The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ArticleFull Access

Genome-Wide Pharmacogenetics of Antidepressant Response in the GENDEP Project

Abstract

Objective

The purpose of this study was to identify genetic variants underlying the considerable individual differences in response to antidepressant treatment. The authors performed a genome-wide association analysis of improvement of depression severity with two antidepressant drugs.

Method

High-quality Illumina Human610-quad chip genotyping data were available for 706 unrelated participants of European ancestry treated for major depression with escitalopram (N=394) or nortriptyline (N=312) over a 12-week period in the Genome-Based Therapeutic Drugs for Depression (GENDEP) project, a partially randomized open-label pharmacogenetic trial.

Results

Single nucleotide polymorphisms in two intergenic regions containing copy number variants on chromosomes 1 and 10 were associated with the outcome of treatment with escitalopram or nortriptyline at suggestive levels of significance and with a high posterior likelihood of true association. Drug-specific analyses revealed a genome-wide significant association between marker rs2500535 in the uronyl 2-sulphotransferase gene and response to nortriptyline. Response to escitalopram was best predicted by a marker in the interleukin-11 (IL11) gene. A set of 72 a priori-selected candidate genes did not show pharmacogenetic associations above a chance level, but an association with response to escitalopram was detected in the interleukin-6 gene, which is a close homologue of IL11.

Conclusions

While limited statistical power means that a number of true associations may have been missed, these results suggest that efficacy of antidepressants may be predicted by genetic markers other than traditional candidates. Genome-wide studies, if properly replicated, may thus be important steps in the elucidation of the genetic basis of pharmacological response.

After 50 years of experience with antidepressant drugs, the large individual variation in antidepressant treatment outcome remains poorly understood. Investigations of biologically related individuals point to a genetic contribution to treatment responsiveness (1), and several polymorphisms in candidate genes have been associated with antidepressant response (2, 3). However, few pharmacogenetic associations have been replicated, and they explain only a small fraction of individual differences in antidepressant response. Since our understanding of the mechanisms of action of antidepressants is incomplete, important genes and regulatory intergenic sequences may have been missed in candidate gene studies. Therefore, a systematic exploration of variation across the genome has the potential to detect further variants that may help in understanding the biology of antidepressant action and predict response to a specific antidepressant in an individual patient. In the present study, we report on a genome-wide pharmacogenetic analysis of more than 500,000 common genetic variants, tested for association with change in depression severity over a 12-week period after commencing treatment with a serotonergic or noradrenergic antidepressant among subjects with moderate to severe depression. We conducted four primary analyses to address general and drug-specific effects of genes on antidepressant response and gene-drug interactions. We used secondary analyses to explore the influence of variants in 72 candidate genes on antidepressant action.

Method

Study Design and Sample

The Genome-Based Therapeutic Drugs for Depression (GENDEP) project (3, 4) is a 12-week partially randomized open-label pharmacogenetic study with two active treatment arms. A total of 811 adult patients (men: N=297; women: N=514) with unipolar depression of at least moderate severity according to ICD-10 or DSM-IV criteria were recruited at nine European centers. Patients were aged 19–72 years and of Caucasian European parentage. Diagnoses were established using the semistructured Schedules for Clinical Assessment in Neuropsychiatry interview (5). Exclusion criteria were personal and family history of schizophrenia or bipolar disorder and current substance dependence. Eligible participants were allocated to treatment with either escitalopram or nortriptyline, which represent different mechanisms of antidepressant action. Escitalopram is a selective inhibitor of the serotonin transporter, with no effect on norepinephrine reuptake (6). Nortriptyline is a tricyclic antidepressant, with an affinity for the norepinephrine transporter that is 100 times higher than for the serotonin transporter (7). Patients with no contraindications were randomly allocated to flexible-dosage nortriptyline (50–150 mg daily) or escitalopram (10–30 mg daily) for 12 weeks. Patients with contraindications for one drug were offered the other. Severity of depression was assessed weekly by three established rating scales (8). A total of 628 participants (77%) completed at least 8 weeks of treatment with the originally allocated antidepressant (4). The GENDEP project was approved by ethics boards of participating centers, and all participants provided written informed consent. Previous investigations of the GENDEP project sample included three candidate gene pharmacogenetic studies (3, 9, 10).

Definition of Phenotype

Of the three scales used in the GENDEP project, the clinician-rated 10-item Montgomery-Åsberg Depression Rating Scale (MADRS) (11) was shown to be the most accurate measure of depression severity (8) and to have the closest agreement with assessment by experienced clinicians (12), and thus it was used as the primary outcome measure. Since improvement in depression severity with treatment is a matter of degree rather than a yes or no phenomenon and there is no indication of bimodal endpoint outcomes (13), continuous measures, such as percentage improvement, are preferable to cutoff-based dichotomous measures of response or remission (14). Percentage change in MADRS scores from baseline to week 12 was selected instead of absolute change, since it shows high correlation with the end-of-treatment score (r=0.84) and low correlation with baseline severity (r=–0.06) and better reflects clinicians' impression of improvement (15). Since the commonly used last-observation-carried-forward procedure introduces bias in the presence of missing data (16), we imputed missing week-12 measurements using the best unbiased estimate from a mixed-effect linear regression model, with fixed linear and quadratic effects of time and random effects of individual and center of recruitment, which are relatively free of bias under a wider range of assumptions (4, 16). Finally, we adjusted for age and recruitment center because outcome was associated with age and varied across centers of recruitment. This adjustment minimized the risk that minor genetic differences as a result of stratification of European populations could be spuriously associated with outcome.

