects. Neurosci.Lett. 2007;416:96–100. doi: 10.1016/j.neulet.2007.01.056. [DOI] [PubMed] [Google Scholar]
- 395.Zai CC, Hwang RW, De LV, Muller DJ, King N, Zai GC, Remington G, Meltzer HY, Lieberman JA, Potkin SG, Kennedy JL. Association study of tardive dyskinesia and twelve DRD2 polymorphisms in schizophrenia patients. Int.J.Neuropsychopharmacol. 2007;10:639–651. doi: 10.1017/S1461145706007152. [DOI] [PubMed] [Google Scholar]
- 396.Zhang A, Xing Q, Wang L, Du J, Yu L, Lin Z, Li X, Feng G, He L. Dopamine transporter polymorphisms and risperidone response in Chinese schizophrenia patients: an association study. Pharmacogenomics. 2007;8:1337–1345. doi: 10.2217/14622416.8.10.1337. [DOI] [PubMed] [Google Scholar]
- 397.Zhang XY, Tan YL, Zhou DF, Haile CN, Cao LY, Xu Q, Shen Y, Kosten TA, Kosten TR. 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;27:246–251. doi: 10.1097/jcp.0b013e3180582412. [DOI] [PubMed] [Google Scholar]
- 398.Grossman I, Sullivan PF, Walley N, Liu Y, Dawson JR, Gumbs C, Gaedigk A, Leeder JS, McEvoy JP, Weale ME, Goldstein DB. Genetic determinants of variable metabolism have little impact on the clinical use of leading antipsychotics in the CATIE study. Genet.Med. 2008;10:720–729. doi: 10.1097/GIM.0b013e3181863239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399.Bozina N, Kuzman MR, Medved V, Jovanovic N, Sertic J, Hotujac L. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J.Psychiatr.Res. 2008;42:89–97. doi: 10.1016/j.jpsychires.2006.10.002. [DOI] [PubMed] [Google Scholar]
- 400.Adams DH, Close S, Farmen M, Downing AM, Breier A, Houston JP. Dopamine receptor D3 genotype association with greater acute positive symptom remission with olanzapine therapy in predominately caucasian patients with chronic schizophrenia or schizoaffective disorder. Hum.Psychopharmacol. 2008;23:267–274. doi: 10.1002/hup.930. [DOI] [PubMed] [Google Scholar]
- 401.Al Hadithy AF, Wilffert B, Stewart RE, Looman NM, Bruggeman R, Brouwers JR, Matroos GE, van OJ, Hoek HW, van Harten PN. Pharmacogenetics of parkinsonism, rigidity, rest tremor, and bradykinesia in African-Caribbean inpatients: differences in association with dopamine and serotonin receptors. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2008;147B:890–897. doi: 10.1002/ajmg.b.30746. [DOI] [PubMed] [Google Scholar]
- 402.Benmessaoud D, Hamdani N, Boni C, Ramoz N, Hamon M, Kacha F, Gorwood P. 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]
- 403.Campbell DB, Ebert PJ, Skelly T, Stroup TS, Lieberman J, Levitt P, Sullivan PF. Ethnic stratification of the association of RGS4 variants with antipsychotic treatment response in schizophrenia. Biol.Psychiatry. 2008;63:32–41. doi: 10.1016/j.biopsych.2007.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404.Crescenti A, Mas S, Gasso P, Parellada E, Bernardo M, Lafuente A. Cyp2d6*3, *4, *5 and *6 polymorphisms and antipsychotic-induced extrapyramidal side-effects in patients receiving antipsychotic therapy. Clin.Exp.Pharmacol.Physiol. 2008;35:807–811. doi: 10.1111/j.1440-1681.2008.04918.x. [DOI] [PubMed] [Google Scholar]
- 405.de LJ, Correa JC, Ruano G, Windemuth A, Arranz MJ, Diaz FJ. Exploring genetic variations that may be associated with the direct effects of some antipsychotics on lipid levels. Schizophr.Res. 2008;98:40–46. doi: 10.1016/j.schres.2007.10.003. [DOI] [PubMed] [Google Scholar]
- 406.Dolzan V, Serretti A, Mandelli L, Koprivsek J, Kastelic M, Plesnicar BK. Acute antipyschotic efficacy and side effects in schizophrenia: association with serotonin transporter promoter genotypes. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:1562–1566. doi: 10.1016/j.pnpbp.2008.05.022. [DOI] [PubMed] [Google Scholar]
- 407.Ellingrod VL, Miller dD, Taylor SF, Moline J, Holman T, Kerr J. Metabolic syndrome and insulin resistance in schizophrenia patients receiving antipsychotics genotyped for the methylenetetrahydrofolate reductase (MTHFR) 677C/T and 1298A/C variants. Schizophr.Res. 2008;98:47–54. doi: 10.1016/j.schres.2007.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408.Gu B, Wang L, Zhang AP, Ma G, Zhao XZ, Li HF, Feng GY, He L, Xing QH. Association between a polymorphism of the HTR3A gene and therapeutic response to risperidone treatment in drug-naive Chinese schizophrenia patients. Pharmacogenet.Genomics. 2008;18:721–727. doi: 10.1097/FPC.0b013e32830500e2. [DOI] [PubMed] [Google Scholar]
