Skip to main content
Dialogues in Clinical Neuroscience logoLink to Dialogues in Clinical Neuroscience
. 2015 Sep;17(3):305–317. doi: 10.31887/DCNS.2015.17.3/emaron

Biological predictors of pharmacological therapy in anxiety disorders

Predictores biológicos para la farmacoterapia en los trastornos de ansiedad

Prédicteurs biologiques du traitement pharmacologique dans les troubles anxieux

Eduard Maron 1,*, David Nutt 2
PMCID: PMC4610615  PMID: 26487811

Abstract

At least one third of patients with anxiety disorders do not adequately respond to available pharmacological treatment. The reason that some patients with anxiety disorders respond well, but others not, to the same classes of medication is not yet fully understood. It is suggested that several biological factors may influence treatment mechanisms in anxiety and therefore could be identified as possible biomarkers predicting treatment response. In this review, we look at current evidence exploring different types of treatment predictors, including neuroimaging, genetic factors, and blood-related measures, which could open up novel perspectives in clinical management of patients with anxiety disorders.

Keywords: biomarker, treatment predictor, brain imaging, gene, anxiety disorder, antidepressant

Introduction

Anxiety disorders, including generalized anxiety disorder (GAD), panic disorder (PD), social anxiety disorder (SAD), simple phobias (SP), obsessive-compulsive disorder (OCD), and post-traumatic stress disorder (PTSD), are the most common psychiatric illnesses experienced, affecting an estimated 18% of people in the United States, according to epidemiological studies.1 Although both OCD and PTSD are still considered by many to be anxiety disorders and have been classified as such in the past, they have been removed from the category in the most recent (5th) edition of the American Psychiatric Association's Diagnostic and Statistical Manual of Mental Disorders (DSM-5) and are now described in different chapters (Obsessive-Compulsive and Related Disorders and Trauma and Stressor-Related Disorders, respectively).2 The age of onset of anxiety disorders is usually in the mid-twenties, and women are twice as likely as men to be diagnosed with an anxiety condition, but how much of this sex difference is due to socioeconomic factors has not been established.3 In additional, anxiety disorders are characterized by a chronic and fluctuating clinical course, and can be seriously disabling diseases causing impairment in social, personal, and occupational functioning, as well as leading to significant loss in quality of life and to an enormous social cost.4 As demonstrated in recent studies, the anxiety disorders are the most costly in the USA, amounting to 46.6 billion, or 31.5% of the total economic costs of mental disorders.5 Therefore, the treatment of these disorders is one of the current problems in medicine today.6 In community studies, patients with anxiety disorders were found to use more medical and psychiatric services than control populations.7 The availability of more effective, relatively low-cost outpatient treatment could substantially reduce the economic and social burden of these common and often crippling disorders.8

Nevertheless, anxiety disorders are notoriously difficult to successfully treat, and a variety of genetic and environmental factors contribute to their development and severity.9 The importance of an improved approach to treating anxiety is highlighted by the inconsistent results seen with the classes of drugs considered to be the contemporary first-line treatment: selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs). Although these agents have been shown to be beneficial for the treatment of certain anxiety disorders, not all patients achieve an adequate clinical therapeutic response. For example, in a 3-year follow-up only 10% of patients with PD were symptom-free10 and only 12% of PD patients were in full remission after 5 years.11 The existence of “nonresponders” indicates that SSRIs and SNRIs are not the failsafe solution to treating anxiety that clinicians have been looking for.3 In addition, SSRIs and SNRIs are associated with complications that can limit their use in some patients, including delays in producing the desired clinical reduction in anxiety or even potential worsening of the anxiety particularly at the start of treatment leading to dropouts.12 Other issues include the risk for discontinuation syndromes in noncompliant patient populations. Furthermore, some SSRIs (eg, citalopram) are not suitable for patients with heart problems.13 Another class of drugs, benzodiazepine (BZD) anxiolytics, has played a central role in the pharmacologic management of anxiety disorders for about 50 years. Although not as widely prescribed as in the past, these compounds nevertheless remain an effective alternative to SSRIs; however, the chronic usage of BZDs is strongly restricted in clinical practice due to dependence risk.14

All things considered, the antidepressants, and first of all the SSRIs, are more preferable pharmacological agents in treatment of anxiety disorders, even if a substantial proportion of patients are not achieving significant improvement and remission during medication. Also it is rather difficult in routine practice to predict which patients will respond well to a pharmacological treatment and which will not. So similarly to in depression, treatment in patients with anxiety disorders is usually chosen on an empirical basis, where clinical approaches are largely trial and error, and when the first treatment does not result in recovery for the patient, there is little proven scientific basis for choosing the next.15 Once treatment has begun, an improvement in clinical symptoms early in the course of therapy generally points towards an eventual good treatment response. However, in most cases, efficacy needs to be evaluated after 6 to 12 weeks of treatment, and a large proportion of patients have persistent symptoms despite a full treatment trial.16 Current treatment guidelines recommend that an initial treatment be tried for long enough a period to determine how much it will benefit a patient.15 On average, at least 4 weeks are needed to attain response, and 6 weeks to attain remission during treatment with an initial SSRI antidepressant, but remission can take 12 weeks or longer.17 Because most patients fail to enter remission with the first antidepressant prescribed,17 they then commonly enter a period of serial trial-and-error treatments using switches between, or combinations of, medications.18 Typically it can take 1 year or more to hit upon a successful treatment.19,20 It is not surprising that using this “hit-or-miss” approach, 26% of those who fail to improve with the first treatment simply stop taking their medication, frequently within the first 2 weeks,21 and up to 42% of patients discontinue medication within the first 30 days.22 Also, this unsuccessful use of antidepressants significantly increases the total cost of treatment that in its turn significantly worsens the overall socioeconomic condition.

There are a vast number of studies that have sought to identify predictors of response to antidepressant medication. Putative predictors include demographic and clinical characteristics, personality traits, biological markers, and psychophysiological features.23,24 Overall, these studies have not as yet yielded findings that were robust enough to be clinically relevant.25,26

However, there is strong belief that further search for biological predictors of treatment response, which are also defined as “treatment biomarkers,” would contribute to the personalized medicine approach, in which biomarkers would guide decision making and help to select the most suitable medication for individual patients. Moreover, incorporation of predicting biomarkers into antidepressant treatment algorithms could speed recovery from disease by shortening or eliminating lengthy and ineffective trials.15 During the last decade there has been intensive ongoing research of biomarkers predicting treatment outcome of antidepressants. To date, the majority of this research was conducted in patients with depression, and there are only a small number of published studies in which treatment biomarkers were explored in patients with any anxiety disorder, which means that biomarker discovery is still on very early stage in anxiety field. Taken into account that the same antidepressant groups, like SSRIs and SNRIs, are highly effective in both depression and anxiety, we will focus here on available data and promising findings from treatment biomarker research in both clinical conditions. Also, keeping in mind that the aforementioned antidepressants may have similar or overlapping treatment mechanisms in depression and anxiety, we suggest that current achievements in biomarker discovery in depression could be applicable for further research in anxiety disorders and may improve our understanding about perspectives in this important area. It should, however, be noted that at the present time none of the biomarkers have sufficiently proven utility to be ready for clinical application, due to their low sensitivity- and specificity.27 Nevertheless, the several classes of biomarkers, including brain structural or functional findings, as well as genomic, proteomic, and metabolomic measures, have been identified by previous research, whereas some of them have shown clear promise for predicting treatment response.15 So, we will review the available evidence supporting the use of different types of treatment predictive biomarkers. For a simplified summary, the key findings are summarized in Table I.

TABLE I. Summary of positive findings for treatment biomarkers in anxiety disorders. ADs, antidepressants; CBT, cognitive behavioral therapy; PE, prolonged exposure therapy; BEP, brief eclectic psychotherapy; w, week; Hip, hippocampus; ACC, anterior cingulate cortex; pgACC, pregenual ACC; rACC, rostral ACC; sACC, subgenual ACC; mlOFC, middle lateral orbitofrontal cortex; mPFC, medial prefrontal cortex; GMV, gray matter volume; dlPFC, dorsolateral prefrontal cortex; BOLD, blood-oxygen-level dependent; OFC, orbitofrontal cortex; iPL, inferior parietal lobe; vACC, ventral anterior cingulate cortex; VFC, ventral frontal cortex; sTG, superior temporal gyrus; TC, temporal cortex; midFC, mid frontal cortex; vmPFC, ventromedial prefrontal cortex; PCC, posterior cingulate cortex; rCBF, regional cerebral blood flow; rCMRGlc, regional glucose metabolism; 99mTc-HMPAO, 99mTc-hexamethylpropyleneamineoxime; 5-HTT, serotonin transporter; 5-HTR1A, serotonin receptor 1A; 5-HTR2A, serotonin receptor 2A; RGS2, regulator of G-protein signaling 2; PACAP, the pituitary adenylate cyclase-activating peptide; CRHR1, corticotropin-releasing hormone receptor 1; DRD3, dopamine receptor D3; NR3C1, nuclear receptor subfamily group C, member 1; PDE1A, phosphodiesterase 1 A; DISP1, dispatched homolog 1; GRIN2B, glutamate receptor subunit epsilon-2; PCDH10, protocadherin 10; GPC6, glypican 6; DHEA, dehydroepiandrosterone; BDNF, brain-derived neurotrophic factor; OCD, obsessive-compulsive disorder; PD, panic disorder; PTSD, post-traumatic stress disorder; GAD, generalized anxiety disorder; Amy, amygdala.