Genotyping

DNA was extracted from blood samples provided by 795 participants and collected in ethylenediaminetetraacetic acid (17). A total of 727 samples that were available in sufficient quantity and quality were sent to the Centre National de Genotypage (Evry Cedex, France) and genotyped using the Illumina Human610-quad bead chip (Illumina, Inc., San Diego). This chip assays more than 610,000 single nucleotide polymorphisms (SNPs) and copy number variant markers selected to provide a comprehensive coverage across populations, and it captures the majority of known common variations in the human genome, based on HapMap (release 23).

Statistical Analysis

After quality control (see the data supplement accompanying the online version of this article), the association between genotypes and adjusted percentage improvement in MADRS scores was performed using linear regression analyses under an additive genetic model, with the first four principal components from population structure analysis used as covariates to control for population stratification (see the data supplement), implemented in PLINK (18). The following four tests were performed: 1) association between genotype and outcome across the whole sample to identify genetic variants associated with improvement in depression severity irrespective of the type of antidepressant treatment; 2) association between genotype and outcome among subjects treated with escitalopram to identify genetic variants associated with the outcome of treatment with a serotonin reuptake inhibitor; 3) association between genotype and outcome among subjects treated with nortriptyline to identify genetic variants associated with the outcome of treatment with a noradrenaline reuptake inhibitor; and 4) interaction between genotype and drug to identify genetic variants that differentially predict the outcome of treatment with escitalopram and nortriptyline. The quality of control for population stratification effects was checked using quantile-quantile plots and genomic control lambda (19).

Frequentist and Bayesian approaches were used to assess the likelihood of results being true or false. To account for multiple testing, genome-wide significance was set at p<5×10–8 (20, 21). However, since many markers associated with important individual differences in outcome may be identified at more modest levels of significance, all associations detected at p<5×10–6 are reported as suggestive findings of interest. To estimate the posterior probability of true positive findings in the context of multiple nonindependent tests, false discovery rate q values were calculated, using the Benjamini and Hochberg step-up procedure (22), which have been shown to retain desirable properties in genome-wide studies and can be interpreted as posterior probabilities of no association at a given locus (23). We included q values alongside p values, and all associations with a q value ≤0.1 in the primary analyses are reported.

The pharmacogenetic analyses had statistical power to detect clinically significant pharmacogenetic effects explaining ≥5% of variance but were expected to miss a large proportion of moderate or weak effects (see the online data supplement).

Exploration of Candidate Genes

We extracted results for 2,801 markers in the coding regions and 20 kilo-base-pair upstream and downstream sequences of 72 candidate genes identified in a literature review (2) as encoding proteins involved in antidepressant action, previously reported to be associated with the outcome of antidepressant treatment or with mental illness in a genome-wide study (see Table 1 in the online data supplement for a list of candidate genes and rationale for their inclusion). Results from these candidate genes that remained significant after correction for the number of markers within a gene are reported.

Results

Genotyping Quality Control and Sample

Illumina Human610-quad genotyping was obtained for 727 subjects. Of the 550,337 SNPs with a minor allele frequency >0.01, a total of 539,391 (98%) were at least 99% complete and retained for analyses. Individual quality control is summarized in Figure 1. There were no sex mismatches, but three individuals with ambiguous genotypic sex were excluded. Three individuals representing outliers on heterozygosity were also excluded. One individual in each of six pairs of related individuals (three first- and three second-degree pairs of relatives) was retained for further analyses. Four individuals representing outliers with genotyping completeness <95% were excluded. After excluding five population structure outliers, 706 of the 811 participants were retained for the main analyses, with a mean genotyping completeness of 99.82%.

Figure 1. Flow Diagram of Depressed Adult Patients Treated With Escitalopram or Nortriptylinea

aSample was drawn from the Genome-Based Therapeutic Drugs for Depression (GENDEP) project.

The clean sample of unrelated subjects of European ancestry passing quality control and included in genome-wide pharmacogenetic analyses is described in Table 1. These 706 included subjects did not differ from the other 105 GENDEP participants on sex, medication history, depression severity, or outcome. There was a difference in age, with included individuals being younger (mean age: 42.0 years [SD=11.7] versus 45.0 years [SD=11.6]; t=2.50, p=0.01), which may have been the result of chance, given the number of comparisons, and should not have influenced results because the outcome was adjusted for age.

