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

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

×

Abstract

Objective:

There is burgeoning interest in augmentation strategies for improving inadequate response to antidepressants. The adjunctive use of standardized pharmaceutical-grade nutrients, known as nutraceuticals, has the potential to modulate several neurochemical pathways implicated in depression. While many studies have been conducted in this area, to date no specialized systematic review (or meta-analysis) has been conducted.

Method:

A systematic search of PubMed, CINAHL, Cochrane Library, and Web of Science was conducted up to December 2015 for clinical trials using adjunctive nutrients for depression. Where sufficient data were available, a random-effects model analyzed the standard mean difference between treatment and placebo in the change from baseline to endpoint, combining the effect size data. Funnel plot and heterogeneity analyses were also performed.

Results:

Primarily positive results were found for replicated studies testing S-adenosylmethionine (SAMe), methylfolate, omega-3 (primarily EPA or ethyl-EPA), and vitamin D, with positive isolated studies for creatine, folinic acid, and an amino acid combination. Mixed results were found for zinc, folic acid, vitamin C, and tryptophan, with nonsignificant results for inositol. No major adverse effects were noted in the studies (aside from minor digestive disturbance). A meta-analysis of adjunctive omega-3 versus placebo revealed a significant and moderate to strong effect in favor of omega-3. Conversely, a meta-analysis of folic acid revealed a nonsignificant difference from placebo. Marked study heterogeneity was found in a Higgins test for both omega-3 and folic acid studies; funnel plots also revealed asymmetry (reflecting potential study bias).

Conclusions:

Current evidence supports adjunctive use of SAMe, methylfolate, omega-3, and vitamin D with antidepressants to reduce depressive symptoms.

Augmentation strategies in depression treatment are being increasingly explored, with a growing recognition that for many people with a depressive disorder, full remission is either short-lived or absent (1). Even in cases of beneficial response, subsyndromal symptoms may still persist (2). Augmentation and combination approaches are often applied in clinical practice to provide an enhanced mood-elevating effect (especially in cases of perceived treatment resistance [3]), and this prescriptive approach may involve multiple agents with antidepressant activity (4). The combining of two or more established antidepressants may target different neurochemical pathways, while augmentation approaches may involve agents that work synergistically with the antidepressant (5). More novel approaches may target nonneurotransmitter pathways to provide adjuvant benefits that may ultimately improve mood (e.g., reduction of inflammation [6]). Augmentation strategies can be initiated either at the start of treatment or later, if there is insufficient response to monotherapy. The coadministration of standardized pharmaceutical-grade nutrients, referred to as nutraceuticals, may provide an effective and safe approach to enhancing antidepressant effects, either by synergistically augmenting a particular activity of an antidepressant medication (e.g., enhancing reuptake inhibition of monoamines) or by providing a range of additional biological effects (7).

While the pathophysiology of depression is complex (and still being unraveled), several key neurobiological mechanisms underpinning the disorder have been considered germane (monoamine impairment, neuroendocrinological changes, reduced neurogenesis, reduction-oxidation reaction [redox] and bioenergetics abnormalities, and cytokine alterations consistent with chronic inflammation) (8, 9). Several nutrients are known to have critical involvement in brain function, and some, such as omega-3, zinc, and folate, have been shown to affect an array of neurobiological processes that may be implicated in depression (10). By targeting these key neurobiological pathways through specific nutraceuticals, such adjunctive treatments have the potential to augment the response to antidepressants.

While the potential of antidepressant augmentation with nutraceuticals is compelling, only recently have enough clinical studies become available to permit a clearer determination of their effectiveness as augmentation agents. To our knowledge, to date there have been no comprehensive systematic reviews of this area or, where sufficient data exist, any specific meta-analysis of these adjunctive studies. Our intention is to provide a comprehensive and critical review of the literature, focusing specifically on the current evidence for adjunctive use of nutraceuticals with antidepressants for clinical depression, and to discuss their potential evidence-based application in clinical practice.