- 409.Gunes A, Dahl ML, Spina E, Scordo MG. Further evidence for the association between 5-HT2C receptor gene polymorphisms and extrapyramidal side effects in male schizophrenic patients. Eur.J.Clin.Pharmacol. 2008;64:477–482. doi: 10.1007/s00228-007-0450-x. [DOI] [PubMed] [Google Scholar]
- 410.Hamdani N, Tabeze JP, Ramoz N, Ades J, Hamon M, Sarfati Y, Boni C, Gorwood P. The CNR1 gene as a pharmacogenetic factor for antipsychotics rather than a susceptibility gene for schizophrenia. Eur.Neuropsychopharmacol. 2008;18:34–40. doi: 10.1016/j.euroneuro.2007.05.005. [DOI] [PubMed] [Google Scholar]
- 411.Ikeda M, Yamanouchi Y, Kinoshita Y, Kitajima T, Yoshimura R, Hashimoto S, O'Donovan MC, Nakamura J, Ozaki N, Iwata N. Variants of dopamine and serotonin candidate genes as predictors of response to risperidone treatment in first-episode schizophrenia. Pharmacogenomics. 2008;9:1437–1443. doi: 10.2217/14622416.9.10.1437. [DOI] [PubMed] [Google Scholar]
- 412.Ji X, Takahashi N, Branko A, Ishihara R, Nagai T, Mouri A, Saito S, Maeno N, Inada T, Ozaki N. An association between serotonin receptor 3B gene (HTR3B) and treatment-resistant schizophrenia (TRS) in a Japanese population. Nagoya J.Med.Sci. 2008;70:11–17. [PubMed] [Google Scholar]
- 413.Ji X, Takahashi N, Saito S, Ishihara R, Maeno N, Inada T, Ozaki N. Relationship between three serotonin receptor subtypes (HTR3A, HTR2A and HTR4) and treatment-resistant schizophrenia in the Japanese population. Neurosci.Lett. 2008;435:95–98. doi: 10.1016/j.neulet.2008.01.083. [DOI] [PubMed] [Google Scholar]
- 414.Kang SG, Choi JE, An H, Park YM, Lee HJ, Han C, Kim YK, Kim SH, Cho SN, Joe SH, Jung IK, Kim L, Lee MS. Manganese superoxide dismutase gene Ala-9Val polymorphism might be related to the severity of abnormal involuntary movements in Korean schizophrenic patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:1844–1847. doi: 10.1016/j.pnpbp.2008.08.013. [DOI] [PubMed] [Google Scholar]
- 415.Kang SG, Choi JE, An H, Lim SW, Lee HJ, Han C, Kim YK, Kim SH, Cho SN, Lee MS, Joe SH, Jung IK, Kim L. No association between the brain-derived neurotrophic factor gene Val66Met polymorphism and tardive dyskinesia in schizophrenic patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:1545–1548. doi: 10.1016/j.pnpbp.2008.05.016. [DOI] [PubMed] [Google Scholar]
- 416.Kang SG, Lee HJ, Choi JE, Park YM, Park JH, Han C, Kim YK, Kim SH, Lee MS, Joe SH, Jung IK, Kim L. Association study between antipsychotics- induced restless legs syndrome and polymorphisms of dopamine D1, D2, D3, and D4 receptor genes in schizophrenia. Neuropsychobiology. 2008;57:49–54. doi: 10.1159/000129667. [DOI] [PubMed] [Google Scholar]
- 417.Kang SG, Lee HJ, Park YM, Choi JE, Han C, Kim YK, Kim SH, Lee MS, Joe SH, Jung IK, Kim L. Possible association between the −2548A/G polymorphism of the leptin gene and olanzapine-induced weight gain. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2008;32:160–163. doi: 10.1016/j.pnpbp.2007.08.002. [DOI] [PubMed] [Google Scholar]
- 418.Kim B, Choi EY, Kim CY, Song K, Joo YH. 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;23:61–67. doi: 10.1002/hup.897. [DOI] [PubMed] [Google Scholar]
- 419.Kohlrausch FB, Salatino-Oliveira A, Gama CS, Lobato MI, Belmonte-de-Abreu P, Hutz MH. G-protein gene 825C>T polymorphism is associated with response to clozapine in Brazilian schizophrenics. Pharmacogenomics. 2008;9:1429–1436. doi: 10.2217/14622416.9.10.1429. [DOI] [PubMed] [Google Scholar]
- 420.Kohlrausch FB, Gama CS, Lobato MI, Belmonte-de-Abreu P, Callegari-Jacques SM, Gesteira A, Barros F, Carracedo A, Hutz MH. Naturalistic pharmacogenetic study of treatment resistance to typical neuroleptics in European-Brazilian schizophrenics. Pharmacogenet.Genomics. 2008;18:599–609. doi: 10.1097/FPC.0b013e328301a763. [DOI] [PubMed] [Google Scholar]