Table 1. Demographic and Clinical Characteristics of Patients in a Genome-Wide Pharmacogenetic Analysisa
Table 1. Demographic and Clinical Characteristics of Patients in a Genome-Wide Pharmacogenetic Analysisa
Enlarge table

Genome-Wide Pharmacogenetic Analysis

Whole sample

The first linear regression analysis tested the association between 539,391 SNP markers and the continuous outcome of age- and recruitment center-adjusted percentage change in MADRS scores under an additive genetic model in the 706 individuals treated with either escitalopram or nortriptyline. A quantile-quantile plot showed a uniform distribution of p values (see the online data supplement), and a genomic control lambda coefficient of 1.007 confirmed no inflation of test statistics. No association was identified at a genome-wide level of significance. The seven strongest associations had suggestive levels of significance and a false-discovery q value <0.1 (Table 2). As seen in Figure 2, these associations were grouped in two intergenic regions on chromosomes 1 and 10. Both associated regions were contained within previously described common copy number variants (24), and the minor alleles were associated with worse response to both antidepressants.

Table 2. Pharmacogenetic Associations Identified at a Genome-Wide or Suggestive Level of Significance in Patients Treated With Escitalopram or Nortriptylinea
Table 2. Pharmacogenetic Associations Identified at a Genome-Wide or Suggestive Level of Significance in Patients Treated With Escitalopram or Nortriptylinea
Enlarge table
Figure 2. Pharmacogenetic Association Analyses (Linear Regression) by Chromosomal Position of Depressed Adult Patients Treated With Escitalopram or Nortriptylinea

aSample was drawn from the Genome-Based Therapeutic Drugs for Depression (GENDEP) project.

The three associated markers on chromosome 1 lie within a 163-kilo-base-pair duplication, which was reported for 98 of 270 HapMap subjects (24). Another six SNPs within this region, all with a minor allele frequency of approximately 0.3, could be imputed with high certainty and were associated with treatment response at similar levels of significance as the genotyped markers (see the online data supplement). Although no known genes overlap these markers, there are three expressed sequence tags that could represent two genes, which are potentially affected. None of the expressed sequence tags was homologous to a known gene, and thus it was not possible to infer their potential function.

The associated markers on chromosome 10 map within several structural variants, including two overlapping copy number variants of >200 kilo base pairs identified in 126/270 HapMap samples. Another 14 imputed SNPs with a minor allele frequency of 6%–7% were associated with treatment response (see the online data supplement). Again, no known genes overlapped these markers, but a single spliced expressed sequence tag (DA737884) was identified, homologous with the mixed-lineage leukemia-translocated MLLT10 gene (a chromatin modifier), located approximately 1 mega base pair downstream.

Escitalopram-treated subjects

A second linear regression analysis tested the association with the continuous outcome of age- and recruitment center-adjusted percentage change in MADRS scores for 394 individuals treated with escitalopram. The quantile-quantile plot and genomic control lambda coefficient of 1.011 showed no inflation of test statistics (see the online data supplement). No marker was associated at a genome-wide level of significance. A synonymous SNP in the coding region of the interleukin-11 gene, IL11, was associated with outcome at a suggestive level of significance (Table 2, Figure 2). This SNP creates new consensus sequences for the exonic splice enhancer factor 2/alternative splicing factor, which could favor an alternative IL11 splice variant (25). It was not in strong linkage disequilibrium with other genotyped or imputed markers.

Nortriptyline-treated subjects

A third linear regression analysis tested the association with the continuous outcome of age- and recruitment center-adjusted percentage change in MADRS scores under an additive genetic model in 312 individuals treated with nortriptyline. A quantile-quantile plot and genomic control lambda coefficient of 0.989 confirmed no inflation of test statistics (see the online data supplement). The intronic rs2500535 SNP within the uronyl 2-sulphotransferase (UST) gene on chromosome 6 was associated with outcome at a genome-wide level of significance and with a very small posterior probability of being a false positive (Table 2, Figure 2). Carriers of the minor A allele at rs2500535 experienced less improvement than carriers of the GG homozygote from week 4 onward. Imputation identified another 11 SNPs, with a minor allele frequency of 4%–6%, associated at genome-wide or suggestive levels of significance. Two of these imputed SNPs (rs2486404 and rs2500525) were predicted to be associated more strongly than the genotyped SNP, after taking into account the uncertainty of imputation. None of the associated markers were located in exons or regions predicted to influence gene function.

Another two genotyped markers associated at suggestive levels of significance (rs4651156 and rs9425322) were contained within an intron of the Ral guanine nucleotide dissociation stimulator-like 1 (RGL1) gene on chromosome 1 (Table 2, Figure 2), encoding a protein involved in G-protein signaling. Imputation identified another five SNPs in this region, with minor allele frequencies between 0.17 and 0.28, associated with response to nortriptyline (see the online data supplement). The genotyped marker rs9425322 causes a guanine-to-adenine transition and is located in an intronic region with strong mammalian conservation. Although this level of intronic conservation suggests regulatory function, the SNP does not directly affect known transcription factor binding sites, and the functional effect of this variant is unknown.

