Abstract
Although weight gain is a serious but variable adverse effect of antipsychotics that has genetic underpinnings, a comprehensive meta-analysis of pharmacogenetics of antipsychotic-related weight gain is missing. In this review, random effects meta-analyses were conducted for dominant and recessive models on associations of specific single nucleotide polymorphisms (SNP) with prospectively assessed antipsychotic-related weight or body mass index (BMI) changes (primary outcome), or categorical increases in weight or BMI (≥7%; secondary outcome). Published studies, identified via systematic database search (last search: December 31, 2014), plus 3 additional cohorts, including 222 antipsychotic-naïve youth, and 81 and 141 first-episode schizophrenia adults, each with patient-level data at 3 or 4 months treatment, were meta-analyzed. Altogether, 72 articles reporting on 46 non-duplicated samples (n = 6700, mean follow-up = 25.1wk) with 38 SNPs from 20 genes/genomic regions were meta-analyzed (for each meta-analysis, studies = 2–20, n = 81–2082). Eleven SNPs from 8 genes were significantly associated with weight or BMI change, and 4 SNPs from 2 genes were significantly associated with categorical weight or BMI increase. Combined, 13 SNPs from 9 genes (Adrenoceptor Alpha-2A [ADRA2A], Adrenoceptor Beta 3 [ADRB3], Brain-Derived Neurotrophic Factor [BDNF], Dopamine Receptor D2 [DRD2], Guanine Nucleotide Binding Protein [GNB3], 5-Hydroxytryptamine (Serotonin) Receptor 2C [HTR2C], Insulin-induced gene 2 [INSIG2], Melanocortin-4 Receptor [MC4R], and Synaptosomal-associated protein, 25kDa [SNAP25]) were significantly associated with antipsychotic-related weight gain (P-values < .05–.001). SNPs in ADRA2A, DRD2, HTR2C, and MC4R had the largest effect sizes (Hedges’ g’s = 0.30–0.80, ORs = 1.47–1.96). Less prior antipsychotic exposure (pediatric or first episode patients) and short follow-up (1–2 mo) were associated with larger effect sizes. Individual antipsychotics did not significantly moderate effect sizes. In conclusion, antipsychotic-related weight gain is polygenic and associated with specific genetic variants, especially in genes coding for antipsychotic pharmacodynamic targets.
Key words: pharmacogenetics, SNP, antipsychotics, weight gain, BMI, meta-analysis
Introduction
Antipsychotics are first-line therapy for schizophrenia-spectrum disorders,1 and frequently used as monotherapy or combined with mood stabilizers or antidepressants for bipolar disorder2 and major depression,3,4 respectively. Despite their efficacy, body weight gain and associated metabolic syndrome are prominent side effects, which increase morbidity and mortality in psychiatric patients.5–7 Many antipsychotics can cause significant weight gain, especially some second-generation antipsychotics (SGAs), like clozapine, olanzapine, and quetiapine.5,8 No consistent clinical predictors of antipsychotic-induced weight gain have been identified, and the pathophysiology of weight gain remains poorly understood.9 Food intake, energy utilization, metabolism, and body weight are regulated by complex interactions among multiple neurotransmitter systems in multiple brain regions, which are pharmacodynamic targets of antipsychotics to some extent.5,10 Genetic factors may play an important role because genome-wide association studies found multiple genes associated with obesity in the general population,11 and functions of proteins that are pharmacodynamic antipsychotic targets may be affected by genetic variants.12,13
Since the late 1990s, pharmacogenetic research has attempted to elucidate genetic underpinnings of antipsychotic-related weight gain. The present study aimed to conduct a comprehensive meta-analysis of the associations of genetic variants with antipsychotic-related weight gain. One key methodological issue in the pharmacogenetics of antipsychotic drug response is that most studies used chronic patient samples.14 Therefore, the present meta-analysis also included data from 3 cohorts of patients with first episode psychosis or minimal prior drug exposure that were largely unpublished, in addition to published studies.
Methods
Literature Search
Two investigators (J-P.Z., R.X.Z., M.N. and/or L.M.) independently conducted an electronic PubMed/Web of Science search (last: December 31, 2014) for pharmacogenetic studies of antipsychotic-related weight change. Combinations of the following key words were used: antipsychotic(s), neuroleptic(s), genetic(s), genomic(s), gene, single nucleotide polymorphism (SNP), polymorphism, weight gain, body mass index (BMI), obesity, and metabolic. We also screened reference lists from identified papers and reviews for additional studies. Inclusion criteria were: (1) humans with mental illness; (2) longitudinal data on body weight or BMI change, or percentage of patients gaining significant weight or BMI within each genotype group after a specified period of antipsychotic treatment; and (3) sufficient data to compute an effect size (ES). Exclusion criteria were: (1) animal or healthy subject studies; (2) cross-sectional studies or longitudinal studies that did not report pre- and post-treatment change in weight or BMI; (3) studies of metabolic syndrome not reporting separate weight or BMI data; (4) studies of SNPs not examined in other studies (“orphan” SNPs); and (5) studies reporting data overlapping with previously published papers (data from the largest report were included). If a SNP was studied in ≥2 independent samples, data from the 3 additional cohorts were added whenever possible so that each SNP was meta-analyzed with ≥5 samples. One study examined candidate genes in association with antipsychotic-related weight gain15 using data from the CATIE trial (Clinical Antipsychotic Trials of Intervention Effectiveness), but it was excluded from the meta-analysis due to its use of different weight gain phenotype. Weight gain was defined in the study as the maximum percent weight change at any time point during the first 18 months of treatment, which was different from all other studies where weight gain was defined as weight change between 2 time points, eg, from baseline to a follow-up time point.
Data Extraction and Outcome Variables
Data were independently extracted by 2 authors (J-P.Z., M.N. and/or L.M.); disagreements were resolved by consensus. For missing information, first and/or last authors were contacted requesting additional/unpublished data.
Primary outcome was change in weight or BMI from baseline until after a specified period of antipsychotic treatment. Whenever ≥1 assessment time points were reported, we picked the one closest to 2–3 months (preferring 3 mo) to increase homogeneity. Change in kg or kg/m2 were preferred but pooled with percent change in weight or BMI if only these were reported. Secondary outcome was the percentage of patients within each genotype group gaining significant weight or BMI during antipsychotic treatment. Most studies used ≥7% body weight gain, but some used ≥5% or ≥10%, which were also pooled.
Additional Cohorts
Three additional cohorts that had published data for at least 1 SNP but for which we obtained patient-level data were also included in the meta-analysis.
In the Second-Generation Antipsychotic Treatment Indications, Effectiveness and Tolerability in Youth (SATIETY sample) study, 222 pediatric patients (age = 13.8±3.6 y; male = 57%) with ≤7 days of antipsychotic lifetime history initiated clinicians’ choice antipsychotics (aripiprazole, olanzapine, quetiapine or risperidone) for the first time and were followed for 3 months.16
In the Zucker Hillside Hospital First Episode Schizophrenia Clinical Trial (ZHH-FE sample), 81 first-episode schizophrenia patients (age = 23.0±4.9 y; male = 75%) were randomized to risperidone or olanzapine and followed for 4 months.17,18
In the European First Episode Schizophrenia Trial (EUFEST sample), 141 first-episode schizophrenia patients (age = 25.6±5.2 y; male = 60%) were randomized to amisulpride, haloperidol, olanzapine, quetiapine or ziprasidone and had weight change data at 3 months follow-up.19
All 3 cohorts were genotyped on approximately 1 million SNPs using the Illumina Omni-1Quad platform, followed by standard quality control procedures (details published previously20). For SNPs included in the meta-analysis, but not genotyped, the SNAP online tool from the Broad Institute was accessed to find proxy SNPs using either the 1000 Genomes Pilot 1 or HapMap 3 CEU population panel, with parameters set as r 2 ≥ .80 and distance ≤100kb.
Statistical Analysis
Outcomes were analyzed separately for each SNP using Comprehensive Meta-Analysis software version 2 (Biostat) whenever ≥2 studies contributed data (otherwise data went into the “orphan” SNP category). For continuous and categorical outcomes, Hedges’ g and OR, ±95% CIs, were calculated as the ES measure. For each SNP with sufficient data, both dominant and recessive genetic models were meta-analyzed in association with weight or BMI changes. For selected SNPs in which results from dominant and recessive models on the primary outcome were suggestive of additive effects of the risk allele, a formal test of additive genetic model was conducted. For each study, a linear regression of BMI change on the number of risk alleles (0, 1, 2) was simulated based on summary statistics (mean, SD, n) from each genotype group in R statistical package, and the regression coefficient was converted to Hedges’ g with corresponding SE (supplementary methods). Pooled ESs were computed with a random effects model to accommodate heterogeneity across included studies.21 In each meta-analysis, a cohort was included only once. Statistical significance of the pooled ES was set at alpha = .05 without multiple testing correction because each SNP was chosen based on previous research, following a hypothesis-driven approach.
Study heterogeneity was assessed using Q and I 2 statistics, with I 2 < 25% representing low, ~50% moderate, and >75% representing high heterogeneity.21 Whenever heterogeneity was present, moderator and meta-regression analyses were conducted to explore moderator effects. Sensitivity analyses were conducted to assess potential influences of any one single study on the pooled ES. Within each meta-analysis, included studies were removed one at a time to check for significant alterations of the pooled ES and associated P-values. Publication bias was assessed with the funnel plot, Egger’s regression test,22 and the “Trim and Fill” method.23
Based on the Venice guideline of systematically assessing cumulative evidence on genetic association,24 each SNP was assigned a category based on 3 criteria: (1) amount of evidence (A: large-scale evidence, total sample size n > 1000; B: moderate amount of evidence, n = 500–1000; C: little evidence, n < 500); (2) replication (A: statistically significant overall ES with no/minimal between-study heterogeneity, I 2 < 25%; B: statistically significant overall ES with moderate to large between-study heterogeneity, I 2 ≥ 25%; C: insignificant overall ES); and (3) evidence of bias (A: statistically significant overall ES without evidence of bias based on “Trim and Fill” method and Egger’s test; B: evidence of bias without significance level change after adjustment, or insignificant overall ES without evidence of bias; C: evidence of bias). To generate an overall index of the strength of evidence, categories of A, B, and C were assigned a score of 3, 2, and 1, respectively, and a total score across the 3 categories was calculated. A total score = 8–9 was considered strong evidence supporting the genotype-phenotype association, a score = 6–7 was considered moderate evidence, and a score ≤5 was considered minimal evidence.
To explore polygenic effects of SNPs, a polygenic risk score was computed in the SATIETY and EUFEST cohorts using an additive genetic model combining top SNPs that were significantly associated with weight gain from the meta-analysis. We also assessed the percent variance explained by the risk score.
Results
Literature Search
The literature search produced 586 unduplicated hits, of which 72 reports (see table 1 for details) met inclusion criteria, entering into the meta-analysis (supplementary figure 1). Sample sizes varied from 32 to 481 and most studies included chronic patients (21 studies included first-episode or antipsychotic-naïve patients). Altogether, the 72 reports referred to 46 independent samples, as several published studies reported on different genes or SNPs from the same cohort. After eliminating redundancy, the total sample size from the 46 samples was 6615. Including the 3 additional cohorts (n = 444; 289 of which were already published), the total independent sample size in 46 samples was 6770. Most studies were short-term, ranging from 4 weeks to 4 months, but 16 studies (22%) had follow-up at ≥1 year (mean follow-up= 25.1±42.0wk, range = 1 mo to 7 y). Most patients were either Caucasian or Asian, including 30 studies (41.7%) with all Asian patients. The most common antipsychotics were olanzapine (52.8%), risperidone (41.7%), and clozapine (40.3%). Thirty-five studies involved monotherapy with a single antipsychotic, including clozapine (studies = 15), olanzapine (studies = 15), risperidone (studies = 4), and iloperidone (studies = 1). Most studies included schizophrenia patients (94.4% of studies), but some also included patients with various psychiatric diagnoses (table 1).
Table 1.
