Review article
Peripheral biomarkers of major depression and antidepressant treatment response: Current knowledge and future outlooks

https://doi.org/10.1016/j.jad.2017.07.001Get rights and content

Highlights

  • We summarize current therapeutic interventions for depression and discuss the need for biomarker identification.

  • We discuss the utility of “omics” screening tools and their applicability to discovering biomarkers.

  • We review studies using blood based “omics” analyses with MDD and treatment response.

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.

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 et al., 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 (Biomarkers Definitions Working Group, 2001, Strimbu and Tavel, 2010). The heterogeneity of MDD necessitates and/or allows for numerous biomarker classifications, as shown in Fig. 1. Diagnostic biomarkers indicate presence and/or future development of disease. Most of the currently-identified biomarkers, described 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.

In this report, we briefly review currently available treatment options 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.

Section snippets

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

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. Fig. 2 denotes the cascade of events necessary for identifying a biomarker, including discovery and validation processing using high- and low-throughput methodology, respectively.

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

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 Fig. 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

Funding

This research was supported in part by the Center for Depression Research and Clinical Care (to MHT) and by the NIMH under Award Number R25MH101078 (to AC).

Financial disclosures

BSG, MKJ, AC, TLM, and MPE report no financial disclosures. JLF has received research funding from the NIA and licensing fees from Regeneron Pharmaceuticals, Janssen Pharmaceuticals, C2N Diagnostics, Treventis Corp., Denali Therapeutics, and ADRx Inc. MHT has received funding support from the Agency for Healthcare Research and Quality (AHRQ), Cyberonics Inc., National Alliance for Research in Schizophrenia and Depression, National Institute of Mental Health (NIMH), National Institute on Drug

Acknowledgements

This research was supported in part by the Center for Depression Research and Clinical Care (to MHT) and by the NIMH under Award Number R25MH101078 (to AC). NIMH had no role in the drafting or review of the manuscript or in the collection or analysis of the data.

References (175)

  • H.A. Garriock et al.

    A genomewide association study of citalopram response in major depressive disorder

    Biol. Psychiatry

    (2010)
  • Y. Gorgulu et al.

    Rapid antidepressant effects of sleep deprivation therapy correlates with serum BDNF changes in major depression

    Brain Res Bull.

    (2009)
  • M.K. Jha et al.

    Can C-reactive protein inform antidepressant medication selection in depressed outpatients? Findings from the CO-MED trial

    Psychoneuroendocrinology

    (2017)
  • M. Kato et al.

    ABCB1 (MDR1) gene polymorphisms are associated with the clinical response to paroxetine in patients with major depressive disorder

    Prog. Neuro-Psychopharmacology and Biological Psychiatry

    (2008)
  • Y.K. Kim et al.

    The influence of stress on neuroinflammation and alterations in brain structure and function in major depressive disorder

    Behav. brain Res.

    (2017)
  • J.B. Kraft et al.

    Analysis of association between the serotonin transporter and antidepressant response in a large clinical sample

    Biol. Psychiatry

    (2007)
  • S.X. Li et al.

    Influence of fluoxetine on the ability of bupropion to modulate extracellular dopamine and norepinephrine concentrations in three mesocorticolimbic areas of rats

    Neuropharmacology

    (2002)
  • P.-Y. Lin et al.

    A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression

    Biol. Psychiatry

    (2010)
  • S. Lista et al.

    Blood and plasma-based proteomic biomarker research in Alzheimer's disease

    Progress. Neurobiol.

    (2013)
  • S. Lucae et al.

    HTR2A gene variation is involved in antidepressant treatment response

    Eur. Neuropsychopharmacol.

    (2010)
  • E. Maron et al.

    Serotonin transporter promoter region polymorphisms do not influence treatment response to escitalopram in patients with major depression

    Eur. Neuropsychopharmacol.

    (2009)
  • R. Abo et al.

    Merging pharmacometabolomics with pharmacogenomics using '1000 Genomes' single-nucleotide polymorphism imputation: selective serotonin reuptake inhibitor response pharmacogenomics

    Pharm. Genom.

    (2012)
  • J.A. Allen et al.

    Lipid raft microdomains and neurotransmitter signalling

    Nat. Rev. Neurosci.

    (2006)
  • S. Anttila et al.