Genotype-drug interaction

A fourth linear regression analysis tested interactions between the type of antidepressant drug and genotypes in their effects on age- and recruitment center-adjusted percentage change in MADRS scores under an additive genetic model. The genomic control lambda coefficient was 0.991. The following two markers interacted with treatment at a suggestive level of significance: 1) rs11666579 in the solute carrier family 27 member 1 (SLC27A1) gene on chromosome 19, encoding a fatty acid transporter, and 2) rs1013696 in an intergenic region 11 kilo base pairs downstream from the nucleolar protein 4 (NOL4) gene on chromosome 18 (Table 2, Figure 2). One additional marker in strong linkage disequilibrium with rs11666579 was identified through imputation (see the online data supplement). The rs2500535 marker in the UST gene, associated with nortriptyline response, was also among the strongest interactions.

Pharmacogenetic Analysis

Candidate genes

Among the 2,801 markers in 72 candidate genes, none was significantly associated with outcome after correction for the number of comparisons (Bonferroni-corrected p value=1.8×10–5). Pharmacogenetic associations that remained significant after correction for the number of markers within a gene are reported (Table 3). The strongest associations were 1) a marker in the norepinephrine transporter gene (SLC6A2) with overall response, 2) markers in a glutamatergic receptor gene (GRIA4) and the interleukin-6 (IL6) gene with response to escitalopram, and 3) markers in the leptin gene (LEP) and another glutamatergic receptor gene (GRIK1) with response to nortriptyline. A marker downstream of the HTR3A gene, encoding a serotonergic receptor, showed a gene-drug interaction because its minor C allele was associated with a poor response to escitalopram and a good response to nortriptyline.

Table 3. Pharmacogenetic Associations Remaining Significant After Correction for the Number of Markers Within Each Candidate Gene in Patients Treated With Escitalopram or Nortriptylinea
Table 3. Pharmacogenetic Associations Remaining Significant After Correction for the Number of Markers Within Each Candidate Gene in Patients Treated With Escitalopram or Nortriptylinea
Enlarge table

Discussion

This genome-wide analysis of the GENDEP project has shown that previously unexpected genomic regions may be more potent predictors of antidepressant response than functional candidate genes. Several genomic loci emerged that were more likely to be associated with response than not, after taking into account the extensive multiple testing performed. The genes encoding uronyl 2-sulphotransferase and IL11 as well as two intergenic regions containing common structural variants were revealed as potentially important for antidepressant action. To our knowledge, none of the markers associated with response in the GENDEP project has previously been reported in genome-wide studies of depression susceptibility (26, 27) or antidepressant response (28). Since the statistical power of the present investigation was limited, it is likely that a number of moderately strong associations have been missed, and integration of data from multiple large samples may reveal further important pharmacogenetic associations.

Irrespective of which antidepressant was used, the outcome of treatment was associated with polymorphisms in two regions on chromosomes 1 and 10. The nearest known gene to the associated locus on chromosome 1 (zinc finger protein 326 [ZNF326]) was 380 kilo base pairs away from the associated markers, and the nearest gene to the locus on chromosome 10 (plexin domain containing 2 [PLXDC2]) was 241 kilo base pairs away from the associated markers. Common copy number polymorphisms that may influence the expression of genes at longer distances through structural changes of the chromatin have been reported in both regions. This finding demonstrates the importance of exploring the whole genome rather than concentrating on coding regions of known genes and their flanking sequences.

Antidepressant-specific analyses revealed a strong genome-wide significant association between a marker in the UST gene and response to nortriptyline. Although the UST gene did not appear on any previous list of candidate genes, the function of uronyl 2-sulphotransferase in the nervous system suggests a mechanism for its involvement in antidepressant action. This enzyme is responsible for the production of oversulfated proteoglycans that are essential for neurogenesis and neuronal migration (29). A knockdown of UST led to severe disturbance of neuronal migration that was rescued by a UST cDNA (30). The apparent delayed onset of the pharmacogenetic effect on response after 4 weeks of treatment was consistent with a neurogenesis-related mechanism. It is presently unclear why such an effect should be specific to nortriptyline, since various types of antidepressants increase neurogenesis (31). Response to nortriptyline was also associated with markers in the RGL1 gene, involved in Ras and Ral signaling pathways of neuronal differentiation and outgrowth (32). Nearby markers in this gene have been associated with conduct disorder and hyperactivity (33). These findings require replication and investigation of the role of neurogenesis-related genes in antidepressant action.

Escitalopram response was predicted by a marker in the gene encoding interleukin-11. Although statistically less robust, this finding was further supported by an association of escitalopram response with a marker in the IL6 gene, an established candidate gene for depression (34) and a close homologue of IL11. Interleukins 6 and 11 are members of a family of neuropoietic cytokines, which are widely expressed in the brain (35) and can potently inhibit serotonin signaling by inducing raphe neurons to produce acetylcholine instead of serotonin (36). A differential expression of these cytokines, or alternative splicing suggested by an exonic splicing enhancer consensus sequence (25), may make some individuals vulnerable to a subtype of depression that may be related to environmental exposures (37) and poor response to serotonergic antidepressants (38). This molecular mechanism is consistent with the role of inflammation in a subtype of depression (39) and could explain the specific moderation by IL6 and IL11 of response to the serotonergic antidepressant escitalopram.