Sample Treatment Predictor Outcome Reference
Neuroimaging biomarkers
PTSD (n=39) CBT (12w) Greater right Hip Response 40
PTSD (n=13) CBT (8w) Larger rACC Response 41
PTSD (n=30) CBT (36) Right sACC Response 42
OCD (n=14) SSRI (12w) Smaller right mlOFC Response 46
OCD (n=15) CBT (12w) Larger right mPFC
OCD (n=15) Surgery ACC Response 47
PD (n=21) SSRI (6w) Total GMV Response 43
PD (n=49) CBT (12w) BOLD in ACC-Amy Response 55
PD (n=14) CBT (4w) Increased BOLD in insula and dlPFC Response 57
PD (n=23) GAD (n=25) CBT (10w) Greater BOLD in cortico-limbic circuitry Response 58
GAD (n=15) SNRI (8w) Greater BOLD in ACC and lower in Amy Response 59, 60
SAnD (n=21) CBT (12w) Greater BOLD in Amy-pgACC Response 62
PTSD (n=21) SSRI (12w) BOLD in precuneus and OFC Response 65
PTSD (n=41) CBT (32w) Greater BOLD in left iPL Response 66
PTSD (n=13) CBT (8w) Greater BOLD in Amy-vACC Non-response 41
OCD (n=25) SSRI (16w) BOLD in right VFC Response 67
OCD (n=17) SSRI (12w) BOLD in leftsTG Response 68
PD (n=15) SSRI (12w) Global cortical rCMRGlc Response 75
SAD (n=72) SSRI (8w) rCBF in Amy, dlPFC and rostral ACC Response 76
SAD (n=15) SSRI (8w) Higher rCBF in left TC and left midFC Non-response 77
SAD (n=12) Tiagabine (6w) Lower rCMRglu in vmPFC Response 78
OCD (n=30) SSRS (24w) Global change in rCBF Response 79
OCD (n=15) SSRS (12w) Higher whole brain 99mTc-HMPAO Response 80
OCD (n=9) SSRS (12w) Higher rCBF in PCC and lower in OFC Response 81
Genetic biomarkers
PD (n=92) SSRI (12w) 5-HTTLPR LL Response 99
PD (n=102) SSRIs (6w) 5-HTR1A-1019GG Response 100
PTSD (n=330) SSRI (12w) 5-HTTLPR LL Response Low dropout 101
SAD (n=32) SSRI (12w) 5-HTTLPR LL Response 102
SAD (n=346) SSRI (10w) RGS2 SNPs Response 104
GAD (n=112) SNRI (24w) PACAP rs2856966 A Response 105
GAD (n=112) SNRI (24w) 5-HTTLPR LL Response 107
GAD (n=156) SNRI (24w) 5-HTR2Ars7997012G Response 109
GAD (n=164) SNRI (6-12w) CRHR1; DRD3; NR3C1; PDE1A Response 111
OCD (n=184) Various ADs (>10w) CYP2D6 non-extensive Number of failed trials 121
OCD (n=804) SSRIs DISP1; GRIN2B, PCDH10; GPC6 Response 122
Blood-based biomarkers
PTSD (n=21) BEP (16w) Increased DHEA Response 116
PTSD (n=28) PE (12w) Decreased cortisol Non-response 117
OCD (n=19) SSRI (8w) Higher whole-blood serotonin Response 118
PD (n=22) SSRI (12w) Lower beta-adrenoceptor affinity Non-response 119
PD (n=42) CBT (10w) Lower BDNF Non-response 120

Neuroimaging

Many of the major advances in biomarker research have arisen from advances in neuroimaging technology, including structural (magnetic resonance imaging [MRI], diffusion tensor imaging [DTI]), functional (functional magnetic resonance imaging [fMRI], positron emission tomography [PET]/single photon emission computed tomography [SPECT], biochemical (magnetic resonance spectroscopy [MRS]), and neurophysiological (electroencephalography [EEG] and magnetoencephalography [MEG]) methodologies.28 These different neuroimaging techniques have all been used to study whether baseline, pretreatment characteristics, or changes in brain functioning and metabolism correlate with symptom improvement following antidepressant treatment.27 For example, the structural brain imaging studies have demonstrated usefulness as pretreatment predictors of antidepressant treatment outcome.15 Recent meta-analyses of structural neuroimaging studies indicate that depressed patients have reduced gray matter volume (GMV) in multiple areas, including the anterior cinguiate cortex (ACC) ,29 subgenual cinguiate cortex,30 and hippocampus.31 The most robust evidence is for the hippocampus, for which larger volumes predicted better response after 8 weeks of pharmacotherapy in two separate samples.32,33 In addition, the GMV in the ACC and posterior cinguiate cortex was also predictive of clinical remission following 8 weeks of fluoxetine treatment.34 In 1997 Mayberg et al35 reported, using PET, that responders to antidepressant treatment showed increased anterior cinguiate metabolism at baseline relative to nonresponders and to healthy controls. The diffusion tensor imaging is another relatively novel method, which applies the structural integrity of the fiber tracts between affected neural areas and has predictive potential for delineating treatment responders. For example, depressive nonresponders to 12 weeks of citalopram36 or escitalopram treatment37 showed a greater prevalence of microstructural abnormalities in white matter pathways connecting the cortex with limbic and paralimbic areas such as the anterior cinguiate. Other studies have also demonstrated that the integrity of these corticolimbic pathways is adversely impacted by adverse life events38 as well as genetic polymorphisms (eg, 5-HTTLPR).39 Despite being based on straightforward quick and simple methodology, only a few structural MRI studies have been designed to explore brain markers of therapeutic response in anxiety disorders. Even those available have mostly applied psychotherapy interventions rather than pharmacological medication and did not directly aim to identify structural brain predictors of the treatment outcome. In part this may reflect the fear that many patients with anxiety disorders have for the confined space of the MRI scanner. However, when studies have been conducted they have been informative. For example, structural MRI studies in PTSD have reported that better response to cognitive behavioral therapy (CBT) is predicted by greater right hippocampal GMV40 and larger rostral ACC volume.41 Both regions are linked to threat processing and fear memory activation, and seem to be involved in mediation of CBT effectiveness. The other study in PTSD showed that symptom improvement was correlated with, and predicted by, cortical thickness in the right subgenual ACC.42 Recently, the relatively small studies in patients with PD have demonstrated that remission following 6 weeks of escitalopram treatment is accompanied by both significant increase in the GMV in the left superior frontal gyrus, but reduction in the right precentral gyrus. Furthermore, the changes in total GMV after remission were correlated with changes in clinical scores.43 Another study using DTI found increased white matter micro-structural integrity reflected by fractional anisotropy in some regions of right uncinate fasciculus and left fronto-occipital fasciculus after escitalopram remission in PD patients.44 Earlier, increases of GMV were showed in left infero-frontal cortex, right fusiform gyrus, and right cerebellum areas in remitted depressive patients with comorbid PD following 6 weeks' medication with duloxetine.45 None of these studies have specifically focused on a predictive effect of brain structural measures on treatment response. The only study to do this was that of Iloexter et al46 who investigated structural MRI correlates as potential pretreatmentbrain markers to predict treatment response in treatment-naive OCD patients, entering a randomized 12-week clinical trial of either fluoxetine or group-based CBT. This study showed that symptom improvement in the fluoxetine treatment group was significantly correlated with smaller pretreatment GMV within the right middle lateral orbitofrontal cortex (OFC), whereas symptom improvement in the CBT treatment group was significantly correlated with larger pretreatment GMV within the right medial prefrontal cortex (mPFC). Although, these findings suggest that pretreatment GMV of distinct brain regions within the lateral OFC and mPFC were differentially correlated with treatment response to fluoxetine versus CBT in OCD patients, the included sample was quiet small and needs replication in larger sets of patients with a prospective design. Additional data is provided by Banks et al47 who revealed features of ACC structure and connectivity that predict clinical response to dorsal anterior cingulotomy for refractory OCD. They suggested that the variability seen in individual responses to a highly consistent, stereotyped procedure may be due to neuroanatomical variation in the patients.

