Introduction
Varied drug response has long been recognized in the treatment of major depressive disorder (MDD) and generalized anxiety disorder (GAD). Of the approximately 3–7% of patients in the United States affected, nearly 50% fail to respond to first-line treatment regimens [
1Anxiety and Depression Association of America. MDD and GAD facts and figures. Silver Springs, MD. 2016; 1–23.
,
2- Stein D.J.
- Stein M.B.
- Pitts C.D.
- Kumar R.
- Hunter B.
Predictors of response to pharmacotherapy in social anxiety disorder: an analysis of 3 placebo-controlled paroxetine trials.
,
3- Trivedi M.H.
- Rush A.J.
- Wisniewski S.R.
- Nierenberg A.A.
- Warden D.
- Ritz L.
- et al.
Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice.
]. Influences such as environmental exposures, nutritional status, co-morbidities, severity of disease, and concomitant medications help to explain some unpredictable drug responses. However, genome wide association studies (GWAS) propose that genetic variation alone accounts for 42% of varied antidepressant response [
4- Tansey K.E.
- Guipponi M.
- Hu X.
- Domenici E.
- Lewis G.
- Malafosse A.
- et al.
Contribution of common genetic variants to antidepressant response.
,
5- Biernacka J.M.
- Sangkuhl K.
- Jenkins G.
- Whaley R.M.
- Barman P.
- Batzler A.
- et al.
The International SSRI Pharmacogenomics Consortium (ISPC): a genome-wide association study of antidepressant treatment response.
,
6- Ising M.
- Lucae S.
- Binder E.B.
- Bettecken T.
- Uhr M.
- Ripke S.
- et al.
A genomewide association study points to multiple loci that predict antidepressant drug treatment outcome in depression.
,
7- Garriock H.A.
- Kraft J.B.
- Shyn S.I.
- Peters E.J.
- Yokoyama J.S.
- Jenkins G.D.
- et al.
A genomewide association study of citalopram response in major depressive disorder.
,
8- Ji Y.
- Biernacka J.M.
- Hebbring S.
- Chai Y.
- Jenkins G.D.
- Batzler A.
- et al.
Pharmacogenomics of selective serotonin reuptake inhibitor treatment for major depressive disorder: genome-wide associations and functional genomics.
,
9- Uher R.
- Perroud N.
- Ng M.Y.
- Hauser J.
- Henigsberg N.
- Maier W.
- et al.
Genome-wide pharmacogenetics of antidepressant response in the GENDEP project.
]. This presents an auspicious principle on which to base the delivery of personalized medicine.
Several classes of antidepressant medication have been shown to benefit individuals with MDD–selective serotonin reuptake inhibitors (SSRIs), selective norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs) are among the most widely used and well-studied [
[10]Working Group on Depressive Disorder. Practice Guideline for the Treatment of Patients with Major Depressive Disorder. American Psychiatric Association.
]. SSRIs are currently the most commonly prescribed drug class for MDD treatment, though even within-class response to treatment varies considerably between patients and identification of the most appropriate medication is a continued challenge [
11- Anderson H.D.
- Pace W.D.
- Libby A.M.
- West D.R.
- Valuck R.J.
Rates of 5 common antidepressant side effects among new adult and adolescent cases of depression: a retrospective US claims study.
,
12- Smith A.J.
- Sketris I.
- Cooke C.
- Gardner D.
- Kisely S.
- Tett S.E.
A comparison of antidepressant use in Nova Scotia, Canada and Australia.
]. Antidepressant response does not show a classic Mendelian model of inheritance, but instead, a moderate number of loci—each with a small effect size—are proposed to be involved in response [
[13]- Porcelli S.
- Drago A.
- Fabbri C.
- Gibiino S.
- Calati R.
- Serretti A.
Pharmacogenetics of antidepressant response.
]. Pharmacogenetics research is actively attempting to link antidepressant treatment response to a portfolio of polymorphisms that correspond to brain circuitry/plasticity [
[14]Psychiatric pharmacogenomics: how to integrate into clinical practice.