A comprehensive list of 72 candidate genes, including those encoding key proteins in monoaminergic and glutamatergic signaling, have produced findings consistent with chance. However, markers in several candidate genes, including IL6, HTR3A, and GRIA4, showed gene-wide significant pharmacogenetic associations that would have been reported as positive results in a candidate gene study. The implication of IL6 in response to serotonergic antidepressants was supported by the pharmacogenetic association with a functionally related, but genomically independent, IL11 gene. The HTR3A gene, encoding the serotonergic 5-HT3A receptor, is an important candidate as effective antidepressants are antagonists of this receptor (40) and colocalize with the receptor on cell surfaces (41). These genes should be considered for exploration in future studies.

A genome-wide pharmacogenetic study faces a number of challenges. The strengths of the GENDEP project include a homogenous sample specifically recruited for a pharmacogenetic investigation, detailed and complete data characterizing response to antidepressants, tight control of population stratification with no overall inflation of test statistics, and high-quality genotyping enabling stringent quality control while retaining most genotyped individuals. On the other hand, our findings are limited by the absence of a placebo-comparison group, which makes it impossible to distinguish between genetic predictors of nonspecific improvement and effects that are common to both antidepressants used in the GENDEP project. Across the four analyses, only one pharmacogenetic association was established at a genome-wide level of significance. The predominance of negative findings could be the result of limited power to detect weak to moderate associations or of the absence of even moderately strong common genetic determinants of antidepressant drug response. A meta-analysis of multiple large samples will be needed to establish the generalizability of findings reported in the present study.

The pharmacogenetic analysis of GENDEP has brought up a number of promising results, including two regions containing structural variants, a genome-wide significant association with nortriptyline response, and a functionally plausible marker associated with response to escitalopram. These results demonstrate the feasibility of genome-wide pharmacogenetic analyses with well-characterized moderately large samples. As in all genetic studies, replications will be required before the value of any particular finding can be fully assessed, but genome-wide studies may thus be important steps in the elucidation of the genetic basis of pharmacological response.

Received July 4, 2009; revision received Sept. 24, 2009; accepted Oct. 22, 2009. From the Medical Research Council Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London; the Laboratory of Psychiatric Genetics, Department of Psychiatry, Poznan University of Medical Sciences, Poznan, Poland; the Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia; the Department of Psychiatry, University of Bonn, Bonn, Germany; the Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark; the Biological Psychiatry Unit and Dual Diagnosis Ward, Istituto Di Ricovero e Cura a Carattere Scientifico, Centro San Giovanni di Dio, Fatebenefratelli, Brescia, Italy; the Central Institute of Mental Health, Division of Genetic Epidemiology in Psychiatry, Mannheim, Germany; the Laboratoire de Psychologie Médicale, Université Libre de Bruxelles and Psy Pluriel, Centre Européen de Psychologie Médicale, Brussels; the Institute of Public Health of the Republic of Slovenia, Ljubljana, Slovenia; the Department of Molecular and Biomedical Sciences, Jozef Stefan Institute, Ljubljana, Slovenia; the Clinic for Chronic Depression, Aarhus University Hospital, Risskov, Denmark; the Neurosciences Centre of Excellence in Drug Discovery, Glaxo­SmithKline R and D, Verona, Italy; the Department of Psychiatry, University of Toronto, Toronto; Computational Biology, GlaxoSmithKline Pharmaceuticals, Stevenage, Hertfordshire, United Kingdom; the Centre National de Genotypage, Evry Cedex, France. Address correspondence and reprint requests to Dr. Uher, P080 SGDP, Institute of Psychiatry, De Crespigny Park, SE5 8AF, London, United Kingdom; (e-mail).

Dr. Henigsberg has participated in clinical trials supported by GlaxoSmithKline and Lundbeck; he has also received honoraria from Lundbeck. Dr. Souery serves on the national advisory boards for AstraZeneca, Bristol-Myers Squibb, Eli Lilly, and Lundbeck. Drs. Muglia and Barnes are employed by GlaxoSmithKline. Drs. Farmer, Aitchison, and McGuffin have received consulting fees and honoraria from Glaxo­SmithKline and Lundbeck. Drs. Uher, Perroud, Ng, Hauser, Maier, Mors, Placentino, Rietschel, Žagar, Czerski, Larsen, Schulze, Zobel, Cohen-Woods, Butler, Lathrop, Breen, Craig, and Lewis and Mr. Jerman and Ms. Pirlo report no financial relationships with commercial interests.

The GENDEP project was supported by a European Commission Framework 6 grant (contract reference: LSHB-CT-2003-503428). Lundbeck provided nortriptyline and escitalopram free of charge for the project. GlaxoSmithKline, the Biomedical Research Centre for Mental Health at the Institute of Psychiatry, King's College London and South London, and the Maudsley National Health Service Foundation Trust (supported by the National Institute for Health Research, Department of Health, United Kingdom) provided support for add-on projects at the London recruitment center. The Medical Research Council, United Kingdom, and GlaxoSmithKline (G0701420) provided support for genotyping. The European Commission Framework, Lundbeck, GlaxoSmithKline, Biomedical Research Centre for Mental Health, Maudsley National Health Service Foundation, and Medical Research Council had no role in the design, conduct, data collection, analysis, interpretation, or writing of this study.