The functional MRI (fMRI) approaches, including task-related and resting-state methods, can provide an additional and more comprehensive view on involvement of anxiety neurocircuits in prediction of treatment outcome. The fMRI measures of neuronal activity in mood-regulating pathways were successfully used in studies exploring brain functional predictors of response to antidepressant medication. For example, Anand et al48 were the first to show that reduced corticolimbic connectivity in depression recovers during the course of treatment and may thus predict antidepressant medication response. The studies, which applied the measure of neuronal response to an emotional task, like processing of facial expressions, also identified that baseline neural reactivity can be used as predictors of treatment outcome. So, increased baseline neuronal reactivity found in depressive patients seems to normalize after successful antidepressant pharmacotherapy49,50 and CBT.51 Also particularly the deficient connections between the amygdala and anterior cinguiate have been found to be ameliorated after 8 weeks of fluoxetine administration,52 and another study showed that the changes in task-related reactivity were complementary- to differences between treatment responders and nonresponders in resting-state connectivity within corticolimbic circuits.53 A recent fMRI study with a large enough sample of patients with PD comorbid with agoraphobia has failed to find any brain regions predictive of CBT outcome.54 However, earlier Leuken et al55 observed that treatment response to CBT in panic patients was associated with an inhibitory functional coupling between the anterior cinguiate cortex and the amygdala, whereas responders and nonresponders were characterized by distinct neuronal activation at baseline time. Their later study in the same sample has demonstrated that inhibitory- ACC-amygdala coupling during fear conditioning was associated with the long variant of the 5-HTTLPR polymorphism in PD responders only. This points toward potential intermediate connectivity- phenotype modulating response to exposure-based CBT.56 A better response to brief CBT was predicted by increased pretreatment activation in bilateral insula and left dorsolateral prefrontal cortex (dlPFC) during threat processing in a recent study by Reinecke et al.57 In addition, the greater activation in cortico-limbic circuitry, including superior frontal gyri, anterior insula, superior temporal and supramarginal gyri and hippocampus, predicted better CBT response in mixed samples of patients with PD and GAD.58 Regarding pharmacological intervention in PD, the remission following escitalopram treatment was associated with changes in regional homogeneity of temporoparietal regions in a recent fMRI study, which, however, did not specifically explore predictive measures of treatment response.43 Pharmaco-fMRI research has not been very fruitful, but there is some evidence in small samples of patients with GAD that greater levels of pretreatment ACC activity and lesser reactivity in the amygdala in anticipation of facial presentation were associated with better reductions in anxiety and worry symptoms after 8 weeks' treatment with venlafaxine. This suggests that ACC-amygdala responsivity could prove useful as a predictor of antidepressant treatment response in GAD.59,60 Interestingly, Pantazatos et al61 reported that SAD is characterized by reduced functional connectivity in left hippocampus-left temporal pole, and this feature has discriminated SAD from both PD and healthy controls and has increased following the 8 weeks' treatment with paroxetine. This study suggests promise for emerging functional connectivity-based biomarkers for SAD diagnosis and pharmacological treatment effects. In addition, greater right amygdala-pregenual ACC (pgACC) connectivity and greater left amygdala-pgACC coupling encompassing medial prefrontal cortex predicted better symptom improvement after CBT in patients with generalized SAD.62 This is in good accordance with previous observations that pretreatment cortical hyperactivity to social threat signals can predict CBT success in this disorder.63,64 The resting-state fMRI study in PTSD patients has revealed that regional spontaneous activity of precuneus and OFC could be a potential prognostic indicator for chronic treatment with antidepressants.65 In another study PTSD responders to psychotherapy showed increased pretreatment activation of the left inferior parietal lobe during contextual cue processing compared with nonresponders. This activation predicted percentage symptom improvement and therefore could be served as a valuable predictive biomarker for PTSD treatment response.66 The earlier fMRI studies had demonstrated that poor improvement after a CBT course is associated with greater bilateral amygdala and ventral anterior cinguiate activation in response to masked fearful faces, suggesting that excessive fear processing of emotional stimuli may be a key factor in limiting responses to psychotherapy of PTSD.41 Finally, promising biomarkers for the effects of SSRIs were recently identified by fMRI approaches in patients with OCD. As was reported, the response to chronic medication with SSRI medications is correlated with changes in connectivity degree in right ventral frontal cortex67 as well as activation in the right cerebellum and in the left superior temporal gyrus.68

Radiotracer brain imaging techniques with SPECT and PET can contribute to further understanding of the biomarkers underlying mechanisms and efficacy of therapeutic intervention.69 A number of studies have examined changes of regional cerebral blood flow (rCBF), regional glucose metabolism (rCMRGlc) or key proteins of neurochemical systems, including serotonin, dopamine, and γ-aminobutyric acid (GABA), in psychiatric patients. However, again, up to now, a very limited number of studies using SPECT or PET have specifically explored the predictive value of the aforementioned functional imaging with regard to the treatment response to antidepressant medication, with the most fruitful findings raised from studies in depression. For example, the treatment response to SSRI antidepressants in depression was strongly predicted by pretreatment binding of serotonin transporters in several mood-related brain regions, including diencephalic structures, bilateral habenula, amygdala-hippocampus complex, and subgenual cingulate cortex70,71 as well as by rCBF and rCMRglu measures in some other brain regions.72,73 In addition, Parsey et al74 have reported that the predictive effect of serotonin 1A receptor binding potential to treatment outcome of antidepressant in depression is modulated by a specific gene variant or polymorphism. As regards the evidence in anxiety disorders, the increase of rCMRGlc was detected in global neocortical areas as well as limbic areas after 12 weeks of escitalopram treatment in PD responders, but not in nonresponders. However, both groups showed pretreatment metabolic reductions which would have unclear predictive value.75 The different treatment-induced coactivations of rCBF between the left amygdala and the dlPFC as well as the rostral ACC were observed among responders and nonresponders to SSRIs and placebo in large samples of SAD and can be considered as useful neuromarkers, differentiating between successful and unsuccessful anxiolytic treatments.76 The earlier study with a small sample of patients with SAD has reported that nonresponders to SSRI medication had higher rCBF at baseline in the anterior and lateral part of the left temporal cortex and the lateral part of the left midfrontal regions as compared with responders.77 Additionally, the magnitude of treatment response to tiagabine, was inversely correlated with pretreatment rCMRglu within vmPFC in patients with generalized SAnD.78 Recently, the large study in OCD has suggested that baseline increased rCBF in forebrain regions and decreased perfusion in posterior brain regions can be potential predictors of treatment response to monotherapy or combined medication with SSRIs.79 Furthermore, the higher whole brain perfusion80 as well as higher rCBF values in posterior cinguiate cortex, but lower in OFC81 have predicted better response to 12 weeks' medication with fluvoxamine in OCD patients, whereas responders, but not nonresponders, showed increased rCBF in the thalamus after treatment, indicating an important role of the thalamic area in drug response.80 In contrast, no pattern of baseline activation that distinguished responders from nonresponders to subsequent SSRI pharmacotherapy was detected in another SPECT perfusion study, which included SAD, OCD, and PTSD samples.82 This study is in good accord with the results of Seedat et al83 who also observed no significant pretreatment differences between PTSD responders and nonresponders in anterior cinguiate perfusion, and even deactivation in the left medial temporal cortex following medication with citalopram, irrespective of clinical response. Interestingly, these conflicting pieces of evidence drawn from imaging studies with pharmacological treatment are in line with results from trials that have applied psychological interventions. So no significant differences in pretreatment rCBF were observed between responders and nonresponders to behavior therapy, however the post-treatment rCBF values in the left medial prefrontal cortex and bilateral middle frontal gyri were significantly lower in the responders than in the nonresponders.84 In addition, no correlation was found between the changes in rCBF and clinical measures in PD patients completed CBT course.85; however, treatment effects of brief eclectic psychotherapy on PTSD symptoms correlated positively with activation in the left superior temporal gyrus, and superior/ middle frontal gyrus.86

As regards SPECT and PET studies of neurochemical systems, several research groups have revealed state changes in availability or density of neurotransmitter receptors and transporters in brain of patients with anxiety disorders; however, the predictive value of these changes on antidepressant response has still not been established.87-89 Finally, other neuroimaging methods, like MR spectroscopy, electroencephalography, and magnetoencephalography, showed promising advantage in detection of biomarkers for pharmacoresponse in depression90-93 and can thus open up new fields in further research of treatment predictors in anxiety disorders.