Study (First Author, Year) | Genes (SNPs) Included in Meta-Analysis | n | Length (wk) | Age | % Male | % Caucasian | Diagnoses | %FE, Drug-Naïve | APs | Outcome Variables |
---|---|---|---|---|---|---|---|---|---|---|
Basile 200171 | HTR2C (rs6318), ADRB3 (rs4994), TNF (rs1800629), HTR2A (rs6313, rs6314) | 80 | 6 | 33.1±8.4 | 65 | 72 | SCZ | NR | CLZ | Weight change |
Basile 200272 | HTR2C (rs3813929) | 80 | 6 | NR | 65 | 72 | SCZ | NR | CLZ | Weight change |
Bishop 200673 | GNB3 (rs5443) | 42 | 6 | 36.0±8.7 | 81 | NR | SCZ | 50% | OLZ | % Weight change |
Brandl 201274 | LEP (rs7799039) | 181 | 6 to 14 | 35.9±10.9 | 65 | 70 | SCZ, SZA | NR | Various APs | % Weight change |
LEPR (rs1137101) | ||||||||||
Calarge 200975 | LEP (rs7799039) | 74 | 2 y | 11.7±2.9 | 91 | 84 | Various diagnoses | NR | RIS | BMI change |
Chowdhury 201276 | MC4R (rs17782313) | 224 | 6 to 14 | 35.6±10.5 | 67 | 70 | SCZ, SZA | NR | Various APs | % Weight change |
Czerwensky 201377 | MC4R (rs17782313) | 173 | 4 | 39.3±14.7 | 37 | NR | Various diagnoses | 28% | Various APs | BMI change |
Weight change | ||||||||||
Czerwensky 201378 | MC4R (rs489693) | 169 | 4 | 39.3±14.7 | 37 | NR | Various diagnoses | 28% | Various APs | BMI change |
Weight change | ||||||||||
Ellingrod 200579 | HTR2C (rs3813929) | 42 | 6 | NR | 81 | 100 | SCZ | NR | OLZ | >10% Weight gain |
Ellingrod 200780 | LEP (rs7799039) | 37 | 6 | 37.0±8.4 | 81 | NR | SCZ | NR | OLZ | BMI change |
LEPR (rs1137101) | ||||||||||
Fernandez 201081 | LEP (rs7799039) | 56 | 14 | 39.1±9.0 | 79 | NR | SCZ | NR | CLZ | Weight change, BMI change |
LEPR (rs1137101) | ||||||||||
Godlewska 200936 | HTR2C (rs3813929, rs518147) | 107 | 6 | 29.3±10.0 | 50 | 100 | SCZ | 34 | OLZ | % BMI change, ≥10% BMI gain |
Herken 200982 | PPARG (rs1801282) | 95 | 6 | 34.4±13.0 | 52 | 100 | SCZ | NR | OLZ | Weight change |
BMI change | ||||||||||
Hoekstra 201083 | HTR2C (rs3813929) | 32 | 8 | 8.7±2.8 | 88 | NR | PDD | NR | RIS | Weight change |
BMI change | ||||||||||
Hong 200184 | HTR2A (rs6313) | 93 | 17 | 37.1±8.2 | 65 | 0 (100% Asian) | SCZ | 0 | CLZ | Weight change |
HTR2C (rs6318) | ||||||||||
HTR6 (T267C) | ||||||||||
Hong 201085 | ANKK1 (rs1800497) | 479 | 4 y | 47.2±13.2 | 61 | 0 (100% Asian) | SCZ | NR | CLZ, OLZ, RIS | ≥7% weight gain |
DRD2 (rs1799978, rs7131056, rs6275, rs2242591) | ||||||||||
Houston 201286 | ANKK1 (rs1800497) | 205 | 8 | NR | NR | 100 | Various diagnoses | NR | OLZ | Weight change |
DRD2 (rs1079598, rs1801028, rs2242591) | ||||||||||
HTR2C (rs3813929, rs518147, rs6318) | ||||||||||
Huang 201187 | TNF (rs1800629) | 500 | 5 y | 43.9±9.1 | 60 | 0 (100% Asian) | SCZ | NR | CLZ, OLZ, RIS | % Weight change |
>7% Weight gain | ||||||||||
Kang 200888 | LEP (rs7799039) | 74 | >3 mo | 47.2±11.6 | 68 | 0 (100% Asian) | SCZ | NR | OLZ | Weight change, ≥7% weight gain |
Kuzman 200889 | HTR2C (rs3813929) | 108 | 4 mo | 30.6±11.5 | 0 | 100 | SCZ | 64 | OLZ, RIS | ≥7% Weight gain |
MDR1 (rs2032582, rs1045642) | ||||||||||
Kuzman 201190 | HTR2C (rs3813929) | 101 | 3 mo | 33.5±10.6 | 0 | 100 | SCZ, SZA, delusional disorder | 100 | OLZ, RIS | BMI change |
MDR1 (rs2032582, rs1045642) | ||||||||||
Laika 201091 | HTR2C (rs3813929) | 56 | 4 | 41.6±15.9 | 50 | 100 | Various diagnoses | NR | OLZ | Weight change, BMI change |
Lane 200692 | HTR2A (rs6313, rs6314) | 123 | 6 | 34.0±9.7 | 55 | 0 (100% Asian) | SCZ | NR | RIS | Weight change |
>7% weight gain | ||||||||||
HTR2C (rs3813929) | ||||||||||
HTR6 (rs1805054) | ||||||||||
DRD2 (rs1799732, rs1801028) | ||||||||||
ANKK1 (rs1800497) | ||||||||||
BDNF (rs6265) | ||||||||||
Le Hellard 200993 | INSIG2 (rs10490624, rs17047764, rs17587100, rs7566605) | 160 | 3 mo | 21.9±8.9 | 61 | 100 | SCZ spectrum disorders | 0 | Various APs | BMI change |
Lencz 201094 | DRD2 (rs1799732) | 58 | 16 | 23.5±4.9 | 76 | 28 | SCZ, SZA, SZP | 100 | RIS, OLZ | Weight change |
Lin 200695 | MDR1 (rs1045642) | 41 | 6 wk | 35.7±8.8 | 80 | 90 | SCZ | NR | OLZ | Weight change |
Malhotra 201220 | MC4R (rs489693) | 139 | 12 | 13.4±3.8 | 58 | 55.4 | Various diagnoses | 100 | RIS, APZ, QTP | BMI change |
Weight change | ||||||||||
73 | 6 | 33.5±8.3 | 62 | 70 | SCZ | 0 | CLZ | BMI change | ||
Weight change | ||||||||||
40 | 6 | 35.2±11. | 55 | 100 | SCZ, SZA | 0 | RIS, APZ, QTP | BMI change | ||
Weight change | ||||||||||
92 | 12 | 26.0±5.2 | 58 | 100 | SCZ, SZA, SZP | 100 | Various APs | BMI change | ||
Weight change | ||||||||||
Miller 200596 | HTR2C (rs3813929) | 41 | 6 mo | 35.6±9.7 | 63 | 85 | SCZ | 0 | CLZ | Weight change |
% BMI change | ||||||||||
>7% BMI gain | ||||||||||
Monteleone 201097 | CNR1 (rs1049353) | 83 | 26.1 | 44±10.5 | 60 | 100 | Various diagnoses | NR | Various APs | ≥7% weight gain |
Mou 200898 | LEP (rs7799039) | 84 | 10 | 24±6.0 | 65 | 0 (100% Asian) | SCZ | 100 | RIS, CPZ | ≥7% weight gain |
Mueller 200599 | SNAP25 (rs1051312, rs3746544, rs8636) | 59 | 14 | 40.1±9.5 | 78 | 25 | SCZ, SZA | 0 | Various Aps | Weight change |
BMI change | ||||||||||
Mueller 2012100 | ANKK1 (rs1800497) | 206 | 6 or 14 | 35.7±10.4 | 68 | 72> >62 | SCZ, SZA | 0 | Various APs | % Weight change |
>7% Weight gain | ||||||||||
DRD2 (rs1799732, rs1799978, rs1079598, rs6275, rs7131056) | ||||||||||
Musil 2008101 | SNAP25 (rs1051312, rs3746544, rs8636) | 162 | 5 | 34.2±12.3 | 57 | 100 | SCZ, SZA | NR | Various APs | BMI change |
Opgen-Rhein 2010102 | HTR2C (rs3813929, rs6318) | 128 | 6 | 38.6±12.0 | 63 | 100 | SCZ, SZA | 17 | Various AP | >7% Weight gain |
INSIG2 (rs17587100, rs10490624, rs17047764, 7566605) | ||||||||||
LEP (rs7799039) | ||||||||||
Park 2006103 | ADRA2A (rs1800544) | 62 | Over 1 y | 46.5±11.1 | 71 | 0 (100% Asian) | SCZ | NR | OLZ | Weight change |
% Weight change | ||||||||||
>10% Weight gain | ||||||||||
Park 2008104 | HTR2C (rs3813929) | 79 | Over 1 y | 46.1±12.1 | 67 | 0 (100% Asian) | SCZ | NR | OLZ | Weight change |
BMI change | ||||||||||
>7% Weight gain | ||||||||||
Park 2009105 | GNB3 (rs5443) | 79 | Over 1 y | 46.6±11.6 | 67 | 0 (100% Asian) | SCZ | NR | OLZ | Weight change |
% Weight change | ||||||||||
BMI change | ||||||||||
>10% Weight gain | ||||||||||
Park 2011106 | CNR1 (rs1049353, rs806368) | 78 | Over 1 y | 46.4±11.6 | 67 | 0 (100% Asian) | SCZ | NR | OLZ | >7% Weight gain |
Perez-Iglesias 2010107 | LEP (rs7799039) | 194 | 1 y | 28.4±8.3 | 58 | 94 | SCZ spectrum disorders | 100 | Various APs | Weight change |
BMI change | ||||||||||
LEPR (rs1137101) | ||||||||||
FTO (rs9939609) | ||||||||||
Popp 2009108 | HTR2C (rs6318) | 102 | 4 | 37.5±13.7 | 45 | 100 | SCZ, SZA | NR | Various APs | BMI change |
Reynolds 200268 | HTR2C (rs3813929) | 123 | 6 and 10 | 26.6±7.7 | 52 | 0 (100% Asian) | SCZ | 100 | Various APs | BMI change |
>7% Weight gain | ||||||||||
Reynolds 2003109 | HTR2C (rs3813929) | 32 | 6 | NR | 66 | 0 (100% Asian) | SCZ | 100 | CLZ | BMI change |
Reynolds 2012110 | FTO (rs9939609) | 93 | 1 y | 25.5±6.7 | 74 | 100 | Psychosis | 100 | Various APs | Weight change |
BMI change | ||||||||||
Ryu 2006111 | LEP (rs7799039) | 71 | 4 | 30.5±7.6 | 45 | 0 (100% Asian) | SCZ | NR | Various APs | BMI change |
Ryu 2007112 | HTR2C (rs3813929) | 84 | 4 | 30.1±7.5 | 46 | 0 (100% Asian) | SCZ | 69 | Various APs | BMI change |
>7% BMI change | ||||||||||
Shao 2008113 | HTR2C (rs3813929, rs518147) | 170 | 1 y | 23.1±5.1 | 35 | 0 (100% Asian) | SCZ | 100 | NR | >7% Weight gain |
Shing 2014114 | FTO (rs9939609) | 218 | 6–14 wk | NR | 66 | 69 | SCZ or SZA | 0 | Various APs | % Weight change |
Sicard 2010115 | HTR2C (rs518147, rs3813929, rs6318) | 205 | Average 10 wk | 35.9±10.1 | 69 | 68 | SCZ or SZA | 0 | Various APs | % Weight change |
>7% Weight gain | ||||||||||
Sickert 2009116 | ADRA2A (rs1800544) | 129 | 10 | 36.5±9.0 | 74 | 50 | SCZ or SZA | 0 | Various Aps | Weight change |
Song 2014117 | FTO (rs9939609) | 237 | 6 mo | 27.5±7.6 | 54 | 0 (100% Asian) | SCZ | 100 | RIS | Weight change |
BMI change | ||||||||||
Souza 2008118 | GNB3 (rs5443) | 208 | 6wk, 14 wk | 35.9±10.3 | 68 | 68 | SCZ or SZA | 0 | Various APs | Weight change |
Srisawat 2014119 | MTHFR (rs1801131, rs1801133) | 182 | 8 wk | 26.2±7.4 | 46 | 0 (100% Asian) | SCZ | 100 | Various Aps | BMI change |
72 | 3 mo | 25.4±6.8 | 74 | 100 | SCZ | 100 | Various APs | BMI change | ||
Steaker 2012120 | PPARG (rs1801282) | 138 | 4 wk | Range17–88 | 46 | NR | Various diagnoses | NR | OLZ | Weight change |
BMI change | ||||||||||
Templeman 2005121 | HTR2C (rs3813929) | 73 | 6wk, 3 mo | 25.2±0.8 | 75 | 100 | Psychosis | 100 | Various APs | BMI change |
LEP (rs7799039) | >7% BMI change | |||||||||
Theisen 2004122 | HTR2C (rs3813929) | 97 | 12 | 22.1±7.7 | 59 | 100 | SCZ spectrum disorders | 0 | CLZ | BMI change |
>7% BMI change | ||||||||||
Thompson 2010123 | HTR2C (rs3813929) | 216 | 4 | NR | NR | NR | SCZ | NR | Iloperidone | Weight change |
Tiwari 2010124 | INSIG2 (rs17587100, rs7566605, rs10490624, rs17047764) | 154 | Average 10 wk | 35.8±9.8 | 71 | 58 | SCZ, SZA | 0 | CLZ, OLZ, RIS, HAL | % Weight change |
Tiwari 2010125 | CNR1 (rs1049353, rs806368) | 183 | Average 10 wk | 36.1±10.2 | 68 | 64 | SCZ, SZA | 0 | Various APs | % Weight change |
Tsai 2002126 | HTR2C (rs3813929) | 80 | 4 mo | 36.7±8.4 | 65 | 0 (100% Asian) | SCZ | 0 | CLZ | BMI change |
SZA | >7% BMI change | |||||||||
Tsai 2003127 | TNF (rs1800629) | 99 | 4 mo | 36.0±8.0 | 66 | 0 (100% Asian) | SCZ | 0 | CLZ | Weight change |
Tsai 2004128 | GNB3 (rs5443) | 87 | 4 mo | 37.0±8.2 | 64 | 0 (100% Asian) | SCZ or SZA | 0 | CLZ | Weight change |
ADRB3 (rs4994) | % Weight change | |||||||||
Tsai 2011129 | BDNF (rs6265) | 481 | >2 y | 43.9±8.9 | 60 | 0 (100% Asian) | SCZ | 0 | CLZ, OLZ, RIS | % Weight change |
Ujike 2008130 | HTR2C (rs3813929, rs6318) | 164 | 4 mo | 51.8±10.9 | 62 | 0 (100% Asian) | SCZ | 0 | OLZ | % BMI change |
HTR2A (rs6213) | ||||||||||
ADRB3 (rs4994) | ||||||||||
GNB3 (rs5443) | ||||||||||
Van Winkel 2010131 | MTHFR (rs1801131, rs1801133) | 104 | 3 mo | 31.3±11.7 | 68 | NR | SCZ or SZA | NR | Various APs | Weight change |
Wang 2005132 | ADRA2A (rs1800544) | 93 | 1 y | 38.4±8.1 | 53 | 0 (100% Asian) | SCZ | 0 | CLZ | Weight change |
>7% Weight gain | ||||||||||
Wang 2005133 | GNB3 (rs5443) | 134 | 1 y | 38.5±8.0 | 60 | 0 (100% Asian) | SCZ | 0 | CLZ | Weight change |
% Weight change | ||||||||||
Wang 2010134 | TNF (rs1800629) | 55 | 8 y | 37.2±7.7 | 49 | 0 (100 Asian) | SCZ | 0 | CLZ | Weight change |
BMI change | ||||||||||
Zai 2012135 | BDNF (rs6265) | 257 | 6 wk | 31.8±7.9 | 76 | 100 | SCZ | 0 | Various APs | Weight Δ |
SZA | >7% Weight gain | |||||||||
Zhang 2003136 | ANKK1 (rs1800497) | 117 | 10 wk | 26.0±8.0 | 50 | 0 (100% Asian) | SCZ | 100% | Various APs | BMI change |
Weight change | ||||||||||
>7% Weight gain | ||||||||||
Zhang 2003137 | LEP (rs7799039) | 128 | 10 wk | 26.0±7.0 | 48 | 0 (100% Asian) | SCZ | 100% | RIS, CPZ | BMI change |
Weight change | ||||||||||
>7% Weight gain | ||||||||||
Zhang 2007138 | LEP (rs7799039) | 102 | Average 7 y | 47.2±6.3 | 66 | 0 (100% Asian) | SCZ | 0 | CLZ | BMI change |
Zhang 2008139 | BDNF (rs6265) | 196 | At least 2 y | NR | 66 | 0 (100% Asian) | SCZ | 100% | Various APs | BMI change |
Notes: AMI, amisulpride; AP, antipsychotic; APZ, aripiprazole; BMI, body mass index; CLZ, clozapine; CPZ, chlorpromazine; FE, first episode; FLU, fluphenazine; HAL, haloperidol; OLZ, olanzapine; QTP, quetiapine; RIS, risperidone; SCZ, schizophrenia; SNP, single nucleotide polymorphism; SULP, sulpiride; SZA, schizoaffective disorder; SZP, schizophreniform disorder; ZIP, ziprasidone; LEP, Leptin; LEPR, Leptin receptor; ADRA2A: Adrenoceptor alpha 2A; ADRB3: Adrenoceptor beta 3; ANKK1: Ankyrin repeat and kinase domain containing 1; BDNF, Brain-derived neurotrophic factor; CNR1, Cannabinoid receptor 1; DRD2, Dopamine receptor D2; FTO, Fat mass and obesity associated; GNB3, Guanine nucleotide binding protein (G protein), beta polypeptide 3; HTR2A, 5-hydroxytryptamine (serotonin) receptor 2A, G protein-coupled; HTR2C, 5-hydroxytryptamine (serotonin) receptor 2C, G protein-coupled; HTR6, 5-hydroxytryptamine (serotonin) receptor 6, G protein-coupled; INSIG2, Insulin-induced gene 2; MC4R, Melanocortin 4 receptor; MDR1 (ABCB1), ATP-binding cassette, sub-family B (MDR/TAP), member 1; MTHFR, Methylenetetrahydrofolate reductase; PPARG, Peroxisome proliferator-activated receptor gamma; SNAP25, Synaptosomal-associated protein, 25kDa; TNF, Tumor necrosis factor.
