Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jun 1.
Published in final edited form as: J Affect Disord. 2017 Jul 5;233:3–14. doi: 10.1016/j.jad.2017.07.001

Peripheral Biomarkers of Major Depression and Antidepressant Treatment Response: Current Knowledge and Future Outlooks

Bharathi S Gadad 1, Manish K Jha 1, Andrew Czysz 1, Jennifer L Furman 1, Taryn L Mayes 1, Michael P Emslie 1, Madhukar H Trivedi 1,#
PMCID: PMC5815949  NIHMSID: NIHMS941050  PMID: 28709695

Abstract

Background

In recent years, we have accomplished a deeper understanding about the pathophysiology of major depressive disorder (MDD). Nevertheless, this improved comprehension has not translated to improved treatment outcome, as identification of specific biologic markers of disease may still be crucial to facilitate a more rapid, successful treatment. Ongoing research explores the importance of screening biomarkers using neuroimaging, neurophysiology, genomics, proteomics, and metabolomics measures.

Results

In the present review, we highlight the biomarkers that are differentially expressed in MDD and treatment response and place a particular emphasis on the most recent progress in advancing technology which will continue the search for blood-based biomarkers.

Limitations

Due to space constraints, we are unable to detail all biomarker platforms, such as neurophysiological and neuroimaging markers, although their contributions are certainly applicable to a biomarker review and valuable to the field.

Conclusions

Although the search for reliable biomarkers of depression and/or treatment outcome is ongoing, the rapidly-expanding field of research along with promising new technologies may provide the foundation for identifying key factors which will ultimately help direct patients toward a quicker and more effective treatment for MDD.

Keywords: depression, biomarkers, biosignatures, genomics, proteomics, metabolomics

1. Introduction

Major Depressive Disorder (MDD) is a prevalent psychiatric disorder associated with varied prognosis, chronic course, and duration of illness with reduced quality of life (Beck et al., 1961; Burton et al., 2015; Daly EJ, 2010). Most MDD patients stay on ineffective medications for too long, switch treatments too early, or simply drop out of care (Burton et al., 2015; Rush et al., 2008; Warden et al., 2007b). Compared to treatment of several other somatic diseases, antidepressant response rates are low, duration to attain therapeutic benefit is long, and treatment-emergent side effect burden is significant (Rush et al., 2011; Trivedi et al., 2006b; Warden et al., 2007a). Furthermore, treatments are selected not based on efficacy, but instead on patient or provider preferences. The factors that ultimately drive these decisions include cost, side effects, tolerability, and/or response during previous episode(s) (Meron et al., 2015). Unlike other specialty fields of medicine, such as breast cancer (Dowsett and Dunbier, 2008), asthma (Lima et al., 2009), macular degeneration (Lee et al., 2009), and multiple sclerosis (Vosslamber et al., 2009), there are no validated biomarkers for depression, thereby stalling the goal of offering precise, targeted treatment for this devastating disorder. Indeed, personalized treatment has the capacity to maximize the likelihood of treatment response or remission, while simultaneously minimizing detrimental side effects (Kessler et al., 2003; Murray et al., 2013).

The search for biomarkers is hindered by the heterogeneity of MDD (Hasler et al., 2004) and the limitation of its current diagnostic categories such as self-reports, measurement based scales, with a lack of understanding of the molecular blood testing compared to other diseases (Insel et al., 2010a). In clinical practice, efforts are made to understand the demographic features, (e.g., gender (Young et al., 2009), race (Friedman et al., 2009), employment status (Warden et al., 2007a)), illness characteristics (e.g., baseline severity of depression (Friedman et al., 2012), duration of illness (Rush et al., 2012), number of previous episodes (Trivedi et al., 2005), age of onset (Zisook et al., 2007), family history of mood disorders (Trivedi et al., 2005), presence of anxious features (Fava et al., 2008), depression symptoms and its subtypes (Friedman et al., 2009), co-morbid psychiatric disorders (Friedman et al., 2009), psychosocial functioning (Vittengl et al., 2009), and social factors (e.g., marital status (Trivedi et al., 2005), level of social support (Lesser et al., 2008), social status (Lesser et al., 2008)). Unfortunately, these have proven to be of limited utility due to the knowledge gap regarding cellular and molecular pathophysiology, blood tests, and events that occur during brain development and maturation in MDD. (Arnow et al., 2015; Bobo et al., 2011; Chan et al., 2012; Sung et al., 2012; Sung et al., 2013; Sung et al., 2015). The underlying biological factors that drive MDD may be better suited to serve as biomarkers for guiding personalized medicine, as they are objective and can be measured externally (2001; Strimbu and Tavel, 2010). The heterogeneity of MDD necessitates and/or allows for numerous biomarker classifications, as shown in Figure 1. Diagnostic biomarkers indicate presence and/or future development of disease. Most of the currently-identified biomarkers, identified below, are predictive, such that baseline levels will provide insight as to whether or not a patient will respond to treatment. Moderators are also characterized at baseline, though provide more detailed information, such that clinicians can predict how a patient will respond to a particular treatment. Mediators define markers that change following treatment initiation and may predict future performance with the same or alternative treatment methodology. To maximize the chances of success, we may also need to go beyond individual biomarkers and venture towards generating multidimensional biomarkers (i.e., biosignatures) by systematically evaluating combinations of both clinical and biological markers.

Figure 1. Biomarkers of major depression.

Figure 1

Biomarkers identified before treatment initiation are classified as diagnostic, predictive, or moderators. Diagnostic markers classify an MDD patient, predictive markers determine overall likelihood of response/remission, and moderators determine likelihood of response/remission with a particular treatment. Mediators are biomarkers collected soon after treatment initiation and help predict overall likelihood of response/remission. Long-term treatment response may also be indicative of ultimate outcome

In this report, we briefly review currently available treatment for depression, though emphasize the necessity for biomarker identification to discriminate depression subtypes and work toward personalized medicine. We present the tools available for biomarker discovery and discuss what these technologies have identified as hits to date. In addition, we discuss our own clinical trial study, EMBARC (Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care), which is exclusively designed to screen numerous putative biomarkers with the aim to identify biosignatures for depression response.

2. Antidepressant Treatment Strategies

Numerous modalities are available to treat individuals with depression. Unfortunately, no treatment is universally effective, although different molecules and neural circuits are targeted, promoting distinct physiological changes. Pharmacological medications continue to be the most commonly-recommended first-line treatment for MDD (Olfson and Marcus, 2009). While there are several ADM classes like selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), monoamine oxidase inhibitors (MAOIs), and others (bupropion and mirtazapine), all similarly target monoamine neurotransmission (Ball et al., 2014; Feighner, 1999; Lin et al., 2011; Saragoussi et al., 2012). Despite the variety of molecular targets, two thirds of MDD patients fail to achieve remission after initial treatment, and almost one third fail to achieve remission even after four consecutive treatment trials (McGrath et al., 2006; Rush et al., 2006a; Rush et al., 2006b; Trivedi et al., 2006a; Warden et al., 2007a).

Outside of the widely-prescribed pharmacological therapies, alternative treatment strategies instead employ indirect mechanisms which may still affect brain physiology, such as psychotherapy, exercise, and somatic treatments. Although their central mechanism(s) of action remain largely unknown, each has demonstrated efficacy in clinical populations. For example, individual or group psychotherapy sessions (e.g., including cognitive-behavioral therapy (CBT), interpersonal therapy (IPT), and behavioral activation) show efficacy in treating depression (Craighead and Dunlop, 2014). Physical activity, including aerobic, anaerobic, and mindfulness ameliorates depressive symptomatology following both acute and chronic sessions. This is demonstrated in numerous studies, although it is important to point out that results are not always consistent, likely due to the heterogeneity of participants and treatment design (Blumenthal et al., 2012; Bridle et al., 2012; Rethorst and Trivedi, 2013; Silveira et al., 2013). Lastly, somatic treatments, including electroconvulsive therapy (ECT), repetitive transcranial magnetic stimulation (rTMS), and vagus nerve stimulation (VNS) have evidence of some efficacy, though its use is often restricted to patients with treatment-resistant or moderate-to-severe depression (Meron et al., 2015).

In each case, these treatment options have demonstrated benefit alone or as an augmentation therapy to previously-described ADMs. The problem persists, however, that even by combining medications or treatment strategies, depressed patients frequently do not achieve response of remission. Discovery of biomarkers will help identify a personalized treatment strategy for each patient and thereby assist with quick and efficacious responsiveness.

3. Biomarker Discovery—Tools and Application

Technological advances over the last few decades has fueled the search for biomarkers which may predict individual response to particular antidepressant treatment strategies. In this section we detail the advanced methodologies with a particular focus on the strategies which enable screening of “Omics” biomarkers. Figure 2 denotes the cascade of events necessary for identifying a biomarker, including discovery and validation processing using high- and low-throughput methodology, respectively. These approaches hold promise, as they enable study of a wide variety of biological processing, ranging from genetic composition to protein breakdown, and any biological entity in between. Below we will review the methodological design and tools for pharmacogenomics, epigenomics, transcriptomics, proteomics, and metabolomics and provide examples of their employment thus far:

Figure 2. Tools and Technologies for the development biomarker candidates.

Figure 2

Biomarker consists of four main phases- discovery, qualification, verification and validation. The tools and associated technologies are listed for pharmacogenomics, epigenetics, transcriptomics, proteomics and metabolomics. “Analytes” and “Samples” refer to the number of different protein targets or samples, respectively that are evaluated in each phase. LC-MS/MS, liquid chromatography tandem mass spectrometry; NGS, Next generation sequencing; PCR, Polymerase chain reaction.

3.1. Pharmacogenomics, Epigenomics and Transcriptomics

Genomics enables the identification of one’s genetic makeup and post-translational modifications, ultimately providing insight regarding a target’s structure and function. Standard large scale genome-wide association studies (GWAS) as well newer, next-generation technologies will serve at the forefront of identifying genetic biomarkers. Large clinical trials [STAR*D (n = 1953) (Garriock et al., 2010), MARS (n = 339) (Ising et al., 2009), GENDEP (n= 706) (Uher et al., 2010), and PGRN-AMPS (n= 529) (Ji et al., 2013)] are harnessing the power of pharmacogenomics to help identify predictors of depression and/or treatment response.

To date, single nucleotide polymorphism (SNP) identification provides the longest list of potential hits in MDD research and treatment response. Current technology enables genotyping of 500,000 to 2.5 million SNPs across the genome (Lohoff, 2010), without a requirement for pre-selection of analytes. Thus, SNP research with GWAS helps to identify new and unbiased pathways involved in mood disorders (Lohoff, 2010). Notable SNP genotyping studies include: Sequenced Treatment Alternatives to Relieve Depression (STAR*D) (Garriock et al., 2010), the Munich Antidepressant Response Signature (MARS) (Ising et al., 2009), the Genome-based Therapeutic Drugs for Depression (GENDEP) (Uher et al., 2010), and the Mayo Clinic Pharmacogenomics Research Network Antidepressant Medication Pharmacogenomics Study (PGRN-AMPS) (Ji et al., 2013), and the ‘1000’ Genomes (Abo et al., 2012). While studies have failed to detect any particular gene of significant in predicting antidepressant response, numerous SNP hits have been identified which mediate several aspects of depression mechanisms and medication metabolism:

  1. Drug Absorption:

    Genetic polymorphisms in the multidrug-resistance gene (MDR1) are associated with both positive (Dong et al., 2009; Gex-Fabry et al., 2008; Kato et al., 2008; Nikisch et al., 2008; Uhr et al., 2008) and negative (Laika et al., 2006; Mihaljevic Peles et al., 2008; Peters et al., 2008) treatment outcomes. P-glycoprotein (P-gp) is the gene product of MDR1, resides at the blood brain barrier, and affects absorption of antidepressants. Thus, antidepressants which are substrates of P-gp (e.g., citalopram, venlafaxine or paroxetine) are particularly susceptible to producing differential treatment outcomes.

  2. Neurotransmitter Transport and Transmission:

    Serotonin: Given the imbalance of monoamine levels associated with MDD, the serotonin (5HT) transporter and receptors have unsurprisingly been studied in numerous trials as a predictor of MDD risk and treatment outcome. Regarding the 5HT transporter, while no significant associations were initially identified with STAR*D participants (Kraft et al., 2007), follow-up analyses have shown differential treatment outcomes associated with the serotonin transporter linked polymorphic region (5-HTTLPR) of the SLC6A4 gene, notably in across races in response to treatment with SSRIs (Mrazek et al., 2009), although other groups have not been able to replicate these data several studies failed to find any association of 5-HTTLPR (Maron et al., 2009; Perlis et al., 2010; Serretti et al., 2013). Discovery and validation of SNPs in the serotonin receptors have been equally as complex. Data from both STAR*D (McMahon et al., 2006; Peters et al., 2009) and MARS (Lucae et al., 2010) indicate an association between the rs7997012 SNP of the serotonin receptor 2A gene and treatment outcome. However, other groups were unable to replicate this finding but found additional polymorphisms relating to treatment outcome (Horstmann et al., 2010; Uher et al., 2009).