]. Theoretically, this will allow the personalization of MDD/GAD treatment by minimizing the use of ‘trial-and-error’ treatment. It is important to note that MDD and GAD likely have an overlapping genetic etiology [
[15]- Camp N.J.
- Lowry M.R.
- Richards R.L.
- Plenk A.M.
- Carter C.
- Hensel C.H.
- et al.
Genome-wide linkage analyses of extended Utah pedigrees identifies loci that influence recurrent, early-onset major depression and anxiety disorders.
]. This, in combination with high rates of comorbidity and ambiguity of onset, provide a strong case for treating MDD and GAD in the same manner [
16The genetics of major depression.
,
17- Kessler R.C.
- Berglund P.
- Demler O.
- Jin R.
- Koretz D.
- Merikangas K.R.
- et al.
The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R).
,
18- Moffitt T.E.
- Harrington H.
- Caspi A.
- Kim-Cohen J.
- Goldberg D.
- Gregory A.M.
- et al.
Depression and generalized anxiety disorder: cumulative and sequential comorbidity in a birth cohort followed prospectively to age 32 years.
].
Much research has focused on pharmacokinetic factors, specifically the liver metabolizing Cytochrome P450 (CYP450) superfamily of enzymes that is responsible for the oxidation of antidepressant medication. The
CYP450 genes mainly involved in antidepressant metabolism encode isoforms in the CYP2D6, CYP2C19, CYP2C9, and CYP3A4/5 enzymes. These genes are highly polymorphic and result in normal (EM-extensive metabolizer), abnormal (IM-intermediate, UM-ultra-rapid, and PM-poor metabolizer), or aberrant metabolizer phenotypes [
[19]- Porcelli S.
- Fabbri C.
- Spina E.
- Serretti A.
- De Ronchi D.
Genetic polymorphisms of cytochrome P450 enzymes and antidepressant metabolism.
]. Metabolizer status was demonstrated to be associated with antidepressant pharmacokinetics in a number of studies and variable rates of metabolism have been shown to increase the potential for adverse drug effects and reduce rates of compliance in patients taking antidepressants [
[20]- Fabbri C.
- Di Girolamo G.
- Serretti A.
Pharmacogenetics of antidepressant drugs: an update after almost 20 years of research.
]. Despite the fact that the Food and Drug Administration (FDA) has incorporated genetic testing information into the labeling of nineteen antidepressants, pharmacogenetics testing has not been incorporated into treatment guidelines because of a gap in consistent evidence linking testing to clinical outcomes, i.e. clinical utility [
21Recommendations from the EGAPP Working Group: testing for cytochrome P450 polymorphisms in adults with nonpsychotic depression treated with selective serotonin reuptake inhibitors.
,
22Food and Drug Administration. Table of pharmacogenomic biomarkers in drug labeling. 2016; 1–84.
].
More recently, candidate gene studies aimed to detect an association between antidepressant response and catecholaminergic genes. The serotonin transporter gene (
SLC6A4) is an obvious candidate as the serotonin transporter is the primary site of SSRI action [
[20]- Fabbri C.
- Di Girolamo G.
- Serretti A.
Pharmacogenetics of antidepressant drugs: an update after almost 20 years of research.
]. A common 44-bp insertion/deletion polymorphism—referred to as the long (L
A) or short (S) forms of
SLC6A4, respectively—was shown to impact transcription and ultimately levels of the serotonin transporter [
[23]- Lesch K.P.
- Bengel D.
- Heils A.
- Sabol S.Z.
- Greenberg B.D.
- Petri S.
- et al.
Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region.
]. In addition to the S allele, a variant of the L allele in which the adenine (L
A) has been replaced with guanine (L
G), is also associated with reduced serotonin transporter levels and is functionally comparable to the S allele [
24- Wendland J.R.
- Martin B.J.
- Kruse M.R.
- Lesch K.-P.
- Murphy D.L.
Simultaneous genotyping of four functional loci of human SLC6A4, with a reappraisal of 5-HTTLPR and rs25531.
,
25- Hu X.
- Oroszi G.
- Chun J.
- Smith T.L.
- Goldman D.