Included Genes and SNPs
Altogether, 38 SNPs from 20 genes/genomic regions on 15 chromosomes were reported in ≥2 independent cohorts, entering into subsequent meta-analyses. Table 2 lists the included SNPs, genes or genomic regions, and other relevant information, as well as proxy SNPs used in the 3 additional cohorts as necessary. The major alleles, minor alleles, and minor allele frequency were based on the 1000 Genome CEU population. Three included SNPs (rs1799732, rs1801028, and rs1051312) did not have proxy SNPs in the 3 additional cohorts, therefore, only published studies were meta-analyzed for these 3 SNPs. The 5-Hydroxytryptamine (Serotonin) Receptor 2C (HTR2C) polymorphism, rs3813929 (−759C/T) was the most studied SNP (published studies = 22). Seven SNPs from Dopamine Receptor D2 (DRD2) were included in the meta-analysis, the most in a single gene.
Table 2.
rs# | # Studies | Gene | SNP | Chr | Position | Region | Major Allele | Minor Allele | MAF | Proxy SNP | R 2 | D′ | Major Allele | Minor Allele | MAF |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
rs1800544 | 3 | ADRA2A | −1291C/G | 10 | 111076745 | C | G | 0.48 | rs521674 | 0.92 | 1 | A | T | 0.26 | |
rs4994 | 3 | ADRB3 | Trp64Arg | 8 | 37823798 | (Missense) | T (Trp) | C (Arg) | 0.10 | ||||||
rs1800497 | 5 | ANKK1 | Taq1A (Glu713Lys) | 11 | 112776038 | Exon 8 | G | A | 0.30 | rs7118900 | 0.90 | 1 | G | A | 0.18 |
rs6265 | 4 | BDNF | Val66Met | 11 | 27658369 | (Missense) | G | A | 0.23 | ||||||
rs1049353 | 3 | CNR1 | 1359G/A | 6 | 88143916 | G | A | 0.14 | |||||||
rs806368 | 2 | CNR1 | 6 | 88140381 | 3′ UTR | T | C | 0.28 | |||||||
rs1799732 | 3 | DRD2 | −141C Ins/Del | 11 | 113475529: 113475530 | Intron | C | — | 0.24 | NA | |||||
rs1079598 | 2 | DRD2 | 11 | 112801484 | Intron 1 | T | C | 0.21 | rs1079594 | 1 | 1 | A | C | 0.13 | |
rs1799978 | 2 | DRD2 | 11 | 113475629 | Intron | A | G | 0.11 | |||||||
rs1801028 | 2 | DRD2 | Ser311Cys | 11 | 112788694 | Exon 7 | C | G | 0.02 | NA | |||||
rs2242591 | 2 | DRD2 | 11 | 112785131 | Downstream | G | A | 0.20 | rs6278 | 1 | 1 | C | A | 0.13 | |
rs6275 | 2 | DRD2 | C939T | 11 | 113412755 | Exon (synon) | C | T | 0.47 | ||||||
rs7131056 | 2 | DRD2 | 11 | 113459052 | C | A | 0.48 | ||||||||
rs9939609 | 3 | FTO | 16 | 53820527 | Intron | T | A | 0.36 | rs3751812 | 1 | 1 | G | T | 0.45 | |
rs5443 | 6 | GNB3 | C825T | 12 | 6954875 | Exon (synon) | C | T | 0.48 | ||||||
rs6313 | 4 | HTR2A | 102T/C | 13 | 46895805 | Intron | C | T | 0.43 | ||||||
rs6314 | 2 | HTR2A | His452Tyr | 13 | 47409034 | Exon | C | T | 0.07 | ||||||
rs3813929 | 22 | HTR2C | −759C/T | X | 113818520 | Upstream, promoter | C | T | 0.12 | ||||||
rs6318 | 7 | HTR2C | Cys23Ser | X | 113871991 | Exon 5 | G | C | 0.17 | ||||||
rs518147 | 4 | HTR2C | −697G/C | X | 113818582 | 5′UTR | G | C | 0.29 | ||||||
rs1805054 | 2 | HTR6 | 267T/C | 1 | 19666020 | C | T | 0.17 | rs1977101 | 1 | 1 | A | G | 0.13 | |
rs10490624 | 3 | INSIG2 | 2 | 118104916 | Intron | A | G | 0.09 | |||||||
rs17047764 | 3 | INSIG2 | 2 | 118868582 | G | C | 0.19 | rs3849327 | 0.94 | 1 | T | C | 0.16 | ||
rs17587100 | 3 | INSIG2 | 2 | 118838410 | A | C | 0.06 | rs17526937 | 1 | 1 | A | G | 0.04 | ||
rs7566605 | 3 | INSIG2 | 2 | 118078449 | G | C | 0.30 | ||||||||
rs7799039 | 12 | LEP | −2548A/G | 7 | 128238730 | G | A | 0.43 | rs10487506 | 1 | 1 | G | A | 0.47 | |
rs1137101 | 4 | LEPR | Q223R | 1 | 65592830 | (Missense) | A | G | 0.41 | ||||||
rs489693 | 5 | MC4R | 18 | 60215554 | C | A | 0.33 | ||||||||
rs17782313 | 2 | MC4R | 18 | 60183864 | T | C | 0.22 | rs476828 | A | G | 0.27 | ||||
rs1045642 | 3 | MDR1 | 3435C/T | 7 | 87509329 | T, C | A | 0.40 | |||||||
rs2032582 | 2 | MDR1 | 2677G/T(A) | 7 | 87531302 | (Missense) | G | A | 0.34 | ||||||
rs1801131 | 3 | MTHFR | 1298A/C | 1 | 11794419 | (Missense) | A | C | 0.23 | ||||||
rs1801133 | 3 | MTHFR | 677C/T | 1 | 11796321 | (Missense) | C | T | 0.33 | ||||||
rs1801282 | 2 | PPARG | Pro12Ala | 3 | 12351626 | Intron (Missense) | C | G | 0.07 | ||||||
rs1051312 | 2 | SNAP25 | DdelI(T/C) | 20 | 10306440 | 3′ UTR | T | C | 0.15 | NA | |||||
rs3746544 | 2 | SNAP25 | MnlI(T/G) | 20 | 10306436 | 3′ UTR | T | G | 0.29 | ||||||
rs8636 | 2 | SNAP25 | TaiI(T/C) | 20 | 10307094 | 3′ UTR | C | T | 0.27 | ||||||
rs1800629 | 4 | TNF | G-308A | 6 | 31575254 | G | A | 0.10 |
Note: MAF, minor allele frequency.
Overview of the Meta-analytic Results
Altogether, 11 SNPs from 8 genes were significantly associated with weight or BMI change, and 4 SNPs from 2 genes were significantly associated with study-defined significant weight gain (table 3). Combined together, 13 SNPs from 9 genes (Adrenoceptor Alpha-2A [ADRA2A], Adrenoceptor Beta 3 [ADRB3], Brain-Derived Neurotrophic Factor [BDNF], DRD2, Guanine Nucleotide Binding Protein [GNB3], HTR2C, Insulin-induced gene 2 [INSIG2], Melanocortin-4 Receptor [MC4R], and Synaptosomal-associated protein, 25kDa [SNAP25]) were significantly associated with antipsychotic-related weight gain (P-values: ≤.05–.001). SNPs in ADRA2A, DRD2, HTR2C, and MC4R had the largest ES (Hedges’ gs = 0.30–0.80, ORs = 1.47–1.96). Forest plots for the significant results are included in supplementary figures 1–5. Heterogeneity across studies was not large for most significant SNPs, except for rs489693 (AA vs C carriers, I 2 = 80%), rs1799732 (Ins/Ins vs Del carriers, I 2 = 62.6%), rs3813929 (CC vs T carriers, I 2 = 65.9%), and rs518147 (GG vs C carrier, I 2 = 57.6%). Publication biases existed for several SNPs. However, the direction of publication bias actually under-estimated the ES for rs6275, rs7131056, and rs17047764 (ie, the corrected ESs became larger). In contrast, adjusting publication bias eliminated the significance for rs3813929 (secondary outcome, adjusted OR = 1.51, 95% CI = 0.91–2.49). Even after adjusting for potential publication bias, the ES was still significant for rs489693 (AA vs C, adjusted Hedges’ g = 0.66, 95% CI = 0.09–1.23). Using the modified Venice guideline, 2 HTR2C SNPs, rs3813929 and rs518147, achieved a score of 8, 3 SNPs from ADRA2A, DRD2 (rs1799732), and GNB3 had a score of 7, and 4 SNPs from DRD2 (rs6275, rs7131056), INSIG2 (rs17047764), and MC4R (rs489693) obtained a score of 6.
Table 3.