- 421.Kwon JS, Kim E, Kang DH, Choi JS, Yu KS, Jang IJ, Shin SG. Taq1A polymorphism in the dopamine D2 receptor gene as a predictor of clinical response to aripiprazole. Eur.Neuropsychopharmacol. 2008;18:897–907. doi: 10.1016/j.euroneuro.2008.07.010. [DOI] [PubMed] [Google Scholar]
- 422.Lafuente A, Bernardo M, Mas S, Crescenti A, Aparici M, Gasso P, Deulofeu R, Mane A, Catalan R, Carne X. Polymorphism of dopamine D2 receptor (TaqIA, TaqIB, and-141C Ins/Del) and dopamine degradation enzyme (COMT G158A, A-278G) genes and extrapyramidal symptoms in patients with schizophrenia and bipolar disorders. Psychiatry Res. 2008;161:131–141. doi: 10.1016/j.psychres.2007.08.002. [DOI] [PubMed] [Google Scholar]
- 423.Lane HY, Liu YC, Huang CL, Chang YC, Wu PL, Huang CH, Tsai GE. RGS4 polymorphisms predict clinical manifestations and responses to risperidone treatment in patients with schizophrenia. J.Clin.Psychopharmacol. 2008;28:64–68. doi: 10.1097/jcp.0b013e3181603f5a. [DOI] [PubMed] [Google Scholar]
- 424.Lavedan C, Volpi S, Polymeropoulos MH, Wolfgang CD. Effect of a ciliary neurotrophic factor polymorphism on schizophrenia symptom improvement in an iloperidone clinical trial. Pharmacogenomics. 2008;9:289–301. doi: 10.2217/14622416.9.3.289. [DOI] [PubMed] [Google Scholar]
- 425.Liou YJ, Wang YC, Chen JY, Chen ML, Chen TT, Bai YM, Lin CC, Liao DL, Lai IC. The coding-synonymous polymorphism rs1045280 (Ser280Ser) in beta-arrestin 2 (ARRB2) gene is associated with tardive dyskinesia in Chinese patients with schizophrenia. Eur.J.Neurol. 2008;15:1406–1408. doi: 10.1111/j.1468-1331.2008.02316.x. [DOI] [PubMed] [Google Scholar]
- 426.Meary A, Brousse G, Jamain S, Schmitt A, Szoke A, Schurhoff F, Gavaudan G, Lancon C, Macquin-Mavier I, Leboyer M, Llorca PM. Pharmacogenetic study of atypical antipsychotic drug response: involvement of the norepinephrine transporter gene. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2008;147B:491–494. doi: 10.1002/ajmg.b.30635. [DOI] [PubMed] [Google Scholar]
- 427.Mou XD, Zhang ZJ, Zhang XR, Shi JB, Sun J. -2548G/A functional polymorphism in the promoter region of leptin gene and antipsychotic agent-induced weight gain in schizophrenic patients: a study of nuclear family-based association. Zhong.Nan.Da.Xue.Xue.Bao.Yi.Xue.Ban. 2008;33:316–320. [PubMed] [Google Scholar]
- 428.Pae CU, Drago A, Kim JJ, Patkar AA, Jun TY, Lee C, Mandelli L, De RD, Paik IH, Serretti A. TAAR6 variation effect on clinic presentation and outcome in a sample of schizophrenic in-patients: an open label study. Eur.Psychiatry. 2008;23:390–395. doi: 10.1016/j.eurpsy.2008.04.004. [DOI] [PubMed] [Google Scholar]
- 429.Park YM, Cho JH, Kang SG, Choi JE, Lee SH, Kim L, Lee HJ. Lack of association between the −759C/T polymorphism of the 5-HT2C receptor gene and olanzapine-induced weight gain among Korean schizophrenic patients. J.Clin.Pharm.Ther. 2008;33:55–60. doi: 10.1111/j.1365-2710.2008.00872.x. [DOI] [PubMed] [Google Scholar]
- 430.Shao P, Zhao JP, Chen JD, Wu RR, He YQ. Association of HTR2C-759C/T and −697G/C polymorphisms with antipsychotic agent-induced weight gain. Zhong.Nan.Da.Xue.Xue.Bao.Yi.Xue.Ban. 2008;33:312–315. [PubMed] [Google Scholar]
- 431.Shinkai T, De LV, Utsunomiya K, Sakata S, Inoue Y, Fukunaka Y, Hwang R, Ohmori O, Kennedy JL, Nakamura J. Functional polymorphism of the human multidrug resistance gene (MDR1) and polydipsia-hyponatremia in schizophrenia. Neuromolecular.Med. 2008;10:362–367. doi: 10.1007/s12017-008-8041-2. [DOI] [PubMed] [Google Scholar]
- 432.Smith RC, Segman RH, Golcer-Dubner T, Pavlov V, Lerer B. Allelic variation in ApoC3, ApoA5 and LPL genes and first and second generation antipsychotic effects on serum lipids in patients with schizophrenia. Pharmacogenomics.J. 2008;8:228–236. doi: 10.1038/sj.tpj.6500474. [DOI] [PubMed] [Google Scholar]
- 433.Souza RP, Romano-Silva MA, Lieberman JA, Meltzer HY, Wong AH, Kennedy JL. Association study of GSK3 gene polymorphisms with schizophrenia and clozapine response. Psychopharmacology (Berl) 2008;200:177–186. doi: 10.1007/s00213-008-1193-9. [DOI] [PubMed] [Google Scholar]