- Schuckit M.A.
An expanded evaluation of the relationship of four alleles to the level of response to alcohol and the alcoholism risk.
]. Patients with variant transporter translation exhibit lower remission rates, increased side effects, and intolerance to SSRIs [
[26]Human serotonin transporter gene (SLC6A4) variants: their contributions to understanding pharmacogenomic and other functional G×G and G×E differences in health and disease.
]. Further, the S and L
G alleles of
SLC6A4 have been correlated with depression and anxiety-related symptomology and antidepressant response in numerous studies [
23- Lesch K.P.
- Bengel D.
- Heils A.
- Sabol S.Z.
- Greenberg B.D.
- Petri S.
- et al.
Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region.
,
27- Luddington N.S.
- Mandadupu A.
- Husk M.
- El-Mallakh R.S.
Clinical implications of genetic variation in the serotonin transporter promoter region: a review.
,
28- Mazzanti C.M.
- Lappalainen J.
- Long J.C.
- et al.
Role of the serotonin transporter promoter polymorphism in anxiety-related traits.
,
29- Hariri A.R.
- Drabant E.M.
- Munoz K.E.
- et al.
A susceptibility gene for affective disorders and the response of the human amygdala.
,
30- Park M.-H.
- Sanders E.
- Howe M.
- et al.
Association of anxiety symptoms in offspring of bipolar parents with serotonin transporter-linked polymorphic region (5-HTTLPR) genotype.
,
31- Schinka J.A.
- Busch R.M.
- Robichaux-Keene N.
A meta-analysis of the association between the serotonin transporter gene polymorphism (5-HTTLPR) and trait anxiety.
]. For example, a study of 36 patients suggested an association between fluoxetine response and
SLC6A4 genotype and identified S allele carriers as being at risk for developing insomnia and agitation with treatment [
[32]- Perlis R.H.
- Mischoulon D.
- Smoller J.W.
- Wan Y.J.
- Lamon-Fava S.
- Lin K.M.
- et al.
Serotonin transporter polymorphisms and adverse effects with fluoxetine treatment.
]. Poor response to citalopram was associated with S/S genotypes [
[33]- Outhred T.
- Das P.
- Dobson-Stone C.
- Felmingham K.L.
- Bryant R.A.
- Nathan P.J.
- et al.
Impact of 5-HTTLPR on SSRI serotonin transporter blockade during emotion regulation: a preliminary fMRI study.
]. Finally, a recent meta-analysis reported an associative model between SSRI response (OR: 1.58; 95% CI: 1.16–2.16, p = .004) and remission (OR: 1.53; 95% CI: 1.14–2.04, p = .004) in Caucasian
SLC6A4 L
A allele carriers [
[34]- Porcelli S.
- Fabbri C.
- Serretti A.
Meta-analysis of serotonin transporter gene promoter polymorphism (5-HTTLPR) association with antidepressant efficacy.
].
Methylenetetrahydrofolate reductase (MTHFR) is a rate limiting enzyme in the production of
l-methylfolate;
l-methylfolate is a critical regulatory molecule in the synthesis of monoamine neurotransmitters associated with mood regulation (i.e. dopamine, norepinephrine, and serotonin) [
[35]L-Methylfolate: a vitamin for your monoamines.
]. Although the
MTHFR gene has not been directly linked to antidepressant response, numerous studies have identified a modest association with depression symptomology and disease [
36- Mischoulon D.
- Lamon-Fava S.
- Selhub J.
- et al.
Prevalence of MTHFR C677T and MS A2756G polymorphisms in major depressive disorder, and their impact on response to fluoxetine treatment.
,
37- Shen X.
- Wu Y.
- Guan T.
- Wang X.
- Qian M.
- Lin M.
- et al.
Association analysis of COMT/MTHFR polymorphisms and major depressive disorder in Chinese Han population.
,
38- Gilbody S.
- Lightfoot T.
- Sheldon T.
Is low folate a risk factor for depression? A meta-analysis and exploration of heterogeneity.
,
39- Lok A.
- Mocking R.J.T.
- Assies J.
- et al.