EudraCT clinical trials registry number, 2004–001723-38 (http://eudract.emea.europa.eu) and International Standard Randomized Controlled Trials registry number, ISRCTN 03693000 (http://www.controlled-trials.com).

The authors thank the following individuals: Maja Bajs, Mara Barreto, Cristian Bonvicini, Desmond Campbell, Elzbieta Cegielska, Monika Dmitrzak-Weglarz, Amanda Elkin, Caterina Giovannini, Joanna M. Gray, Cerisse Gunasinghe, Bhanu Gupta, Susanne Höfels, Petra Kalember, Pawel Kapelski, Zrnka Kovacic, Dejan Kozel, Anna Leszczynska-Rodziewicz, Sylvie Linotte, Andrej Marusic, Julien Mendlewicz, Metka Paragi, Laura Pedrini, Jorge Perez, Ute Pfeiffer, Aleksandra Rajewska-Rager, Luciana Rillosi, Christine Schmäl, Anna Schuhmacher, Maria Skibinska, Rebecca Smith, Anne Schinkel Stamp, Jana Strohmaier, Jerneja Sveticic, Aleksandra Szczepankiewicz, Alenka Tancic, Sandra Weber, and Richard J. Williamson.

References

1 Franchini L , Serretti A , Gasperini M , Smeraldi E : Familial concordance of fluvoxamine response as a tool for differentiating mood disorder pedigrees. J Psychiatr Res 1998; 32:255–259 CrossrefGoogle Scholar

2 Kato M , Serretti A : Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol Psychiatry (Epub ahead of print, Nov 4, 2008) Google Scholar

3 Uher R , Huezo-Diaz P , Perroud N , Smith R , Rietschel M , Mors O , Hauser J , Maier W , Kozel D , Henigsberg N , Barreto M , Placentino A , Dernovsek MZ , Schulze T , Kalember P , Zobel A , Czerski P , Larsen ER , Souery D , Govannini C , Gray JM , Lewis CM , Farmer A , Aitchison KJ , McGuffin P , Craig I : Genetic predictors of antidepressant response in the GENDEP project. Pharmacogenomics J 2009; 9:225–233 Crossref, MedlineGoogle Scholar

4 Uher R , Maier W , Hauser J , Marusic A , Schmael C , Mors O , Henigsberg N , Souery D , Placentino A , Rietschel M , Zobel A , Dmitrzak-Weglarz M , Petrovic A , Jorgensen L , Kalember P , Giovannini C , Barreto M , Elkin A , Landau S , Farmer A , Aitchison KJ , McGuffin P : Differential efficacy of escitalopram and nortriptyline on dimensional measures of depression. Br J Psychiatry 2009; 194:252–259 Crossref, MedlineGoogle Scholar

5 Wing JK , Sartorius N , Ustin TB : Diagnosis and Clinical Measurement in Psychiatry: A Reference Manual for SCAN. Geneva, World Health Organization, 1998 Google Scholar

6 Sanchez C , Bergqvist PB , Brennum LT , Gupta S , Hogg S , Larsen A , Wiborg O : Escitalopram, the S–(+)–enantiomer of citalopram, is a selective serotonin reuptake inhibitor with potent effects in animal models predictive of antidepressant and anxiolytic activities. Psychopharmacology (Berl) 2003; 167:353–362 Google Scholar

7 Sanchez C , Hyttel J : Comparison of the effects of antidepressants and their metabolites on reuptake of biogenic amines and on receptor binding. Cell Mol Neurobiol 1999; 19:467–489 Crossref, MedlineGoogle Scholar

8 Uher R , Farmer A , Maier W , Rietschel M , Hauser J , Marusic A , Mors O , Elkin A , Williamson RJ , Schmael C , Henigsberg N , Perez J , Mendlewicz J , Janzing JG , Zobel A , Skibinska M , Kozel D , Stamp AS , Bajs M , Placentino A , Barreto M , McGuffin P , Aitchison KJ : Measuring depression: comparison and integration of three scales in the GENDEP study. Psychol Med 2008; 38:289–300 Crossref, MedlineGoogle Scholar

9 Huezo-Diaz P , Uher R , Smith R , Rietschel M , Henigsberg N , Marusic A , Mors O , Maier W , Hauser J , Souery D , Placentino A , Zobel A , Larsen EJ , Czerski PM , Gupta G , Hoda F , Perroud N , Farmer A , Craig I , Aitchison KJ , McGuffin P : Moderation of antidepressant response by the serotonin transporter gene. Br J Psychiatry 2009; 195:30–38 Crossref, MedlineGoogle Scholar

10 Perroud N , Aitchison KJ , Uher R , Smith R , Huezo-Diaz P , Marusic A , Maier W , Mors O , Placentino A , Henigsberg N , Rietschel M , Hauser J , Souery D , Kapelski P , Bonvicini C , Zobel A , Jorgensen L , Petrovic A , Kalember P , Schulze TG , Gupta B , Gray JM , Lewis CM , Farmer AE , McGuffin P , Craig I : Genetic predictors of increase in suicidal ideation during antidepressant treatment in the GENDEP project. Neuropsychopharmacology 2009; 34:2517–2528 Crossref, MedlineGoogle Scholar