    Dopamine: A SNP at codon 158 of Catechol –O- Methyl Transferase (COMT) gene (rs4680) results in a valine-methionine substitution (i.e. met/met genotype) and is associated with decreased COMT activity (Chen et al., 2004; Lachman et al., 1996). COMT is the main catalytic enzyme of dopamine in the brain (Gogos et al., 1998; Kaenmaki et al., 2010; Sesack et al., 1998), and thus, this SNP has been reported to have favorable outcomes with antidepressants in some studies (Baune et al., 2008; Benedetti et al., 2009; Benedetti et al., 2010; Kocabas et al., 2010; Spronk et al., 2011; Tsai et al., 2009), but not others (Arias et al., 2006; Perlis et al., 2009; Serretti et al., 2013; Szegedi et al., 2005). In contrast, treatment-resistant patients with the val/val genotype higher response with ECT (Anttila et al., 2007), which was replicated in female patients (Domschke et al., 2010).

    Glutamate receptor: Polymorphisms in glutamate ionotropic kainite receptor (GRIK) 4 gene were shown to be associated with treatment response in samples from STAR*D (Paddock et al., 2007), MARS (Horstmann et al., 2010) and others (Pu et al., 2013). Specifically, the rs1954787 SNP was most robust in STAR*D and was associated with the same directionality with the MARS and Chinese Han trials. However, other groups have failed to replicate these associations (Perlis et al., 2010; Serretti et al., 2012).

  3. Monoamine Metabolism:

    Type A Monoamine Oxidase-A (MAO-A) catabolizes monoamine transmitters, serotonin, norepinephrine, and dopamine, and therefore is a candidate player in the onset, progression, and treatment of mood disorders, including depression. Combined with decreased levels of serotonin and norepinephrine, a genetic polymorphism which increases MAO-A expression is proposed as a key factor associated with MDD (Naoi et al., 2017). Further evidence indicates MAO-A as a promising biomarker; the SNP-rs6323 is predictive of treatment response with mirtazapine in women with bipolar disorder (Tadic et al., 2007). Studies examining fluoxetine and bupropion in Mexican and Caucasian populations, however, have shown no association with outcome (Peters et al., 2004; Tiwari et al., 2013).

    Cytochrome P450 enzymes (CYPs) expression and function are altered by monoaminergic neurotransmission. Commercially-available kits enable identification of SNPs in P450 enzymes which may be used to classify individuals as extensive metabolizers, intermediate metabolizers, poor metabolizers or ultra-rapid metabolizers (Porcelli et al., 2011). While these polymorphisms can be helpful in predicting adverse effects due to drug metabolism (D’Empaire et al., 2011; Lim et al., 2014; Porcelli et al., 2011), their association with SSRI treatment response in STAR*D (Mihaljevic Peles et al., 2008; Peters et al., 2008), GENDEP (Hodgson et al., 2014; Hodgson et al., 2015) and other studies (Grasmader et al., 2004; Serretti et al., 2009; Shams et al., 2006) has at best been either negative or weakly positive (Lobello et al., 2010).

  4. Immune-Regulation

    FK506 binding protein 5 (FKBP5): The FKBP5 (rs1360780) gene, and particularly its epigenetic variants, is strongly associated with morphological brain changes in regions regulating emotions (Han et al., 2017) and displays a predisposition to developing MDD. FKBP5 genetic variants (including rs1360780, rs4713916, and rs3800373) have also shown a strong link in Caucasian subjects with various antidepressants treatments (Binder et al., 2004), which has been replicated by some groups (Binder et al., 2008; Gawlik et al., 2006). Following treatment with escitalopram or duloxetine, no association is observed with genetic variation and response (Perlis et al., 2010; Uher et al., 2009).

  5. Multiple single nucleotide polymorphisms:

    Given the discordance among studies investigating individual SNPs, an alternative approach is to investigate the interactive role of multiple SNPs. In a cohort of MDD outpatients of Asian ethnicity, Lim et al. found that responsiveness to SSRI could be predicted with 87% accuracy with a model incorporating SNPs of GRIK 2 (rs543196) and glutamate decarboxylase 1 (rs3828275 of GAD1), and haplotypes of tryptophan hydroxylase 2 (TPH2) and 5-HTTLPR (Lim et al., 2014). Their model was not accurate for predicting response to non-SSRI medications, however, supporting the notion that multiple SNP analyses could predict differential response to antidepressants.

Other genomics tools, including epigenetic modification, whole exome/gene sequencing, and transcriptomics/RNA Seq (i.e., “next generation sequencing”) are relatively new technologies, and thus their employment with MDD biomarker research is limited. However, early preliminary studies with MDD diagnostics suggest their feasibility and applicability:

Epigenetics refers to the study of heritable phenotypic traits which occur without any alterations in DNA sequence (Berger et al., 2009), such as post translational modification of histones, DNA methylation, and/or microRNA expression. Epigenetics has been used recently in depression research, both in clinical trials and in animal model studies (Maze et al., 2014; Oh et al., 2015). Oh et al. conducted DNA modification analysis in white blood cells from monozygotic twins discordant for MDD, in brain prefrontal cortex with MDD, and control subjects (total n = 304) using microarray fine mapping. Domschke et al. observed a higher methylation status of the SLC6A4 gene in MDD patients who showed a better response following 6 weeks treatment with escitalopram (Domschke et al., 2014). Epigenome-wide association studies (EWAS) have shown that the tricyclic antidepressants, amitriptyline and imipramine, as well as the SSRI paroxetine, reduced DNA methylation in rat primary astrocytes (Menke and Binder, 2014). Lopez et al. found that reduced histone H3 lysine 27 trimethylation (H3K27me3) significantly correlated with improvement in depressive symptoms and peripheral blood BDNF mRNA levels (Lopez et al., 2013). Thus, while the study of predictive biomarkers is still developing, preliminary studies demonstrate an association of epigenetic modifications with MDD.

Immediately downstream of epigenetics is transcriptomics, the study of gene transcription, which may help to identify pathways associated with the pathophysiology of mood disorders by examining gene expression and novel splice transcripts. For example, a preliminary study by Jansen et al. demonstrated strong gene expression differences between current MDD and control participants [current MDD (N=882), remitted MDD (N=635) and control (N=331)]. Further, RNA sequencing allowed them to associate the robustly-expressed MDD genes interleukin-6 signaling and natural killer cell pathways (Jansen et al., 2016) In an exploratory study of transcriptomic biomarkers with whole-genome expression of MDD remitters versus non-responders, Hennings et al. initially discovered messenger ribonucleic acid (mRNA) transcripts of interest and then used a replication sample of 142 patients from MARS study (Hennings et al., 2015). They found that lower pre-treatment mRNA levels of retinoid-related orphan receptor alpha (RORa), germinal center expressed transcript 2 (GCET2), and chitinase 3-like protein 2 (CHI3L2) were associated with greater likelihood of antidepressant response.

Pharmacogenomics-to-date has provided an interesting framework for future studies. Cumulative evidence supports the involvement of some genes and molecular pathways in MDD and antidepressant efficacy, notably those involved with monoamine transport and metabolism (SLC6A4, HTR2A, and cytochrome P450 genes). However, many of these hits demonstrate difficulty in validation. Although reasons for this could be due to retrospective analysis of biomarkers, low sample size, variations in data collection, or subjective clinical evaluation, there is a clear need for further characterization before they may be translated into the clinic.

3.2 Proteomics

Following gene transcription, a protein may be studied, either individually or in combination with others. The levels of several proteins, notably inflammatory factors, are implicated with MDD and are increasingly associated with treatment outcome. Proteomics methodology may be performed with a high-throughput discovery setup or targeted quantitation. Common techniques are based on size characterization or antibody/aptamer binding.

Two Dimensional Gel Electrophoresis (2DE) with Mass Spectrometry (MS) is a common methodology available to study the proteome, and has frequently been used with psychiatric disorders such as depression, schizophrenia, and bipolar disorder, as well as in studies of neurodegenerative disorders such as Alzheimer’s disease (Lista et al., 2013; Martins-de-Souza et al., 2010; Schirle et al., 2012). In recent years, using 2D ITRAQ LC-MS with MDD samples, Xu et al. showed that several proteins (e.g., apolipoprotein D, B-100, ceruloplasmin histidine rich glyocprotein, semophorin, and α-2-macroglobulin) are significantly up- or downregulated (Xu et al., 2012). 2DE–MS-based proteomics is not without limitations, however. This technique shows difficulty with detecting proteins that are low-abundance, acidic, basic, or have an extremely high or low molecular weight.

Shotgun Proteomics is an alternative to the direct MS-based approach described above and can separate and identify thousands of proteins in one experiment. The liquid chromatography–tandem mass spectrometry (LC–MS) technique combines chromatographic steps in a high-throughput manner prior to MS analyses. Use of shotgun proteomics with MDD is limited, although it is capable of revealing differentially expressed proteins not found with 2DE methods (Martins-de-Souza et al., 2010; Schirle et al., 2012). Among the few MDD studies, shotgun LC-MS analysis of MDD patient samples with and without psychosis showed significant differences in proteomic profiles (Martins-de-Souza et al., 2010; Schirle et al., 2012). Mass spectrometry proteomics technology carries the beneficial capability of screening of a specimen’s entire proteome, but sacrifices significant quantification power in the process. Furthermore, sample preparation in MS is not well-standardized and data interpretation is complicated due to the sheer amount of proteins assayed. As such, this technique requires more characterization before a reliable biomarker may be identified and translated to the clinic.

Multiplex Proteomics

The latest introduction of advanced multiplex luminex based technologies allow measurement of multiple analytes in individual small-volume samples, revolutionizing proteomic analyses. Different platforms include rules based medicine (RBM), bioplexes, meso-scale discovery (MSD), somalogic, and others which are suitable for developing convenient, rapid, sensitive, and specific assays for a wide range of diseases including major depression (Chen and Zhu, 2006; Xu et al., 2012). The technology employs multiplexed dye-coded microspheres, coated with specific capture reagents (antibodies and/or DNA-based aptamers) which may bind numerous analytes within a single biological sample. To date, multiplex technology varies from screening 10–1100 analytes in the same sample. This approach minimizes sampling errors, required sample volume, and cost for assay reagents. Multiplex-based technologies have been implicated in screening for putative biomarkers in Alzheimer’s disease, Parkinson’s disease, cancer, and infectious disorders, as well as for psychiatric disorders like schizophrenia, depression, and bipolar (Chen and Zhu, 2006).

Inflammatory biomarkers, growth factors, and lipids have recently been studied with the rules based medicine (RBM) multiplex discovery platform (Bot et al., 2015; Diniz et al., 2015). Bot et al. conducted studies on 1589 participants from the Netherlands Study of Depression and Anxiety and found differences in protein level across current MDD, remitted MDD, and healthy control participants. The analytes predominantly associated with diverse cell communication and signal transduction processes, immune response, and protein metabolism (Bot et al., 2015). In a subsequent RBM study by Diniz et al., remitted MDD participants displayed differential expression of 24 proteins related to regulation of immune-inflammatory activity, intracellular signaling, cell survival, and protein and lipid homeostasis (Diniz et al., 2015). Several studies have evaluated peripheral blood levels of neurotrophic factors, but ultimately found no evidence for their utility as biomarker(s) of differential antidepressant treatment response (Brunoni et al., 2014; Buttenschon et al., 2015; Gorgulu and Caliyurt, 2009; Matrisciano et al., 2009; Molendijk et al., 2014; Ninan et al., 2014).

Antibody-based assays like western blots or enzyme linked immunosorbent assays (ELISA) have been around much longer than the other described proteomic assays, are very well-optimized, and have significant quantification advantages. ELISA technology may be used to quantify one specific protein or may be multiplexed to analyze several at the same time. They are frequently used in biomarker research and have produced many potential proteins of interest, particularly in inflammatory cascades. So far, meta-analyses have shown serum TNF-α, IL-6 CRP, BDNF and IL-1β levels as consistent proteomic markers associated with MDD and treatment response. Perhaps the most well-characterized inflammatory marker, however, is C-reactive protein (CRP), a marker of global inflammation, which has been reported as a biomarker of treatment response in several studies. Low (<1 mg/mL) baseline levels of CRP successfully identified patients who would respond better to escitalopram than nortriptyline in the GENDEP study (Uher et al., 2014). Correspondingly, higher baseline CRP levels were associated with greater reduction in depression severity with nortriptyline treatment. Raison et al. found that elevated high sensitivity CRP (> 5 mg/ml) at baseline was associated with a significantly greater likelihood of treatment response with infliximab as compared to placebo (Raison et al., 2013). In a more recent study, Jha et al. found that MDD patients with low baseline CRP levels (<1 mg/L) respond better to SSRI monotherapy, and patients with higher levels of CRP respond better to combination therapy of bupropion and SSRI (Jha et al., 2017).