    Catechol-O-methyltransferase (COMT) polymorphisms predict treatment response in electroconvulsive therapy

    Pharm. J.

    (2007)
  • B.A. Arnow et al.

    Depression subtypes in predicting antidepressant response: a report from the iSPOT-D trial

    Am. J. Psychiatry

    (2015)
  • M. Arns et al.

    Frontal and rostral anterior cingulate (rACC) theta EEG in depression: implications for treatment outcome?

    Eur. Neuropsychopharmacol.

    (2015)
  • J.A. Ascher et al.

    Bupropion: a review of its mechanism of antidepressant activity

    J. Clin. Psychiatry

    (1995)
  • J. Assies et al.

    Plasma and erythrocyte fatty acid patterns in patients with recurrent depression: a matched case-control study

    PLoS One

    (2010)
  • S. Ball et al.

    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)
  • B.T. Baune et al.

    Association of the COMT val158met variant with antidepressant treatment response in major depression

    Neuropsychopharmacol.: Off. Publ. Am. Coll. Neuropsychopharmacol.

    (2008)
  • A. Beck et al.

    An inventory for measuring depression

    Arch. General. Psychiatry

    (1961)
  • F. Benedetti et al.

    The catechol-O-methyltransferase Val(108/158)Met polymorphism affects antidepressant response to paroxetine in a naturalistic setting

    Psychopharmacol. (Berl.)

    (2009)
  • S.L. Berger et al.

    An operational definition of epigenetics

    Genes Dev.

    (2009)
  • E.B. Binder et al.

    Polymorphisms in FKBP5 are associated with increased recurrence of depressive episodes and rapid response to antidepressant treatment

    Nat. Genet

    (2004)
  • E.B. Binder et al.

    Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults

    Jama

    (2008)
  • Biomarkers Definitions Working Group

    Biomarkers and surrogate endpoints: preferred definitions and conceptual framework

    Clin. Pharmacol. Ther.

    (2001)
  • J.A. Blumenthal et al.

    Is exercise a viable treatment for depression?

    ACSMs Health Fit. J.

    (2012)
  • M. Bot et al.

    Serum proteomic profiling of major depressive disorder

    Transl. Psychiatry

    (2015)
  • C. Bridle et al.

    Effect of exercise on depression severity in older people: systematic review and meta-analysis of randomised controlled trials

    Br. J. Psychiatry

    (2012)
  • C. Burton et al.

    Restarting antidepressant treatment following early discontinuation-a primary care database study

    Fam. Pract.

    (2015)
  • H.N. Chan et al.

    Correlates and outcomes of depressed out-patients with greater and fewer anxious symptoms: a CO-MED report

    Int J. Neuropsychopharmacol.

    (2012)
  • H.W. Chase et al.

    Accounting for dynamic fluctuations across time when examining fMRI test-retest reliability: analysis of a reward paradigm in the EMBARC study

    PloS One

    (2015)
  • C.S. Chen et al.

    Protein microarrays

    BioTechniques

    (2006)
  • W.E. Craighead et al.

    Combination psychotherapy and antidepressant medication treatment for depression: for whom, when, and how

    Annu. Rev. Psychol.

    (2014)
  • A.H. Czysz et al.

    G-protein signaling, lipid rafts and the possible sites of action for the antidepressant effects of n-3 polyunsaturated fatty acids

    CNS Neurol. Disord. Drug Targets

    (2013)
  • T.M. Daly EJ et al.

    Health-related quality of life in depression: a STAR*D report

    Ann. Clin. Psychiatry

    (2010)
  • I. D'Empaire et al.

    Antidepressant treatment and altered CYP2D6 activity: are pharmacokinetic variations clinically relevant?

    J. Psychiatr. Pract.

    (2011)
  • B.S. Diniz et al.

    Plasma biosignature and brain pathology related to persistent cognitive impairment in late-life depression

    Mol. Psychiatry

    (2015)
  • K. Domschke et al.

    COMT val158met influence on electroconvulsive therapy response in major depression

    Am. J. Med. Genet. Part B: Neuropsychiatr. Genet.

    (2010)
  • K. Domschke et al.

    Serotonin transporter gene hypomethylation predicts impaired antidepressant treatment response

    Int J. Neuropsychopharmacol.

    (2014)
  • Cited by (0)

    View full text