Genetics

There is a belief that responsiveness to or tolerability of treatment may be influenced by inherited factors. In particular, pharmacogenetic investigations have suggested that differences in antidepressant efficacy can be determined by certain genetic variations inpatients. These studies have multiplied in the past decade due to the improved technology for measuring gene variants with the potential impact that finding reliable genetic predictors might markedly have on everyday clinical practice.94,95 However, the best strategy to detect the key markers involved has not been clearly identified, since both candidate gene studies and genome-wide association studies (GWAS) have provided results which fell short of the expectations.96 To date the majority of candidate-gene studies in the antidepressant field have been conducted in patients with depression and have investigated metabolism-related genes, those that code for receptors and transporters, and those related to second-messenger systems. The most promising results in the pharmacokinetic field have been reported for genetic variations of genes coding for CYP2D6 and Pglycoprotein, although comparative evidence between different drugs is present only for the CYP2D6 gene variants.97 A complicating factor in relation to drug actions in the brain is that the number of potential pharmacodynamic targets appears to be quite large. The most important of a very long list currently appear to be genes coding for tryptophan hydroxylase, catecholO-methyltransf erase (COMT), monoamine oxidase A (MAOA), serotonin transporter (5-HTT), norepinephrine transporter (NET), dopamine transporter (DAT), monoamine receptors (5-HT1A, 5-HT2A, 5-HT6, 5-HT3A, 5-HT3B, β1 adrenoceptor), dopamine (DA) receptors, G protein β3 subunit, corticotropin-releasing hormone (CRH) receptor I (CRHR1), glucocorticoid receptor, angiotensin-converting enzyme, circadian locomotor output cycles kaput (CLOCK), nitric oxide synthase, interleukin (IL)-iβ, and brain-derived neurotrophic factor (BDNF).98

Recently, the GWAS approach has been proposed as having a powerful advantage in overcoming the major limitations of candidate gene studies, as they allow us to genotype the hundreds of thousands of polymorphisms across the whole genome, skipping the need for a priori hypothesis and providing novel information about involvement of genetic variation in the phenotype or process of interest. The available GWAS so far, and their meta-analyses, suggest that some single nucleotide polymorphisms' (SNPs) including in 5-HT2A, UBE3C, uronyl-2-sulfotransferase and interleukin-11 genes, can be useful markers of treatment response to antidepressant medication in depressive patients.15 In contrast to the wealth of pharmacogenetic studies in depression, the research of genetic markers of treatment response in anxiety disorders is still limited to single, nonreplicated candidate-gene trials in relatively small samples. So, two available studies in PD have demonstrated that better response to SSRI treatment is predicted by L-form of 5-HTTLPR and the 5-HT1A receptor -1019C/G polymorphism, respectively.99,100 The LL genotype of 5-HTTLPR was also associated with greater responsiveness to sertraline treatment and with lower dropouts due to adverse events among PTSD patients.101 In addition, reduction in social anxiety symptoms during SSRI treatment was significantly associated with 5 HTTLPR genotype in very small samples.102 However none of three gene candidates, (5-HTTLPR, COMT and TPH2) predicted the response to CBT in another, larger study.103 Recently, the most powerful study (in 346 patients) with SAnD has found that two of the four RGS2 SNPs predicted remission to sertraline treatment, what suggests that this gene can be a biomarker of the likelihood of substantially benefiting from SSRI medication among patients with social phobia.104 An intensive search for genetic predictors has been also conducted in GAD, where a few genes, including the pituitary adenylate cyclase-activating peptide (PACAP), 5-HTTLPR, the serotonin 2A receptor gene (HTR2A), corticotropin-releasing hormone receptor 1 (CRHRl), dopamine receptor D3 (DRD3), nuclear receptor subfamily group C, member 1 (NR3C1) and phosphodiesterase 1A (PDE1A), were found as potential markers predicting treatment response to venlafaxine XR or duloxetine medication.105-111 However, the most intriguing pharmacogenetic findings involving both pharmacokinetic and pharmacodynamic lines of evidence have emerged from studies in OCD and have been reviewed in more detail in very comprehensive paper by Zai et al.112 As they summarized, only two CYP450 liver enzyme genes, CYP2D6 and CYP2C19, have been studied in relation to the antidepressant response in OCD. This showed that nonresponders appear to be more common among non-extensive metabolizers according to genetic status of CYP2D6, suggesting that genes regulating metabolism of drugs play an important role in treatment response. As regarding the pharmacodynamic studies in OCD, then available data are still inconsistent, preliminary, or not yet replicated in independent, well-powered samples. Among various candidate genes a number of those related to serotonin, glutamate, dopamine systems and neurotrophic factors have been identified as promising genetic predictors of treatment response to antidepressants in OCD.112 Furthermore, OCD remains the only single anxiety disorder, where the GWAS approach was applied to detect novel biomarkers of treatment response. Many new loci were identified as top hits in the recent GWAS of antidepressant response in OCD patients, including the GRIN2B, GPC6, DISP1, ANKFN1, ARRDC4, TIAM1, PCDH1D, LOCT30101, and PCDH10 genes; however, a great deal of further research is required to clarify their functional status and their potential role in the treatment response.112

Rapid progress in genomic research and bioinformatics promises to provide the technology for far more explorative and powerful approaches in discovery of novel biomarkers, for example via transcriptional and microRNA analyzing. So, microarray studies of peripheral gene transcription signatures have suggested a shared expression of the majority of genes in brain and peripheral blood, and have identified unique gene expression profiles in patients with depression and anxiety disorders, and healthy subjects vulnerable to anxiety challenge tests. In our recent study we used the Illumina microarray platform for whole genome expression profiling in depressive patients receiving 12 weeks of medication with escitalopram. We found that the magnitude of the expression of some specific genes, YWHAZ, NR2C2, ZNF641 and FKBP1A, responsible for neurotrophin, immune and actin-related processes, differentiated responders and nonresponders to medication and could be used as earlier predictors of treatment outcome.113 In addition, we conducted the first application of full exome sequencing for the analysis of pharmacogenomics on antidepressant treatment in depressive patients and discovered that the A allele of rs41271330 variation in the bone morphogenetic protein (BMP5) gene is a strong predictor of treatment resistance to escitalopram therapy in depression.114 Taken together these both methods seem highly- promising in further search of treatment biomarkers in anxiety disorders.

Blood-based biomarkers

Plasma appears to be a rational source for proteomic and metabolomic measurements in psychiatric conditions because it is easily accessible, and several molecules from the brain are transported across the blood-brain barrier and reach the circulation. However, the blood-based biomarkers of treatment response in psychiatric disorders remain in very early stages of development and none have demonstrated reliability for predicting pharmacological outcome.15 This is particularly true for anxiety disorders, where only a few studies have been performed on plasma-based treatment predictors. Due to the strong involvement of stressrelated systems in PTSD neurobiology, the neuroendocrine measures of treatment response have been used in PTSD patients and provided some proof of concept.115 For example, Olff et al116 found an increase in dehydroepiandrosterone (DHEA) levels among those who responded to treatment for PTSD. Another study found that nonresponders to treatment for PTSD resulting from the 9/11 World Trade Center terrorist attacks had significant declines in post-treatment cortisol levels that distinguished them from responders.117 The measuring of peripheral serotonergic parameters, like whole-blood serotonin concentration, platelet serotonin transporter, 5-HT2A receptor binding characteristics and platelet IP3 content, is the oldest classical approach, which has identified some predictors of clinical outcome of the treatment in OCD patients medicated with SSRIs. In particular, Delorme et al118 have reported that higher whole-blood serotonin concentration is predictive of better improvement to 8 weeks' treatment with SSRIs in OCD patients. In addition the β-adrenoceptor affinity (1/Kd) was decreased and adaptively normalized after treatment with paroxetine in the acute panic patients. Also a low pretreatment β-adrenoceptor affinity was found to predict the treatment response to paroxetine in patients with PD, and was suggested as a biomarker of pharmacological outcome in PD.119 Finally, a lower concentration of serum brain-derived neurotrophic factor (BDNF) was associated with poor response to CBT in patients with PD, suggesting that BDNF might contribute to therapeutic response in panic disorder. However, further clinical trials with pharmacological intervention are required to confirm this suggestion.120

Conclusions

Overall, there is increasing effort to find reliable biomarkers for prediction of treatment outcome in anxiety disorders. However, as with research in disorders close to anxiety disorders such as depression, there are still not any biologic or genetic predictors of sufficient clinical utility to inform the selection of a specific pharmacological compound for an individual patient, because of low sensitivity and specificity of suggested biomarkers.26 Moreover, the multiple limitations of available studies make the picture of biological predictors of pharmacological response in anxiety far from complete. Most of these studies have been conducted in small samples and had clear shortcomings in their clinical design and treatment outcome assessment. Furthermore, different anxiety disorders are not equally covered by research interest and some particular phenotypes have been almost ignored by investigators to date. All of these limitations significantly complicate our understanding about the role of biomarkers in treatment prediction. Nevertheless, there is strong belief that advanced approaches, including neuroimaging and genetic and proteomic techniques, as well as their combination, should contribute to novel discovery of treatment predictors in anxiety disorders. The one facilitating factor for further research in this area could be previous achievements in depression due to the fact that both conditions, depression and anxiety, are highly comorbid with each other, and both respond well to the same classes of antidepressant medications. So, the existing data obtained by research in depression may stimulate further projects and studies in anxiety disorders. First of all, it will help to validate already known biological predictors of therapeutic outcome to pharmacological intervention, and secondly it will direct us toward the identification of unique biomarkers underlying treatment response in various phenotypes of anxiety.

Contributor Information

Eduard Maron, Department of Psychiatry, North Estonia Medical Centre, Tallinn, Estonia; Department of Psychiatry, University of Tartu, Tartu, Estonia ; Faculty of Medicine, Department of Medicine, Centre for Neuropsychopharmacology, Division of Brain Sciences, Imperial College London, London, UK.

David Nutt, Faculty of Medicine, Department of Medicine, Centre for Neuropsychopharmacology, Division of Brain Sciences, Imperial College London, London, UK.