Aside from CRP, ELISAs have been used to quantify other potential biomarkers, including protein p11 in natural killer (NK) cells and monocytes. Svenningsson et al. found that a reduction in p11 after 1 – 2 weeks of citalopram treatment was significantly correlated with subsequent reduction in depression severity (Svenningsson et al., 2014). Levels of many peripheral blood inflammatory cytokines are reduced with SSRI treatment (Hannestad et al., 2011). Janssen et al. found that antidepressants modulate cytokine functioning and directly influence treatment outcome in MDD (Janssen et al., 2010). Further, they showed that antidepressants normalize serum levels of cytokines including interleukin IL-6, IL-1β, tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ). Maes et al. examined the effects of clomipramine, sertraline, and trazadone on the stimulated production of IFN-γ and IL-10, and observed that all three antidepressants significantly increased the IFN-γ/IL-10 ratio (Maes, 2001). Lastly, a recent study by Gadad et al. identified only two of 31 potential inflammatory markers (Eotaxin/CCL11 and IFNγ) that significantly changed pre- to post- antidepressant treatment in the CO-MED study (Gadad et al., 2017, in press; Rush et al., 2011). Interestingly, increased levels of Eotaxin was associated with remission, whereas decreased IFNγ was associated with non-remission. Thus, antibody-based assays certainly provide a feasible platform for biomarker research, although their utility is somewhat hindered by the unavailability of protein-specific antibodies, low throughput, and high cost.

Interestingly, proteomics techniques are commonly combined in biomarker research. Oftentimes, a non-targeted approach (i.e., shotgun or multiplex) is used to identify putative markers, and ELISA or Western blot is used to validate the potential hits. This discovery/validation setup is crucial for biomarker research, as it is necessary to demonstrate that the differences found in a limited set of samples are applicable to a broader cohort. Lee et al. identified 10 proteins that were consistently upregulated or downregulated in MDD (n=5). Validation with ELISA demonstrated consistency with three of these: ceruloplasmin, inter-alpha-trypsin inhibitor heavy chain H4, and complement component 1qC (Lee et al., 2015). In addition, Stelzhammaer et al. used multiplex immunoassays and LC-MS with serum samples from MDD cohorts (first onset and antidepressant drug naïve) and matched controls independently. Results identified several possible biomarkers, including cytokines and interleukins, BDNF, cortisol, angiotensin-converting enzyme, and enzymes participating in the oxidative stress response (Stelzhammer et al., 2014).

Validation/Standardization

As proteomics is the most developed ‘omics platform thus far, its limitations are becoming well-characterized. 2DE, LC-MS, and shotgun or multiplex proteomics/metabolomics are used to evaluate the global expression in an individual biological sample. Although a powerful tool for simultaneously evaluating a wealth of markers, sensitivity and specificity are compromised. With ELISA, although relatively sensitive, attention must be paid closely to the possibility for inconsistent results. Samples are generally run simultaneously to prevent within-study variation, but assay detection can differ widely based on the company or antibody employed. Even within individuals, protein levels are susceptible to change with time of day or fasting status. Thus, for biomarker identification to be universally accepted and implemented in the clinic, all procedures, from collection of specimens to analysis of results, must be standardized. These markers have been consistently been reproduced and validated in several clinical trials.

3.3 Metabolomics

Metabolites are the final products of interactions between gene expression, protein function and the cellular environment (Fernie et al., 2004). Thus, metabolite profiling holds great promise for the identification of pathways involved in antidepressant response and pathophysiology of depression (Kaddurah-Daouk and Krishnan, 2009). In contrast to studies of DNA, RNA, or proteins, there is no building block equivalent like nucleic acids or amino acids in the metabolome, and the chemical diversity of metabolites makes their study particularly challenging. Like proteins, some individual metabolites can be assayed through various detection methods: although the majority are currently detected via mass spectrometry.

Mass Spectrometry (MS) was initiated as a semi-quantitative method for providing either targeted or largescale metabolome analyses. Moreover, high-performance liquid chromatography, gas chromatography, and targeted electrochemistry based MS platforms have also been widely used to quantify abundant metabolic biomarkers in serum, plasma, and CSF from MDD participants (Martins-de-Souza, 2014). Many early depression metabolomic studies focused on broad metabolite classes, such as lipids (lipidomics). This was in part due to technologic limitations (Piomelli et al., 2007), although there was a known connection between lipids and neuronal signaling and disease (Allen et al., 2006; Donati and Rasenick, 2008). A meta-analysis of 14 studies comparing the total n-3 polyunsaturated fatty acids (n-3 PUFAs) levels in serum, plasma or erythrocytes in depressed vs non-depressed individuals demonstrated a significantly lower amount of n-3 PUFA in depressed populations (Lin et al., 2010). These findings have led to numerous clinical trials demonstrating efficacy of particular n-3 PUFAs as adjunct therapy for depression (Gertsik et al., 2012). Investigation into their mechanism of action has also generated further basic science inquiry about the pathophysiology of depression (Czysz and Rasenick, 2013). As lipidomics technologies have improved, however, research has expanded beyond n-3 PUFAs. In a large study of plasma from 742 participants with records of depression and anxiety related HADS-A/D and CES-D scores were compared to 148 phospho- and sphingolipids. Most notably the ratio of sphingomyelin 23:1 to sphingomyelin 16:0 was inversely related to depression severity (Demirkan et al., 2013). Furthermore, plasma and erythrocytes from 65 control and 137 MDD (19% currently depressed) participants in the DELTA study demonstrated lower levels of mono unsaturated and saturated fatty acids in depressed patients (Assies et al., 2010). Additionally, a small study of bipolar depressed patients investigated metabolomic changes following ketamine or esketamine treatment, and found more evidence for lipid changes (Rotroff et al., 2016). Targeted electrochemistry based metabolomics platform (LCECA) profiled serum samples from MDD patients and found that metabolites of the tryptophan metabolism pathway (e.g., kynurenine) were differentially regulated (Kaddurah-Daouk et al., 2013). When 800 metabolites were screened across plasma from depressed, remitted, or never depressed patients, results revealed that GABA, glycerate, citrate, glycerol, and 9,12,octadecadienoate were reduced in the currently depressed cohort (Paige, 2007).

More recent investigations have looked at a wider array of metabolites outside of lipids, including amino acids, hormones, and biogenic amines (Martins-de-Souza, 2014). These studies have aimed either to differentiate depressed from non-depressed patients (Paige, 2007) or predict patient response to drug therapy (Gupta et al., 2016; Kaddurah-Daouk et al., 2013; Kaddurah-Daouk et al., 2011).

Nuclear Magnetic Resonance Spectroscopy (NMR)

The initiation of high-resolution proton nuclear magnetic resonance spectroscopy (1H-NMR) provided a means of analyzing several thousands of metabolites with high throughput (Abo et al., 2012; Kaddurah-Daouk and Krishnan, 2009). Aside from blood, urine samples have been used for metabolomics analysis. Tian et al. measured small endogenous metabolites using NMR and (Tian et al., 2014)reported that creatinine, taurine, 2-oxoglutarate, and xanthurenic acid increased significantly after treatment with the Chinese medication, xiaoyaosan, in MDD participants (Tian et al., 2014).

Multiplex-based metabolites screening is an advanced and quantitative method with the ability to quickly provide potential metabolic biomarker signatures (Rotroff et al., 2016). In combination with the targeted metabolomics kits currently available, it may be used for either exploratory or targeted analyses and provides reproducibility (Rotroff et al., 2016).

The above studies are somewhat limited by the heterogeneous mix of patients and/or pharmacotherapies. Other work has investigated metabolomics following treatment with specific antidepressants, including sertraline (Gupta et al., 2016; Kaddurah-Daouk et al., 2013; Kaddurah-Daouk et al., 2011), escitalopram (Ji et al., 2011), ketamine, and esketamine (Villasenor et al., 2014). Metabolite profiling of tryptophan metabolism in a 4-week double-blind placebo-controlled study showed that response to either sertraline or placebo was associated with metabolomics changes when post-treatment was compared to pre-treatment. Specifically, 5-methoxytryptophol and melatonin levels increased, while the kynurenine:melatonin and 3-hydroxykynurenine:melatonin ratios decreased (Zhu et al., 2013). Using the same samples in a separate study, Kaddurah-Daouk et al. found that pre-treatment levels of tryptophan, phenylalanine, purine, and tocopherol could predict responders versus non-responders (Kaddurah-Daouk et al., 2011). In a subsequent study, they found that a pre- to post-treatment decrease in branched chain amino acid (valine, leucine, and isoleucine) levels correlated with improvement in depression severity, notably in patients treated with sertraline (Kaddurah-Daouk et al., 2013). Other amino acids (glycine, glutamic acid, aspartic acid, asparagine) and the small molecule, hydroxylamine, have also been implicated as predictors of changes in QIDS-C score following escitalopram treatment (Ji et al., 2011). As mentioned above, the stability of many molecules is weak, and metabolites are especially susceptible given their quick turnover. Additionally, several mentioned above (fatty acids) are highly variable depending on fasting state. Thus, it will be of utmost importance to standardize collection materials should metabolomics of adipokines be a biomarker hit.

3.4 Data Driven Informatics

Bioinformatics, the study of information processing in biological systems, gained widespread prominence with the large volume of data arising from the human genome project (Hogeweg, 2011). Bioinformatics tools are now widely used to analyze the biological systems through various -omics studies like genomics, transcriptomics, epigenomics, proteomics, and metabolomics. The ongoing human connectome project (HCP; http://humanconnectome.org) is scanning 1200 healthy adults and utilizing robust informatics tools to analyze this large volume of data to map the neural network and understand the functional connections in the human brain (Toga et al., 2012). Arns et al. has brought these large datasets into the public domain, enabling the possibility of crowdsourcing biomarker discovery (Arns et al., 2015).

4. The Future of Biomarker Identification via Clinical Trial Analyses

The limitations of current diagnostic criteria for psychiatric illnesses have led to the Research Domain Criteria (RDoC) project by the NIMH (Insel et al., 2010b). Especially in the depression field, where clinical syndrome-based subtyping has failed to personalize treatment, the framework postulated in RDoC provides an exciting and promising future. RDoC aims to classify high level domains (or subtypes) from a heterogeneous population by integrating assessments from numerous systems, including emotional, cognitive, motivational, social behavioral, and potentially others, such as biological and physiological systems.

Among the biggest limitations of current depression biomarker research is the potpourri of studies looking at specific biomarker classes (e.g., genetics, metabolites, etc.) and focusing on one or two mutations, proteins, etc. rather than. When analyzed separately, there is an inability to detect how biomarkers may interact or synergize to promote the ultimate depression phenotype. Thus, to address the aims of RDoC and to hone in on specific subtypes, future clinical trials should take an integrative approach to biomarker research. Studies need to recruit large numbers of participants to accurately represent the population at-large. With the wealth of data which may ultimately be collected, both discovery and targeted analyses should be performed. Most of the tools described above are high-throughput, thereby enabling the identification of new ‘hits’. Also desired, will be the replication and validation of previously-implicated markers, such as those described above. Most importantly, however, future studies should attempt to identify multiple features (i.e., a biosignature) which together most-accurately predict response. This is the goal of the Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care (EMBARC) study (Trivedi et al., 2016).

The EMBARC study design, including inclusion and exclusion criteria, may be found at https://clinicaltrials.gov/ct2/show/NCT01407094. In summary, sertraline (an SSRI; (Bolden-Watson and Richelson, 1993; Owens et al., 2001)) and bupropion (a dopamine/norepinephrine reuptake inhibitor; (Ascher et al., 1995)) were selected as antidepressant medications for their different mechanisms of action. Placebo was included to establish the changes in clinical and biological markers that occur in the absence of pharmacologically active treatment (Dong and Blier, 2001; Li et al., 2002; Nomikos et al., 1989; Trivedi et al., 2016). The study includes two stages, with each stage lasting 8 weeks. At baseline, the study participants complete self-report clinical instruments and undergo neuroimaging, neurophysiological, and behavioral testing, then provide a blood sample. In addition to self-reporting, a research psychiatrist or psychologist completes clinician-rated instruments for clinical phenotyping. Metrics are repeated at a 1 week visit, and blood is collected at weeks 4, 8, 9 (if medication is switched), 12, and 16 (Trivedi et al., 2016).

4.1 Clinical and biological markers collected in EMBARC

Comprehensive clinical phenotyping in EMBARC is done with structured diagnostic assessments and self-report instruments to evaluate ongoing symptoms, antidepressant treatment history, trauma history, social functioning, sexual functioning, personality, and intelligence (Greenberg et al., 2015; Trivedi et al., 2016). Behavioral assessments include psychomotor slowing (choice reaction time and word fluency task), cognitive control (Flanker task), working memory (A not B task), and reward responsiveness (probabilistic reward task). Neuroimaging includes structural assessments (DTI and three dimensional high resolution Magnetization-Prepared Rapid Gradient-Echo, MP-RAGE), functional imaging while performing challenge tasks (e.g., implicit emotion processing and regulation; and reward processing), and resting state imaging (e.g., blood-oxygen-level dependent (BOLD) and arterial spin labeling (ASL) (Webb et al., 2016). Neurophysiological assessments include resting EEG with eyes opened and closed, Low Resolution Electromagnetic Tomography (LORETA) to localize theta activity, and measurement of Loudness Dependence of Auditory Evoked Potentials (LDAEP) after presentation of 1000 Hz tones at 5 different intensity levels using a headphone. While imaging and EEG analysis have already begun to demonstrate the capacity for subtyping depression (Chase et al., 2015; Webb et al., 2016). Plasma and serum samples will next be analyzed using genomic, proteomic, and metabolomics technologies to potentially identify additional biomarkers.