11 Montgomery SA , Asberg M : A new depression scale designed to be sensitive to change. Br J Psychiatry 1979; 134:382–389 Crossref, MedlineGoogle Scholar

12 Mulder RT , Joyce PR , Frampton C : Relationships among measures of treatment outcome in depressed patients. J Affect Disord 2003; 76:127–135 Crossref, MedlineGoogle Scholar

13 Uher R , Muthen B , Souery D , Mors O , Jaracz J , Placentino A , Petrovic A , Zobel A , Henigsberg N , Rietschel M , Aitchison KJ , Farmer A , McGuffin P : Trajectories of change in depression severity during treatment with antidepressants. Psychol Med (Epub ahead of print, Oct 29, 2009) MedlineGoogle Scholar

14 Streiner DL : Breaking up is hard to do: the heartbreak of dichotomizing continuous data. Can J Psychiatry 2002; 47:262–266 Crossref, MedlineGoogle Scholar

15 Leucht S , Kane JM , Etschel E , Kissling W , Hamann J , Engel RR : Linking the PANSS, BPRS, and CGI: clinical implications. Neuropsychopharmacology 2006; 31:2318–2325 Crossref, MedlineGoogle Scholar

16 Lane P : Handling drop-out in longitudinal clinical trials: a comparison of the LOCF and MMRM approaches. Pharm Stat 2008; 7:93–106 Crossref, MedlineGoogle Scholar

17 Freeman B , Smith N , Curtis C , Huckett L , Mill J , Craig IW : DNA from buccal swabs recruited by mail: evaluation of storage effects on long-term stability and suitability for multiplex polym­erase chain reaction genotyping. Behav Genet 2003; 33:67–72 Crossref, MedlineGoogle Scholar

18 Purcell S , Neale B , Todd-Brown K , Thomas L , Ferreira MA , Bender D , Maller J , Sklar P , De Bakker PI , Daly MJ , Sham PC : PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007; 81:559–575 Crossref, MedlineGoogle Scholar

19 Bacanu SA , Devlin B , Roeder K : Association studies for quantitative traits in structured populations. Genet Epidemiol 2002; 22:78–93 Crossref, MedlineGoogle Scholar

20 Dudbridge F , Gusnanto A : Estimation of significance thresholds for genomewide association scans. Genet Epidemiol 2008; 32:227–234 Crossref, MedlineGoogle Scholar

21 Pe'er I , Yelensky R , Altshuler D , Daly MJ : Estimation of the multiple testing burden for genomewide association studies of nearly all common variants. Genet Epidemiol 2008; 32:381–385 Crossref, MedlineGoogle Scholar

22 Benjamini Y , Hochberg Y : Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B 1995; 57:289–300 Google Scholar

23 Storey JD , Tibshirani R : Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 2003; 100:9440–9445 Crossref, MedlineGoogle Scholar

24 Redon R , Ishikawa S , Fitch KR , Feuk L , Perry GH , Andrews TD , Fiegler H , Shapero MH , Carson AR , Chen W , Cho EK , Dallaire S , Freeman JL , Gonzalez JR , Gratacos M , Huang J , Kalaitzopoulos D , Komura D , MacDonald JR , Marshall CR , Mei R , Montgomery L , Nishimura K , Okamura K , Shen F , Somerville MJ , Tchinda J , Valsesia A , Woodwark C , Yang F , Zhang J , Zerjal T , Zhang J , Armengol L , Conrad DF , Estivill X , Tyler-Smith C , Carter NP , Aburatani H , Lee C , Jones KW , Scherer SW , Hurles ME : Global variation in copy number in the human genome. Nature 2006; 444:444–454 Crossref, MedlineGoogle Scholar

25 Cartegni L , Wang J , Zhu Z , Zhang MQ , Krainer AR : ESEfinder: a web resource to identify exonic splicing enhancers. Nucleic Acids Res 2003; 31:3568–3571 Crossref, MedlineGoogle Scholar

26 Muglia P , Tozzi F , Galwey NW , Francks C , Upmanyu R , Kong XQ , Antoniades A , Domenici E , Perry J , Rothen S , Vandeleur CL , Mooser V , Waeber G , Vollenweider P , Preisig M , Lucae S , Muller-Myhsok B , Holsboer F , Middleton LT , Roses AD : Genome-wide association study of recurrent major depressive disorder in two European case-control cohorts. Mol Psychiatry (Epub ahead of print, Dec 23, 2008) Google Scholar

27 Sullivan PF , de Geus EJ , Willemsen G , James MR , Smit JH , Zandbelt T , Arolt V , Baune BT , Blackwood D , Cichon S , Coventry WL , Domschke K , Farmer A , Fava M , Gordon SD , He Q , Heath AC , Heutink P , Holsboer F , Hoogendijk WJ , Hottenga JJ , Hu Y , Kohli M , Lin D , Lucae S , Macintyre DJ , Maier W , McGhee KA , McGuffin P , Montgomery GW , Muir WJ , Nolen WA , Nothen MM , Perlis RH , Pirlo K , Posthuma D , Rietschel M , Rizzu P , Schosser A , Smit AB , Smoller JW , Tzeng JY , van Dyck R , Verhage M , Zitman FG , Martin NG , Wray NR , Boomsma DI , Penninx BW : Genome-wide association for major depressive disorder: a possible role for the presynaptic protein piccolo. Mol Psychiatry 2009; 14:359–375 Crossref, MedlineGoogle Scholar