- 434.Spellmann I, Muller N, Musil R, Zill P, Douhet A, Dehning S, Cerovecki A, Bondy B, Moller HJ, Riedel M. Associations of SNAP-25 polymorphisms with cognitive dysfunctions in Caucasian patients with schizophrenia during a brief trail of treatment with atypical antipsychotics. Eur.Arch.Psychiatry Clin.Neurosci. 2008;258:335–344. doi: 10.1007/s00406-007-0800-9. [DOI] [PubMed] [Google Scholar]
- 435.Srivastava V, Deshpande SN, Nimgaonkar VL, Lerer B, Thelma B. Genetic correlates of olanzapine-induced weight gain in schizophrenia subjects from north India: role of metabolic pathway genes. Pharmacogenomics. 2008;9:1055–1068. doi: 10.2217/14622416.9.8.1055. [DOI] [PubMed] [Google Scholar]
- 436.Wang L, Fang C, Zhang A, Du J, Yu L, Ma J, Feng G, Xing Q, He L. The --1019 C/G polymorphism of the 5-HT(1)A receptor gene is associated with negative symptom response to risperidone treatment in schizophrenia patients. J.Psychopharmacol. 2008;22:904–909. doi: 10.1177/0269881107081522. [DOI] [PubMed] [Google Scholar]
- 437.Webb BT, Sullivan PF, Skelly T, van den Oord EJ. Model-based gene selection shows engrailed 1 is associated with antipsychotic response. Pharmacogenet.Genomics. 2008;18:751–759. doi: 10.1097/FPC.0b013e32830162bc. [DOI] [PubMed] [Google Scholar]
- 438.Xu MQ, St CD, Feng GY, Lin ZG, He G, Li X, He L. BDNF gene is a genetic risk factor for schizophrenia and is related to the chlorpromazine-induced extrapyramidal syndrome in the Chinese population. Pharmacogenet.Genomics. 2008;18:449–457. doi: 10.1097/FPC.0b013e3282f85e26. [DOI] [PubMed] [Google Scholar]
- 439.Xuan J, Zhao X, He G, Yu L, Wang L, Tang W, Li X, Gu N, Feng G, Xing Q, He L. Effects of the dopamine D3 receptor (DRD3) gene polymorphisms on risperidone response: a pharmacogenetic study. Neuropsychopharmacology. 2008;33:305–311. doi: 10.1038/sj.npp.1301418. [DOI] [PubMed] [Google Scholar]
- 440.Zai CC, Romano-Silva MA, Hwang R, Zai GC, Deluca V, Muller DJ, King N, Voineskos AN, Meltzer HY, Lieberman JA, Potkin SG, Remington G, Kennedy JL. Genetic study of eight AKT1 gene polymorphisms and their interaction with DRD2 gene polymorphisms in tardive dyskinesia. Schizophr.Res. 2008;106:248–252. doi: 10.1016/j.schres.2008.08.036. [DOI] [PubMed] [Google Scholar]
- 441.Need AC, Keefe RS, Ge D, Grossman I, Dickson S, McEvoy JP, Goldstein DB. Pharmacogenetics of antipsychotic response in the CATIE trial: a candidate gene analysis. Eur.J.Hum.Genet. 2009;17:946–957. doi: 10.1038/ejhg.2008.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 442.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. 2009 doi: 10.1002/ajmg.b.30981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 443.Al Hadithy AF, Ivanova SA, Pechlivanoglou P, Semke A, Fedorenko O, Kornetova E, Ryadovaya L, Brouwers JR, Wilffert B, Bruggeman R, Loonen AJ. Tardive dyskinesia and DRD3, HTR2A and HTR2C gene polymorphisms in Russian psychiatric inpatients from Siberia. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2009;33:475–481. doi: 10.1016/j.pnpbp.2009.01.010. [DOI] [PubMed] [Google Scholar]
- 444.Al Hadithy AF, Wilffert B, Bruggeman R, Stewart RE, Brouwers JR, Matroos GE, Hoek HW, van Harten PN. Lack of association between antipsychotic-induced Parkinsonism or its subsymptoms and rs4606 SNP of RGS2 gene in African-Caribbeans and the possible role of the medication: the Curacao extrapyramidal syndromes study X. Hum.Psychopharmacol. 2009;24:123–128. doi: 10.1002/hup.997. [DOI] [PubMed] [Google Scholar]
- 445.Angelucci F, Bernardini S, Gravina P, Bellincampi L, Trequattrini A, Di IF, Vanni D, Federici G, Caltagirone C, Bossu P, Spalletta G. Delusion symptoms and response to antipsychotic treatment are associated with the 5-HT2A receptor polymorphism (102T/C) in Alzheimer's disease: a 3-year follow-up longitudinal study. J.Alzheimers.Dis. 2009;17:203–211. doi: 10.3233/JAD-2009-1031. [DOI] [PubMed] [Google Scholar]
- 446.Barlas IO, Cetin M, Erdal ME, Semiz UB, Basoglu C, Ay ME, Herken H, Uzun O. Lack of association between DRD3 gene polymorphism and response to clozapine in Turkish schizoprenia patients. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2009;150B:56–60. doi: 10.1002/ajmg.b.30770. [DOI] [PubMed] [Google Scholar]