Gene | rs# | SNP | Genotype Comparison | BMI or Weight Change | BMI or Weight Change >7% or 10% | Category & Score | ||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Hedges’ g (95% CI) | P | # Study (Total n) | I 2 | “T&F” | OR (95% CI) | P | # Study (Total n) | I 2 | “T&F” | |||||
ADRA2A | rs1800544 | −1291C/G | CC vs G | −0.22 (−0.39, −0.05) | .01 | 6 (645) | 0% | 0 | 0.50 (0.24, 1.05) | .07 | 5 (516) | 61% | +2 | B,B,A |
GG vs C | 0.30 (0.09, 0.51) | .01 | 6 (645) | 24% | 0 | 1.74 (0.79, 3.85) | .17 | 5 (516) | 58% | 0 | 7 | |||
Additive (G) | 0.20 (0.06, 0.33) | .004 | 6 (645) | 30% | 0 | |||||||||
ADRB3 | rs4994 | Trp64Arg | Trp/Trp vs Arg | −0.20 (−0.48, 0.09) | .18 | 6 (680) | 54% | +1 | 1.10 (0.44, 2.77) | .83 | 3 (358) | 47% | 0 | B,B,C |
Arg/Arg vs Trp | 0.84 (0.20, 1.47) | .01 | 2 (235) | 0% | NA | 5 | ||||||||
ANKK1 | rs1800497 | Taq1A (Glu713Lys) | CC vs T | 0.05 (−0.09, 0.19) | .51 | 7 (842) | 0% | 0 | 0.97 (0.75, 1.25) | .80 | 7 (1181) | 0% | +3 | B,C,C |
TT vs C | −0.09 (−0.44, 0.26) | .61 | 7 (842) | 35% | +2 | 0.93 (0.64, 1.35) | .70 | 7 (1179) | 0% | −3 | 4 | |||
Additive (C) | 0.05 (−0.09, 0.19) | .46 | 7 (842) | 9% | −2 | |||||||||
BDNF | rs6265 | Val66Met (G/A) | AA vs G | 0.06 (−0.41, 0.53) | .81 | 7 (1393) | 82% | +2 | 0.79 (0.37, 1.68) | .53 | 5 (609) | 0% | −1 | A,C,C |
GG vs A | 0.13 (−0.07, 0.32) | .21 | 7 (1393) | 61% | 0 | 1.49 (1.02, 2.18) | .04 | 5 (609) | 0% | 0 | 5 | |||
CNR1 | rs1049353 | 1359 G/A | AA vs G | −0.08 (−0.51, 0.36) | .74 | 4 (534) | 0% | 0 | 1.31 (0.51, 3.39) | .57 | 5 (522) | 0% | −3 | B,C,C |
GG vs A | 0.04 (−0.14, 0.23) | .64 | 4 (534) | 0% | +1 | 1.04 (0.69, 1.58) | .84 | 5 (522) | 0% | +3 | 4 | |||
rs806368 | AA vs G | 0.84 (0.53, 1.35) | .47 | 2 (251) | 0% | NA | C,C,B | |||||||
GG vs A | 0.92 (0.50, 1.71) | .80 | 2 (251) | 0% | NA | 4 | ||||||||
DRD2 | rs1799732 | −141C Ins/Del | Del/Del vs Ins | 0.36 (−0.07, 0.79) | .10 | 3 (305) | 0% | 0 | 1.94 (0.65, 5.76) | .23 | 2 (247) | 0% | NA | C,A,A |
Ins/Ins vs Del | −0.44 (−0.86, −0.02) | .04 | 3 (305) | 63% | 0 | 0.63 (0.27, 1.46) | .28 | 2 (247) | 46% | NA | 7 | |||
Additive (Del) | 0.31 (0.07, 0.54) | .01 | 3 (305) | 0% | 0 | |||||||||
rs1079598 | CC vs T | −0.07 (−0.75, 0.61) | .85 | 5 (595) | 55% | 0 | 2.28 (0.74, 7.07) | .15 | 4 (485) | 0% | −1 | B,C,C | ||
TT vs C | 0.06 (−0.13, 0.24) | .53 | 5 (595) | 0% | +1 | 0.72 (0.45, 1.13) | .15 | 4 (485) | 0% | +2 | 4 | |||
rs1799978 | AA vs G | −0.19 (−0.43, 0.06) | .13 | 4 (486) | 0% | +1 | 0.88 (0.62, 1.26) | .49 | 5 (940) | 7% | +3 | C,C,C | ||
3 | ||||||||||||||
rs1801028 | Ser311Cys | CC vs T | 0.17 (−0.33, 0.66) | .50 | 2 (241) | 0% | NA | C,C,C | ||||||
3 | ||||||||||||||
rs2242591 | AA vs G | −0.17 (−0.91, 0.58) | .66 | 4 (480) | 51% | +1 | 0.84 (0.54, 1.31) | .44 | 4 (814) | 0% | −1 | C,C,C | ||
GG vs A | 0.10 (−0.11, 0.31) | .34 | 4 (480) | 0% | 0 | 1.07 (0.77, 1.48) | .71 | 4 (814) | 0% | +1 | 3 | |||
Additive (G) | 0.09 (−0.13, 0.32) | .42 | 4 (480) | 17% | 0 | |||||||||
rs6275 | C939T | CC vs T | −0.35 (−0.54, −0.16) | <.001 | 4 (482) | 0% | −1 | 0.79 (0.59, 1.07) | .13 | 5 (942) | 0% | −1 | C,A,B | |
TT vs C | 0.29 (0.04, 0.53) | .02 | 4 (482) | 5% | −1 | 1.39 (0.86, 2.22) | .18 | 5 (942) | 32% | 0 | 6 | |||
Additive (T) | 0.25 (0.09, 0.41) | .002 | 4 (482) | 0% | −1 | |||||||||
rs7131056 | AA vs C | 0.14 (−0.14, 0.43) | .32 | 4 (480) | 47% | −1 | 1.15 (0.62, 2.14) | .66 | 5 (939) | 68% | 0 | C,A,B | ||
CC vs A | −0.31 (−0.51, −0.12) | .002 | 4 (480) | 0% | −1 | 0.78 (0.52, 1.15) | .20 | 5 (939) | 31% | 0 | 6 | |||
Additive (A) | 0.19 (0.03, 0.34) | .02 | 4 (480) | 0% | 0 | |||||||||
FTO | rs9939609 | AA vs T | 0.03 (−0.25, 0.31) | .85 | 6 (790) | 49% | −1 | 0.72 (0.18, 2.92) | .65 | 3 (364) | 66% | −1 | A,C,C | |
TT vs A | −0.01 (−0.15, 0.12) | .85 | 7 (1027) | 0% | 0 | 1.52 (0.96, 2.42) | .08 | 3 (364) | 0% | +2 | 5 | |||
GNB3 | rs5443 | C825T | CC vs T | −0.18 (−0.38, 0.02) | .08 | 10 (1004) | 44% | 0 | 0.74 (0.49, 1.13) | .16 | 4 (443) | 0% | −2 | A,B,B |
TT vs C | 0.28 (0.08, 0.48) | .006 | 8 (865) | 32% | 0 | 1.20 (0.71, 2.03) | .49 | 4 (443) | 2% | +2 | 7 | |||
Additive (T) | 0.18 (0.04, 0.32) | .01 | 8 (865) | 30% | 0 | |||||||||
HTR2A | rs6313 | 102T/C | CC vs T | −0.12 (−0.28, 0.04) | .16 | 7 (814) | 0% | +1 | 0.79 (0.48, 1.29) | .34 | 4 (481) | 10% | +1 | B,C,C |
TT vs C | 0.11 (−0.05, 0.27) | .19 | 7 (814) | 0% | 0 | 1.17 (0.71, 1.92) | .54 | 4 (481) | 6% | 0 | 4 | |||
Additive (T) | 0.08 (−0.04, 0.20) | .17 | 7 (814) | 0% | −2 | |||||||||
rs6314 | His452Tyr | His/His vs Tyr | −0.05 (−0.42, 0.33) | .81 | 5 (563) | 69% | 0 | 0.91 (0.48, 1.71) | .76 | 4 (485) | 41% | 0 | B,C,B | |
Tyr/Tyr vs His | 0.48 (−0.69, 1.64) | .42 | 3 (324) | 68% | 0 | 1.62 (0.23, 11.38) | .63 | 2 (246) | 32% | NA | 5 | |||
HTR2C | rs3813929 | −759C/T | CC vs T | 0.23 (0.04, 0.42) | .02 | 20 (2082) | 66% | 0 | 1.96 (1.19, 3.22) | .009 | 18 (1738) | 67% | −4 | A,B,A |
8 | ||||||||||||||
rs6318 | Cys23Ser | GG vs C | 0.10 (−0.10, 0.29) | .34 | 9 (1111) | 35% | +2 | 1.47 (1.03, 2.11) | .04 | 5 (687) | 0% | −1 | A,C,C | |
5 | ||||||||||||||
rs518147 | −697G/C | GG vs C | 0.18 (0.02, 0.34) | .03 | 5 (671) | 0% | 0 | 1.86 (1.03, 3.35) | .04 | 5 (659) | 58% | 0 | B,A,A | |
8 | ||||||||||||||
HTR6 | rs1805054 | 267T/C | CC vs T | −0.03 (−0.33, 0.28) | .87 | 5 (576) | 48% | +2 | 0.92 (0.47, 1.80) | .82 | 3 (361) | 30% | 0 | B,C,C |
TT vs C | −0.11 (−0.41, 0.20) | .50 | 3 (338) | 0% | +2 | 0.33 (0.04, 3.03) | .33 | 1 (123) | NA | NA | 4 | |||
INSIG2 | rs10490624 | TT vs C | 0.07 (−0.19, 0.33) | .61 | 5 (666) | 44% | +1 | 0.96 (0.54, 1.71) | .89 | 4 (485) | 24% | +2 | B,C,C | |
4 | ||||||||||||||
rs17047764 | CC vs G | 0.31 (0.00, 0.61) | .05 | 5 (665) | 23% | +2 | 1.55 (0.55, 4.39) | .41 | 3 (360) | 0% | −2 | B,A,C | ||
GG vs C | −0.17 (−0.36, 0.02) | .08 | 5 (665) | 10% | 0 | 1.20 (0.81, 1.78) | .37 | 4 (485) | 0% | +1 | 6 | |||
Additive (C) | 0.19 (0.03, 0.35) | .02 | 5 (665) | 10% | 0 | |||||||||
rs17587100 | AA vs C | 0.05 (−0.22, 0.32) | .73 | 5 (665) | 23% | 0 | 0.65 (0.26, 1.60) | .34 | 4 (489) | 43% | +2 | B,C,B | ||
5 | ||||||||||||||
rs7566605 | CC vs G | 0.06 (−0.19, 0.31) | .64 | 5 (655) | 0% | +2 | 0.86 (0.36, 2.05) | .74 | 4 (481) | 34% | −1 | B,C,C | ||
GG vs C | 0.10 (−0.06, 0.27) | .22 | 5 (655) | 11% | 0 | 0.87 (0.59, 1.27) | .46 | 4 (481) | 0% | +1 | 4 | |||
LEP | rs7799039 | −2548A/G | AA vs G | −0.08 (−0.30, 0.13) | .45 | 11 (1138) | 51% | 0 | 1.24 (0.50, 3.05) | .64 | 4 (414) | 74% | +2 | A,C,C |
GG vs A | 0.16 (−0.01, 0.32) | .06 | 10 (967) | 25% | 0 | 0.73 (0.33, 1.60) | .43 | 3 (340) | 5% | +2 | 5 | |||
Additive (G) | 0.07 (−0.06, 0.19) | .27 | 7 (763) | 0% | 0 | |||||||||
LEPR | rs1137101 | Q223R | AA vs G | 0.03 (−0.14, 0.19) | .74 | 7 (682) | 0% | +3 | B,C,C | |||||
GG vs A | 0.09 (−0.11, 0.30) | .38 | 6 (653) | 0% | +1 | 4 | ||||||||
MC4R | rs489693 | AA vs C | 0.80 (0.20, 1.41) | .009 | 6 (583) | 80% | −1 | B,B,B | ||||||
CC vs A | −.28 (−0.53, −0.03) | .03 | 6 (583) | 52% | +1 | 6 | ||||||||
Additive (A) | 0.30 (0.04, 0.57) | .03 | 6 (583) | 67% | −1 | |||||||||
rs17782313 | CC vs T | 0.14 (−0.29, 0.56) | .53 | 5 (735) | 49% | +1 | B,B,C | |||||||
TT vs C | −0.25 (−0.52, 0.02) | .07 | 5 (735) | 68% | 0 | 5 | ||||||||
Additive (C) | 0.19 (−0.05, 0.42) | .12 | 5 (735) | 64% | 0 | |||||||||
MDR1 | rs1045642 | 3435C/T | CC vs T | −0.02 (−0.22, 0.17) | .81 | 5 (499) | 0% | 0 | 0.87 (0.55, 1.36) | .53 | 4 (467) | 0% | +1 | C,C,C |
TT vs C | −0.04 (−0.26, 0.18) | .71 | 5 (499) | 0% | +2 | 0.83 (0.36, 1.89) | .65 | 4 (467) | 61% | 0 | 3 | |||
rs2032582 | 2677G/T(A) | GG vs T | 0.06 (−0.13, 0.25) | .54 | 4 (462) | 0% | +1 | 1.01 (0.53, 1.91) | .98 | 4 (469) | 58% | 0 | C,C,C | |
TT vs G | −0.06 (−0.32, 0.20) | .68 | 4 (462) | 0% | +1 | 0.79 (0.44, 1.39) | .41 | 4 (469) | 2% | 0 | 3 | |||
MTHFR | rs1801131 | 1298A/C | AA vs C | 0.07 (−0.08, 0.22) | .36 | 6 (707) | 0% | 0 | 1.36 (0.86, 2.15) | .19 | 3 (359) | 0% | +2 | B,C,C |
CC vs A | −0.16 (−0.57, 0.24) | .43 | 6 (707) | 43% | −2 | 0.57 (0.09, 3.77) | .56 | 3 (359) | 77% | 0 | 4 | |||
rs1801133 | 677C/T | CC vs T | 0.17 (−0.08, 0.41) | .19 | 6 (705) | 58% | 0 | 0.99 (0.49, 1.99) | .98 | 3 (357) | 49% | +1 | B,C,B | |
TT vs C | −0.01 (−0.24, 0.22) | .92 | 6 (705) | 0% | 0 | 1.16 (0.50, 2.70) | .74 | 3 (357) | 0% | −2 | 5 | |||
PPARG | rs1801282 | Pro12Ala | Pro/Pro vs Ala | 0.09 (−0.15, 0.33) | .47 | 5 (622) | 23% | 0 | 0.79 (0.49, 1.30) | .35 | 4 (529) | 0% | +1 | B,C,B |
5 | ||||||||||||||
SNAP25 | rs1051312 | DdelI(T/C) | TT vs C | −0.58 (−0.87, −0.29) | <.001 | 2 (218) | 0% | NA | C,A,C | |||||
5 | ||||||||||||||
rs3746544 | MnlI(T/G) | GG vs T | 0.03 (−0.27, 0.33) | .86 | 4 (416) | 0% | +1 | 1.40 (0.66, 2.99) | .38 | 3 (361) | 9% | 0 | C,C,C | |
TT vs G | 0.09 (−0.24, 0.43) | .59 | 4 (416) | 63% | +1 | 0.73 (0.46, 1.15) | .17 | 3 (361) | 0% | 0 | 3 | |||
rs8636 | TaiI(T/C) | CC vs T | 0.19 (−0.10, 0.47) | .20 | 4 (418) | 48% | 0 | 0.88 (0.56, 1.38) | .57 | 3 (364) | 0% | 0 | C,C,C | |
TT vs C | −0.09 (−0.41, 0.23) | .58 | 4 (418) | 0% | +2 | 1.40 (0.67, 2.93) | .38 | 3 (364) | 0% | 0 | 3 | |||
TNF-alpha | rs1800629 | G-308A | AA vs G | 0.21 (−0.49, 0.90) | .56 | 4 (796) | 18% | 0 | 0.23 (0.01, 4.53) | .34 | 1 (500) | NA | NA | B,C,B |
GG vs A | 0.15 (−0.08, 0.37) | .20 | 7 (1090) | 44% | 0 | 1.10 (0.65, 1.87) | .73 | 4 (862) | 43% | 0 | 5 |
Note: “T&F”: “Trim and Fill” method to assess potential publication bias. 0 = no missing study (no evidence of publication bias); negative value = # missing studies favoring the major or minor allele carriers in the comparison; positive value = # missing studies favoring the homozygotes in the comparison. Bolded results: P ≤ .05. “Additive”: additive genetic model with the risk allele in the parenthesis. “Category & Score”: categories and scores of the strength of evidence based on the modified Venice guideline.24
Specific Meta-analytic Results
5-Hydroxytryptamine (Serotonin) Receptor 2C Gene.