28 Ising M , Lucae S , Binder EB , Bettecken T , Uhr M , Ripke S , Kohli MA , Hennings JM , Horstmann S , Kloiber S , Menke A , Bondy B , Rupprecht R , Domschke K , Baune BT , Arolt V , Rush AJ , Holsboer F , Muller-Myhsok B : A genomewide association study points to multiple loci that predict antidepressant drug treatment outcome in depression. Arch Gen Psychiatry 2009; 66:966–975 Crossref, MedlineGoogle Scholar

29 Akita K , von Holst A , Furukawa Y , Mikami T , Sugahara K , Faissner A : Expression of multiple chondroitin/dermatan sulfotransferases in the neurogenic regions of the embryonic and adult central nervous system implies that complex chondroitin sulfates have a role in neural stem cell maintenance. Stem Cells 2008; 26:798–809 Crossref, MedlineGoogle Scholar

30 Ishii M , Maeda N : Oversulfated chondroitin sulfate plays critical roles in the neuronal migration in the cerebral cortex. J Biol Chem 2008; 283:32610–32620 Crossref, MedlineGoogle Scholar

31 Duman RS , Monteggia LM : A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006; 59:1116–1127 Crossref, MedlineGoogle Scholar

32 Goi T , Rusanescu G , Urano T , Feig LA : Ral-specific guanine nucleotide exchange factor activity opposes other Ras effectors in PC12 cells by inhibiting neurite outgrowth. Mol Cell Biol 1999; 19:1731–1741 Crossref, MedlineGoogle Scholar

33 Anney RJ , Lasky-Su J , O'Dushlaine C , Kenny E , Neale BM , Mulligan A , Franke B , Zhou K , Chen W , Christiansen H , Rias-Vasquez A , Banaschewski T , Buitelaar J , Ebstein R , Miranda A , Mulas F , Oades RD , Roeyers H , Rothenberger A , Sergeant J , Sonuga-Barke E , Steinhausen H , Asherson P , Faraone SV , Gill M : Conduct disorder and ADHD: evaluation of conduct problems as a categorical and quantitative trait in the International Multicentre ADHD Genetics Study. Am J Med Genet B Neuropsychiatr Genet 2008; 147B:1369–1378 Crossref, MedlineGoogle Scholar

34 Bull SJ , Huezo-Diaz P , Binder EB , Cubells JF , Ranjith G , Maddock C , Miyazaki C , Alexander N , Hotopf M , Cleare AJ , Norris S , Cassidy E , Aitchison KJ , Miller AH , Pariante CM : Functional polymorphisms in the interleukin-6 and serotonin transporter genes, and depression and fatigue induced by interferon-alpha and ribavirin treatment. Mol Psychiatry 2009; 14:1095–1104 Crossref, MedlineGoogle Scholar

35 Watanabe D , Yoshimura R , Khalil M , Yoshida K , Kishimoto T , Taga T , Kiyama H : Characteristic localization of gp130 (the signal-transducing receptor component used in common for IL–6/IL–11/CNTF/LIF/OSM) in the rat brain. Eur J Neurosci 1996; 8:1630–1640 Crossref, MedlineGoogle Scholar

36 Rudge JS , Eaton MJ , Mather P , Lindsay RM , Whittemore SR : CNTF induces raphe neuronal precursors to switch from a serotonergic to a cholinergic phenotype in vitro. Mol Cell Neurosci 1996; 7:204–221 Crossref, MedlineGoogle Scholar

37 Danese A , Moffitt TE , Pariante CM , Ambler A , Poulton R , Caspi A : Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Arch Gen Psychiatry 2008; 65:409–415 Crossref, MedlineGoogle Scholar

38 Uher R : The implications of gene-environment interactions in depression: Will cause inform cure? Mol Psychiatry 2008; 13:1070–1078 Crossref, MedlineGoogle Scholar

39 Miller AH , Maletic V , Raison CL : Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry 2009; 65:732–741 Crossref, MedlineGoogle Scholar

40 Eisensamer B , Rammes G , Gimpl G , Shapa M , Ferrari U , Hapfelmeier G , Bondy B , Parsons C , Gilling K , Zieglgansberger W , Holsboer F , Rupprecht R : Antidepressants are functional antagonists at the serotonin type 3 (5-HT3) receptor. Mol Psychiatry 2003; 8:994–1007 Crossref, MedlineGoogle Scholar

41 Eisensamer B , Uhr M , Meyr S , Gimpl G , Deiml T , Rammes G , Lambert JJ , Zieglgansberger W , Holsboer F , Rupprecht R : Antidepressants and antipsychotic drugs colocalize with 5-HT3 receptors in raft-like domains. J Neurosci 2005; 25:10198–10206 Crossref, MedlineGoogle Scholar