- 447.Calarge CA, Ellingrod VL, Zimmerman B, Acion L, Sivitz WI, Schlechte JA. Leptin gene −2548G/A variants predict risperidone-associated weight gain in children and adolescents. Psychiatr.Genet. 2009;19:320–327. doi: 10.1097/ypg.0b013e3283328e06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 448.Chen SF, Shen YC, Chen CH. HTR2A A-1438G/T102C polymorphisms predict negative symptoms performance upon aripiprazole treatment in schizophrenic patients. Psychopharmacology (Berl) 2009;205:285–292. doi: 10.1007/s00213-009-1538-z. [DOI] [PubMed] [Google Scholar]
- 449.Chen SF, Shen YC, Chen CH. Effects of the DRD3 Ser9Gly polymorphism on aripiprazole efficacy in schizophrenic patients as modified by clinical factors. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2009;33:470–474. doi: 10.1016/j.pnpbp.2009.01.007. [DOI] [PubMed] [Google Scholar]
- 450.Consoli G, Lastella M, Ciapparelli A, Dell'Osso MC, Ciofi L, Guidotti E, Danesi R, Dell'Osso L, Del TM, Di PA. ABCB1 polymorphisms are associated with clozapine plasma levels in psychotic patients. Pharmacogenomics. 2009;10:1267–1276. doi: 10.2217/pgs.09.51. [DOI] [PubMed] [Google Scholar]
- 451.Du J, Zhang A, Wang L, Xuan J, Yu L, Che R, Li X, Gu N, Lin Z, Feng G, Xing Q, He L. Relationship between response to risperidone, plasma concentrations of risperidone and CYP3A4 polymorphisms in schizophrenia patients. J.Psychopharmacol. 2009 doi: 10.1177/0269881109104932. [DOI] [PubMed] [Google Scholar]
- 452.Gasso P, Mas S, Bernardo M, Alvarez S, Parellada E, Lafuente A. A common variant in DRD3 gene is associated with risperidone-induced extrapyramidal symptoms. Pharmacogenomics.J. 2009 doi: 10.1038/tpj.2009.26. [DOI] [PubMed] [Google Scholar]
- 453.Godlewska BR, Olajossy-Hilkesberger L, Ciwoniuk M, Olajossy M, Marmurowska-Michalowska H, Limon J, Landowski J. Olanzapine-induced weight gain is associated with the −759C/T and −697G/C polymorphisms of the HTR2C gene. Pharmacogenomics.J. 2009;9:234–241. doi: 10.1038/tpj.2009.18. [DOI] [PubMed] [Google Scholar]
- 454.Greenbaum L, Smith RC, Rigbi A, Strous R, Teltsh O, Kanyas K, Korner M, Lancet D, Ben-Asher E, Lerer B. Further evidence for association of the RGS2 gene with antipsychotic-induced parkinsonism: protective role of a functional polymorphism in the 3'-untranslated region. Pharmacogenomics.J. 2009;9:103–110. doi: 10.1038/tpj.2008.6. [DOI] [PubMed] [Google Scholar]
- 455.Gregoor JG, van der Weide J, Mulder H, Cohen D, van Megen HJ, Egberts AC, Heerdink ER. Polymorphisms of the LEP- and LEPR gene and obesity in patients using antipsychotic medication. J.Clin.Psychopharmacol. 2009;29:21–25. doi: 10.1097/JCP.0b013e31819359be. [DOI] [PubMed] [Google Scholar]
- 456.Gunes A, Melkersson KI, Scordo MG, Dahl ML. Association between HTR2C and HTR2A polymorphisms and metabolic abnormalities in patients treated with olanzapine or clozapine. J.Clin.Psychopharmacol. 2009;29:65–68. doi: 10.1097/JCP.0b013e31819302c3. [DOI] [PubMed] [Google Scholar]
- 457.Gupta M, Bhatnagar P, Grover S, Kaur H, Baghel R, Bhasin Y, Chauhan C, Verma B, Manduva V, Mukherjee O, Purushottam M, Sharma A, Jain S, Brahmachari SK, Kukreti R. Association studies of catechol-O-methyltransferase (COMT) gene with schizophrenia and response to antipsychotic treatment. Pharmacogenomics. 2009;10:385–397. doi: 10.2217/14622416.10.3.385. [DOI] [PubMed] [Google Scholar]
- 458.Herken H, Erdal M, Aydin N, Sengul C, Karadag F, Barlas O, Akin F. The association of olanzapine-induced weight gain with peroxisome proliferator-activated receptor-gamma2 Pro12Ala polymorphism in patients with schizophrenia. DNA Cell Biol. 2009;28:515–519. doi: 10.1089/dna.2009.0893. [DOI] [PubMed] [Google Scholar]
- 459.Jaquenoud SE, Knezevic B, Morena GP, Harenberg S, Oneda B, Crettol S, Ansermot N, Baumann P, Eap CB. ABCB1 and cytochrome P450 polymorphisms: clinical pharmacogenetics of clozapine. J.Clin.Psychopharmacol. 2009;29:319–326. doi: 10.1097/JCP.0b013e3181acc372. [DOI] [PubMed] [Google Scholar]
- 460.Kang SG, Lee HJ, Choi JE, An H, Rhee M, Kim L. Association study between glutathione S-transferase GST-M1, GST-T1, and GST-P1 polymorphisms and tardive dyskinesia. Hum.Psychopharmacol. 2009;24:55–60. doi: 10.1002/hup.988. [DOI] [PubMed] [Google Scholar]