The one-carbon-cycle and methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism in recurrent major depressive disorder; influence of antidepressant use and depressive state?.
]. Two
MTHFR polymorphisms, C677T and A1298C, result in diminished enzyme activity, and moreover the T allele of C677T has been associated with decreased
l-methylfolate levels [
[40]- Bjelland I.
- Tell G.S.
- Vollset S.E.
- Refsum H.
- Ueland P.M.
Folate, vitamin B12, homocysteine, and the MTHFR 677C->T polymorphism in anxiety and depression: the Hordaland Homocysteine Study.
]. As MDD has an established association with low serum folate levels [
38- Gilbody S.
- Lightfoot T.
- Sheldon T.
Is low folate a risk factor for depression? A meta-analysis and exploration of heterogeneity.
,
40- Bjelland I.
- Tell G.S.
- Vollset S.E.
- Refsum H.
- Ueland P.M.
Folate, vitamin B12, homocysteine, and the MTHFR 677C->T polymorphism in anxiety and depression: the Hordaland Homocysteine Study.
], folate augmentation in patients unresponsive to SSRI/SNRI treatment improved patient adherence [
41- Wade R.L.
- Kindermann S.L.
- Hou Q.
- Thase M.E.
Comparative assessment of adherence measures and resource use in SSRI/SNRI-treated patients with depression using second-generation antipsychotics or L-methylfolate as adjunctive therapy.
,
42- Papakostas G.I.
- Shelton R.C.
- Zajecka J.M.
- Etemad B.
- Rickels K.
- Clain A.
- et al.
L-methylfolate as adjunctive therapy for SSRI-resistant major depression: results of two randomized, double-blind, parallel-sequential trials.
]. Further, a meta-analysis of 15,315 participants reported a significant relationship between folate status and depression (OR: 1.55; 95% CI: 1.26–1.91; p < .05) [
[38]- Gilbody S.
- Lightfoot T.
- Sheldon T.
Is low folate a risk factor for depression? A meta-analysis and exploration of heterogeneity.
].
l-Methylfolate has been efficaciously used as an adjunctive therapy for patients with inadequate or poor SSRI response and was shown to improve adherence and decrease cost of care [
41- Wade R.L.
- Kindermann S.L.
- Hou Q.
- Thase M.E.
Comparative assessment of adherence measures and resource use in SSRI/SNRI-treated patients with depression using second-generation antipsychotics or L-methylfolate as adjunctive therapy.
,
42- Papakostas G.I.
- Shelton R.C.
- Zajecka J.M.
- Etemad B.
- Rickels K.
- Clain A.
- et al.
L-methylfolate as adjunctive therapy for SSRI-resistant major depression: results of two randomized, double-blind, parallel-sequential trials.
]. Therefore, an indirect link to
MTHFR polymorphisms and antidepressant treatment outcome is likely.
Pharmacogenetic testing has the potential to reduce antidepressant discontinuation due to adverse events and increase overall efficacy. Ideally, pharmacogenetics would inform individualized decisions by identifying DNA variants that predict outcomes. Promising evidence, including increased quality of life and reduced depression/anxiety scores, were reported with assay-guided treatment of MDD patients [
[43]- Brennan F.X.
- Gardner K.R.
- Lombard J.
- et al.
A naturalistic study of the effectiveness of pharmacogenetic testing to guide treatment in psychiatric patients with mood and anxiety disorders.
]. To date, only two randomized controlled trials (RCTs) have been conducted to investigate the impact of pharmacogenetics testing on antidepressant outcome [
44- Winner J.G.
- Carhart J.M.
- Altar A.
- Allen J.D.
- Dechairo B.M.
A prospective, randomized, double-blind study assessing the clinical impact of integrated pharmacogenomic testing for major depressive disorder.
,
45Improved antidepressant remission in major depression via a pharmacokinetic pathway polygene pharmacogenetic report.
]; one reported a two-fold increase in depression symptom relief while the other reported a greater chance of disease remission with pharmacogenetics testing usage (2.52-fold; 95% CI: 1.71–3.73; Z: 4.66, p < .0001). Despite these promising results, a systematic review of guided-treatment
versus usual care deemed current evidence inconclusive and condemns the widespread use of pharmacogenetics testing at the onset of MDD treatment [
[46]- Peterson K.