HTR2C was the most studied gene, and 3 SNPs were included in the meta-analysis. The most studied SNP, rs3813929, was reported in 22 studies with additional information from the SATIETY and EUFEST sample, totaling 24 studies. The ZHH FE sample did not have any T-allele carrier genotype, and was not included in the analysis. Among these studies, 20 studies reported continuous weight or BMI change data and 18 reported categorical weight gain data. Study duration ranged from 4 weeks to 1 year. The C allele was associated with significantly more weight gain than the T allele. Because the gene is located in the X chromosome, C hemizygosity in males is equivalent to the CC genotype in females. Because the T allele is relatively rare (frequency = 12%), it was not possible to meta-analyze TT vs C carriers. The pooled ES from 20 studies was small (Hedges’ g = 0.23, P = .017, with significant heterogeneity; table 3; supplementary figure 1). Although meta-regression analysis showed that studies with larger sample sizes had smaller ES (P = .003), this finding might be confounded by the fact that larger studies tended to be in chronic patients. Therefore, a series of subgroup analyses was conducted to further dissect the heterogeneity.
When studies were divided into subgroups based on treatment duration, the short-term studies (4–8wk) produced larger ESs, (Hedges’ g = 0.44, P = .004, studies=12, n = 1209). Although there was no evidence of publication bias, the heterogeneity across studies was still high (I 2 = 76.1%). When further classifying samples into chronic vs first-episode patients, the first-episode samples with short-term follow-up produced the largest ES (Hedge’s g = 0.67, P = .002, studies = 6, n = 589) compared to chronic samples (Hedge’s g = 0.21, P = .27, studies = 6, n = 620), with a significant between-group difference (Q = 9.74, df = 1, P = .002; supplementary figure 2). In contrast, the studies with longer-term duration (3–4 mo, and 6 mo to 1 y) did not produce significant pooled ESs, and there was no difference between first-episode and chronic samples, although the overall trend favored the CC genotype. When mixing studies with different treatment lengths, the pooled ES in first-episode studies (Hedges’ g = 0.47, P = .02, studies = 8, n = 810) was significantly higher than in chronic samples (Hedges’ g = 0.14, P = .18, studies=12, n = 1280), Q = 4.75, df = 1, P = .03). Results for the categorical outcome variable were similar, but to some extent, less significant. Moderator analyses of race, sex, and specific antipsychotic were not significant.
The above-mentioned results were similar for rs518147 (table 3), but overall ES’s were smaller. Due to the small number of included studies, no meta-regression or moderator analysis was conducted. The meta-analytic findings for rs6318 were inconsistent, in that the primary outcome was not significant, but the secondary outcome was significant with evidence of publication bias (table 3).
Dopamine Receptor D2 Gene.
Three of seven SNPs in DRD2 included in the analysis showed significant associations with weight gain (table 3). For rs1799732, an additive model of the risk allele (deletion of C) was identified, ie, each additional risk allele was associated with a 0.31 SD of extra weight gain, P = .01. For rs6275, the T-allele was the risk allele because TT homozygotes gained more weight than C carriers (Hedges’ g = 0.29, P = .02) and CC homozygotes gained less weight than T carriers (Hedges’ g = −0.35, P < .01). Both heterogeneity and publication bias were minimal. Similar findings were observed for rs7131056 (table 3). Due to the small number of studies, moderator analyses were not performed.
ADRA2A Gene and GNB3 Gene.
One SNP in ADRA2A, rs1800544, was significantly associated with weight gain across 6 studies, with the G allele increasing the risk. Both heterogeneity and publication bias were minimal (table 3; supplementary figure 4). Similarly, 1 SNP in GNB3, rs5443, was associated with weight gain across 10 studies. TT homozygotes of this SNP gained significantly more weight than the C carriers (Hedges’ g = 0.26, P = .01; table 3; supplementary figure 5). Heterogeneity was small, and there was no evidence of publication bias. When analyzing 5 short-term studies only (treatment durations ≤3 mo), the pooled ES became even larger (Hedges’ g = 0.39, P < .001, studies = 5, n = 486).
MC4R Gene and INSIG2 Gene.
Two SNPs near MC4R were included in the meta-analysis, and one of them (rs489693) was significantly associated with weight gain in 6 studies. AA homozygotes of this SNP gained more weight than the C allele carriers (Hedges’ g = 0.80, P = .009; table 3; supplementary figure 3). Heterogeneity across studies was high, and there was publication bias. However, even after adjusting for potentially missing studies, the association remained significant (Hedges’ g = 0.66, P < .05). The ZHH FE cohort was an extreme outlier. The pooled ES became more significant after dropping this sample (Hedges’ g = 1.05, P = 1.9×10−7). There was no significant moderator variable. Similarly, only 1 of 4 SNPs in INSIG2, rs17047764, was significantly associated with weight gain (P = .048, table 3). After adjusting for potential publication bias, the pooled ES remained significant.
Polygenic risk scores (PRS) were computed combining 6 top SNPs from HTR2C, DRD2, ADRA2A, GNB3, MC4R, and INSIG2. Number of risk alleles in each SNP (ie, 0, 1, or 2) was multiplied by its pooled ES from the additive model (table 3), and the sum of the 6 products was the PRS. This PRS explained 5.6% of the total variance in weight gain in each of the SATIETY and EUFEST cohorts, Ps < .01 (supplementary figure 6).
ADRB3, BDNF, and SNAP25.
One SNP from each of these 3 genes was significantly associated with weight gain, but either the sample size was small (studies = 2 for rs4994 in ADRB3 and rs1051312 in SNAP25), or the primary outcome was not significant (rs6265 in BDNF; table 3).
Discussion
To our best knowledge, this is the first comprehensive meta-analytic review of pharmacogenetics of antipsychotic-related weight gain that examined quantitatively multiple genetic variants. We investigated 38 SNPs in 20 genes or genetic regions distributed in 15 chromosomes in association with antipsychotic-related weight gain in 6770 patients from 46 non-overlapping samples published in 72 reports and including patient-level data from 3 cohorts providing unpublished data on 33 SNPs that were added to the meta-analysis. We found that 13 SNPs from 9 genes (ADRA2A, ADRB3, BDNF, DRD2, GNB3, HTR2C, INSIG2, MC4R, and SNAP25) were significantly associated with antipsychotic-related weight gain. Among these genes, HTR2C was most consistently associated with antipsychotic-related weight gain, and there was moderate evidence supporting the association of ADRA2A, DRD2, GNB3, MC4R, and INSIG2, based on the modified Venice guidelines. Relationships of other genes (ADRB3, BDNF and SNAP25) with antipsychotic-related weight gain were less consistent. Finally, polygenic scores using 6 SNPs seemed to explain a small proportion of weight gain.
With the widespread use of SGAs, weight gain and related metabolic syndrome have become a significant public health issue.25 Weight gain is especially prominent in young and antipsychotic-naïve patients.5,16,26 Extensive effort has been made to understand the pathophysiological mechanisms of drug-related weight gain, and genetic variations seem to play a significant role.10,13 The present study aimed at providing a comprehensive and quantitative review of the literature on pharmacogenetics of antipsychotic drug-related weight gain. Previously, meta-analyses of pharmacogenetic association of single genes with antipsychotic-related weight gain have been published, but methodological issues limited their conclusions. First, many previous meta-analyses included cross-sectional studies where obesity or 1-time assessments of weight were correlated with a genetic variant. Body weight is determined by multiple factors including genetic, behavioral and medication effects.9 Without longitudinal assessments of weight change during antipsychotic treatment, the alleged genotype-phenotype association may be confounded by other, unmeasured variables. For example, it has been shown in multiple GWAS that FTO is associated with obesity in the general populations,27 and most of these studies had 1-time measurements of obesity. However, in longitudinal studies of weight gain, FTO was not associated with antipsychotic-related weight gain in 7 studies (table 3). In the present meta-analysis, we included only longitudinal studies. Second, previous meta-analyses tended to pool studies of different treatment durations. ESs of a particular genetic variant on weight gain may be different at different time points. Moreover, antipsychotic-related weight gain is asymptotic and it is unclear when the weight gain begins to plateau, which also depends on the degree of prior antipsychotic-related weight gain. Mixing studies of different treatment durations may bias assessments of the true effect size. In the present review, we preferred time points closest to 2–3 months and we analyzed specific time points whenever enough studies were available. In addition, previous meta-analyses tended to mix studies with chronic and antipsychotic-naïve or first-episode samples with minimal prior antipsychotic exposure. Studying patients with minimal drug exposure in pharmacogenetics minimizes order effects and increases the signal-to-noise ratio.14 In the present review, we added unpublished data from 3 first-episode/drug-naïve cohorts, and separated chronic samples from first-episode/antipsychotic-naïve samples whenever possible.
Results for Pharmacodynamic Targets of Antipsychotic Drugs
HTR2C showed the most consistent association with antipsychotic-related weight gain, with multiple SNPs showing this association and in a large number of studies, including both primary (continuous variable) and secondary (categorical) outcomes. Not only is it one of the first studied genes in antipsychotic-related weight gain, it is also one of the most studied genes. The HTR2C gene encodes the 2C subtype of serotonin receptor (5-HT2C) and is located on the Chromosome Xq24. Experimental studies demonstrated the relevance of 5-HT2C receptors in regulating appetite and food intake,28 and 5-HT2C agonists, such as dexfenfluramine and lorcaserin, can decrease food intake, resulting in significant weight loss.29 5-HTR2C antagonists, including many antipsychotics, may increase food intake, despite satiety, causing weight gain in animal models.30,31 Mice deficient in 5-HT2C develop hyperphagia leading to obesity.32,33 The most studied SNP in HTR2C, rs3813929, is located in the promoter region of the gene and may play a role in regulating gene expression. Several studies found that the T-allele is associated with increased transcriptional activity of the gene, compared to the C allele.34,35 Although the C-allele is the major allele in the population, the T-allele seems to be protective against antipsychotic-related weight gain in the meta-analysis, perhaps by enhancing gene expression of HTR2C, which partially counterbalances the 5-HT2C antagonistic antipsychotic effect. This SNP is located in the beginning of a CpG island (83 CpG count, chromosome X:113818520-113819453, UCSC Genome Browser) in the promoter region of HTR2C, suggesting that DNA methylation pattern variation may play a role in the SNP effect. Another SNP, rs518147, is only 62bp away from rs3813929. These 2 SNPs may be in high linkage disequilibrium,36 representing probably the same signal. The third SNP in the HTR2C gene, rs6318, is located in an exon about 150kb from the first 2 SNPs. It is a missense SNP resulting in cysteine to serine substitution in position 23 of the protein sequence, which may disrupt a disulfide bridge affecting the receptor function.37
DRD2 had a robust association with antipsychotic-related weight gain. Three of seven SNPs included in the meta-analysis showed a significant relationship, although the number of studies and total sample sizes were not large. Being the main pharmacodynamic target of antipsychotics,38 variations in DRD2 function are plausible in explaining antipsychotic-related adverse events. At least 2 of the 3 SNPs in DRD2, located in Chromosome 11q23.2, rs1799732 (−141C Ins/Del) and rs6275, may be functional polymorphisms. rs1799732 represents a deletion (vs insertion) of cytosine at position −141, located in the 5′ promoter region of DRD2. In vitro data showed that cell lines transfected with the Del allele were less active in a luciferase reporter assay than cell lines transfected with the Ins allele.39 In vivo data with PET imaging also suggested that this polymorphism may influence D2 receptor density in the striatum of healthy volunteers unexposed to antipsychotics.40 A previous meta-analysis demonstrated that rs1799732 is associated with antipsychotic efficacy.41 rs6275 (C939T) is a synonymous polymorphism and is in close proximity and high linkage disequilibrium with rs6277 (C957T). Although not resulting in an amino acid sequence change of the D2 receptor protein, the T-allele of rs6277 is associated with down-regulated D2 receptors in the striatum42 and decreased DRD2 mRNA stability and half-life.43 Dopaminergic pathways are involved in the brain reward circuitry and modulate motivation, sense of well-being, and feeding behavior.44 Many DRD2 polymorphisms are associated with drug addiction, nicotine consumption, and eating disorders.45 In animal studies, D2 receptor availability in the striatum was significantly lower in obese than lean rats. In human studies, the availability of the striatal dopamine transporter was negatively correlated with BMI in healthy volunteers.46 These results led researchers to hypothesize that a hypodopaminergic reward circuitry, which may be caused by the D2 antagonism from antipsychotic treatment, results in abnormal over-eating and obesity.44 It is possible that certain variants of DRD2, such as the Del allele of rs1799732 or the T-allele of rs6275 and rs6277, may already produce fewer D2 receptors, and the subsequent over-eating behavior and weight gain are exacerbated by the use of antipsychotics.