- 461.Kobylecki CJ, Jakobsen KD, Hansen T, Jakobsen IV, Rasmussen HB, Werge T. CYP2D6 genotype predicts antipsychotic side effects in schizophrenia inpatients: a retrospective matched case-control study. Neuropsychobiology. 2009;59:222–226. doi: 10.1159/000223734. [DOI] [PubMed] [Google Scholar]
- 462.Kwon JS, Joo YH, Nam HJ, Lim M, Cho EY, Jung MH, Choi JS, Kim B, Kang DH, Oh S, Park T, Hong KS. Association of the glutamate transporter gene SLC1A1 with atypical antipsychotics-induced obsessive-compulsive symptoms. Arch.Gen.Psychiatry. 2009;66:1233–1241. doi: 10.1001/archgenpsychiatry.2009.155. [DOI] [PubMed] [Google Scholar]
- 463.Laika B, Leucht S, Heres S, Schneider H, Steimer W. Pharmacogenetics and olanzapine treatment: CYP1A2*1F and serotonergic polymorphisms influence therapeutic outcome. Pharmacogenomics.J. 2009 doi: 10.1038/tpj.2009.32. [DOI] [PubMed] [Google Scholar]
- 464.Le HS, Theisen FM, Haberhausen M, Raeder MB, Ferno J, Gebhardt S, Hinney A, Remschmidt H, Krieg JC, Mehler-Wex C, Nothen 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. doi: 10.1038/sj.mp.4002133. [DOI] [PubMed] [Google Scholar]
- 465.Lee HY, Kim DJ, Lee HJ, Choi JE, Kim YK. No association of serotonin transporter polymorphism (5-HTTVNTR and 5-HTTLPR) with characteristics and treatment response to atypical antipsychotic agents in schizophrenic patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2009;33:276–280. doi: 10.1016/j.pnpbp.2008.11.013. [DOI] [PubMed] [Google Scholar]
- 466.Liou YJ, Chen ML, Wang YC, Chen JY, Liao DL, Bai YM, Lin CC, Chen TT, Mo GH, Lai IC. Analysis of genetic variations in the RGS9 gene and antipsychotic-induced tardive dyskinesia in schizophrenia. Am.J.Med.Genet.B Neuropsychiatr.Genet. 2009;150B:239–242. doi: 10.1002/ajmg.b.30796. [DOI] [PubMed] [Google Scholar]
- 467.Mossner R, Schuhmacher A, Kuhn KU, Cvetanovska G, Rujescu D, Zill P, Quednow BB, Rietschel M, Wolwer W, Gaebel W, Wagner M, Maier W. Functional serotonin 1A receptor variant influences treatment response to atypical antipsychotics in schizophrenia. Pharmacogenet.Genomics. 2009;19:91–94. doi: 10.1097/FPC.0b013e328311a917. [DOI] [PubMed] [Google Scholar]
- 468.Mulder H, Cohen D, Scheffer H, Gispen-de WC, Arends J, Wilmink FW, Franke B, Egberts AC. HTR2C gene polymorphisms and the metabolic syndrome in patients with schizophrenia: a replication study. J.Clin.Psychopharmacol. 2009;29:16–20. doi: 10.1097/JCP.0b013e3181934462. [DOI] [PubMed] [Google Scholar]
- 469.Park SW, Lee JG, Kong BG, Lee SJ, Lee CH, Kim JI, Kim YH. Genetic association of BDNF val66met and GSK-3beta-50T/C polymorphisms with tardive dyskinesia. Psychiatry Clin.Neurosci. 2009;63:433–439. doi: 10.1111/j.1440-1819.2009.01976.x. [DOI] [PubMed] [Google Scholar]
- 470.Sickert L, Muller DJ, Tiwari AK, Shaikh S, Zai C, De SR, De LV, Meltzer HY, Lieberman JA, Kennedy JL. Association of the alpha2A adrenergic receptor −1291C/G polymorphism and antipsychotic-induced weight gain in European-Americans. Pharmacogenomics. 2009;10:1169–1176. doi: 10.2217/pgs.09.43. [DOI] [PubMed] [Google Scholar]
- 471.Volpi S, Potkin SG, Malhotra AK, Licamele L, Lavedan C. Applicability of a genetic signature for enhanced iloperidone efficacy in the treatment of schizophrenia. J.Clin.Psychiatry. 2009;70:801–809. doi: 10.4088/jcp.08m04391. [DOI] [PubMed] [Google Scholar]
- 472.Wei Z, Wang L, Xuan J, Che R, Du J, Qin S, Xing Y, Gu B, Yang L, Li H, Li J, Feng G, He L, Xing Q. Association analysis of serotonin receptor 7 gene (HTR7) and risperidone response in Chinese schizophrenia patients. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2009;33:547–551. doi: 10.1016/j.pnpbp.2009.02.008. [DOI] [PubMed] [Google Scholar]
- 473.Wilffert B, Al Hadithy AF, Sing VJ, Matroos G, Hoek HW, van OJ, Bruggeman R, Brouwers JR, van Harten PN. 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;23:652–659. doi: 10.1177/0269881108091594. [DOI] [PubMed] [Google Scholar]