4.2 Statistical innovations in EMBARC

Mixed effects models will be used to analyze individual variables for their role as mediators or moderators. The sample will then be randomly divided into exploratory and validation samples for generation and testing a differential treatment response index (DTRI) by using a combination of variables and interaction among variables that best predict treatment outcome (Chase et al., 2015; Trivedi et al., 2016). Based on the exploratory sample derived from stage 1 of the study, DTRI can be generated for levels of improvement expected with Sertraline treatment (data not shown). The validation sample will next be used to test DTRI. Side effect indices (IndexSE) will be generated in a similar manner using exploratory and validation samples.

4.3 Limitations of EMBARC

Generalizability of findings from EMBARC may be limited to the subgroup of MDD patients who meet the eligibility criteria of this study, especially due to restrictions on age of onset, chronicity, co-morbid psychiatric disorders, exclusionary medications and presence of treatment resistance. Similar to results from other clinical trials, we cannot be certain that the peripheral changes observed are in direct relation to changes observed centrally. Certainly, there are several ways in which peripheral molecules enter the brain (e.g., a weakened blood brain barrier, active transport, cerebrospinal fluid-lymph node interaction (Kim and Won, 2017{Robson, 2017 #366)}, although the intricacies and nuances of how the two systems directly relate remains elusive. Lastly, EMBARC was designed to generate candidate clinical and biological markers, and not to test a priori hypotheses. Thus, it may lack the adequate power necessary to draw definitive conclusions about mediators or moderators. Lack of assessment of clinical and biological markers of treatment outcomes with other antidepressant treatments like psychotherapy, exercise, novel antidepressant medications (e.g., ketamine), and somatic treatments is another limitation of EMBARC. A naturalistic follow-up study with longer term assessment may help to evaluate functional recovery, risk of relapse or recurrence, and efficacy of subtype treatment matching.

5. Conclusions and Future Perspectives

The ineffective treatment of depression necessitates biomarker discovery. We are equipped with complex and high throughput technologies, which combined with large-scale clinical trials, should enable detection of depression biosignatures and ultimately a higher change of remission. As demonstrated in Figure 3, to date, biomarker research has begun spanning the genome, proteome, and metabolome, and the utility of these technologies will only continue to grow. Presently there exists a potpourri of studies in each of these fields: SNP studies have identified genes related to monoaminergic and glutamatergic signaling. Protein studies have the most robust evidence for disturbances in immunologic pathways including interleukin IL-6, IL-1β, TNF-α, and IFNγ. Finally, metabolomics studies have identified a variety of candidate metabolites related to depression and drug response. A limited number of these results have been validated in additional patient cohorts, however. As a result, no individual or collection of biomarkers have translated into clinical practice for either diagnosis of depression or guidance of treatment selection. The heterogeneous pathology driving depression makes biomarker discovery particularly challenging, though provides the capability to hone in on numerous underlying biomarkers (i.e., a biosignature) to define specific subgroups. Robust techniques are necessary, as is the continuing refinement of both measurement and analytical tools for biomarker discovery. These technological advances combined with an increasing identification of putative biomarkers will help tailor depression treatment to individual patients, ultimately leading to faster and more efficacious treatment.

Figure 3. ‘Omics based approach in major depression and treatment matching.

Figure 3

Along with imaging and physiology, ‘omics’ and clinical phenotype are major components which may lead to treatment matching via unique biosignature. ‘Omics include: genomics, proteomics, transcriptomics, epigenomics, and metabolomics. Phenotype includes social, clinical and behavioral. Overall, evaluation of these features may enable identification of different subtypes of depression, which may improve treatment matching.