- Dieperink E.
- Anderson J.
- Boundy E.
- Ferguson L.
- Helfand M.
Rapid evidence review of the comparative effectiveness, harms, and cost-effectiveness of pharmacogenomics-guided antidepressant treatment versus usual care for major depressive disorder.
].
The utility of pharmacogenetics testing remains unclear though—in part because of a relative lack of RCTs and an abundance of small cohort, statistically under powered studies—because the method by which pharmacogenetic testing influences clinical treatment is not well-established [
47Abandoning personalization to get to precision in the pharmacotherapy of depression.
,
48Pharmacogenetic testing in psychiatry: not (quite) ready for primetime.
]. We therefore examined data from a naturalistic study of a commercial pharmacogenetic test to characterize how likely clinicians were to make test-concordant medication changes, and whether outcomes improved when assay-congruent medication regimens were implemented. As a means to thoroughly address this gap in the literature and realistically assess the utility of pharmacogenetics testing in the treatment of MDD/GAD, we aimed to (i) determine if pharmacogenetics testing influenced clinician decision-making and prescribing patterns, and, (ii) identify putative genetic predictors of treatment outcome.
Materials and methods
Patient cohort
A post-hoc analysis was performed on genotyping and outcomes data from a previously conducted clinical trial (ClinicalTrails.gov: NCT01507155) [
[43]- Brennan F.X.
- Gardner K.R.
- Lombard J.
- et al.
A naturalistic study of the effectiveness of pharmacogenetic testing to guide treatment in psychiatric patients with mood and anxiety disorders.
]. Original study design stipulated that adult patients must be diagnosed with a psychiatric disorder by a mental health care specialist who, for the purpose of this trial, ordered Genecept pharmacogenetic testing (n = 1024). Study participants were required to have the ability to complete electronic informed consents and be able to comprehend/complete online questionnaires. For the present study, primary diagnoses other than MDD (n = 297) or GAD (n = 171) were excluded. There were 468 patients in total evaluated in this analysis. Each of the 468 patients were evaluated by the clinician-reported outcome scales, but just 86 (18.4%) patients completed all of the patient-reported outcome questionnaires at each time point. Only patients that had full and complete data sets were included in this study (observed cases analysis). Clinicians were defined as mental health care professionals with the ability to prescribe medication and order a pharmacogenetics test, i.e. possession of a valid national provider identifier (NPI) number and prescribing privileges.
Genecept reporting
All clinicians were given information about trial design and goals and were willing clinicial participants. Clinicians were provided with a Genecept Report (Genomind King of Prussia, PA, USA) for each study participant at a one-month follow-up visit (to baseline visit). The report included genotyping results for ten genes (SLC6A4, MTHFR, 5HT2C, COMT, CACNA1C, DRD2, ANK3, CYP2D6, CYP2C19, CYP3A4) and details the implications of each genetic result on the use of a variety of FDA approved medications in the following classes: antidepressants, mood stabilizers/anticonvulsants, typical antipsychotics, atypical antipsychotics, anxiolytics, stimulants, nonsteroidal anti-inflammatory drugs (NSAIDs) and analgesics.
Genotypic processing
Of the ten genes that the Genecept Assay addressed,
SLC6A4 and
MTHFR were the genes directly associated with antidepressant treatment. As the assay was designed to assess how one would respond to a variety of drug classes, limiting the evaluation to just genes that effect response to antidepressants was adequate for the purposes of this analysis. Patients were binned into groups based on their
SLC6A4 genotypes and then again separately by their
MTHFR genotypes. For both genes, bins were dependent on phenotypic association with SSRI response. For
SLC6A4, bins consisted of either ‘wild-type (WT)’ (n = 125) or ‘at risk for poor SSRI response’ (n = 334) genotypes (
SLC6A4 WT: L
A/L
A;
SLC6A4 risk: L
G/L
G, L
G/S, L
G/L
A, L
A/S, S/S). Similarly, for
MTHFR, bins consisted of either ‘WT’ (n = 195) or ‘at risk for poor treatment outcome’ (n = 272) genotypes (
MTHFR WT: C/C;
MTHFR risk: C/T, T/T). See
Table 1.