- 474.Zai CC, Tiwari AK, Basile V, De LV, Muller DJ, King N, Voineskos AN, Remington G, Meltzer HY, Lieberman JA, Potkin SG, Kennedy JL. Association study of tardive dyskinesia and five DRD4 polymorphisms in schizophrenia patients. Pharmacogenomics.J. 2009;9:168–174. doi: 10.1038/tpj.2009.2. [DOI] [PubMed] [Google Scholar]
- 475.Zai CC, Tiwari AK, Basile V, De LV, Muller DJ, Voineskos AN, Remington G, Meltzer HY, Lieberman JA, Potkin SG, Kennedy JL. Oxidative stress in tardive dyskinesia: Genetic association study and meta-analysis of NADPH quinine oxidoreductase 1 (NQO1) and Superoxide dismutase 2 (SOD2, MnSOD) genes. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2009 doi: 10.1016/j.pnpbp.2009.09.020. [DOI] [PubMed] [Google Scholar]
- 476.Zuo L, Luo X, Krystal JH, Cramer J, Charney DS, Gelernter J. The efficacies of clozapine and haloperidol in refractory schizophrenia are related to DTNBP1 variation. Pharmacogenet.Genomics. 2009;19:437–446. doi: 10.1097/FPC.0b013e32832b9cfc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 477.McClay JL, Adkins DE, Aberg 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. 2009 doi: 10.1038/mp.2009.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 478.Alkelai A, Greenbaum L, Rigbi A, Kanyas K, Lerer B. Genome-wide association study of antipsychotic-induced parkinsonism severity among schizophrenia patients. Psychopharmacology (Berl) 2009;206:491–499. doi: 10.1007/s00213-009-1627-z. [DOI] [PubMed] [Google Scholar]
- 479.Aberg K, Adkins DE, Bukszar J, Webb BT, Caroff SN, Miller DD, Sebat J, Stroup S, Fanous AH, Vladimirov VI, McClay JL, Lieberman JA, Sullivan PF, van den Oord EJ. Genome-wide Association Study of Movement-Related Adverse Antipsychotic Effects. Biol.Psychiatry. 2009 doi: 10.1016/j.biopsych.2009.08.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 480.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. doi: 10.1038/mp.2008.56. [DOI] [PubMed] [Google Scholar]
- 481.Volpi S, Heaton C, Mack K, Hamilton JB, Lannan R, Wolfgang CD, Licamele L, Polymeropoulos MH, Lavedan C. Whole genome association study identifies polymorphisms associated with QT prolongation during iloperidone treatment of schizophrenia. Mol.Psychiatry. 2009;14:1024–1031. doi: 10.1038/mp.2008.52. [DOI] [PubMed] [Google Scholar]
- 482.Arranz MJ, de LJ. Pharmacogenetics and pharmacogenomics of schizophrenia: a review of last decade of research. Mol.Psychiatry. 2007;12:707–747. doi: 10.1038/sj.mp.4002009. [DOI] [PubMed] [Google Scholar]
- 483.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. doi: 10.1097/01213011-200503000-00003. [DOI] [PubMed] [Google Scholar]
- 484.Miyamoto S, Duncan GE, Marx CE, Lieberman JA. Treatments for schizophrenia: a critical review of pharmacology and mechanisms of action of antipsychotic drugs. Mol.Psychiatry. 2005;10:79–104. doi: 10.1038/sj.mp.4001556. [DOI] [PubMed] [Google Scholar]
- 485.Lieberman JA. Dopamine partial agonists: a new class of antipsychotic. CNS Drugs. 2004;18:251–267. doi: 10.2165/00023210-200418040-00005. [DOI] [PubMed] [Google Scholar]
- 486.Mamo D, Kapur S, Shammi CM, Papatheodorou G, Mann S, Therrien F, Remington G. A PET study of dopamine D2 and serotonin 5-HT2 receptor occupancy in patients with schizophrenia treated with therapeutic doses of ziprasidone. Am.J.Psychiatry. 2004;161:818–825. doi: 10.1176/appi.ajp.161.5.818. [DOI] [PubMed] [Google Scholar]
- 487.Arinami T, Gao M, Hamaguchi H, Toru M. A functional polymorphism in the promoter region of the dopamine D2 receptor gene is associated with schizophrenia. Hum.Mol.Genet. 1997;6:577–582. doi: 10.1093/hmg/6.4.577. [DOI] [PubMed] [Google Scholar]
- 488.Jonsson EG, Nothen MM, Grunhage F, Farde L, Nakashima Y, Propping P, Sedvall GC. Polymorphisms in the dopamine D2 receptor gene and their relationships to striatal dopamine receptor density of healthy volunteers. Mol.Psychiatry. 1999;4:290–296. doi: 10.1038/sj.mp.4000532. [DOI] [PubMed] [Google Scholar]
- 489.Goldman D, Urbanek M, Guenther D, Robin R, Long JC. A functionally deficient DRD2 variant [Ser311Cys] is not linked to alcoholism and substance abuse. Alcohol. 1998;16:47–52. doi: 10.1016/s0741-8329(97)00176-6. [DOI] [PubMed] [Google Scholar]