References

  1. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95. doi: 10.1067/mcp.2001.113989. [DOI] [PubMed] [Google Scholar]
  2. Abo R, Hebbring S, Ji Y, Zhu H, Zeng ZB, Batzler A, Jenkins GD, Biernacka J, Snyder K, Drews M, Fiehn O, Fridley B, Schaid D, Kamatani N, Nakamura Y, Kubo M, Mushiroda T, Kaddurah-Daouk R, Mrazek DA, Weinshilboum RM. Merging pharmacometabolomics with pharmacogenomics using ‘1000 Genomes’ single-nucleotide polymorphism imputation: selective serotonin reuptake inhibitor response pharmacogenomics. Pharmacogenet Genomics. 2012;22:247–253. doi: 10.1097/FPC.0b013e32835001c9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allen JA, Halverson-Tamboli RA, Rasenick MM. Lipid raft microdomains and neurotransmitter signalling. Nature Reviews Neuroscience. 2006;8:128–140. doi: 10.1038/nrn2059. [DOI] [PubMed] [Google Scholar]
  4. Anttila S, Huuhka K, Huuhka M, Illi A, Rontu R, Leinonen E, Lehtimaki T. Catechol-O-methyltransferase (COMT) polymorphisms predict treatment response in electroconvulsive therapy. The pharmacogenomics journal. 2007;8:113–116. doi: 10.1038/sj.tpj.6500468. [DOI] [PubMed] [Google Scholar]
  5. Arias B, Serretti A, Lorenzi C, Gasto C, Catalan R, Fananas L. Analysis of COMT gene (Val 158 Met polymorphism) in the clinical response to SSRIs in depressive patients of European origin. J Affect Disord. 2006;90:251–256. doi: 10.1016/j.jad.2005.11.008. [DOI] [PubMed] [Google Scholar]
  6. Arnow BA, Blasey C, Williams LM, Palmer DM, Rekshan W, Schatzberg AF, Etkin A, Kulkarni J, Luther JF, Rush AJ. Depression Subtypes in Predicting Antidepressant Response: A Report From the iSPOT-D Trial. American Journal of Psychiatry. 2015;172:743–750. doi: 10.1176/appi.ajp.2015.14020181. [DOI] [PubMed] [Google Scholar]
  7. Arns M, Etkin A, Hegerl U, Williams LM, DeBattista C, Palmer DM, Fitzgerald PB, Harris A, deBeuss R, Gordon E. Frontal and rostral anterior cingulate (rACC) theta EEG in depression: Implications for treatment outcome? European Neuropsychopharmacology. 2015 doi: 10.1016/j.euroneuro.2015.03.007. [DOI] [PubMed] [Google Scholar]
  8. Ascher JA, Cole JO, Colin JN, Feighner JP, Ferris RM, Fibiger HC, Golden RN, Martin P, Potter WZ, Richelson E, et al. Bupropion: a review of its mechanism of antidepressant activity. The Journal of clinical psychiatry. 1995;56:395–401. [PubMed] [Google Scholar]
  9. Assies J, Pouwer F, Lok A, Mocking RJ, Bockting CL, Visser I, Abeling NG, Duran M, Schene AH. Plasma and erythrocyte fatty acid patterns in patients with recurrent depression: a matched case-control study. PLoS One. 2010;5:e10635. doi: 10.1371/journal.pone.0010635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ball S, Classi P, Dennehy EB. What happens next?: a claims database study of second-line pharmacotherapy in patients with major depressive disorder (MDD) who initiate selective serotonin reuptake inhibitor (SSRI) treatment. Ann Gen Psychiatry. 2014;13:8. doi: 10.1186/1744-859X-13-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Baune BT, Hohoff C, Berger K, Neumann A, Mortensen S, Roehrs T, Deckert J, Arolt V, Domschke K. Association of the COMT val158met variant with antidepressant treatment response in major depression. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2008;33:924–932. doi: 10.1038/sj.npp.1301462. [DOI] [PubMed] [Google Scholar]
  12. Beck A, Ward C, Mendelson M, Mock J, Erbaugh J. An inventory for measuring depression. Archives of general psychiatry. 1961;4:561–571. doi: 10.1001/archpsyc.1961.01710120031004. [DOI] [PubMed] [Google Scholar]
  13. Benedetti F, Colombo C, Pirovano A, Marino E, Smeraldi E. The catechol-O-methyltransferase Val(108/158)Met polymorphism affects antidepressant response to paroxetine in a naturalistic setting. Psychopharmacology (Berl) 2009;203:155–160. doi: 10.1007/s00213-008-1381-7. [DOI] [PubMed] [Google Scholar]
  14. Benedetti F, Dallaspezia S, Colombo C, Lorenzi C, Pirovano A, Smeraldi E. Effect of catechol-O-methyltransferase Val(108/158)Met polymorphism on antidepressant efficacy of fluvoxamine. European psychiatry: the journal of the Association of European Psychiatrists. 2010;25:476–478. doi: 10.1016/j.eurpsy.2009.12.007. [DOI] [PubMed] [Google Scholar]
  15. Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev. 2009;23:781–783. doi: 10.1101/gad.1787609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Binder EB, Bradley RG, Liu W, Epstein MP, Deveau TC, Mercer KB, Tang Y, Gillespie CF, Heim CM, Nemeroff CB, Schwartz AC, Cubells JF, Ressler KJ. Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults. Jama. 2008;299:1291–1305. doi: 10.1001/jama.299.11.1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Binder EB, Salyakina D, Lichtner P, Wochnik GM, Ising M, Putz B, Papiol S, Seaman S, Lucae S, Kohli MA, Nickel T, Kunzel HE, Fuchs B, Majer M, Pfennig A, Kern N, Brunner J, Modell S, Baghai T, Deiml T, Zill P, Bondy B, Rupprecht R, Messer T, Kohnlein O, Dabitz H, Bruckl T, Muller N, Pfister H, Lieb R, Mueller JC, Lohmussaar E, Strom TM, Bettecken T, Meitinger T, Uhr M, Rein T, Holsboer F, Muller-Myhsok B. Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment. Nat Genet. 2004;36:1319–1325. doi: 10.1038/ng1479. [DOI] [PubMed] [Google Scholar]
  18. Blumenthal JA, Smith PJ, Hoffman BM. Is Exercise a Viable Treatment for Depression? ACSMs Health Fit J. 2012;16:14–21. doi: 10.1249/01.FIT.0000416000.09526.eb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bobo WV, Chen H, Trivedi MH, Stewart JW, Nierenberg AA, Fava M, Kurian BT, Warden D, Morris DW, Luther JF, Husain MM, Cook IA, Lesser IM, Kornstein SG, Wisniewski SR, Rush AJ, Shelton RC. Randomized comparison of selective serotonin reuptake inhibitor (escitalopram) monotherapy and antidepressant combination pharmacotherapy for major depressive disorder with melancholic features: a CO-MED report. J Affect Disord. 2011;133:467–476. doi: 10.1016/j.jad.2011.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bolden-Watson C, Richelson E. Blockade by newly-developed antidepressants of biogenic amine uptake into rat brain synaptosomes. Life Sci. 1993;52:1023–1029. doi: 10.1016/0024-3205(93)90194-8. [DOI] [PubMed] [Google Scholar]
  21. Bot M, Chan MK, Jansen R, Lamers F, Vogelzangs N, Steiner J, Leweke FM, Rothermundt M, Cooper J, Bahn S, Penninx BW. Serum proteomic profiling of major depressive disorder. Transl Psychiatry. 2015;5:e599. doi: 10.1038/tp.2015.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bridle C, Spanjers K, Patel S, Atherton NM, Lamb SE. Effect of exercise on depression severity in older people: systematic review and meta-analysis of randomised controlled trials. Br J Psychiatry. 2012;201:180–185. doi: 10.1192/bjp.bp.111.095174. [DOI] [PubMed] [Google Scholar]
  23. Brunoni AR, Machado-Vieira R, Zarate CA, Jr, Vieira EL, Vanderhasselt MA, Nitsche MA, Valiengo L, Bensenor IM, Lotufo PA, Gattaz WF, Teixeira AL. BDNF plasma levels after antidepressant treatment with sertraline and transcranial direct current stimulation: results from a factorial, randomized, sham-controlled trial. Eur Neuropsychopharmacol. 2014;24:1144–1151. doi: 10.1016/j.euroneuro.2014.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Burton C, Cochran AJ, Cameron IM. Restarting antidepressant treatment following early discontinuation-a primary care database study. Family practice. 2015 doi: 10.1093/fampra/cmv063. [DOI] [PubMed] [Google Scholar]
  25. Buttenschon HN, Foldager L, Elfving B, Poulsen PH, Uher R, Mors O. Neurotrophic factors in depression in response to treatment. J Affect Disord. 2015;183:287–294. doi: 10.1016/j.jad.2015.05.027. [DOI] [PubMed] [Google Scholar]
  26. Chan HN, Rush AJ, Nierenberg AA, Trivedi M, Wisniewski SR, Balasubramani GK, Friedman ES, Gaynes BN, Davis L, Morris D, Fava M. Correlates and outcomes of depressed out-patients with greater and fewer anxious symptoms: a CO-MED report. Int J Neuropsychopharmacol. 2012;15:1387–1399. doi: 10.1017/S1461145711001660. [DOI] [PubMed] [Google Scholar]
  27. Chase HW, Fournier JC, Greenberg T, Almeida JR, Stiffler R, Zevallos CR, Aslam H, Cooper C, Deckersbach T, Weyandt S, Adams P, Toups M, Carmody T, Oquendo MA, Peltier S, Fava M, McGrath PJ, Weissman M, Parsey R, McInnis MG, Kurian B, Trivedi MH, Phillips ML. Accounting for Dynamic Fluctuations across Time when Examining fMRI Test-Retest Reliability: Analysis of a Reward Paradigm in the EMBARC Study. PloS one. 2015;10:e0126326. doi: 10.1371/journal.pone.0126326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chen CS, Zhu H. Protein microarrays. BioTechniques. 2006;40:423, 425, 427. doi: 10.2144/06404TE01. passim. [DOI] [PubMed] [Google Scholar]
  29. Chen J, Lipska BK, Halim N, Ma QD, Matsumoto M, Melhem S, Kolachana BS, Hyde TM, Herman MM, Apud J, Egan MF, Kleinman JE, Weinberger DR. Functional analysis of genetic variation in catechol-O-methyltransferase (COMT): effects on mRNA, protein, and enzyme activity in postmortem human brain. Am J Hum Genet. 2004;75:807–821. doi: 10.1086/425589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Craighead WE, Dunlop BW. Combination psychotherapy and antidepressant medication treatment for depression: for whom, when, and how. Annual review of psychology. 2014;65:267–300. doi: 10.1146/annurev.psych.121208.131653. [DOI] [PubMed] [Google Scholar]
  31. Czysz AH, Rasenick MM. G-protein signaling, lipid rafts and the possible sites of action for the antidepressant effects of n-3 polyunsaturated fatty acids. CNS & neurological disorders drug targets. 2013;12:466–473. doi: 10.2174/1871527311312040005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. D’Empaire I, Guico-Pabia CJ, Preskorn SH. Antidepressant treatment and altered CYP2D6 activity: are pharmacokinetic variations clinically relevant? J Psychiatr Pract. 2011;17:330–339. doi: 10.1097/01.pra.0000405363.95881.01. [DOI] [PubMed] [Google Scholar]
  33. Daly EJ, TM, Wisniewski SR, Nierenberg AA, Gaynes BN, Warden D, Morris DW, Luther JF, Farabaugh A, Cook I, Rush AJ. Health-related quality of life in depression: a STAR*D report. ANNALS OF CLINICAL PSYCHIATRY. 2010;22:43–55. [PubMed] [Google Scholar]
  34. Demirkan A, Isaacs A, Ugocsai P, Liebisch G, Struchalin M, Rudan I, Wilson JF, Pramstaller PP, Gyllensten U, Campbell H, Schmitz G, Oostra BA, van Duijn CM. Plasma phosphatidylcholine and sphingomyelin concentrations are associated with depression and anxiety symptoms in a Dutch family-based lipidomics study. J Psychiatr Res. 2013;47:357–362. doi: 10.1016/j.jpsychires.2012.11.001. [DOI] [PubMed] [Google Scholar]
  35. Diniz BS, Sibille E, Ding Y, Tseng G, Aizenstein HJ, Lotrich F, Becker JT, Lopez OL, Lotze MT, Klunk WE, Reynolds CF, Butters MA. Plasma biosignature and brain pathology related to persistent cognitive impairment in late-life depression. Molecular psychiatry. 2015;20:594–601. doi: 10.1038/mp.2014.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Domschke K, Tidow N, Schwarte K, Deckert J, Lesch KP, Arolt V, Zwanzger P, Baune BT. Serotonin transporter gene hypomethylation predicts impaired antidepressant treatment response. Int J Neuropsychopharmacol. 2014;17:1167–1176. doi: 10.1017/S146114571400039X. [DOI] [PubMed] [Google Scholar]
  37. Domschke K, Zavorotnyy M, Diemer J, Nitsche S, Hohoff C, Baune BT, Deckert J, Arolt V, Zwanzger P. COMT val158met influence on electroconvulsive therapy response in major depression. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics. 2010;153B:286–290. doi: 10.1002/ajmg.b.30949. [DOI] [PubMed] [Google Scholar]
  38. Donati RJ, Rasenick MM. Lipid rafts, G proteins and the etiology of and treatment for depression: progress toward a depression biomarker. Future Neurol. 2008;3:511–514. [Google Scholar]
  39. Dong J, Blier P. Modification of norepinephrine and serotonin, but not dopamine, neuron firing by sustained bupropion treatment. Psychopharmacology (Berl) 2001;155:52–57. doi: 10.1007/s002130000665. [DOI] [PubMed] [Google Scholar]
  40. Dowsett M, Dunbier AK. Emerging biomarkers and new understanding of traditional markers in personalized therapy for breast cancer. Clin Cancer Res. 2008;14:8019–8026. doi: 10.1158/1078-0432.CCR-08-0974. [DOI] [PubMed] [Google Scholar]
  41. Fava M, Rush AJ, Alpert JE, Balasubramani GK, Wisniewski SR, Carmin CN, Biggs MM, Zisook S, Leuchter A, Howland R, Warden D, Trivedi MH. Difference in treatment outcome in outpatients with anxious versus nonanxious depression: a STAR*D report. Am J Psychiatry. 2008;165:342–351. doi: 10.1176/appi.ajp.2007.06111868. [DOI] [PubMed] [Google Scholar]
  42. Feighner JP. Mechanism of action of antidepressant medications. The Journal of clinical psychiatry. 1999;60(Suppl 4):4–11. discussion 12–13. [PubMed] [Google Scholar]
  43. Fernie AR, Trethewey RN, Krotzky AJ, Willmitzer L. Metabolite profiling: from diagnostics to systems biology. Nat Rev Mol Cell Biol. 2004;5:763–769. doi: 10.1038/nrm1451. [DOI] [PubMed] [Google Scholar]
  44. Friedman ES, Davis LL, Zisook S, Wisniewski SR, Trivedi MH, Fava M, Rush AJ. Baseline depression severity as a predictor of single and combination antidepressant treatment outcome: results from the CO-MED trial. Eur Neuropsychopharmacol. 2012;22:183–199. doi: 10.1016/j.euroneuro.2011.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Friedman ES, Wisniewski SR, Gilmer W, Nierenberg AA, Rush AJ, Fava M, Zisook S, Balasubramani GK, Trivedi MH. Sociodemographic, clinical, and treatment characteristics associated with worsened depression during treatment with citalopram: results of the NIMH STAR(*)D trial. Depress Anxiety. 2009;26:612–621. doi: 10.1002/da.20568. [DOI] [PubMed] [Google Scholar]