One SNP in ADRA2A, rs1800544, was consistently associated with antipsychotic-related weight gain. It is located in the upstream of ADRA2A, and may be a binding site for transcription factors (ENCODE, UCSC Genome Browser). Just like 5-HT2C and D2, the alpha-2A receptor is a pharmacodynamic target of many antipsychotics, especially SGAs, including risperidone, olanzapine, and clozapine.47 Interestingly, the adrenergic system also innervates adipose tissue.48 Beta-3 adrenergic receptors in the brown adipose tissue are involved in production of heat (thermogenesis or fat burning),49 whereas alpha-receptors have an inhibitory effect on lipolysis in the adipose tissue.50 The SNP of interest, rs1800544, was associated with body fat accumulation51 in a large epidemiological study and affected plasma concentrations of glucose, insulin and cortisol in a human experimental study.52 However, it is not clear how the different alleles affect receptor density. Notably, the allele frequency is different between racial groups in that G is the minor allele in CEU (minor allele frequency [MAF] = 27.5%) but the C-allele is the minor allele in Asians (MAF = 27.5%) and Africans (23.7%). To add to the complexity, some of the included studies did not specify whether genotyping was done along the positive or negative DNA strand. In the present meta-analysis, an effort was made to align the correct allele across studies. Nevertheless, caution is warranted when interpreting these findings.
Results for Genes Implicated in Obesity in the General Population
GNB3 also seems be consistently associated with antipsychotic-related weight gain. In 10 studies (n = 1004), the TT homozygotes of rs5443 gained more weight than the C-allele carriers, with minimal heterogeneity across studies and no evidence of publication bias. Heterotrimeric G-proteins are important regulators of intracellular signaling pathways53 and the beta-3 subunit is encoded by the GNB3 gene. The SNP, rs5443 (C825T), is located on exon 10 of the GNB3 gene. The T-allele is associated with alternative splicing of GNB3 transcription, which results in enhanced signal transduction,54 and the T-allele carries a higher risk of cardiovascular disease55 and obesity56 in the general population. Increased signaling by G-proteins stimulates adipogenesis and may lead to obesity.56 This SNP seems to be associated with antidepressant efficacy,57 but it is unclear whether antipsychotics interact with the G-protein subunit directly. It is possible that the SNP moderates the effect of antipsychotics on weight gain. Further studies are warranted to elucidate underlying mechanisms.
Another gene that has moderate evidence for its association with antipsychotic-related weight gain is MC4R, located in Chromosome 18q21. In several genome-wide association studies,58–60 a genomic locus near MC4R was associated with obesity in the general population. MC4R mutations have been linked to extreme early-onset obesity,61 and MC4R knockout mice develop obesity.62 The SNP, rs489693, was approaching genome-wide significance in association with antipsychotic-related weight gain in a pediatric antipsychotic-naïve cohort, and this effect was replicated in 3 independent samples.20 The present meta-analysis added 1 unpublished cohort and 1 published study to the 4 samples. Despite heterogeneity and potential publication bias, AA homozygotes gained significantly more weight than C carriers, with a moderate effect size. MC4R neurons in the hypothalamic paraventricular nucleus, activated by α-melanocyte stimulating hormone (α-MSH) produced by the pro-opiomelanocortin (POMC)-expressing neurons, can induce decreased food intake and increased energy expenditure.63 POMC neurons are partly controlled by serontonergic signals via the 2-HT2C receptors.64 Studies have shown that deficits in either MC4R,61 POMC,65 prohormone convertase 1/3 (one of the key enzymes that converts POMC to α-MSH, encoded by the PCSK1 gene),66,67 or 5-HT2C68 resulted in obesity or drug-induced weight gain. Therefore, the POMC pathway may be an important mechanism. In addition, MC4R seems to interact with multiple other neurotransmitter pathways, including dopamine, leptin, and BDNF, in regulating appetite, eating, and energy homeostasis.69
In contrast to MC4R, other genes that have been significantly associated with obesity in the general population or that are involved in appetite regulation and energy homeostasis, including FTO, LEP (Leptin), LEPR (Leptin receptor), BDNF and INSIG2, were not consistently associated with antipsychotic-related weight gain. In the present meta-analysis, genes that are direct pharmacodynamic targets of antipsychotics were more likely significantly associated with weight gain, except for MC4R and GNB3. Perhaps, genes that are involved in metabolic regulation, energy homeostasis, and appetite control may impact upon the downstream effects of antipsychotic actions, therefore, it may be more difficult to find significant associations with antipsychotic-induced adverse events.
Several limitations of this meta-analysis must be considered. First, many results were heterogeneous across studies, but for most SNPs the number of studies was not large enough to perform moderator or meta-regression analyses. However, we were able to conduct these analyses for a few SNPs with ≥8 studies. The most significant finding is that pooled ESs tended to be larger in studies with short-term follow-up and those including patients with minimal prior antipsychotic exposure. This finding was true for both HTR2C and GNB3. The diminishing gene effects during longer-term antipsychotic treatment and follow-up could be explained by a diminishing weight gain signal overall and/or greater contributions of behavioral and environmental factors, including changes in diet and exercise or antipsychotic adherence. Thus, although we attempted to examine more homogeneous samples in terms of patient populations, prior antipsychotic exposure and follow-up duration, the analyzed studies remained heterogeneous. One source of heterogeneity may come from variability in ancestry background, which may present different allele structures and was not well accounted for in meta-analysis. We have attempted to run subgroup analysis in different racial groups whenever possible, but no significant difference was found. Future studies should always include a 2- to 3-month time point, and a greater focus on antipsychotic-naïve and first episode patients would be helpful. Interestingly, the pooled effect size for rs7700039 (−2548A/G) in LEP was only marginally significant in 10 studies, and examining the studies with short-term durations and first-episode patients failed to improve the overall ES, suggesting that perhaps LEP does not play an important role in antipsychotic-related weight gain. Additionally, we detected a significant publication bias for several results, as is expected in mostly single-gene studies. However, the overall bias seemed conservative, as generally the ES increased when potentially missing studies were imputed. Another issue is that although we attempted to evaluate the strength of evidence on the phenotype-genotype association based on the modified Venice criteria, it needs to be acknowledged that the Venice criteria was not designed for pharmacogenetic studies. Therefore, interpretation of these findings should be cautious. Finally, the antipsychotics used in each study were heterogeneous, which may limit the detection of pharmacogenetic signals. Many studies examined clozapine and olanzapine, which confer the highest weigh gain liability and are pharmacologically different from other antipsychotics. The power of detecting a signal may be increased by studying a single agent or agents with similar weight gain properties.20
In summary, the present meta-analysis attempted to overcome the limitations of previous reviews and comprehensively examined all genetic variants deemed relevant in association with antipsychotic-related weight gain. Several genes, including HTR2C, DRD2, ADRA2A, MC4R and GNB3 seem to be consistently associated with antipsychotic-related weight gain. ES were larger in patients with minimal prior antipsychotic exposure and in short-term studies. Despite promising findings, the ES of individual SNPs and genes are too small to fulfill the promise of personalized medicine. Because antipsychotic-related weight gain is likely polygenic and affected by environmental factors,9 future studies should carefully consider study design issues14,70 and explore combining multiple genetic markers and relevant clinical factors to improve clinical prediction.
Supplementary Material
Supplementary material is available at http://schizophreniabulletin.oxfordjournals.org.
Funding
This work was supported in part by the National Institutes of Health (K23MH097108 to J-P.Z., R21MH099868 to T.L., P30MH090590 to J.M.K. and P50MH080173 to A.K.M.) and by NARSAD Young Investigator Grants to J-P.Z. from the Brain & Behavior Research Foundation.
Supplementary Material
Acknowledgments
J-P.Z. has received grant support from the National Institute of Mental Health, Brain & Behavioral Research Foundation, and Genomind, Inc. T.L. has received grant support from the National Institute of Mental Health, the US-Israel Binational Science Fund, and the Brain & Behavioral Research Foundation. D.G.R. has been a consultant to Asubio, Shire and Otsuka, and he has received grants from Bristol Meyers Squibb, Janssen, and Otsuka. J.M.K. has been a consultant for Alkermes, Amgen, Bristol-Myers Squibb, Eli Lilly, EnVivo Pharmaceuticals (Forum), Forest, Genentech, H. Lundbeck. Intracellular Therapies, Janssen Pharmaceutica, Johnson and Johnson, Merck, Novartis, Otsuka, Pierre Fabre, Reviva, Roche, Sunovion and Teva. Dr. Kane has received honoraria for lectures from Bristol-Myers Squibb, Janssen, Genentech, Lundbeck and Otsuka. J.M.K. is a Shareholder in MedAvante, Inc and the Vanguard Research Group. A.K.M. has received grant support from the National Institute of Mental Health and the Brain & Behavioral Research Foundation. A.K.M. is a consultant to Genomind, Inc and Forum Pharmaceuticals. C.U.C. has been a consultant and/or advisor to or has received honoraria from AbbVie, Acadia, Actavis, Alkermes, Eli Lilly, Forum, Genentech, Gerson Lehrman Group, IntraCellular Therapies, Janssen/J&J, Lundbeck, MedAvante, Medscape, Otsuka, Pfizer, ProPhase, Reviva, Roche, Sunovion, Supernus, Takeda and Teva. He has received grant support from Bristol-Myers Squibb, Otsuka, Lundbeck and Takeda.