REFERENCES

  • 1.Kessler RC., Chiu WT., Dernier O., Merikangas KR., Walters EE. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617–627. doi: 10.1001/archpsyc.62.6.617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Washington, DC: American Psychiatric Association; 2013 [Google Scholar]
  • 3.Farb DH., Ratner MH. Targeting the modulation of neural circuitry for the treatment of anxiety disorders. Pharmacol Rev. 2014;66:1002–1032. doi: 10.1124/pr.114.009126. [DOI] [PubMed] [Google Scholar]
  • 4.Mendlowicz MV., Stein MB. Quality of life in individuals with anxiety disorders. Am J Psychiatry. 2000;157:669–682. doi: 10.1176/appi.ajp.157.5.669. [DOI] [PubMed] [Google Scholar]
  • 5.Rice DP., Miller LS. Health economics and cost implications of anxiety and other mental disorders in the United States. Br J Psychiatry. 1998;34 (suppl):4–9. [PubMed] [Google Scholar]
  • 6.Whooly MA., Simon GE. Managing depression in medical outpatients. N Engl J Med. 2000;343:1942–1950. doi: 10.1056/NEJM200012283432607. [DOI] [PubMed] [Google Scholar]
  • 7.Markowitz JS., Weissman MM., Ouellette R., Lish JD., Klerman GL. Quality of life in panic disorder. Arch Gen Psychiatry. 1989;46:984–992. doi: 10.1001/archpsyc.1989.01810110026004. [DOI] [PubMed] [Google Scholar]
  • 8.DuPont RL., Rice DP., Miller LS., Shiraki SS., Rowland CR., Harwood HJ. Economic costs of anxiety disorders. Anxiety. 1996;2:167–172. doi: 10.1002/(SICI)1522-7154(1996)2:4<167::AID-ANXI2>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 9.Scott KA., Hoban AE., Clarke G., Moloney GM., Dinan TG., Cryan JF. Thinking small: towards microRNA-based therapeutics for anxiety disorders. Expert Opin Investig Drugs. 2015;8:1–14. doi: 10.1517/13543784.2014.997873. [DOI] [PubMed] [Google Scholar]
  • 10.Noyes R Jr., Crowe RR., Harris EL., Hamra BJ., McChesney CM., Chaudhry DR. Relationship between panic disorder and agoraphobia. A family study. Arch Gen Psychiatry. 1986;43:227–232. doi: 10.1001/archpsyc.1986.01800030037004. [DOI] [PubMed] [Google Scholar]
  • 11.Faravelli C., Paterniti S., Scarpato A. 5-year prospective, naturalistic follow-up study of panic disorder. Compr Psychiatry. 1995;36:271–277. doi: 10.1016/s0010-440x(95)90072-1. [DOI] [PubMed] [Google Scholar]
  • 12.Ravindran LN., Stein MB. The pharmacologic treatment of anxiety disorders: a review of progress. J Clin Psychiatry. 2010;71:839–854. doi: 10.4088/JCP.10r06218blu. [DOI] [PubMed] [Google Scholar]
  • 13.Wang SM., Pae CU. How much to worry about the FDA warning in the use of citalopram? Expert Rev Neurother. 2013;13:883–886. doi: 10.1586/14737175.2013.820450. [DOI] [PubMed] [Google Scholar]
  • 14.Argyropoulos SV., Nutt DJ. The use of benzodiazepines in anxiety and other disorders. Eur Neuropsychopharmacol. 1999;6:S407–S412. doi: 10.1016/s0924-977x(99)00052-8. [DOI] [PubMed] [Google Scholar]
  • 15.Leuchter AF., Cook IA., Hamilton SP., et al Biomarkers to predict antidepressant response. Curr Psychiatry Rep. 2010;12:553–562. doi: 10.1007/s11920-010-0160-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fu CH., Steiner H., Costa freda SG. Predictive neural biomarkers of clinical response in depression: a meta-analysis of functional and structural neuroimaging studies of pharmacological and psychological therapies. Neurobiol Dis. 2013;52:75–83. doi: 10.1016/j.nbd.2012.05.008. [DOI] [PubMed] [Google Scholar]
  • 17.Trivedi MH., Fava M., Wisniewski SR., et al Medication augmentation after the failure of SSRIs for depression. N Engl J Med. 2006;354:1243–1252. doi: 10.1056/NEJMoa052964. [DOI] [PubMed] [Google Scholar]
  • 18.Rush AJ., Warden D., Wisniewski SR., et al STAR*D: revising conventional wisdom. CNS Drugs. 2009;23:627–647. doi: 10.2165/00023210-200923080-00001. [DOI] [PubMed] [Google Scholar]
  • 19.Rush AJ. Limitations in efficacy of antidepressant monotherapy. J Clin Psychiatry. 2007;68:8–10. [PubMed] [Google Scholar]
  • 20.Keitner Gl., Ryan CE., Miller IW., Norman WH. Recovery and major depression: factors associated with twelve-month outcome. Am J Psychiatry. 1992;149:93–99. doi: 10.1176/ajp.149.1.93. [DOI] [PubMed] [Google Scholar]
  • 21.Warden D., Trivedi MH., Wisniewski SR., et al Predictors of attrition during initial (citalopram) treatment for depression: a STAR*D report. Am. J Psychiatry. 2007;164:1189–1197. doi: 10.1176/appi.ajp.2007.06071225. [DOI] [PubMed] [Google Scholar]
  • 22.Olfson M., Marcus SC., Tedeschi M., Wan GJ. Continuity of antidepressant treatment for adults with depression in the United States. Am J Psychiatry. 2006;163:101–108. doi: 10.1176/appi.ajp.163.1.101. [DOI] [PubMed] [Google Scholar]
  • 23.Bagby RM., Ryder AG., Cristi C. Psychosocial and clinical predictors of response to pharmacotherapy for depression. J Psychiatry Neurosci. 2002;27:250–257. [PMC free article] [PubMed] [Google Scholar]
  • 24.Joyce PR., Mulder RT., Cloninger CR. Temperament predicts clomipramine and desipramine response in major depression. J Affect Disord. 1994;30:35–46. doi: 10.1016/0165-0327(94)90149-x. [DOI] [PubMed] [Google Scholar]
  • 25.Lerer B., Macciardi F. Pharmacogenetics of antidepressant and moodstabilizing drugs: a review of candidate-gene studies and future research directions. Int J Neuropsychopharmacol. 2002;5:255–275. doi: 10.1017/S1461145702002936. [DOI] [PubMed] [Google Scholar]
  • 26.Szadoczky E., Rozsa S., Zambori J., Furedi J. Predictors for 2-year outcome of major depressive episode. J Affect Disord. 2004;83:49–57. doi: 10.1016/j.jad.2004.05.001. [DOI] [PubMed] [Google Scholar]
  • 27.Labermaier C., Masana M., Muller MB. Biomarkers predicting antidepressant treatment response: how can we advance the field? Dis Markers. 2013;35:23–31. doi: 10.1155/2013/984845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Niciu MJ., Mathews DC., Nugent AC., et al Developing biomarkers in mood disorders research through the use of rapid-acting antidepressants. Depress Anxiety. 2014;31:297–307. doi: 10.1002/da.22224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Koolschijn PC., van Haren NE., Lensvelt-Mulders GJ., Hulshoff Pol HE., Kahn RS. Brain volume abnormalities in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Hum Brain Mapp. 2009;30:3719–3735. doi: 10.1002/hbm.20801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hajek T., Kozeny J., Kopecek M., Alda M., Hoschl C. Reduced subgenual cinguiate volumes in mood disorders: a meta-analysis. J Psychiatry Neurosci. 2008;33:91–99. [PMC free article] [PubMed] [Google Scholar]
  • 31.McKinnon M., Yucel K., Nazarov A., MacQueen GM. A meta-analysis examining clinical predictors of hippocampal volume in patients with major depressive disorder. J Psychiatry Neurosci. 2009;34:41–54. [PMC free article] [PubMed] [Google Scholar]
  • 32.MacQueen GM., Yucel K., Taylor VH., Macdonald K., Joffe R. Posterior hippocampal volumes are associated with remission rates in patients with major depressive disorder. Biol Psychiatry. 2008;64:880–888. doi: 10.1016/j.biopsych.2008.06.027. [DOI] [PubMed] [Google Scholar]
  • 33.Vakili K., Pillay SS., Later B., et al Hippocampal volume in primary unipolar major depression: a magnetic resonance imaging study. Biol Psychiatry. 2000;47:1087–1090. doi: 10.1016/s0006-3223(99)00296-6. [DOI] [PubMed] [Google Scholar]
  • 34.Costafreda SG., Chu C., Ashburner J., Fu CH. Prognostic and diagnostic potential of the structural neuroanatomy of depression. PLoS One. 2009;4:e6353. doi: 10.1371/journal.pone.0006353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mayberg HS., Brannan SK., Mahurin RK., et al Cinguiate function in depression: a potential predictor of treatment response. Neuroreport. 1997;8:1057–1061. doi: 10.1097/00001756-199703030-00048. [DOI] [PubMed] [Google Scholar]
  • 36.Alexopoulos GS., Kiosses DN., Choi SJ., Murphy CF., Lim KO. Frontal white matter microstructure and treatment response of late-life depression: a preliminary study. Am J Psychiatry. 2002;159:1929–1932. doi: 10.1176/appi.ajp.159.11.1929. [DOI] [PubMed] [Google Scholar]