  46. Gadad BS, Jha MK, Grannemann B, Mayes TL, Trivedi MH. Proteomics profiling reveals inflammatory biomarkers of antidepressant treatment response: Findings from the CO-MED trial. Journal of Psychiatric Research. 2017 doi: 10.1016/j.jpsychires.2017.05.012. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Garriock HA, Kraft JB, Shyn SI, Peters EJ, Yokoyama JS, Jenkins GD, Reinalda MS, Slager SL, McGrath PJ, Hamilton SP. A genomewide association study of citalopram response in major depressive disorder. Biological psychiatry. 2010;67:133–138. doi: 10.1016/j.biopsych.2009.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Gawlik M, Moller-Ehrlich K, Mende M, Jovnerovski M, Jung S, Jabs B, Knapp M, Stoeber G. Is FKBP5 a genetic marker of affective psychosis? A case control study and analysis of disease related traits. BMC psychiatry. 2006;6:52. doi: 10.1186/1471-244X-6-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Gertsik L, Poland RE, Bresee C, Rapaport MH. Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder. Journal of Clinical Psychopharmacology. 2012;32:61–64. doi: 10.1097/JCP.0b013e31823f3b5f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, Karayiorgou M. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci U S A. 1998;95:9991–9996. doi: 10.1073/pnas.95.17.9991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gorgulu Y, Caliyurt O. Rapid antidepressant effects of sleep deprivation therapy correlates with serum BDNF changes in major depression. Brain Res Bull. 2009;80:158–162. doi: 10.1016/j.brainresbull.2009.06.016. [DOI] [PubMed] [Google Scholar]
  52. Grasmader K, Verwohlt PL, Rietschel M, Dragicevic A, Muller M, Hiemke C, Freymann N, Zobel A, Maier W, Rao ML. Impact of polymorphisms of cytochrome-P450 isoenzymes 2C9, 2C19 and 2D6 on plasma concentrations and clinical effects of antidepressants in a naturalistic clinical setting. Eur J Clin Pharmacol. 2004;60:329–336. doi: 10.1007/s00228-004-0766-8. [DOI] [PubMed] [Google Scholar]
  53. Greenberg T, Chase HW, Almeida JR, Stiffler R, Zevallos CR, Aslam HA, Deckersbach T, Weyandt S, Cooper C, Toups M, Carmody T, Kurian B, Peltier S, Adams P, McInnis MG, Oquendo MA, McGrath PJ, Fava M, Weissman M, Parsey R, Trivedi MH, Phillips ML. Moderation of the Relationship Between Reward Expectancy and Prediction Error-Related Ventral Striatal Reactivity by Anhedonia in Unmedicated Major Depressive Disorder: Findings From the EMBARC Study. The American journal of psychiatry. 2015;172:881–891. doi: 10.1176/appi.ajp.2015.14050594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Gupta M, Neavin D, Liu D, Biernacka J, Hall-Flavin D, Bobo WV, Frye MA, Skime M, Jenkins GD, Batzler A, Kalari K, Matson W, Bhasin SS, Zhu H, Mushiroda T, Nakamura Y, Kubo M, Wang L, Kaddurah-Daouk R, Weinshilboum RM. TSPAN5, ERICH3 and selective serotonin reuptake inhibitors in major depressive disorder: pharmacometabolomics-informed pharmacogenomics. Molecular psychiatry. 2016;21:1717–1725. doi: 10.1038/mp.2016.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Han KM, Won E, Sim Y, Kang J, Han C, Kim YK, Kim SH, Joe SH, Lee MS, Tae WS, Ham BJ. Influence of FKBP5 polymorphism and DNA methylation on structural changes of the brain in major depressive disorder. Sci Rep. 2017;7:42621. doi: 10.1038/srep42621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hannestad J, DellaGioia N, Bloch M. The Effect of Antidepressant Medication Treatment on Serum Levels of Inflammatory Cytokines: A Meta-Analysis. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2011;36:2452–2459. doi: 10.1038/npp.2011.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hasler G, Drevets WC, Manji HK, Charney DS. Discovering endophenotypes for major depression. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2004;29:1765–1781. doi: 10.1038/sj.npp.1300506. [DOI] [PubMed] [Google Scholar]
  58. Hennings JM, Uhr M, Klengel T, Weber P, Putz B, Touma C, Czamara D, Ising M, Holsboer F, Lucae S. RNA expression profiling in depressed patients suggests retinoid-related orphan receptor alpha as a biomarker for antidepressant response. Transl Psychiatry. 2015;5:e538. doi: 10.1038/tp.2015.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hodgson K, Tansey K, Dernovsek MZ, Hauser J, Henigsberg N, Maier W, Mors O, Placentino A, Rietschel M, Souery D, Smith R, Craig IW, Farmer AE, Aitchison KJ, Belsy S, Davis OS, Uher R, McGuffin P. Genetic differences in cytochrome P450 enzymes and antidepressant treatment response. J Psychopharmacol. 2014;28:133–141. doi: 10.1177/0269881113512041. [DOI] [PubMed] [Google Scholar]
  60. Hodgson K, Tansey KE, Uher R, Dernovsek MZ, Mors O, Hauser J, Souery D, Maier W, Henigsberg N, Rietschel M, Placentino A, Craig IW, Aitchison KJ, Farmer AE, Dobson RJ, McGuffin P. Exploring the role of drug-metabolising enzymes in antidepressant side effects. Psychopharmacology (Berl) 2015 doi: 10.1007/s00213-015-3898-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hogeweg P. The Roots of Bioinformatics in Theoretical Biology. PLoS Comput Biol. 2011;7:e1002021. doi: 10.1371/journal.pcbi.1002021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Horstmann S, Lucae S, Menke A, Hennings JM, Ising M, Roeske D, Muller-Myhsok B, Holsboer F, Binder EB. Polymorphisms in GRIK4, HTR2A, and FKBP5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2010;35:727–740. doi: 10.1038/npp.2009.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Insel T, Cuthbert B, Garvey M, Heinssen R, Pine DS, Quinn K, Sanislow C, Wang P. Research Domain Criteria (RDoC): Toward a New Classification Framework for Research on Mental Disorders. American Journal of Psychiatry. 2010a;167:748–751. doi: 10.1176/appi.ajp.2010.09091379. [DOI] [PubMed] [Google Scholar]
  64. Insel T, Cuthbert B, Garvey M, Heinssen R, Pine DS, Quinn K, Sanislow C, Wang P. Research domain criteria (RDoC): toward a new classification framework for research on mental disorders. The American journal of psychiatry. 2010b;167:748–751. doi: 10.1176/appi.ajp.2010.09091379. [DOI] [PubMed] [Google Scholar]
  65. 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. Archives of general psychiatry. 2009;66:966–975. doi: 10.1001/archgenpsychiatry.2009.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Jansen R, Penninx BW, Madar V, Xia K, Milaneschi Y, Hottenga JJ, Hammerschlag AR, Beekman A, van der Wee N, Smit JH, Brooks AI, Tischfield J, Posthuma D, Schoevers R, van Grootheest G, Willemsen G, de Geus EJ, Boomsma DI, Wright FA, Zou F, Sun W, Sullivan PF. Gene expression in major depressive disorder. Mol Psychiatry. 2016;21:339–347. doi: 10.1038/mp.2015.57. [DOI] [PubMed] [Google Scholar]
  67. Janssen DG, Caniato RN, Verster JC, Baune BT. A psychoneuroimmunological review on cytokines involved in antidepressant treatment response. Hum Psychopharmacol. 2010;25:201–215. doi: 10.1002/hup.1103. [DOI] [PubMed] [Google Scholar]
  68. Jha MK, Minhajuddin A, Gadad BS, Greer T, Grannemann B, Soyombo A, Mayes TL, Rush AJ, Trivedi MH. Can C-reactive protein inform antidepressant medication selection in depressed outpatients? Findings from the CO-MED trial. Psychoneuroendocrinology. 2017;78:105–113. doi: 10.1016/j.psyneuen.2017.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ji Y, Biernacka JM, Hebbring S, Chai Y, Jenkins GD, Batzler A, Snyder KA, Drews MS, Desta Z, Flockhart D, Mushiroda T, Kubo M, Nakamura Y, Kamatani N, Schaid D, Weinshilboum RM, Mrazek DA. Pharmacogenomics of selective serotonin reuptake inhibitor treatment for major depressive disorder: genome-wide associations and functional genomics. The pharmacogenomics journal. 2013;13:456–463. doi: 10.1038/tpj.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Ji Y, Hebbring S, Zhu H, Jenkins GD, Biernacka J, Snyder K, Drews M, Fiehn O, Zeng Z, Schaid D, Mrazek DA, Kaddurah-Daouk R, Weinshilboum RM. Glycine and a glycine dehydrogenase (GLDC) SNP as citalopram/escitalopram response biomarkers in depression: pharmacometabolomics-informed pharmacogenomics. Clin Pharmacol Ther. 2011;89:97–104. doi: 10.1038/clpt.2010.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kaddurah-Daouk R, Bogdanov MB, Wikoff WR, Zhu H, Boyle SH, Churchill E, Wang Z, Rush AJ, Krishnan RR, Pickering E, Delnomdedieu M, Fiehn O. Pharmacometabolomic mapping of early biochemical changes induced by sertraline and placebo. Transl Psychiatry. 2013;3:e223. doi: 10.1038/tp.2012.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kaddurah-Daouk R, Boyle SH, Matson W, Sharma S, Matson S, Zhu H, Bogdanov MB, Churchill E, Krishnan RR, Rush AJ, Pickering E, Delnomdedieu M. Pretreatment metabotype as a predictor of response to sertraline or placebo in depressed outpatients: a proof of concept. Transl Psychiatry. 2011;1:e26. doi: 10.1038/tp.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kaddurah-Daouk R, Krishnan KR. Metabolomics: a global biochemical approach to the study of central nervous system diseases. Neuropsychopharmacology. 2009;34:173–186. doi: 10.1038/npp.2008.174. [DOI] [PubMed] [Google Scholar]
  74. Kaenmaki M, Tammimaki A, Myohanen T, Pakarinen K, Amberg C, Karayiorgou M, Gogos JA, Mannisto PT. Quantitative role of COMT in dopamine clearance in the prefrontal cortex of freely moving mice. J Neurochem. 2010;114:1745–1755. doi: 10.1111/j.1471-4159.2010.06889.x. [DOI] [PubMed] [Google Scholar]
  75. Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, Rush AJ, Walters EE, Wang PS. The Epidemiology of Major Depressive Disorder. JAMA: The Journal of the American Medical Association. 2003;289:3095–3105. doi: 10.1001/jama.289.23.3095. [DOI] [PubMed] [Google Scholar]
  76. Kim YK, Won E. The influence of stress on neuroinflammation and alterations in brain structure and function in major depressive disorder. Behavioural brain research. 2017;329:6–11. doi: 10.1016/j.bbr.2017.04.020. [DOI] [PubMed] [Google Scholar]
  77. Kocabas NA, Faghel C, Barreto M, Kasper S, Linotte S, Mendlewicz J, Noro M, Oswald P, Souery D, Zohar J, Massat I. The impact of catechol-O-methyltransferase SNPs and haplotypes on treatment response phenotypes in major depressive disorder: a case-control association study. International clinical psychopharmacology. 2010;25:218–227. doi: 10.1097/YIC.0b013e328338b884. [DOI] [PubMed] [Google Scholar]
  78. Kraft JB, Peters EJ, Slager SL, Jenkins GD, Reinalda MS, McGrath PJ, Hamilton SP. Analysis of association between the serotonin transporter and antidepressant response in a large clinical sample. Biological psychiatry. 2007;61:734–742. doi: 10.1016/j.biopsych.2006.07.017. [DOI] [PubMed] [Google Scholar]
  79. Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, Weinshilboum RM. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics. 1996;6:243–250. doi: 10.1097/00008571-199606000-00007. [DOI] [PubMed] [Google Scholar]
  80. Lee AY, Raya AK, Kymes SM, Shiels A, Brantley MA., Jr Pharmacogenetics of complement factor H (Y402H) and treatment of exudative age-related macular degeneration with ranibizumab. Br J Ophthalmol. 2009;93:610–613. doi: 10.1136/bjo.2008.150995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lee J, Joo EJ, Lim HJ, Park JM, Lee KY, Park A, Seok A, Lee H, Kang HG. Proteomic analysis of serum from patients with major depressive disorder to compare their depressive and remission statuses. Psychiatry Investig. 2015;12:249–259. doi: 10.4306/pi.2015.12.2.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Lesser I, Rosales A, Zisook S, Gonzalez C, Flores D, Trivedi M, Sciolla A, Luther J, Wisniewski S, Alpert J, Cook I, Rush AJ, Epstein M. Depression outcomes of Spanish- and english-speaking Hispanic outpatients in STAR*D. Psychiatr Serv. 2008;59:1273–1284. doi: 10.1176/ps.2008.59.11.1273. [DOI] [PubMed] [Google Scholar]
  83. Li SX, Perry KW, Wong DT. Influence of fluoxetine on the ability of bupropion to modulate extracellular dopamine and norepinephrine concentrations in three mesocorticolimbic areas of rats. Neuropharmacology. 2002;42:181–190. doi: 10.1016/s0028-3908(01)00160-5. [DOI] [PubMed] [Google Scholar]
  84. Lim S-W, Won H-H, Kim H, Myung W, Kim S, Kim K-K, Carroll BJ, Kim J-W, Kim DK. Genetic Prediction of Antidepressant Drug Response and Nonresponse in Korean Patients. PloS one. 2014;9:e107098. doi: 10.1371/journal.pone.0107098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lima JJ, Blake KV, Tantisira KG, Weiss ST. Pharmacogenetics of asthma. Curr Opin Pulm Med. 2009;15:57–62. doi: 10.1097/MCP.0b013e32831da8be. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Lin HC, Erickson SR, Balkrishnan R. Physician prescribing patterns of innovative antidepressants in the United States: the case of MDD patients 1993–2007. Int J Psychiatry Med. 2011;42:353–368. doi: 10.2190/PM.42.4.b. [DOI] [PubMed] [Google Scholar]
  87. Lin P-Y, Huang S-Y, Su K-P. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biological psychiatry. 2010;68:140–147. doi: 10.1016/j.biopsych.2010.03.018. [DOI] [PubMed] [Google Scholar]
  88. Lista S, Faltraco F, Prvulovic D, Hampel H. Blood and plasma-based proteomic biomarker research in Alzheimer’s disease. Progress in neurobiology. 2013;101–102:1–17. doi: 10.1016/j.pneurobio.2012.06.007. [DOI] [PubMed] [Google Scholar]