Table 1Participant demographics and allele frequencies.
AStudy participants that listed gender as ‘unknown’ are as follows: SLC6A4 WT = 4; SLC6A4 Risk = 21; MTHFR WT = 12; MTHFR Risk = 13. BNumber of participants who were prescribed treatment regimens congruent with Genecept Pharmacogenetics Testing. Mann-Whitney U Test used to compare age, previous trials, medication changes, CGI-S, and CGI-I; two-sample Z-Test used to compare gender and congruency proportions; *p-value <.05. All numbers are mean values unless otherwise noted.
Outcome measures
Outcomes data were collected at three separate time points:
baseline (standard MDD/GAD treatment),
1-month from assay results received, and
3-month follow-up. Subjects were stratified into groups based on whether their clinician’s treatment choice (between receiving assay results and 3-month follow-up) was congruent with Genecept assay results. Three patient-reported scales were utilized to measure outcomes: The Quick Inventory of Depressive Symptoms (QIDS-SR) [
[49]- Rush A.J.
- Guillion C.M.
- Basco M.R.
- et al.
The inventory of depressive symptomatology (IDS): psychometric properties.
], the Quality of Life Enjoyment and Satisfaction Questionnaire Short Form (Q-LES-Q-SF) [
[50]Quality of life enjoyment and satisfaction questionnaire-short form for quality of life assessments in clinical practice: a psychometric study.
], and the Udvalg for Kliniske Undersøgelser Side Effect Rating Scale (UKU) [
[51]- Lingjaerde O.
- Ahlfors U.G.
- Bech P.
- et al.
The UKU side effect rating scale: a new comprehensive rating scale for psychotropic drugs and a cross-sectional study of side effects in neuroleptic-treated patients.
]. Each of the questionnaires was made available and administered through an online portal. The QIDS-SR scale ranges from 0 to 27, with 0 indicating no depression. The Q-LES-Q-SF scale ranges from 0 to 100, with higher scores indicating a greater satisfaction with life. The UKU scale measures the degree of side effects and ranges from 0 to 100, with 0 indicating low side effects. Clinicians simultaneously reported a Clinical Global Impression (CGI) score for severity at baseline (CGI-S) and for improvement at 3-months (CGI-I).
Tracking clinical treatment regimens
Clinicians reported primary diagnoses for each patient at the baseline time point. Clinicians simultaneously logged each patient’s treatment regimen. Changes to treatment regimens were reported in electronic surveys administered at each subsequent time point. Time point data was compared and treatment choices were considered congruent if they (i) discontinued an assay-indicated risk medication, or, (ii) initiated a medication indicated as a therapeutic option. Incongruent decisions included continuing or initiating treatment using an assay-indicated risk medication or failing to initiate an indicated therapeutic option. In many cases, the clinicians made more than a single change to their patient’s treatment. Collectively, these changes were considered congruent only if each of the changes met the criteria listed above for congruency. If at least one of the changes included administering or continuing use of a treatment deemed as a risk by the assay, then the treatment regimen was considered incongruent with the report.
Statistical analysis
Two-way repeated-measures analysis of variance (ANOVA) models, with Tukey post-hoc tests, were used to analyze the relationship between risk genotypes, assay congruency, and treatment outcome. This analysis evaluated whether changes to outcome measures—QIDS-SR, Q-LES-Q-SF, or UKU—were the result of independent variables of treatment action (relative to genotype) and of time, or to their interaction. Analyses were performed using R version 3.3.2 (http://cran.r-project.org). A two-sample Z-test was used to detect a difference between congruency proportion measures. Additionally, two major comparisons were investigated. Of the subjects carrying the MTHFR risk allele, a comparison between folate-supplemented SSRI/SNRI treatment and SSRI/SNRI treatment without folate supplementation was made. For participants with a risk genotype for SLC6A4, those taking an SSRI were compared to those alternatively prescribed a miscellaneous antidepressant/SNRI. Miscellaneous antidepressants included bupropion, mirtazapine, and nefazodone.