References
- 1. Kane JM, Correll CU. Pharmacologic treatment of schizophrenia. Dialogues Clin Neurosci. 2010;12:345–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Galling B, Garcia MA, Osuchukwu U, Hagi K, Correll CU. Safety and tolerability of antipsychotic-mood stabilizer co-treatment in the management of acute bipolar disorder: results from a systematic review and exploratory meta-analysis. Expert Opin Drug Saf. 2015;14:1181–1199. [DOI] [PubMed] [Google Scholar]
- 3. American Psychiatric Association. Practice Guideline for the Treatment of Patients With Major Depressive Disorder [Internet]. Washington, DC: American Psychiatric Association; 2010. www.psychiatryonline.com Accessed July 15, 2015. [Google Scholar]
- 4. Farahani A, Correll CU. Are antipsychotics or antidepressants needed for psychotic depression? A systematic review and meta-analysis of trials comparing antidepressant or antipsychotic monotherapy with combination treatment. J Clin Psychiatry. 2012;73:486–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. De Hert M, Detraux J, van Winkel R, Correll CU. Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nat Rev Endocrinol. 2011;8:114–126. [DOI] [PubMed] [Google Scholar]
- 6. Correll CU, Joffe BI, Rosen LM, Sullivan TB, Joffe RT. Cardiovascular and cerebrovascular risk factors and events associated with second-generation antipsychotic compared to antidepressant use in a non-elderly adult sample: results from a claims-based inception cohort study. World Psychiatry. 2015;14:56–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Correll CU, Detraux J, De Lepeleire J, De Hert M. Effects of antipsychotics, antidepressants and mood stabilizers on risk for physical diseases in people with schizophrenia, depression and bipolar disorder. World Psychiatry. 2015;14:119–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Allison DB, Mentore JL, Heo M, et al. Antipsychotic-induced weight gain: a comprehensive research synthesis. Am J Psychiatry. 1999;156:1686–1696. [DOI] [PubMed] [Google Scholar]
- 9. Correll CU, Lencz T, Malhotra AK. Antipsychotic drugs and obesity. Trends Mol Med. 2011;17:97–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Correll CU, Malhotra AK. Pharmacogenetics of antipsychotic-induced weight gain. Psychopharmacology (Berl). 2004;174:477–489. [DOI] [PubMed] [Google Scholar]
- 11. Fall T, Ingelsson E. Genome-wide association studies of obesity and metabolic syndrome. Mol Cell Endocrinol. 2014;382:740–757. [DOI] [PubMed] [Google Scholar]
- 12. Zhang J-P, Malhotra AK. Pharmacogenetics and antipsychotics: therapeutic efficacy and side effects prediction. Expert Opin Drug Metabol Toxicol. 2011;7:9–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Lett TA, Wallace TJ, Chowdhury NI, Tiwari AK, Kennedy JL, Müller DJ. Pharmacogenetics of antipsychotic-induced weight gain: review and clinical implications. Mol Psychiatry. 2012;17:242–266. [DOI] [PubMed] [Google Scholar]
- 14. Malhotra AK, Zhang JP, Lencz T. Pharmacogenetics in psychiatry: translating research into clinical practice. Mol Psychiatry. 2012;17:760–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. 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] [PMC free article] [PubMed] [Google Scholar]
- 16. Correll CU, Manu P, Olshanskiy V, Napolitano B, Kane JM, Malhotra AK. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302:1765–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Robinson DG, Woerner MG, Napolitano B, et al. Randomized comparison of olanzapine versus risperidone for the treatment of first-episode schizophrenia: 4-month outcomes. Am J Psychiatry. 2006;163:2096–2102. [DOI] [PubMed] [Google Scholar]
- 18. Lencz T, Robinson DG, Xu K, et al. DRD2 promoter region variation as a predictor of sustained response to antipsychotic medication in first-episode schizophrenia patients. Am J Psychiatry. 2006;163:529–531. [DOI] [PubMed] [Google Scholar]
- 19. Kahn RS, Fleischhacker WW, Boter H, et al. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371:1085–1097. [DOI] [PubMed] [Google Scholar]
- 20. Malhotra AK, Correll CU, Chowdhury NI, et al. Association between common variants near the Melanocortin 4 Receptor Gene and severe antipsychotic drug-induced weight gain. Arch Gen Psychiatry. 2012;69:904–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Borenstein M, Hedges LV, Higgins JPT, Rothstein HR. Introduction to Meta-Analysis. Chichester, UK: John Wiley & Sons, Ltd; 2009. [Google Scholar]
- 22. Egger M, Smith GD, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315:629–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Duval SJ, Tweedie RL. A non-parametric “trim and fill” method of assessing publication bias in meta-analysis. J Am Stat Assoc. 2000;95:89–98. [Google Scholar]
- 24. Ioannidis JP, Boffetta P, Little J, et al. Assessment of cumulative evidence on genetic associations: interim guidelines. Int J Epidemiol. 2008;37:120–132. [DOI] [PubMed] [Google Scholar]
- 25. DE Hert M, Correll CU, Bobes J, et al. Physical illness in patients with severe mental disorders. I. Prevalence, impact of medications and disparities in health care. World Psychiatry. 2011;10:52–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Fleischhacker WW, Siu CO, Bodén R, Pappadopulos E, Karayal ON, Kahn RS; EUFEST Study Group Metabolic risk factors in first-episode schizophrenia: baseline prevalence and course analysed from the European First-Episode Schizophrenia Trial. Int J Neuropsychopharmacol. 2013;16:987–995. [DOI] [PubMed] [Google Scholar]
- 27. Fall T, Ingelsson E. Genome-wide association studies of obesity and metabolic syndrome. Mol Cell Endocrinol. 2014;382:740–757. [DOI] [PubMed] [Google Scholar]
- 28. Vickers SP, Easton N, Webster LJ, et al. Oral administration of the 5-HT2Creceptor agonist, mCPP, reduces body weight gain in rats over 28 days as a result of maintained hypophagia. Psychopharmacology (Berl). 2003;167:274–280. [DOI] [PubMed] [Google Scholar]
- 29. Smith SR, Weissman NJ, Anderson CM, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363:245–256. [DOI] [PubMed] [Google Scholar]
- 30. Simansky KJ. Serotonergic control of the organization of feeding and satiety. Behav Brain Res. 1996;73:37–42. [DOI] [PubMed] [Google Scholar]
- 31. Bonhaus DW, Weinhardt KK, Taylor M, et al. RS-102221: a novel high affinity and selective, 5-HT2C receptor antagonist. Neuropharmacology. 1997;36:621–629. [DOI] [PubMed] [Google Scholar]
- 32. Tecott LH, Sun LM, Akana SF, et al. Eating disorder and epilepsy in mice lacking 5-HT2c serotonin receptors. Nature. 1995;374:542–546. [DOI] [PubMed] [Google Scholar]
- 33. Nonogaki K, Strack AM, Dallman MF, Tecott LH. Leptin-independent hyperphagia and type 2 diabetes in mice with a mutated serotonin 5-HT2C receptor gene. Nature Med. 1998;4:1152–1156. [DOI] [PubMed] [Google Scholar]
- 34. Yuan X, Yamada K, Ishiyama-Shigemoto S, Koyama W, Nonaka K. Identification of polymorphic loci in the promoter region of the serotonin 5-HT2C receptor gene and their association with obesity and type II diabetes. Diabetologia. 2000;43:373–376. [DOI] [PubMed] [Google Scholar]
- 35. Buckland PR, Hoogendoorn B, Guy CA, Smith SK, Coleman SL, O’Donovan MC. Low gene expression conferred by association of an allele of the 5-HT2C receptor gene with antipsychotic-induced weight gain. Am J Psychiatry. 2005;162:613–615. [DOI] [PubMed] [Google Scholar]
- 36. Godlewska BR, Olajossy-Hilkesberger L, Ciwoniuk M, et al. 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] [PubMed] [Google Scholar]
- 37. Drago A, Serretti A. Focus on HTR2C: a possible suggestion for genetic studies of complex disorders. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:601–637. [DOI] [PubMed] [Google Scholar]
- 38. Kapur S, Mamo D. Half a century of antipsychotics and still a central role for dopamine D2 receptors. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:1081–1090. [DOI] [PubMed] [Google Scholar]
- 39. 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] [PubMed] [Google Scholar]
- 40. Ritchie T, Noble EP. Association of seven polymorphisms of the D2 dopamine receptor gene with brain receptor-binding characteristics. Neurochemical Res. 2003;28:73–82. [DOI] [PubMed] [Google Scholar]
- 41. Zhang JP, Lencz T, Malhotra AK. D2 receptor genetic variation and clinical response to antipsychotic drug treatment: a meta-analysis. Am J Psychiatry. 2010;167:763–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Hirvonen MM, Laakso A, Någren K, Rinne JO, Pohjalainen T, Hietala J. C957T polymorphism of dopamine D2 receptor gene affects striatal DRD2 in vivo availability by changing the receptor affinity. Synapse. 2009;63:907–912. [DOI] [PubMed] [Google Scholar]
- 43. Duan J, Wainwright MS, Comeron JM, et al. Synonymous mutations in the human dopamine receptor D2 (DRD2) affect mRNA stability and synthesis of the receptor. Hum Mol Genet. 2003;12:205–216. [DOI] [PubMed] [Google Scholar]
- 44. Blum K, Thanos PK, Gold MS. Dopamine and glucose, obesity, and reward deficiency syndrome. Front Psychol. 2014;5:919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Hamdi A, Porter J, Prasad C. Decreased striatal D2 dopamine receptors in obese Zucker rats: changes during aging. Brain Res. 1992;589:338–340. [DOI] [PubMed] [Google Scholar]
- 46. Chen PS, Yang YK, Yeh TL, et al. Correlation between body mass index and striatal dopamine transporter availability in healthy volunteers--a SPECT study. NeuroImage. 2008;40:275–279. [DOI] [PubMed] [Google Scholar]
- 47. 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] [PubMed] [Google Scholar]
- 48. Snitker S, Macdonald I, Ravussin E, Astrup A. The sympathetic nervous system and obesity: role in aetiology and treatment. Obes Rev. 2000;1:5–15. [DOI] [PubMed] [Google Scholar]
- 49. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359. [DOI] [PubMed] [Google Scholar]
- 50. van Baak MA. The peripheral sympathetic nervous system in human obesity. Obes Rev. 2001;2:3–14. [DOI] [PubMed] [Google Scholar]
- 51. Garenc C, Perusse L, Chagnon YC, et al. The alpha 2-adrenergic receptor gene and body fat content and distribution: the HERITAGE Family Study. Mol Med. 2002;8:88–94. [PMC free article] [PubMed] [Google Scholar]
- 52. Rosmond R, Bouchard C, Bjorntorp P. A C-1291G polymorphism in the alpha2A-adrenergic receptor gene (ADRA2A) promoter is associated with cortisol escape from dexamethasone and elevated glucose levels. J Inter Med. 2002;251:252–257. [DOI] [PubMed] [Google Scholar]
- 53. Kozasa T, Hajicek N, Chow CR, Suzuki N. Signalling mechanisms of RhoGTPase regulation by the heterotrimeric G proteins G12 and G13. J Biochem. 2011;150:357–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Siffert W, Rosskopf D, Siffert G, et al. Association of a human G-protein beta3 subunit variant with hypertension. Nat Genet. 1998;18:45–48. [DOI] [PubMed] [Google Scholar]
- 55. Zethelius B, Berglund L, Sundstrom J, et al. Use of multiple biomarkers to improve the prediction of death from cardiovascular causes. N Engl J Med. 2008;358:2107–2116. [DOI] [PubMed] [Google Scholar]
- 56. Siffert W. G protein polymorphisms in hypertension, atherosclerosis, and diabetes. Annu Rev Med. 2005;56:17–28. [DOI] [PubMed] [Google Scholar]
- 57. Hu Q, Zhang SY, Liu F, et al. Influence of GNB3 C825T polymorphism on the efficacy of antidepressants in the treatment of major depressive disorder: a meta-analysis. J Affect Disord. 2014;172C:103–109. [DOI] [PubMed] [Google Scholar]
- 58. Chambers JC, Elliott P, Zabaneh D, et al. Common genetic variation near MC4R is associated with waist circumference and insulin resistance. Nat Genet. 2008;40:716–718. [DOI] [PubMed] [Google Scholar]
- 59. Loos RJ, Lindgren CM, Li S, et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet. 2008;40:768–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Zobel DP, Andreasen CH, Grarup N, et al. Variants near MC4R are associated with obesity and influence obesity-related quantitative traits in a population of middle-aged people: studies of 14,940 Danes. Diabetes. 2009;58:757–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Farooqi IS, O’Rahilly S. Monogenic obesity in humans. Annu Rev Med. 2005;56:443–458. [DOI] [PubMed] [Google Scholar]
- 62. Huszar D, Lynch CA, Fairchild-Huntress V, et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88:131–141. [DOI] [PubMed] [Google Scholar]
- 63. Kim JD, Leyva S, Diano S. Hormonal regulation of the hypothalamic melanocortin system. Front Physiol. 2014;5:480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Yeo GS, Heisler LK. Unraveling the brain regulation of appetite: lessons from genetics. Nature Neurosci. 2012;15:1343–1349. [DOI] [PubMed] [Google Scholar]
- 65. Krude H, Gruters A. Implications of proopiomelanocortin (POMC) mutations in humans: the POMC deficiency syndrome. Trends Endocrinol Metab. 2000;11:15–22. [DOI] [PubMed] [Google Scholar]
- 66. Benzinou M, Creemers JW, Choquet H, et al. Common nonsynonymous variants in PCSK1 confer risk of obesity. Nat Genet. 2008;40:943–945. [DOI] [PubMed] [Google Scholar]
- 67. Kilpelainen TO, Bingham SA, Khaw KT, Wareham NJ, Loos RJ. Association of variants in the PCSK1 gene with obesity in the EPIC-Norfolk study. Hum Mol Genet. 2009;18:3496–3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. 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] [PubMed] [Google Scholar]
- 69. Harrold JA, Williams G. Melanocortin-4 receptors, beta-MSH and leptin: key elements in the satiety pathway. Peptides. 2006;27:365–371. [DOI] [PubMed] [Google Scholar]
- 70. Hamilton SP. The Promise of Psychiatric Pharmacogenomics. Biol Psychiatry. 2015;77:29–35. [DOI] [PubMed] [Google Scholar]
- 71. 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]