  • 37.Alexopoulos GS., Murphy CF., Gunning-Dixon FM., et al Microstructural white matter abnormalities and remission of geriatric depression. Am: J Psychiatry. 2008;165:238–244. doi: 10.1176/appi.ajp.2007.07050744. [DOI] [PubMed] [Google Scholar]
  • 38.Choi J., Jeong B., Rohan ML., Polcari AM., Teicher MH. Preliminary evidence for white matter tract abnormalities in young adults exposed to parental verbal abuse. Biol Psychiatry. 2009;65:227–234. doi: 10.1016/j.biopsych.2008.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Alexopoulos GS., Murphy CF., Gunning-Dixon FM., et al Serotonin transporter polymorphisms, microstructural white matter abnormalities and remission of geriatric depression. J Affect Disord. 2009;119:132–141. doi: 10.1016/j.jad.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Levy-Gigi E., Szabo C., Kelemen O., Keri S. Association among clinical response, hippocampal volume, and FKBP5 gene expression in individuals with posttraumatic stress disorder receiving cognitive behavioral therapy. Biol Psychiatry. 2013;74:793–800. doi: 10.1016/j.biopsych.2013.05.017. [DOI] [PubMed] [Google Scholar]
  • 41.Bryant RA., Felmingham K., Whitford TJ., et al Rostral anterior cinguiate volume predicts treatment response to cognitive-behavioural therapy for posttraumatic stress disorder. J Psychiatry Neurosci. 2008;33:142–146. [PMC free article] [PubMed] [Google Scholar]
  • 42.Dickie EW., Brunet A., Akerib V., Armony JL. Anterior cinguiate cortical thickness is a stable predictor of recovery from post-traumatic stress disorder. Psychol Med. 2013;43:645–653. doi: 10.1017/S0033291712001328. [DOI] [PubMed] [Google Scholar]
  • 43.Lai CH., Wu YT. Changes in regional homogeneity of parieto-temporal regions in panic disorder patients who achieved remission with antidepressant treatment. J Affect Disord. 2013;151:709–714. doi: 10.1016/j.jad.2013.08.006. [DOI] [PubMed] [Google Scholar]
  • 44.Lai CH., Wu YT., Yu PL., Yuan W. Improvements in white matter microstructural integrity of right uncinate fasciculus and left fronto-occipital fasciculus of remitted first-episode medication-naive panic disorder patients. J Affect Disord. 2013;150:330–336. doi: 10.1016/j.jad.2013.04.014. [DOI] [PubMed] [Google Scholar]
  • 45.Lai CH., Hsu YY. A subtle grey-matter increase in first-episode, drugnaive major depressive disorder with panic disorder after 6 weeks' duloxetine therapy. Int J Neuropsychopharmacol. 2011;14:225–235. doi: 10.1017/S1461145710000829. [DOI] [PubMed] [Google Scholar]
  • 46.Hoexter MQ., Dougherty DD., Shavitt RG., et al Differential prefrontal gray matter correlates of treatment response to fluoxetine or cognitivebehavioral therapy in obsessive-compulsive disorder. Eur Neuropsychopharmacol. 2013;23:569–580. doi: 10.1016/j.euroneuro.2012.06.014. [DOI] [PubMed] [Google Scholar]
  • 47.Banks GP., Mikell CB., Youngerman BE., et al Neuroanatomical characteristics associated with response to dorsal anterior cingulotomy for obsessive-compulsive disorder. JAMA Psychiatry. 2015;72:127–135. doi: 10.1001/jamapsychiatry.2014.2216. [DOI] [PubMed] [Google Scholar]
  • 48.Anand A., Li Y., Wang Y., et al Antidepressant effect on connectivity of the mood-regulating circuit: an FMRI study. Neuropsychopharmacology. 2005;30:1334–1344. doi: 10.1038/sj.npp.1300725. [DOI] [PubMed] [Google Scholar]
  • 49.Fu CH., Williams SC., Cleare AJ., et al Attenuation of the neural response to sad faces in major depression by antidepressant treatment: a prospective, event-related functional magnetic resonance imaging study. Arch Gen Psychiatry. 2004;61:877–889. doi: 10.1001/archpsyc.61.9.877. [DOI] [PubMed] [Google Scholar]
  • 50.Sheline Yl., Barch DM., Donnelly JM., Ollinger JM., Snyder AZ., Mintun MA. Increased amygdala response to masked emotional faces in depressed subjects resolves with antidepressant treatment: an fMRI study. Biol Psychiatry. 2001;50:651–658. doi: 10.1016/s0006-3223(01)01263-x. [DOI] [PubMed] [Google Scholar]
  • 51.Fu CH., Williams SC., Cleare AJ., Scott J., et al Neural responses to sad facial expressions in major depression following cognitive behavioral therapy. Biol Psychiatry. 2008;64:505–512. doi: 10.1016/j.biopsych.2008.04.033. [DOI] [PubMed] [Google Scholar]
  • 52.Chen CH., Suckling J., Ooi C., et al Functional coupling of the amygdala in depressed patients treated with antidepressant medication. Neuropsychopharmacology. 2008;33:1909–1918. doi: 10.1038/sj.npp.1301593. [DOI] [PubMed] [Google Scholar]
  • 53.Anand A., Li Y., Wang Y., Gardner K., Lowe MJ. Reciprocal effects of antidepressant treatment on activity and connectivity of the mood regulating circuit: an FMRI study. J Neuropsychiatry Clin Neurosci. 2007;19:274–282. doi: 10.1176/appi.neuropsych.19.3.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hahn T., Kircher T., Straube B., et al Predicting treatment response to cognitive behavioral therapy in panic disorder with agoraphobia by integrating local neural information. JAMA. Psychiatry. 2015;72:68–74. doi: 10.1001/jamapsychiatry.2014.1741. [DOI] [PubMed] [Google Scholar]
  • 55.Lueken U., Straube B., Konrad C., et al Neural substrates of treatment response to cognitive-behavioral therapy in panic disorder with agoraphobia. Am J Psychiatry. 2013;170:1345–1355. doi: 10.1176/appi.ajp.2013.12111484. [DOI] [PubMed] [Google Scholar]
  • 56.Lueken U., Straube B., Wittchen HU., et al Therapygenetics: anterior cinguiate cortex-amygdala coupling is associated with 5-HTTLPR and treatment response in panic disorder with agoraphobia. J Neural Transm. 2015;122:135–144. doi: 10.1007/s00702-014-1311-2. [DOI] [PubMed] [Google Scholar]
  • 57.Reinecke A., Thilo K., Filippini N., Croft A., Harmer CJ. Predicting rapid response to cognitive-behavioural treatment for panic disorder: the role of hippocampus, insula, and dorsolateral prefrontal cortex. Behav Res Ther. 2014;62:120–128. doi: 10.1016/j.brat.2014.07.017. [DOI] [PubMed] [Google Scholar]
  • 58.Ball TM., Stein MB., Ramsawh HJ., Campbell-Sills L., Paulus MP. Singlesubject anxiety treatment outcome prediction using functional neuroimaging. Neuropsychopharmacology. 2014;39:1254–1261. doi: 10.1038/npp.2013.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Whalen PJ., Johnstone T., Somerville LH., et al A functional magnetic resonance imaging predictor of treatment response to venlafaxine in generalized anxiety disorder. Biol Psychiatry. 2008;63:858–863. doi: 10.1016/j.biopsych.2007.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Nitschke JB., Sarinopoulos I., Oathes DJ., et al Anticipatory activation in the amygdala and anterior cinguiate in generalized anxiety disorder and prediction of treatment response. Am J Psychiatry. 2009;166:302–310. doi: 10.1176/appi.ajp.2008.07101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pantazatos SP., Talati A., Schneier FR., Hirsch J. Reduced anterior temporal and hippocampal functional connectivity during face processing discriminates individuals with social anxiety disorder from healthy controls and panic disorder, and increases following treatment. Neuropsychopharmacology. 2014;39:425–434. doi: 10.1038/npp.2013.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Klumpp H., Keutmann MK., Fitzgerald DA., Shankman SA., Phan KL. Resting state amygdala-prefrontal connectivity predicts symptom change after cognitive behavioral therapy in generalized social anxiety disorder. Biol Mood Anxiety Disord. 2014;4:14. doi: 10.1186/s13587-014-0014-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Doehrmann O., Ghosh SS., Polli FE., et al Predicting treatment response in social anxiety disorder from functional magnetic resonance imaging. JAMA. Psychiatry. 2013;70:87–97. doi: 10.1001/2013.jamapsychiatry.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Klumpp H., Fitzgerald DA., Phan KL. Neural predictors and mechanisms of cognitive behavioral therapy on threat processing in social anxiety disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2013;45:83–91. doi: 10.1016/j.pnpbp.2013.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhu H., Qiu C., Meng Y., et al Altered spontaneous neuronal activity in chronic posttraumatic stress disorder patients before and after a 12-week paroxetine treatment. J Affect Disord. 2015;174:257–264. doi: 10.1016/j.jad.2014.11.053. [DOI] [PubMed] [Google Scholar]