  89. Lobello KW, Preskorn SH, Guico-Pabia CJ, Jiang Q, Paul J, Nichols AI, Patroneva A, Ninan PT. Cytochrome P450 2D6 phenotype predicts antidepressant efficacy of venlafaxine: a secondary analysis of 4 studies in major depressive disorder. The Journal of clinical psychiatry. 2010;71:1482–1487. doi: 10.4088/JCP.08m04773blu. [DOI] [PubMed] [Google Scholar]
  90. Lohoff FW. Overview of the genetics of major depressive disorder. Curr Psychiatry Rep. 2010;12:539–546. doi: 10.1007/s11920-010-0150-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lopez JP, Mamdani F, Labonte B, Beaulieu MM, Yang JP, Berlim MT, Ernst C, Turecki G. Epigenetic regulation of BDNF expression according to antidepressant response. Mol Psychiatry. 2013;18:398–399. doi: 10.1038/mp.2012.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lucae S, Ising M, Horstmann S, Baune BT, Arolt V, Muller-Myhsok B, Holsboer F, Domschke K. HTR2A gene variation is involved in antidepressant treatment response. Eur Neuropsychopharmacol. 2010;20:65–68. doi: 10.1016/j.euroneuro.2009.08.006. [DOI] [PubMed] [Google Scholar]
  93. Maes M. The immunoregulatory effects of antidepressants. Hum Psychopharmacol. 2001;16:95–103. doi: 10.1002/hup.191. [DOI] [PubMed] [Google Scholar]
  94. Maron E, Tammiste A, Kallassalu K, Eller T, Vasar V, Nutt DJ, Metspalu A. Serotonin transporter promoter region polymorphisms do not influence treatment response to escitalopram in patients with major depression. Eur Neuropsychopharmacol. 2009;19:451–456. doi: 10.1016/j.euroneuro.2009.01.010. [DOI] [PubMed] [Google Scholar]
  95. Martins-de-Souza D. Proteomics, metabolomics, and protein interactomics in the characterization of the molecular features of major depressive disorder. Dialogues Clin Neurosci. 2014;16:63–73. doi: 10.31887/DCNS.2014.16.1/dmartins. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Martins-de-Souza D, Harris LW, Guest PC, Turck CW, Bahn S. The role of proteomics in depression research. European archives of psychiatry and clinical neuroscience. 2010;260:499–506. doi: 10.1007/s00406-009-0093-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Matrisciano F, Bonaccorso S, Ricciardi A, Scaccianoce S, Panaccione I, Wang L, Ruberto A, Tatarelli R, Nicoletti F, Girardi P, Shelton RC. Changes in BDNF serum levels in patients with major depression disorder (MDD) after 6 months treatment with sertraline escitalopram, or venlafaxine. J Psychiatr Res. 2009;43:247–254. doi: 10.1016/j.jpsychires.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Maze I, Shen L, Zhang B, Garcia BA, Shao N, Mitchell A, Sun H, Akbarian S, Allis CD, Nestler EJ. Analytical tools and current challenges in the modern era of neuroepigenomics. Nat Neurosci. 2014;17:1476–1490. doi: 10.1038/nn.3816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. McGrath PJ, Stewart JW, Fava M, Trivedi MH, Wisniewski SR, Nierenberg AA, Thase ME, Davis L, Biggs MM, Shores-Wilson K, Luther JF, Niederehe G, Warden D, Rush AJ. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry. 2006;163:1531–1541. doi: 10.1176/ajp.2006.163.9.1531. quiz 1666. [DOI] [PubMed] [Google Scholar]
  100. McMahon FJ, Buervenich S, Charney D, Lipsky R, Rush AJ, Wilson AF, Sorant AJ, Papanicolaou GJ, Laje G, Fava M, Trivedi MH, Wisniewski SR, Manji H. Variation in the gene encoding the serotonin 2A receptor is associated with outcome of antidepressant treatment. Am J Hum Genet. 2006;78:804–814. doi: 10.1086/503820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Menke A, Binder EB. Epigenetic alterations in depression and antidepressant treatment. Dialogues Clin Neurosci. 2014;16:395–404. doi: 10.31887/DCNS.2014.16.3/amenke. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Meron D, Hedger N, Garner M, Baldwin DS. Transcranial direct current stimulation (tDCS) in the treatment of depression: systematic review and meta-analysis of efficacy and tolerability. Neuroscience & Biobehavioral Reviews. 2015;57:46–62. doi: 10.1016/j.neubiorev.2015.07.012. [DOI] [PubMed] [Google Scholar]
  103. Mihaljevic Peles A, Bozina N, Sagud M, Rojnic Kuzman M, Lovric M. MDR1 gene polymorphism: therapeutic response to paroxetine among patients with major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1439–1444. doi: 10.1016/j.pnpbp.2008.03.018. [DOI] [PubMed] [Google Scholar]
  104. Molendijk ML, Spinhoven P, Polak M, Bus BA, Penninx BW, Elzinga BM. Serum BDNF concentrations as peripheral manifestations of depression: evidence from a systematic review and meta-analyses on 179 associations (N=9484) Molecular psychiatry. 2014;19:791–800. doi: 10.1038/mp.2013.105. [DOI] [PubMed] [Google Scholar]
  105. Mrazek DA, Rush AJ, Biernacka JM, O’Kane DJ, Cunningham JM, Wieben ED, Schaid DJ, Drews MS, Courson VL, Snyder KA, Black JL, 3rd, Weinshilboum RM. SLC6A4 variation and citalopram response. American journal of medical genetics. Part B, Neuropsychiatric genetics: the official publication of the International Society of Psychiatric Genetics. 2009;150B:341–351. doi: 10.1002/ajmg.b.30816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Murray CJ, Atkinson C, Bhalla K, Birbeck G, Burstein R, Chou D, Dellavalle R, Danaei G, Ezzati M, Fahimi A, Flaxman D, Foreman Gabriel S, Gakidou E, Kassebaum N, Khatibzadeh S, Lim S, Lipshultz SE, London S, Lopez MacIntyre MF, Mokdad AH, Moran A, Moran AE, Mozaffarian D, Murphy T, Naghavi M, Pope C, Roberts T, Salomon J, Schwebel DC, Shahraz S, Sleet DA, Murray Abraham J, Ali MK, Atkinson C, Bartels DH, Bhalla K, Birbeck G, Burstein R, Chen H, Criqui MH, Dahodwala Jarlais Ding EL, Dorsey ER, Ebel BE, Ezzati M, Fahami Flaxman S, Flaxman AD, Gonzalez-Medina D, Grant B, Hagan H, Hoffman H, Kassebaum N, Khatibzadeh S, Leasher JL, Lin J, Lipshultz SE, Lozano R, Lu Y, Mallinger L, McDermott MM, Micha R, Miller TR, Mokdad AA, Mokdad AH, Mozaffarian D, Naghavi M, Narayan KM, Omer SB, Pelizzari PM, Phillips D, Ranganathan D, Rivara FP, Roberts T, Sampson U, Sanman E, Sapkota A, Schwebel DC, Sharaz S, Shivakoti R, Singh GM, Singh D, Tavakkoli M, Towbin JA, Wilkinson JD, Zabetian A, Murray Abraham J, Ali MK, Alvardo M, Atkinson C, Baddour LM, Benjamin EJ, Bhalla K, Birbeck G, Bolliger I, Burstein R, Carnahan E, Chou D, Chugh SS, Cohen A, Colson KE, Cooper LT, Couser W, Criqui MH, Dabhadkar KC, Dellavalle RP, Jarlais Dicker D, Dorsey ER, Duber H, Ebel BE, Engell RE, Ezzati M, Felson DT, Finucane MM, Flaxman S, Flaxman AD, Fleming T, Foreman Forouzanfar MH, Freedman G, Freeman MK, Gakidou E, Gillum RF, Gonzalez-Medina D, Gosselin R, Gutierrez HR, Hagan H, Havmoeller R, Hoffman H, Jacobsen KH, James SL, Jasrasaria R, Jayarman S, Johns N, Kassebaum N, Khatibzadeh S, Lan Q, Leasher JL, Lim S, Lipshultz SE, London S, Lopez Lozano R, Lu Y, Mallinger L, Meltzer M, Mensah GA, Michaud C, Miller TR, Mock C, Moffitt TE, Mokdad AA, Mokdad AH, Moran A, Naghavi M, Narayan KM, Nelson RG, Olives C, Omer SB, Ortblad K, Ostro B, Pelizzari PM, Phillips D, Raju M, Razavi H, Ritz B, Roberts T, Sacco RL, Salomon J, Sampson U, Schwebel DC, Shahraz S, Shibuya K, Silberberg D, Singh JA, Steenland K, Taylor JA, Thurston GD, Vavilala MS, Vos T, Wagner GR, Weinstock MA, Weisskopf MG, Wulf S, Murray The state of US health, 1990–2010: burden of diseases, injuries, and risk factors. Jama. 2013;310:591–608. doi: 10.1001/jama.2013.13805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Naoi M, Maruyama W, Shamoto-Nagai M. Type A monoamine oxidase and serotonin are coordinately involved in depressive disorders: from neurotransmitter imbalance to impaired neurogenesis. J Neural Transm (Vienna) 2017 doi: 10.1007/s00702-017-1709-8. [DOI] [PubMed] [Google Scholar]
  108. Ninan PT, Shelton RC, Bao W, Guico-Pabia CJ. BDNF, interleukin-6, and salivary cortisol levels in depressed patients treated with desvenlafaxine. Prog Neuropsychopharmacol Biol Psychiatry. 2014;48:86–91. doi: 10.1016/j.pnpbp.2013.09.016. [DOI] [PubMed] [Google Scholar]
  109. Nomikos GG, Damsma G, Wenkstern D, Fibiger HC. Acute effects of bupropion on extracellular dopamine concentrations in rat striatum and nucleus accumbens studied by in vivo microdialysis. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 1989;2:273–279. doi: 10.1016/0893-133x(89)90031-6. [DOI] [PubMed] [Google Scholar]
  110. Oh G, Wang SC, Pal M, Chen ZF, Khare T, Tochigi M, Ng C, Yang YA, Kwan A, Kaminsky ZA, Mill J, Gunasinghe C, Tackett JL, Gottesman, Willemsen G, de Geus EJ, Vink JM, Slagboom PE, Wray NR, Heath AC, Montgomery GW, Turecki G, Martin NG, Boomsma DI, McGuffin P, Kustra R, Petronis A. DNA modification study of major depressive disorder: beyond locus-by-locus comparisons. Biol Psychiatry. 2015;77:246–255. doi: 10.1016/j.biopsych.2014.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Olfson M, Marcus SC. National patterns in antidepressant medication treatment. Archives of general psychiatry. 2009;66:848–856. doi: 10.1001/archgenpsychiatry.2009.81. [DOI] [PubMed] [Google Scholar]
  112. Owens MJ, Knight DL, Nemeroff CB. Second-generation SSRIs: human monoamine transporter binding profile of escitalopram and R-fluoxetine. Biological psychiatry. 2001;50:345–350. doi: 10.1016/s0006-3223(01)01145-3. [DOI] [PubMed] [Google Scholar]
  113. Paddock S, Laje G, Charney D, Rush AJ, Wilson AF, Sorant AJ, Lipsky R, Wisniewski SR, Manji H, McMahon FJ. Association of GRIK4 with outcome of antidepressant treatment in the STAR*D cohort. Am J Psychiatry. 2007;164:1181–1188. doi: 10.1176/appi.ajp.2007.06111790. [DOI] [PubMed] [Google Scholar]
  114. Paige LAMMWKKRRK-DRDC. A preliminary metabolomic analysis of older adults with and without depression. International Journal of Geriatric Psychiatry. 2007;22:418–423. doi: 10.1002/gps.1690. [DOI] [PubMed] [Google Scholar]
  115. Perlis RH, Fijal B, Adams DH, Sutton VK, Trivedi MH, Houston JP. Variation in Catechol-O-Methyltransferase Is Associated with Duloxetine Response in a Clinical Trial for Major Depressive Disorder. Biological psychiatry. 2009;65:785–791. doi: 10.1016/j.biopsych.2008.10.002. [DOI] [PubMed] [Google Scholar]
  116. Perlis RH, Fijal B, Dharia S, Heinloth AN, Houston JP. Failure to replicate genetic associations with antidepressant treatment response in duloxetine-treated patients. Biological psychiatry. 2010;67:1110–1113. doi: 10.1016/j.biopsych.2009.12.010. [DOI] [PubMed] [Google Scholar]
  117. Peters EJ, Slager SL, Jenkins GD, Reinalda MS, Garriock HA, Shyn SI, Kraft JB, McGrath PJ, Hamilton SP. Resequencing of serotonin-related genes and association of tagging SNPs to citalopram response. Pharmacogenet Genomics. 2009;19:1–10. doi: 10.1097/FPC.0b013e3283163ecd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Peters EJ, Slager SL, Kraft JB, Jenkins GD, Reinalda MS, McGrath PJ, Hamilton SP. Pharmacokinetic genes do not influence response or tolerance to citalopram in the STAR*D sample. PLoS One. 2008;3:e1872. doi: 10.1371/journal.pone.0001872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Peters EJ, Slager SL, McGrath PJ, Knowles JA, Hamilton SP. Investigation of serotonin-related genes in antidepressant response. Molecular psychiatry. 2004;9:879–889. doi: 10.1038/sj.mp.4001502. [DOI] [PubMed] [Google Scholar]
  120. Piomelli D, Astarita G, Rapaka R. A neuroscientist’s guide to lipidomics. Nature Reviews Neuroscience. 2007;8:743–754. doi: 10.1038/nrn2233. [DOI] [PubMed] [Google Scholar]
  121. Porcelli S, Fabbri C, Spina E, Serretti A, De Ronchi D. Genetic polymorphisms of cytochrome P450 enzymes and antidepressant metabolism. Expert Opin Drug Metab Toxicol. 2011;7:1101–1115. doi: 10.1517/17425255.2011.597740. [DOI] [PubMed] [Google Scholar]
  122. Pu M, Zhang Z, Xu Z, Shi Y, Geng L, Yuan Y, Zhang X, Reynolds GP. Influence of genetic polymorphisms in the glutamatergic and GABAergic systems and their interactions with environmental stressors on antidepressant response. Pharmacogenomics. 2013;14:277–288. doi: 10.2217/pgs.13.1. [DOI] [PubMed] [Google Scholar]
  123. Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, Haroon E, Miller AH. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA psychiatry. 2013;70:31–41. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Rethorst CD, Trivedi MH. Evidence-based recommendations for the prescription of exercise for major depressive disorder. J Psychiatr Pract. 2013;19:204–212. doi: 10.1097/01.pra.0000430504.16952.3e. [DOI] [PubMed] [Google Scholar]
  125. Rotroff DM, Corum DG, Motsinger-Reif A, Fiehn O, Bottrel N, Drevets WC, Singh J, Salvadore G, Kaddurah-Daouk R. Metabolomic signatures of drug response phenotypes for ketamine and esketamine in subjects with refractory major depressive disorder: new mechanistic insights for rapid acting antidepressants. Transl Psychiatry. 2016;6:e894. doi: 10.1038/tp.2016.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Rush AJ, Trivedi MH, Stewart JW, Nierenberg AA, Fava M, Kurian BT, Warden D, Morris DW, Luther JF, Husain MM, Cook IA, Shelton RC, Lesser IM, Kornstein SG, Wisniewski SR. Combining medications to enhance depression outcomes (CO-MED): acute and long-term outcomes of a single-blind randomized study. The American journal of psychiatry. 2011;168:689–701. doi: 10.1176/appi.ajp.2011.10111645. [DOI] [PubMed] [Google Scholar]
  127. Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, Niederehe G, Thase ME, Lavori PW, Lebowitz BD, McGrath PJ, Rosenbaum JF, Sackeim HA, Kupfer DJ, Luther J, Fava M. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006a;163:1905–1917. doi: 10.1176/ajp.2006.163.11.1905. [DOI] [PubMed] [Google Scholar]