Discussion
Study results suggest that receiving Genecept pharmacogenetic testing improves MDD/GAD patient outcomes as measured through patient-reported scales for depression, side effect severity, and quality of life. Intriguingly, improvements in quality of life scores were significantly higher in
SLC6A4 risk patients receiving SNRI/miscellaneous antidepressants
versus SCL6A4 risk patients receiving SSRIs (
Fig. 3). Amongst the 468 patients tested, 50.6% of their clinicians prescribed assay-congruent treatment regimens, decreased SSRI prescription rate by 17.6%, and, increased SNRI prescription rate by 25.2%, miscellaneous antidepressant rate by 9.89%, and folate derivative prescription rate by 431% (
Fig. 1,
Fig. 2). At baseline, we did not find a significant difference between participant demographics or clinical global impression scores for
SLC6A4 WT
versus risk patients, nor
MTHFR WT
versus risk patients (
Table 1). Over the course of the 3-month clinical trial, SSRIs represented the most discontinued drug class though discontinuation was higher in patients with
SLC6A4 risk genotypes. Similarly, folate derivatives represented the most added drug class throughout the trial though addition was higher in patients with risk
MTHFR risk genotypes (
Table 2).
The question of whether genetic markers can help predict response to medication has major implications for personalizing treatment for depression and anxiety. A genome-wide complex trait analysis estimated the contribution of common polymorphisms to antidepressant response to be 42% (SE = 0.18; p = .009) in patients with MDD [
[4]- Tansey K.E.
- Guipponi M.
- Hu X.
- Domenici E.
- Lewis G.
- Malafosse A.
- et al.
Contribution of common genetic variants to antidepressant response.
]. Further supporting the principle that antidepressant response is a complex trait with substantial genetic influence is the low clinical efficacy of antidepressant medication– one in three patients does not fully recover from depression even after several treatment trials [
52- Hennings J.M.
- Owashi T.
- Binder E.B.
- et al.
Clinical characteristics and treatment outcome in a representative sample of depressed inpatients – findings from the Munich Antidepressant Response Signature (MARS) project.
,
53- Rush J.A.
- Trivedi M.H.
- Wisniewski S.R.
- et al.
Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report.
]. However, two separate genome-wide analyses (GWA) focusing on pharmacodynamic genes did not detect a single SNP significantly associated with SSRI response [
5- Biernacka J.M.
- Sangkuhl K.
- Jenkins G.
- Whaley R.M.
- Barman P.
- Batzler A.
- et al.
The International SSRI Pharmacogenomics Consortium (ISPC): a genome-wide association study of antidepressant treatment response.
,
6- Ising M.
- Lucae S.
- Binder E.B.
- Bettecken T.
- Uhr M.
- Ripke S.
- et al.
A genomewide association study points to multiple loci that predict antidepressant drug treatment outcome in depression.
]. This suggests that the effects of genetic variation on antidepressant response are multifaceted and warrant the use of observational trials to simultaneously evaluate genetic components that, in combination, wield an implication about the larger whole.
Genecept pharmacogenetic testing evaluates multiple pharmacodynamic and pharmacokinetic genes relevant to psychiatric treatment outcome and is therefore a plausible avenue to personalize MDD drug treatment for an individual. In a retrospective study of health claims data, authors report enhanced medication adherence and outpatient cost savings with Genecept assay-guided treatment [
[54]- Fagerness J.
- Fonseca E.
- Hess G.P.
- et al.
Pharmacogenetic-guided psychiatric intervention associated with increased adherence and cost savings.
]. A subsequent naturalistic study reported improved patient outcomes with Genecept assay-guided treatment for a mixed-diagnosis patient cohort [
[43]- Brennan F.X.
- Gardner K.R.
- Lombard J.
- et al.
A naturalistic study of the effectiveness of pharmacogenetic testing to guide treatment in psychiatric patients with mood and anxiety disorders.