  • 66.van Rooij SJ., Geuze E., Kennis M., Rademaker AR., Vink M. Neural correlates of inhibition and contextual cue processing related to treatment response in PTSD. Neuropsychopharmacology. 2015;40:667–675. doi: 10.1038/npp.2014.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shin DJ., Jung WH., He Y., et al The effects of pharmacological treatment on functional brain connectome in obsessive-compulsive disorder. Biol Psychiatry. 2014;75:606–614. doi: 10.1016/j.biopsych.2013.09.002. [DOI] [PubMed] [Google Scholar]
  • 68.Sanematsu H., Nakao T., Yoshiura T., et al Predictors of treatment response to fluvoxamine in obsessive-compulsive disorder: an fMRI study. J Psychiatr Res. 2010;44:193–200. doi: 10.1016/j.jpsychires.2009.08.007. [DOI] [PubMed] [Google Scholar]
  • 69.Deckersbach T., Dougherty DD., Rauch SL. Functional imaging of mood and anxiety disorders. J Neuroimaging. 2006;16:1–10. doi: 10.1177/1051228405001474. [DOI] [PubMed] [Google Scholar]
  • 70.Kugaya A., Sanacora G., Staley JK., et al Brain serotonin transporter availability predicts treatment response to selective serotonin reuptake inhibitors. Biol Psychiatry. 2004;56:497–502. doi: 10.1016/j.biopsych.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 71.Lanzenberger R., Kranz GS., Haeusler D., et al Prediction of SSRI treatment response in major depression based on serotonin transporter interplay between median raphe nucleus and projection areas. Neuroimage. 2012;63:874–881. doi: 10.1016/j.neuroimage.2012.07.023. [DOI] [PubMed] [Google Scholar]
  • 72.Brockmann H., Zobel A., Joe A., et al The value of HMPAO SPECT in predicting treatment response to citalopram in patients with major depression. Psychiatry Res. 2009;173:107–112. doi: 10.1016/j.pscychresns.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 73.Milak MS., Parsey RV., Lee L., et al Pretreatment regional brain glucose uptake in the midbrain on PET may predict remission from a major depressive episode after three months of treatment. Psychiatry Res. 2009;173:63–70. doi: 10.1016/j.pscychresns.2008.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Parsey RV., Olvet DM., Oquendo MA., Huang YY., Ogden RT., Mann JJ. Higher 5-HT1A receptor binding potential during a major depressive episode predicts poor treatment response: preliminary data from a naturalistic study. Neuropsychopharmacology. 2006;31:1745–1749. doi: 10.1038/sj.npp.1300992. [DOI] [PubMed] [Google Scholar]
  • 75.Kang EH., Park JE., Lee KH., Cho YS., Kim JJ., Yu BH. Regional brain metabolism and treatment response in panic disorder patients: an [18F]FDGPET study. Neuropsychobiology. 2012;66:106–111. doi: 10.1159/000337740. [DOI] [PubMed] [Google Scholar]
  • 76.Faria V., Ahs F., Appel L., et al Amygdala-frontal couplings characterizing SSRI and placebo response in social anxiety disorder. Int J Neuropsychopharmacol. 2014;17:1149–1157. doi: 10.1017/S1461145714000352. [DOI] [PubMed] [Google Scholar]
  • 77.Van der Linden G., van Heerden B., Warwick J., et al Functional brain imaging and pharmacotherapy in social phobia: single photon emission computed tomography before and after treatment with the selective serotonin reuptake inhibitor citalopram. Prog Neuropsychopharmacol Biol Psychiatry. 2000;24:419–438. doi: 10.1016/s0278-5846(99)00109-8. [DOI] [PubMed] [Google Scholar]
  • 78.Evans KC., Simon NM., Dougherty DD., et al A PET study of tiagabine treatment implicates ventral medial prefrontal cortex in generalized social anxiety disorder. Neuropsychopharmacology. 2009;34:390–398. doi: 10.1038/npp.2008.69. [DOI] [PubMed] [Google Scholar]
  • 79.Wen SL., Cheng MH., Cheng MF., Yue JH., Wang H. Pharmacotherapy response and regional cerebral blood flow characteristics in patients with obsessive-compulsive disorder. Behav Brain Fund. 2013;9:31. doi: 10.1186/1744-9081-9-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ho Pian KL., van Megen HJ., Ramsey NF., et al Decreased thalamic blood flow in obsessive-compulsive disorder patients responding to fluvoxamine. Psychiatry Res. 2005;138:89–97. doi: 10.1016/j.pscychresns.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 81.Rauch SL., Shin LM., Dougherty DD., Alpert NM., Fischman AJ., Jenike MA. Predictors of fluvoxamine response in contamination-related obsessive compulsive disorder: a PET symptom provocation study. Neuropsychopharmacology. 2002;27:782–791. doi: 10.1016/S0893-133X(02)00351-2. [DOI] [PubMed] [Google Scholar]
  • 82.Carey PD., Warwick J., Niehaus DJ., et al Single photon emission computed tomography (SPECT) of anxiety disorders before and after treatment with citalopram. BMC Psychiatry. 2004;4:30. doi: 10.1186/1471-244X-4-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Seedat S., Warwick J., van Heerden B., et al Single photon emission computed tomography in posttraumatic stress disorder before and after treatment with a selective serotonin reuptake inhibitor. J Affect Disord. 2004;80:45–53. doi: 10.1016/S0165-0327(03)00047-8. [DOI] [PubMed] [Google Scholar]
  • 84.Yamanishi T., Nakaaki S., Omori IM., et al Changes after behavior therapy among responsive and nonresponsive patients with obsessivecompulsive disorder. Psychiatry Res. 2009;172:242–250. doi: 10.1016/j.pscychresns.2008.07.004. [DOI] [PubMed] [Google Scholar]
  • 85.Seo HJ., Choi YH., Chung YA., Rho W., Chae JH. Changes in cerebral blood flow after cognitive behavior therapy in patients with panic disorder: a SPECT study. Neuropsychiatr Dis Treat. 2014;10:661–669. doi: 10.2147/NDT.S58660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lindauer RJ., Booij J., Habraken JB., et al Effects of psychotherapy on regional cerebral blood flow during trauma imagery in patients with post-traumatic stress disorder: a randomized clinical trial. Psychol Med. 2008;38:543–554. doi: 10.1017/S0033291707001432. [DOI] [PubMed] [Google Scholar]
  • 87.Maron E., Nutt D., Shlik J. Neuroimaging of serotonin system in anxiety disorders. Curr Pharm Des. 2012;18:5699–5708. doi: 10.2174/138161212803530844. [DOI] [PubMed] [Google Scholar]
  • 88.Stengler-Wenzke K., Muller U., Barthel H., Angermeyer MC., Sabri O., Hesse S. Serotonin transporter imaging with [123I]beta-CIT SPECT before and after one year of citalopram treatment of obsessive-compulsive disorder. Neuropsychobiology. 2006;53:40–45. doi: 10.1159/000090702. [DOI] [PubMed] [Google Scholar]
  • 89.Warwick JM., Carey PD., Cassimjee N., et al Dopamine transporter binding in social anxiety disorder: the effect of treatment with escitalopram. Metab Brain Dis. 2012;27:151–158. doi: 10.1007/s11011-012-9280-3. [DOI] [PubMed] [Google Scholar]
  • 90.Grimm S., Luborzewski A., Schubert F., et al Region-specific glutamate changes in patients with unipolar depression. J Psychiatr Res. 2012;46:1059–1065. doi: 10.1016/j.jpsychires.2012.04.018. [DOI] [PubMed] [Google Scholar]
  • 91.Olbrich S., Arns M. EEG biomarkers in major depressive disorder: discriminative power and prediction of treatment response. Int Rev Psychiatry. 2013;25:604–618. doi: 10.3109/09540261.2013.816269. [DOI] [PubMed] [Google Scholar]
  • 92.Cornwell BR., Salvadore G., Furey M., et al Synaptic potentiation is critical for rapid antidepressant response to ketamine in treatment-resistant major depression. Biol Psychiatry. 2012;72:555–561. doi: 10.1016/j.biopsych.2012.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Li CT., Chen LF., Tu PC., Wang SJ., Chen MH., Su TP., et al Impaired prefronto-thalamic functional connectivity as a key feature of treatmentresistant depression: a combined MEG, PET and rTMS study. PLoS One. 2013;8:e70089. doi: 10.1371/journal.pone.0070089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pickar D., Rubinow K. Pharmacogenomics of psychiatric disorders. Trends Pharmacol Sci. 2001;22:75–83. doi: 10.1016/s0165-6147(00)01603-5. [DOI] [PubMed] [Google Scholar]