- 72. Basile VS, Masellis M, De Luca V, Meltzer HY, Kennedy JL. 759C/T genetic variation of 5HT(2C) receptor and clozapine-induced weight gain. Lancet. 2002;360:1790–1791. [DOI] [PubMed] [Google Scholar]
- 73. 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]
- 74. Brandl EJ, Frydrychowicz C, Tiwari AK, et al. Association study of polymorphisms in leptin and leptin receptor genes with antipsychotic-induced body weight gain. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38:134–141. [DOI] [PubMed] [Google Scholar]
- 75. 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] [PMC free article] [PubMed] [Google Scholar]
- 76. Chowdhury NI, Tiwari AK, Souza RP, et al. Genetic association study between antipsychotic-induced weight gain and the melanocortin-4 receptor gene. Pharmacogenomics J. 2013;13:272–279. [DOI] [PubMed] [Google Scholar]
- 77. Czerwensky F, Leucht S, Steimer W. Association of the common MC4R rs17782313 polymorphism with antipsychotic-related weight gain. J Clin Psychopharmacol. 2013;33:74–79. [DOI] [PubMed] [Google Scholar]
- 78. Czerwensky F, Leucht S, Steimer W. MC4R rs489693: a clinical risk factor for second generation antipsychotic-related weight gain? Int J Neuropsychopharmacol. 2013;16:2103–2109. [DOI] [PubMed] [Google Scholar]
- 79. Ellingrod VL, Perry PJ, Ringold JC, et al. 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] [PubMed] [Google Scholar]
- 80. 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]
- 81. Fernandez E, Carrizo E, Fernandez V, et al. Polymorphisms of the LEP- and LEPR genes, metabolic profile after prolonged clozapine administration and response to the antidiabetic metformin. Schizophrenia Res. 2010;121:213–217. [DOI] [PubMed] [Google Scholar]
- 82. Herken H, Erdal M, Aydin N, et al. 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] [PubMed] [Google Scholar]
- 83. Hoekstra PJ, Troost PW, Lahuis BE, et al. Risperidone-induced weight gain in referred children with autism spectrum disorders is associated with a common polymorphism in the 5-hydroxytryptamine 2C receptor gene. J Child Adolesc Psychopharmacol. 2010;20:473–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. 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] [PubMed] [Google Scholar]
- 85. Hong CJ, Liou YJ, Bai YM, Chen TT, Wang YC, Tsai SJ. Dopamine receptor D2 gene is associated with weight gain in schizophrenic patients under long-term atypical antipsychotic treatment. Pharmacogenet Genomics. 2010;20:359–366. [DOI] [PubMed] [Google Scholar]
- 86. Houston JP, Kohler J, Bishop JR, et al. Pharmacogenomic associations with weight gain in olanzapine treatment of patients without schizophrenia. J Clin Psychiatry. 2012;73:1077–1086. [DOI] [PubMed] [Google Scholar]
- 87. Huang HH, Wang YC, Wu CL, et al. TNF-alpha -308 G>A polymorphism and weight gain in patients with schizophrenia under long-term clozapine, risperidone or olanzapine treatment. Neurosci Lett. 2011;504:277–280. [DOI] [PubMed] [Google Scholar]
- 88. Kang SG, Lee HJ, Park YM, et al. 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] [PubMed] [Google Scholar]
- 89. Kuzman MR, Medved V, Bozina N, Hotujac L, Sain I, Bilusic H. The influence of 5-HT(2C) and MDR1 genetic polymorphisms on antipsychotic-induced weight gain in female schizophrenic patients. Psychiatry Res. 2008;160:308–315. [DOI] [PubMed] [Google Scholar]
- 90. Kuzman MR, Medved V, Bozina N, Grubišin J, Jovanovic N, Sertic J. Association study of MDR1 and 5-HT2C genetic polymorphisms and antipsychotic-induced metabolic disturbances in female patients with schizophrenia. Pharmacogenomics J. 2011;11:35–44. [DOI] [PubMed] [Google Scholar]
- 91. Laika B, Leucht S, Heres S, Schneider H, Steimer W. Pharmacogenetics and olanzapine treatment: CYP1A2*1F and serotonergic polymorphisms influence therapeutic outcome. Pharmacogenomics J. 2010;10:20–29. [DOI] [PubMed] [Google Scholar]
- 92. Lane HY, Liu YC, Huang CL, et al. Risperidone-related weight gain: genetic and nongenetic predictors. J Clin Psychopharmacol. 2006;26:128–134. [DOI] [PubMed] [Google Scholar]
- 93. Le Hellard S, Theisen FM, Haberhausen M, et al. Association between the insulin-induced gene 2 (INSIG2) and weight gain in a German sample of antipsychotic-treated schizophrenic patients: perturbation of SREBP-controlled lipogenesis in drug-related metabolic adverse effects? Mol Psychiatry. 2009;14:308–317. [DOI] [PubMed] [Google Scholar]
- 94. Lencz T, Robinson DG, Napolitano B, et al. DRD2 promoter region variation predicts antipsychotic-induced weight gain in first episode schizophrenia. Pharmacogenet Genomics. 2010;20:569–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lin YC, Ellingrod VL, Bishop JR, Miller DD. The relationship between P-glycoprotein (PGP) polymorphisms and response to olanzapine treatment in schizophrenia. Ther Drug Monit. 2006;28:668–672. [DOI] [PubMed] [Google Scholar]
- 96. 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] [PubMed] [Google Scholar]
- 97. Monteleone P, Milano W, Petrella C, Canestrelli B, Maj M. Endocannabinoid Pro129Thr FAAH functional polymorphism but not 1359G/A CNR1 polymorphism is associated with antipsychotic-induced weight gain. J Clin Psychopharmacol. 2010;30:441–445. [DOI] [PubMed] [Google Scholar]
- 98. 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]
- 99. Müller DJ, Klempan TA, De Luca V, et al. The SNAP-25 gene may be associated with clinical response and weight gain in antipsychotic treatment of schizophrenia. Neurosci Lett. 2005;379:81–89. [DOI] [PubMed] [Google Scholar]
- 100. Müller DJ, Zai CC, Sicard M, et al. Systematic analysis of dopamine receptor genes (DRD1-DRD5) in antipsychotic-induced weight gain. Pharmacogenomics J. 2012;12:156–164. [DOI] [PubMed] [Google Scholar]
- 101. Musil R, Spellmann I, Riedel M, et al. SNAP-25 gene polymorphisms and weight gain in schizophrenic patients. J Psychiatr Res. 2008;42:963–970. [DOI] [PubMed] [Google Scholar]
- 102. Opgen-Rhein C, Brandl EJ, Muller DJ, et al. Association of HTR2C, but not LEP or INSIG2, genes with antipsychotic-induced weight gain in a German sample. Pharmacogenomics. 2010;11:773–780. [DOI] [PubMed] [Google Scholar]
- 103. Park YM, Chung YC, Lee SH, et al. Weight gain associated with the alpha2a-adrenergic receptor -1,291 C/G polymorphism and olanzapine treatment. Am J Med Genet B Neuropsychiatr Genet. 2006;141B:394–397. [DOI] [PubMed] [Google Scholar]
- 104. Park YM, Cho JH, Kang SG, et al. 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] [PubMed] [Google Scholar]
- 105. Park YM, Chung YC, Lee SH, et al. G-protein beta3 subunit gene 825C/T polymorphism is not associated with olanzapine-induced weight gain in Korean schizophrenic patients. Psychiatry Investig. 2009;6:39–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Park YM, Choi JE, Kang SG, et al. Cannabinoid type 1 receptor gene polymorphisms are not associated with olanzapine-induced weight gain. Hum Psychopharmacol. 2011;26:332–337. [DOI] [PubMed] [Google Scholar]
- 107. Perez-Iglesias R, Mata I, Amado JA, et al. Effect of FTO, SH2B1, LEP, and LEPR polymorphisms on weight gain associated with antipsychotic treatment. J Clin Psychopharmacol. 2010;30:661–666. [DOI] [PubMed] [Google Scholar]
- 108. Popp J, Leucht S, Heres S, Steimer W. DRD4 48bp VNTR but not 5-HT 2C Cys23Ser receptor polymorphism is related to antipsychotic-induced weight gain. Pharmacogenomics J. 2009;9:71–77. [DOI] [PubMed] [Google Scholar]
- 109. 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] [PubMed] [Google Scholar]
- 110. Reynolds GP, Yevtushenko OO, Gordon S, Arranz B, San L, Cooper SJ. The obesity risk gene FTO influences body mass in chronic schizophrenia but not initial antipsychotic drug-induced weight gain in first-episode patients. Int J Neuropsychopharmacol. 2013;16:1421–1425. [DOI] [PubMed] [Google Scholar]
- 111. Ryu S, Jang WS, Cho EY, Kim SK, Lee D, Hong KS. Association study of -2548A/g polymorphism of leptin gene with antipsychotics-induced weight gain. Korean J Psychopharmacol. 2006;17:423–428. [Google Scholar]
- 112. Ryu S, Cho EY, Park T, et al. -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] [PubMed] [Google Scholar]
- 113. 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]
- 114. Shing EC, Tiwari AK, Brandl EJ, et al. Fat mass- and obesity-associated (FTO) gene and antipsychotic-induced weight gain: an association study. Neuropsychobiology. 2014;69:59–63. [DOI] [PubMed] [Google Scholar]
- 115. Sicard MN, Zai CC, Tiwari AK, et al. Polymorphisms of the HTR2C gene and antipsychotic-induced weight gain: an update and meta-analysis. Pharmacogenomics. 2010;11:1561–1571. [DOI] [PubMed] [Google Scholar]
- 116. Sickert L, Müller DJ, Tiwari AK, et al. Association of the alpha 2A adrenergic receptor -1291C/G polymorphism and antipsychotic-induced weight gain in European-Americans. Pharmacogenomics. 2009;10:1169–1176. [DOI] [PubMed] [Google Scholar]
- 117. Song X, Pang L, Feng Y, et al. Fat-mass and obesity-associated gene polymorphisms and weight gain after risperidone treatment in first episode schizophrenia. Behav Brain Funct. 2014;10:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Souza RP, De Luca V, Muscettola G, et al. Association of antipsychotic induced weight gain and body mass index with GNB3 gene: a meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1848–1853. [DOI] [PubMed] [Google Scholar]
- 119. Srisawat U, Reynolds GP, Zhang ZJ, et al. Methyle netetrahydrofolate reductase (MTHFR) 677C/T polymorphism is associated with antipsychotic-induced weight gain in first-episode schizophrenia. Int J Neuropsychopharmacol. 2014;17:485–490. [DOI] [PubMed] [Google Scholar]
- 120. Staeker J, Leucht S, Steimer W. Peroxisome proliferator-activated receptor gamma (PPARG) Pro12Ala: lack of association with weight gain in psychiatric inpatients treated with olanzapine or clozapine. Mol Diagn Ther. 2012;16:93–98. [DOI] [PubMed] [Google Scholar]
- 121. 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] [PubMed] [Google Scholar]
- 122. Theisen FM, Hinney A, Bromel T, et al. Lack of association between the -759C/T polymorphism of the 5-HT2C receptor gene and clozapine-induced weight gain among German schizophrenic individuals. Psychiatr Genet. 2004;14:139–142. [DOI] [PubMed] [Google Scholar]
- 123. Thompson A, Lavedan C, Volpi S. Absence of weight gain association with the HTR2C -759C/T polymorphism in patients with schizophrenia treated with iloperidone. Psychiatry Res. 2010;175:271–273. [DOI] [PubMed] [Google Scholar]
- 124. Tiwari AK, Zai CC, Meltzer HY, Lieberman JA, Müller DJ, Kennedy JL. Association study of polymorphisms in insulin induced gene 2 (INSIG2) with antipsychotic-induced weight gain in European and African-American schizophrenia patients. Hum Psychopharmacol. 2010;25:253–259. [DOI] [PubMed] [Google Scholar]
- 125. Tiwari AK, Zai CC, Likhodi O, et al. A common polymorphism in the cannabinoid receptor 1 (CNR1) gene is associated with antipsychotic-induced weight gain in Schizophrenia. Neuropsychopharmacology. 2010;35:1315–1324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. 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] [PubMed] [Google Scholar]
- 127. 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] [PubMed] [Google Scholar]
- 128. 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] [PubMed] [Google Scholar]
- 129. Tsai A, Liou YJ, Hong CJ, Wu CL, Tsai SJ, Bai YM. Association study of brain-derived neurotrophic factor gene polymorphisms and body weight change in schizophrenic patients under long-term atypical antipsychotic treatment. Neuromolecular Med. 2011;13:328–333. [DOI] [PubMed] [Google Scholar]
- 130. Ujike H, Nomura A, Morita Y, et al. Multiple genetic factors in olanzapine-induced weight gain in schizophrenia patients: a cohort study. J Clin Psychiatry. 2008;69:1416–1422. [DOI] [PubMed] [Google Scholar]
- 131. van Winkel R, Moons T, Peerbooms O, et al. MTHFR genotype and differential evolution of metabolic parameters after initiation of a second generation antipsychotic: an observational study. Int Clin Psychopharmacol. 2010;25:270–276. [DOI] [PubMed] [Google Scholar]
- 132. 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 (Vienna). 2005;112:1463–1468. [DOI] [PubMed] [Google Scholar]
- 133. 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] [PubMed] [Google Scholar]
- 134. Wang YC, Bai YM, Chen JY, Lin CC, Lai IC, Liou YJ. Genetic association between TNF-alpha -308 G>A polymorphism and longitudinal weight change during clozapine treatment. Hum Psychopharmacol. 2010;25:303–309. [DOI] [PubMed] [Google Scholar]
- 135. Zai GC, Zai CC, Chowdhury NI, et al. The role of brain-derived neurotrophic factor (BDNF) gene variants in antipsychotic response and antipsychotic-induced weight gain. Prog Neuropsychopharmacol Biol Psychiatry. 2012;39:96–101. [DOI] [PubMed] [Google Scholar]
- 136. Zhang ZJ, Yao ZJ, Zhang XB, et al. No association of antipsychotic agent-induced weight gain with a DA receptor gene polymorphism and therapeutic response. Acta Pharmacol Sinica. 2003;24:235–240. [PubMed] [Google Scholar]
- 137. Zhang ZJ, Yao ZJ, Mou XD, et al. [Association of -2548G/A functional polymorphism in the promoter region of leptin gene with antipsychotic agent-induced weight gain]. Zhonghua Yi Xue Za Zhi. 2003;83:2119–2123. [PubMed] [Google Scholar]
- 138. Zhang XY, Tan YL, Zhou DF, et al. Association of clozapine-induced weight gain with a polymorphism in the leptin promoter region in patients with chronic schizophrenia in a Chinese population. J Clin Psychopharmacol. 2007;27:246–251. [DOI] [PubMed] [Google Scholar]
- 139. Zhang XY, Zhou DF, Wu GY, et al. BDNF levels and genotype are associated with antipsychotic-induced weight gain in patients with chronic schizophrenia. Neuropsychopharmacology. 2008;33:2200–2205. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.