]. However, these previous studies failed to isolate collective biomarkers specific to the clinical utility of MDD/GAD treatment.
Emphasis on patients with genetic predispositions to treatment resistant depression (TRD) is of grave importance and the most profound indications for the use of pharmacogenetic testing are in TRD cases [
14Psychiatric pharmacogenomics: how to integrate into clinical practice.
,
46- Peterson K.
- Dieperink E.
- Anderson J.
- Boundy E.
- Ferguson L.
- Helfand M.
Rapid evidence review of the comparative effectiveness, harms, and cost-effectiveness of pharmacogenomics-guided antidepressant treatment versus usual care for major depressive disorder.
]. For this group of patients it is particularly important to determine the supposed adequacy and outcome of treatment; 10–30% of the depressive population receiving treatment do not respond adequately to antidepressants [
55Approaches to understanding and addressing treatment-resistant depression: a scoping review.
,
56Treatment-resistant depression: therapeutic trends, challenges, and future directions.
]. This is especially troublesome as the burden of TRD is substantial: chronic unremitting depressive disease confers personal suffering and disability and carries an associated employee healthcare cost double that of nonresistant employees [
[57]- Greenberg P.
- Corey-Lisle P.K.
- Birnbaum H.
- Marynchenko M.
- Claxton A.
Economic implications of treatment-resistant depression among employees.
]. Patients with
MTHFR risk genotypes represent a population of patients at risk for poor SSRI response. This is supported by the concept that deficient folate levels impact catecholaminergic pathways [
[35]L-Methylfolate: a vitamin for your monoamines.
] and can correspondingly be seen in our reported evidence:
MTHFR risk patients demonstrated a significantly increased number of medication changes (i.e. failed medication trials) (
Table 1). Fundamentally,
SLC6A4 risk patients also represent a patient population at risk for poor SSRI response due to the mode of action of SSRIs and their reliance on adequate serotonin transporter levels [
[33]- Outhred T.
- Das P.
- Dobson-Stone C.
- Felmingham K.L.
- Bryant R.A.
- Nathan P.J.
- et al.
Impact of 5-HTTLPR on SSRI serotonin transporter blockade during emotion regulation: a preliminary fMRI study.
].
As in most studies including patients with MDD, heterogeneity between individual patients limits the power of genetic analyses. Discordances in cohort samples, including differences in diagnosis criteria and treatment regimens, further increase this phenotypic heterogeneity. Thus, the overall cohort is likely to include subgroups with unique susceptibility factors for clinical depression contingent on unidentified biological factors and environmental aspects not controlled for (
e.g. chaotic home-life or dietary choices). Additional study limitations include a lack of ancestral data and limited statistical power due to a relatively undersized patient cohort. As the power to detect associations was low, corrections of multiple testing were not performed, as these would increase the probability of producing false negative results. Lastly, hard endpoints on which to base clinical success, such as suicide or hospitalization, are infrequently encountered in psychiatric prospective studies of short follow-up duration [
[58]Expectations, validity, and reality in pharmacogenetics.
]. In an attempt to rectify this constraint, data were collected at three separate time points over a 3-month time period, each taking into account surrogate hard-endpoints more appropriate for a short-term trial. Future studies should aim to perform longer-term evaluations, incorporate hard-endpoints, and report data on ‘time to effectiveness’ to gauge the duration of time to treatment success/failure for antidepressant medications.
An important issue often unaddressed by studies evaluating pharmacogenetic testing is the issue of whether reported genetic information leads to better clinical outcomes (i.e. having clinical utility). The present study sought to address this by determining whether testing warranted not only changes in clinician prescribing behavior, but specifically assay-congruent changes. Moreover, this study identified distinct genetic components relevant to MDD/GAD treatment outcome—as opposed to a grouping of known pharmaco-psychiatric genes—and reports SLC6A4 and MTHFR as putative biological predictors of quality of life.
Article info
Publication history
Published online: December 13, 2017
Accepted:
November 29,
2017
Received in revised form:
September 8,
2017
Received:
July 27,
2017
Copyright
© 2017 The Authors. Published by Elsevier Inc.