  • 95.Serretti A., Artioli P., Quartesan R. Pharmacogenetics in the treatment of depression: pharmacodynamic studies. Pharmacogenet Genomics. 2005;15:61–67. doi: 10.1097/01213011-200502000-00001. [DOI] [PubMed] [Google Scholar]
  • 96.Fabbri C., Di Girolamo G., Serretti A. Pharmacogenetics of antidepressant drugs: an update after almost 20 years of research. Am J Med Genet B Neuropsychiatr Genet. 2013;162B:487–520. doi: 10.1002/ajmg.b.32184. [DOI] [PubMed] [Google Scholar]
  • 97.Porcelli S., Drago A., Fabbri C., Gibiino S., Calati R., Serretti A. Pharmacogenetics of antidepressant response. J Psychiatry Neurosci. 2011;36:87–113. doi: 10.1503/jpn.100059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kato M., Serretti A. Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol Psychiatry. 2010;15:473–500. doi: 10.1038/mp.2008.116. [DOI] [PubMed] [Google Scholar]
  • 99.Perna G., Favaron E., Di Bella D., Bussi R., Bellodi L. Antipanic efficacy of paroxetine and polymorphism within the promoter of the serotonin transporter gene. Neuropsychopharmacology. 2005;30:2230–2235. doi: 10.1038/sj.npp.1300822. [DOI] [PubMed] [Google Scholar]
  • 100.Yevtushenko OO., Oros MM., Reynolds GP. Early response to selective serotonin reuptake inhibitors in panic disorder is associated with a functional 5-HT1A receptor gene polymorphism. J Affect Disord. 2010;123:308–311. doi: 10.1016/j.jad.2009.09.007. [DOI] [PubMed] [Google Scholar]
  • 101.Mushtaq D., Ali A., Margoob MA., Murtaza I., Andrade C. Association between serotonin transporter gene promoter-region polymorphism and 4- and 12-week treatment response to sertraline in posttraumatic stress disorder. J Affect Disord. 2012;136:955–962. doi: 10.1016/j.jad.2011.08.033. [DOI] [PubMed] [Google Scholar]
  • 102.Stein MB., Seedat S., Gelernter J. Serotonin transporter gene promoter polymorphism predicts SSRI response in generalized social anxiety disorder. Psychopharmacology (Berl). 2006;187:68–72. doi: 10.1007/s00213-006-0349-8. [DOI] [PubMed] [Google Scholar]
  • 103.Andersson E., Ruck C., Lavebratt C., Hedman, et al Genetic polymorphisms in monoamine systems and outcome of cognitive behavior therapy for social anxiety disorder. PLoS One. 2013;8:e79015. doi: 10.1371/journal.pone.0079015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Stein MB., Keshaviah A., Haddad SA., et al Influence of RGS2 on sertraline treatment for social anxiety disorder. Neuropsychopharmacology. 2014;39:1340–1346. doi: 10.1038/npp.2013.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Cooper AJ., Narasimhan S., Rickels K., Lohoff FW. Genetic polymorphisms in the PACAP and PAC1 receptor genes and treatment response to venlafaxine XR in generalized anxiety disorder. Psychiatry Res. 2013;210:1299–1300. doi: 10.1016/j.psychres.2013.07.038. [DOI] [PubMed] [Google Scholar]
  • 106.Cooper AJ., Rickels K., Lohoff FW. Association analysis between the A118G polymorphism in the OPRM1 gene and treatment response to venlafaxine XR in generalized anxiety disorder. Hum Psychopharmacol. 2013;28:258–262. doi: 10.1002/hup.2317. [DOI] [PubMed] [Google Scholar]
  • 107.Lohoff FW., Narasimhan S., Rickels K. Interaction between polymorphisms in serotonin transporter (SLC6A4) and serotonin receptor 2A (HTR2A) genes predict treatment response to venlafaxine XR in generalized anxiety disorder. Pharmacogenomics J. 2013;13:464–469. doi: 10.1038/tpj.2012.33. [DOI] [PubMed] [Google Scholar]
  • 108.Narasimhan S., Aquino TD., Multani PK., Rickels K., Lohoff FW. Variation in the catechol-O-methyltransferase (COMT) gene and treatment response to venlafaxine XR in generalized anxiety disorder. Psychiatry Res. 2012;198:112–115. doi: 10.1016/j.psychres.2011.12.034. [DOI] [PubMed] [Google Scholar]
  • 109.Lohoff FW., Aquino TD., Narasimhan S., Multani PK., Etemad B., Rickels K. Serotonin receptor 2A (HTR2A) gene polymorphism predicts treatment response to venlafaxine XR in generalized anxiety disorder. Pharmacogenomics J. 2013;13:21–26. doi: 10.1038/tpj.2011.47. [DOI] [PubMed] [Google Scholar]
  • 110.Narasimhan S., Aquino TD., Hodge R., Rickels K., Lohoff FW. Association analysis between the Val66Met polymorphism in the brain-derived neurotrophic factor (BDNF) gene and treatment response to venlafaxine XR in generalized anxiety disorder. Neurosci Lett. 2011;503:200–202. doi: 10.1016/j.neulet.2011.08.035. [DOI] [PubMed] [Google Scholar]
  • 111.Perlis RH., Fijal B., Dharia S., Houston JP. Pharmacogenetic investigation of response to duloxetine treatment in generalized anxiety disorder. Pharmacogenomics J. 2013;13:280–285. doi: 10.1038/tpj.2011.62. [DOI] [PubMed] [Google Scholar]
  • 112.Zai G., Brandl EJ., Muller DJ., Richter MA., Kennedy JL. Pharmacogenetics of antidepressant treatment in obsessive-compulsive disorder: an update and implications for clinicians. Pharmacogenomics. 2014;45:1147–1157. doi: 10.2217/pgs.14.83. [DOI] [PubMed] [Google Scholar]
  • 113.Maron E., Pettai K., Tammiste A., et al Are there specific genes underlying treatment resistance in depression? New evidence from genome-wide analysis. Paper presented at: 22nd ECNP Congress 2009. (S.13.01.) [Google Scholar]
  • 114.Tammiste A., Jiang T., Fischer K., et al Whole-exome sequencing identifies a polymorphism in the BMP5 gene associated with SSRI treatment response in major depression. J Psychopharmacol. 2013;27:915–920. doi: 10.1177/0269881113499829. [DOI] [PubMed] [Google Scholar]
  • 115.Lehrner A., Yehuda R. Biomarkers of PTSD: military applications and considerations. Eur J Psychotraumatol. 2014;14:5. doi: 10.3402/ejpt.v5.23797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Olff M., de Vries GJ., Guzelcan Y., Assies J., Gersons BP. Changes in Cortisol and DHEA plasma levels after psychotherapy for PTSD. Psychoneuroendocrinology. 2007;32:619–626. doi: 10.1016/j.psyneuen.2007.04.001. [DOI] [PubMed] [Google Scholar]
  • 117.Yehuda R., Bierer LM., Sarapas C., Makotkine I., Andrew R., Seckl JR. Cortisol metabolic predictors of response to psychotherapy for symptoms of PTSD in survivors of the World Trade Center attacks on September 11, 2001. Psychoneuroendocrinology. 2009;34:1304–1313. doi: 10.1016/j.psyneuen.2009.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Delorme R., Chabane N., Callebert J., et al Platelet serotonergic predictors of clinical improvement in obsessive compulsive disorder. J Clin Psychopharmacol. 2004;24:18–23. doi: 10.1097/01.jcp.0000104905.75206.50. [DOI] [PubMed] [Google Scholar]
  • 119.Lee IS., Kim KJ., Kang EH., Yu BH. beta-adrenoceptor affinity as a biological predictor of treatment response to paroxetine in patients with acute panic disorder. J Affect Disord. 2008;110:156–160. doi: 10.1016/j.jad.2007.12.007. [DOI] [PubMed] [Google Scholar]
  • 120.Kobayashi K., Shimizu E., Hashimoto K., et al Serum brain-derived neurotrophic factor (BDNF) levels in patients with panic disorder: as a biological predictor of response to group cognitive behavioral therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29:658–663. doi: 10.1016/j.pnpbp.2005.04.010. [DOI] [PubMed] [Google Scholar]
  • 121.Brandl EJ., Tiwari AK., Zhou X., et al Influence of CYP2D6 and CYP2C19 gene variants on antidepressant response in obsessive-compulsive disorder. Pharmacogenomics J. 2014;14:176–181. doi: 10.1038/tpj.2013.12. [DOI] [PubMed] [Google Scholar]
  • 122.Qin H., Samuels JF., Wang Y., Zhu Y., et al Whole-genome association analysis of treatment response in obsessive-compulsive disorder. Mol Psychiatry. In press. doi: 10.1038/mp.2015.32. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Dialogues in Clinical Neuroscience are provided here courtesy of Taylor & Francis

RESOURCES