  128. Rush AJ, Trivedi MH, Wisniewski SR, Stewart JW, Nierenberg AA, Thase ME, Ritz L, Biggs MM, Warden D, Luther JF, Shores-Wilson K, Niederehe G, Fava M, Team SDS. Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for depression. The New England journal of medicine. 2006b;354:1231–1242. doi: 10.1056/NEJMoa052963. [DOI] [PubMed] [Google Scholar]
  129. Rush AJ, Wisniewski SR, Warden D, Luther JF, Davis LL, Fava M, Nierenberg AA, Trivedi MH. Selecting among second-step antidepressant medication monotherapies: predictive value of clinical, demographic, or first-step treatment features. Arch Gen Psychiatry. 2008;65:870–880. doi: 10.1001/archpsyc.65.8.870. [DOI] [PubMed] [Google Scholar]
  130. Rush AJ, Wisniewski SR, Zisook S, Fava M, Sung SC, Haley CL, Chan HN, Gilmer WS, Warden D, Nierenberg AA, Balasubramani GK, Gaynes BN, Trivedi MH, Hollon SD. Is prior course of illness relevant to acute or longer-term outcomes in depressed out-patients? A STAR*D report. Psychol Med. 2012;42:1131–1149. doi: 10.1017/S0033291711002170. [DOI] [PubMed] [Google Scholar]
  131. Saragoussi D, Chollet J, Bineau S, Chalem Y, Milea D. Antidepressant switching patterns in the treatment of major depressive disorder: a General Practice Research Database (GPRD) Study. Int J Clin Pract. 2012;66:1079–1087. doi: 10.1111/j.1742-1241.2012.03015.x. [DOI] [PubMed] [Google Scholar]
  132. Schirle M, Bantscheff M, Kuster B. Mass spectrometry-based proteomics in preclinical drug discovery. Chemistry & biology. 2012;19:72–84. doi: 10.1016/j.chembiol.2012.01.002. [DOI] [PubMed] [Google Scholar]
  133. Serretti A, Calati R, Massat I, Linotte S, Kasper S, Lecrubier Y, Sens-Espel R, Bollen J, Zohar J, Berlo J, Lienard P, De Ronchi D, Mendlewicz J, Souery D. Cytochrome P450 CYP1A2, CYP2C9, CYP2C19 and CYP2D6 genes are not associated with response and remission in a sample of depressive patients. International clinical psychopharmacology. 2009;24:250–256. doi: 10.1097/YIC.0b013e32832e5b0d. [DOI] [PubMed] [Google Scholar]
  134. Serretti A, Chiesa A, Crisafulli C, Massat I, Linotte S, Calati R, Kasper S, Bailer U, Lecrubier Y, Fink M, Antonijevic I, Forray C, Snyder L, Bollen J, Zohar J, De Ronchi D, Souery D, Mendlewicz J. Failure to replicate influence of GRIK4 and GNB3 polymorphisms on treatment outcome in major depression. Neuropsychobiology. 2012;65:70–75. doi: 10.1159/000329553. [DOI] [PubMed] [Google Scholar]
  135. Serretti A, Fabbri C, Pellegrini S, Porcelli S, Politi P, Bellino S, Menchetti M, Mariotti V, Demi C, Martinelli V, Cappucciati M, Bozzatello P, Brignolo E, Brambilla P, Pae CU, Balestrieri M, De Ronchi D. No effect of serotoninergic gene variants on response to interpersonal counseling and antidepressants in major depression. Psychiatry Investig. 2013;10:180–189. doi: 10.4306/pi.2013.10.2.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Sesack SR, Hawrylak VA, Guido MA, Levey AI. Cellular and subcellular localization of the dopamine transporter in rat cortex. Adv Pharmacol. 1998;42:171–174. doi: 10.1016/s1054-3589(08)60720-6. [DOI] [PubMed] [Google Scholar]
  137. Shams ME, Arneth B, Hiemke C, Dragicevic A, Muller MJ, Kaiser R, Lackner K, Hartter S. CYP2D6 polymorphism and clinical effect of the antidepressant venlafaxine. J Clin Pharm Ther. 2006;31:493–502. doi: 10.1111/j.1365-2710.2006.00763.x. [DOI] [PubMed] [Google Scholar]
  138. Silveira H, Moraes H, Oliveira N, Coutinho ES, Laks J, Deslandes A. Physical exercise and clinically depressed patients: a systematic review and meta-analysis. Neuropsychobiology. 2013;67:61–68. doi: 10.1159/000345160. [DOI] [PubMed] [Google Scholar]
  139. Spronk D, Arns M, Barnett KJ, Cooper NJ, Gordon E. An investigation of EEG, genetic and cognitive markers of treatment response to antidepressant medication in patients with major depressive disorder: a pilot study. J Affect Disord. 2011;128:41–48. doi: 10.1016/j.jad.2010.06.021. [DOI] [PubMed] [Google Scholar]
  140. Stelzhammer V, Haenisch F, Chan MK, Cooper JD, Steiner J, Steeb H, Martins-de-Souza D, Rahmoune H, Guest PC, Bahn S. Proteomic changes in serum of first onset, antidepressant drug-naive major depression patients. Int J Neuropsychopharmacol. 2014;17:1599–1608. doi: 10.1017/S1461145714000819. [DOI] [PubMed] [Google Scholar]
  141. Strimbu K, Tavel JA. What are Biomarkers? Current opinion in HIV and AIDS. 2010;5:463–466. doi: 10.1097/COH.0b013e32833ed177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Sung SC, Haley CL, Wisniewski SR, Fava M, Nierenberg AA, Warden D, Morris DW, Kurian BT, Trivedi MH, Rush AJ. The impact of chronic depression on acute and long-term outcomes in a randomized trial comparing selective serotonin reuptake inhibitor monotherapy versus each of 2 different antidepressant medication combinations. The Journal of clinical psychiatry. 2012;73:967–976. doi: 10.4088/JCP.11m07043. [DOI] [PubMed] [Google Scholar]
  143. Sung SC, Wisniewski SR, Balasubramani GK, Zisook S, Kurian B, Warden D, Trivedi MH, Rush AJ. Does early-onset chronic or recurrent major depression impact outcomes with antidepressant medications? A CO-MED trial report. Psychol Med. 2013;43:945–960. doi: 10.1017/S0033291712001742. [DOI] [PubMed] [Google Scholar]
  144. Sung SC, Wisniewski SR, Luther JF, Trivedi MH, Rush AJ. Pre-treatment insomnia as a predictor of single and combination antidepressant outcomes: a CO-MED report. J Affect Disord. 2015;174:157–164. doi: 10.1016/j.jad.2014.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Svenningsson P, Berg L, Matthews D, Ionescu DF, Richards EM, Niciu MJ, Malinger A, Toups M, Manji H, Trivedi MH, Zarate CA, Greengard P. Preliminary evidence that early reduction in p11 levels in natural killer cells and monocytes predicts the likelihood of antidepressant response to chronic citalopram. Molecular psychiatry. 2014;19:962–964. doi: 10.1038/mp.2014.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Szegedi A, Rujescu D, Tadic A, Muller MJ, Kohnen R, Stassen HH, Dahmen N. The catechol-O-methyltransferase Val108/158Met polymorphism affects short-term treatment response to mirtazapine, but not to paroxetine in major depression. The pharmacogenomics journal. 2005;5:49–53. doi: 10.1038/sj.tpj.6500289. [DOI] [PubMed] [Google Scholar]
  147. Tadic A, Muller MJ, Rujescu D, Kohnen R, Stassen HH, Dahmen N, Szegedi A. The MAOA T941G polymorphism and short-term treatment response to mirtazapine and paroxetine in major depression. American journal of medical genetics. Part B, Neuropsychiatric genetics: the official publication of the International Society of Psychiatric Genetics. 2007;144B:325–331. doi: 10.1002/ajmg.b.30462. [DOI] [PubMed] [Google Scholar]
  148. Tian JS, Peng GJ, Gao XX, Zhou YZ, Xing J, Qin XM, Du GH. Dynamic analysis of the endogenous metabolites in depressed patients treated with TCM formula Xiaoyaosan using urinary (1)H NMR-based metabolomics. Journal of ethnopharmacology. 2014;158(Pt A):1–10. doi: 10.1016/j.jep.2014.10.005. [DOI] [PubMed] [Google Scholar]
  149. Tiwari AK, Zai CC, Sajeev G, Arenovich T, Muller DJ, Kennedy JL. Analysis of 34 candidate genes in bupropion and placebo remission. Int J Neuropsychopharmacol. 2013;16:771–781. doi: 10.1017/S1461145712000843. [DOI] [PubMed] [Google Scholar]
  150. Toga AW, Clark KA, Thompson PM, Shattuck DW, Van Horn JD. Mapping the human connectome. Neurosurgery. 2012;71:1–5. doi: 10.1227/NEU.0b013e318258e9ff. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, Ritz L, Nierenberg AA, Lebowitz BD, Biggs MM, Luther JF, Shores-Wilson K, Rush AJ, Team SDS. Medication augmentation after the failure of SSRIs for depression. The New England journal of medicine. 2006a;354:1243–1252. doi: 10.1056/NEJMoa052964. [DOI] [PubMed] [Google Scholar]
  152. Trivedi MH, McGrath PJ, Fava M, Parsey RV, Kurian BT, Phillips ML, Oquendo MA, Bruder G, Pizzagalli D, Toups M, Cooper C, Adams P, Weyandt S, Morris DW, Grannemann BD, Ogden RT, Buckner R, McInnis M, Kraemer HC, Petkova E, Carmody TJ, Weissman MM. Establishing moderators and biosignatures of antidepressant response in clinical care (EMBARC): Rationale and design. J Psychiatr Res. 2016;78:11–23. doi: 10.1016/j.jpsychires.2016.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Trivedi MH, Morris DW, Pan JY, Grannemann BD, John Rush A. What moderator characteristics are associated with better prognosis for depression? Neuropsychiatr Dis Treat. 2005;1:51–57. doi: 10.2147/nedt.1.1.51.52298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, Norquist G, Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK, Fava M. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006b;163:28–40. doi: 10.1176/appi.ajp.163.1.28. [DOI] [PubMed] [Google Scholar]
  155. Tsai SJ, Gau YT, Hong CJ, Liou YJ, Yu YW, Chen TJ. Sexually dimorphic effect of catechol-O-methyltransferase val158met polymorphism on clinical response to fluoxetine in major depressive patients. J Affect Disord. 2009;113:183–187. doi: 10.1016/j.jad.2008.04.017. [DOI] [PubMed] [Google Scholar]
  156. 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 TG, Kalember P, Zobel A, Czerski PM, Larsen ER, Souery D, Giovannini C, Gray JM, Lewis CM, Farmer A, Aitchison KJ, McGuffin P, Craig I. Genetic predictors of response to antidepressants in the GENDEP project. The pharmacogenomics journal. 2009;9:225–233. doi: 10.1038/tpj.2009.12. [DOI] [PubMed] [Google Scholar]
  157. Uher R, Perroud N, Ng MY, Hauser J, Henigsberg N, Maier W, Mors O, Placentino A, Rietschel M, Souery D, Zagar T, Czerski PM, Jerman B, Larsen ER, Schulze TG, Zobel A, Cohen-Woods S, Pirlo K, Butler AW, Muglia P, Barnes MR, Lathrop M, Farmer A, Breen G, Aitchison KJ, Craig I, Lewis CM, McGuffin P. Genome-wide pharmacogenetics of antidepressant response in the GENDEP project. The American journal of psychiatry. 2010;167:555–564. doi: 10.1176/appi.ajp.2009.09070932. [DOI] [PubMed] [Google Scholar]
  158. Uher R, Tansey KE, Dew T, Maier W, Mors O, Hauser J, Dernovsek MZ, Henigsberg N, Souery D, Farmer A, McGuffin P. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. The American journal of psychiatry. 2014;171:1278–1286. doi: 10.1176/appi.ajp.2014.14010094. [DOI] [PubMed] [Google Scholar]
  159. Villasenor A, Ramamoorthy A, Silva dos Santos M, Lorenzo MP, Laje G, Zarate C, Jr, Barbas C, Wainer IW. A pilot study of plasma metabolomic patterns from patients treated with ketamine for bipolar depression: evidence for a response-related difference in mitochondrial networks. Br J Pharmacol. 2014;171:2230–2242. doi: 10.1111/bph.12494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Vittengl JR, Clark LA, Jarrett RB. Deterioration in psychosocial functioning predicts relapse/recurrence after cognitive therapy for depression. Journal of affective disorders. 2009;112:135–143. doi: 10.1016/j.jad.2008.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Vosslamber S, van Baarsen LG, Verweij CL. Pharmacogenomics of IFN-beta in multiple sclerosis: towards a personalized medicine approach. Pharmacogenomics. 2009;10:97–108. doi: 10.2217/14622416.10.1.97. [DOI] [PubMed] [Google Scholar]
  162. Warden D, Rush AJ, Trivedi MH, Fava M, Wisniewski SR. The STAR*D Project results: a comprehensive review of findings. Curr Psychiatry Rep. 2007a;9:449–459. doi: 10.1007/s11920-007-0061-3. [DOI] [PubMed] [Google Scholar]
  163. Warden D, Trivedi MH, Wisniewski SR, Davis L, Nierenberg AA, Gaynes BN, Zisook S, Hollon SD, Balasubramani GK, Howland R, Fava M, Stewart JW, Rush AJ. Predictors of attrition during initial (citalopram) treatment for depression: a STAR*D report. Am J Psychiatry. 2007b;164:1189–1197. doi: 10.1176/appi.ajp.2007.06071225. [DOI] [PubMed] [Google Scholar]
  164. Webb CA, Dillon DG, Pechtel P, Goer FK, Murray L, Huys QJ, Fava M, McGrath PJ, Weissman M, Parsey R, Kurian BT, Adams P, Weyandt S, Trombello JM, Grannemann B, Cooper CM, Deldin P, Tenke C, Trivedi M, Bruder G, Pizzagalli DA. Neural Correlates of Three Promising Endophenotypes of Depression: Evidence from the EMBARC Study. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2016;41:454–463. doi: 10.1038/npp.2015.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Xu HB, Zhang RF, Luo D, Zhou Y, Wang Y, Fang L, Li WJ, Mu J, Zhang L, Zhang Y, Xie P. Comparative proteomic analysis of plasma from major depressive patients: identification of proteins associated with lipid metabolism and immunoregulation. Int J Neuropsychopharmacol. 2012;15:1413–1425. doi: 10.1017/S1461145712000302. [DOI] [PubMed] [Google Scholar]
  166. Young EA, Kornstein SG, Marcus SM, Harvey AT, Warden D, Wisniewski SR, Balasubramani GK, Fava M, Trivedi MH, John Rush A. Sex differences in response to citalopram: a STAR*D report. J Psychiatr Res. 2009;43:503–511. doi: 10.1016/j.jpsychires.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Zhu H, Bogdanov MB, Boyle SH, Matson W, Sharma S, Matson S, Churchill E, Fiehn O, Rush JA, Krishnan RR, Pickering E, Delnomdedieu M, Kaddurah-Daouk R Pharmacometabolomics Research N. Pharmacometabolomics of response to sertraline and to placebo in major depressive disorder - possible role for methoxyindole pathway. PLoS One. 2013;8:e68283. doi: 10.1371/journal.pone.0068283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Zisook S, Lesser I, Stewart JW, Wisniewski SR, Balasubramani GK, Fava M, Gilmer WS, Dresselhaus TR, Thase ME, Nierenberg AA, Trivedi MH, Rush AJ. Effect of age at onset on the course of major depressive disorder. The American journal of psychiatry. 2007;164:1539–1546. doi: 10.1176/appi.ajp.2007.06101757. [DOI] [PubMed] [Google Scholar]

RESOURCES