- 490.Bakker PR, van Harten PN, van OJ. Antipsychotic-induced tardive dyskinesia and polymorphic variations in COMT, DRD2, CYP1A2 and MnSOD genes: a meta-analysis of pharmacogenetic interactions. Mol.Psychiatry. 2008;13:544–556. doi: 10.1038/sj.mp.4002142. [DOI] [PubMed] [Google Scholar]
- 491.Lundstrom K, Turpin MP. Proposed schizophrenia-related gene polymorphism: expression of the Ser9Gly mutant human dopamine D3 receptor with the Semliki Forest virus system. Biochem.Biophys.Res.Commun. 1996;225:1068–1072. doi: 10.1006/bbrc.1996.1296. [DOI] [PubMed] [Google Scholar]
- 492.Bakker PR, van Harten PN, van OJ. Antipsychotic-induced tardive dyskinesia and the Ser9Gly polymorphism in the DRD3 gene: a meta analysis. Schizophr.Res. 2006;83:185–192. doi: 10.1016/j.schres.2006.01.010. [DOI] [PubMed] [Google Scholar]
- 493.Meltzer HY. Role of serotonin in the action of atypical antipsychotic drugs. Clin.Neurosci. 1995;3:64–75. [PubMed] [Google Scholar]
- 494.Meltzer HY, Li Z, Kaneda Y, Ichikawa J. Serotonin receptors: their key role in drugs to treat schizophrenia. Prog.Neuropsychopharmacol.Biol.Psychiatry. 2003;27:1159–1172. doi: 10.1016/j.pnpbp.2003.09.010. [DOI] [PubMed] [Google Scholar]
- 495.Kroeze WK, Kristiansen K, Roth BL. Molecular biology of serotonin receptors structure and function at the molecular level. Curr.Top.Med.Chem. 2002;2:507–528. doi: 10.2174/1568026023393796. [DOI] [PubMed] [Google Scholar]
- 496.Arranz MJ, Munro J, Sham P, Kirov G, Murray RM, Collier DA, Kerwin RW. Meta-analysis of studies on genetic variation in 5-HT2A receptors and clozapine response. Schizophr.Res. 1998;32:93–99. doi: 10.1016/s0920-9964(98)00032-2. [DOI] [PubMed] [Google Scholar]
- 497.Lerer B, Segman RH, Tan EC, 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. doi: 10.1017/S1461145705005389. [DOI] [PubMed] [Google Scholar]
- 498.De LV, Mueller DJ, de BA, Kennedy JL. Association of the HTR2C gene and antipsychotic induced weight gain: a meta-analysis. Int.J.Neuropsychopharmacol. 2007;10:697–704. doi: 10.1017/S1461145707007547. [DOI] [PubMed] [Google Scholar]
- 499.Roche Molecular systems I. AmpliChip CYP450 Test for in Vitro Diagnostic Use. Roche Diagnostics. 2006 [Google Scholar]
- 500.Katsanis SH, Javitt G, Hudson K. Public health. A case study of personalized medicine. Science. 2008;320:53–54. doi: 10.1126/science.1156604. [DOI] [PubMed] [Google Scholar]
- 501.Recommendations from the EGAPP Working Group: testing for cytochrome P450 polymorphisms in adults with nonpsychotic depression treated with selective serotonin reuptake inhibitors. Genet.Med. 2007;9:819–825. doi: 10.1097/gim.0b013e31815bf9a3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 502.Wagner JA. Overview of biomarkers and surrogate endpoints in drug development. Dis.Markers. 2002;18:41–46. doi: 10.1155/2002/929274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 503.FDA. Guidance for Industry: Pharmacogenomic data submission. FDA. 2005 [Google Scholar]
- 504.Frueh FW, Amur S, Mummaneni P, Epstein RS, Aubert RE, DeLuca TM, Verbrugge RR, Burckart GJ, Lesko LJ. Pharmacogenomic biomarker information in drug labels approved by the United States food and drug administration: prevalence of related drug use. Pharmacotherapy. 2008;28:992–998. doi: 10.1592/phco.28.8.992. [DOI] [PubMed] [Google Scholar]
- 505.Arranz MJ, Kapur S. Pharmacogenetics in psychiatry: are we ready for widespread clinical use? Schizophr.Bull. 2008;34:1130–1144. doi: 10.1093/schbul/sbn114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 506.van der WJ, Steijns LS, van Weelden MJ. The effect of smoking and cytochrome P450 CYP1A2 genetic polymorphism on clozapine clearance and dose requirement. Pharmacogenetics. 2003;13:169–172. doi: 10.1097/00008571-200303000-00006. [DOI] [PubMed] [Google Scholar]
- 507.Desai HD, Seabolt J, Jann MW. Smoking in patients receiving psychotropic medications: a pharmacokinetic perspective. CNS.Drugs. 2001;15:469–494. doi: 10.2165/00023210-200115060-00005. [DOI] [PubMed] [Google Scholar]
- 508.Spina E, de LJ. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin.Pharmacol.Toxicol. 2007;100:4–22. doi: 10.1111/j.1742-7843.2007.00017.x. [DOI] [PubMed] [Google Scholar]