Method

Search Strategy

PubMed, CINAHL (Cumulative Index of Nursing and Allied Health Literature), Web of Science, and Cochrane Library databases were searched up to December 2015. We looked for human clinical trials by using the search terms “Depression,” “Major Depressive Disorder,” “Major Depression,” “Mood,” “Antidepressant,” and “SSRI” in combination with the search terms “Adjunct,” “Adjunctive,” “Adjuvant,” “Augmentation,” and “Add-on” in combination with a range of nutraceutical search terms and 14 individual nutrients known to be important for neurological function, e.g., omega-3, folic acid, amino acids, vitamins, and minerals. Clinical trial registers were searched for relevant studies to cross-reference with the literature review and to locate unpublished data. A forward search of the identified articles was subsequently performed by using a Web of Science cited reference search, in addition to hand-searching the literature and reference lists, contacting authors and academic personnel for studies in the area, and searching the Internet for “gray” literature (data, published or unpublished, that is not readily accessible through the main databases).

Study Inclusion Criteria

We reviewed studies that reported uncontrolled, controlled, or quasi-experimental human studies that used any adjunctive (i.e., combined with pharmacotherapy) nutrient-based intervention for either diagnosed major depressive disorder (primary diagnosis or comorbid with another condition, e.g., diabetes, cardiovascular disease, cancer) or ongoing depression (defined as current use of antidepressant medication and a moderate or above-threshold level of depressive symptoms according to a validated scale, e.g., a score above 17 on the Hamilton Depression Rating Scale [HAM-D]). Further, the depression had to be either clinician-diagnosed or a primary health issue (i.e., not depressive symptoms studied as a secondary outcome for any other primary medical condition). For a study to be considered an “adjunctive study,” more than 95% of the study participants must have been taking antidepressant medication. Studies that were open-label without a control (e.g., an antidepressant-only group or single-blind dose variation design) must have recruited participants with “nonresponsive” or “treatment-resistant” depression (defined as no response to current or additionally previous antidepressant medication used for that depressive episode). Use of a recognized depression assessment scale was required for the primary depression outcome. The studies were required to have a duration of treatment of at least 21 days and a total sample size of >10 per arm. Case studies were not included. Only English-language articles since 1960 were included. No criteria were set for gender, age, or ethnicity of participants. All articles that did not meet these criteria were excluded. Studies were selected for final inclusion by means of consensus within the research group.

Study Tabulation

Studies were tabulated in four separate groups: one-carbon cycle nutraceuticals, omega-3, tryptophan, and other nutraceuticals. These were evaluated for dose, age, sample size, method, coprescribed antidepressant medication, diagnostic criteria, percentage of female participants, completion rate, and whether the study revealed a significant effect in favor of treatment or control condition (full data appear in the supplement accompanying the online version of this article). In the tables presenting results, “+” is used to indicate a statistically significant reduction in depression rating scores in the treatment group compared with the control group at study endpoint, and “n.s.“ is used to indicate no significant difference in depression rating scores between the treatment and control groups at study endpoint. For open-label studies, “+” is employed if the treatment significantly reduced depression symptoms on the depression rating scale over the course of the trial compared with baseline, and “n.s.“ is used if the treatment did not significantly reduce depression symptoms on the depression rating scale over the duration of the trial compared to baseline. This information is also summarized in text.

Statistical Analysis

A review of the studies was employed to determine whether multiple studies with acceptable homogeneity and adequate data were available to apply a meta-analysis. Homogeneity sufficient for meta-analysis was defined as the availability of more than two randomized controlled trials studying a homogeneous isolated nutraceutical compound at a therapeutic dose for 21 days or more for the adjunctive treatment of major depressive disorder. Such trials were available only for omega-3 and folic acid. A random-effects model was used, as the directional effect of omega-3 and folic acid in depression has not yet been established. The model analyzed the standard mean difference between treatment and placebo in the change between baseline and endpoint, combining the effect size (Hedges g) data. Data were analyzed by means of Comprehensive Meta-Analysis software, version 3 (Biostat, Englewood, N.J., 2015). The pooled effect size was determined by using a 95% confidence interval, while significance was determined by z tests (significance was defined as a p value of <0.05). Sensitivity analyses were conducted by comparing the results in a fixed-effects model and by removing studies with lesser homogeneity. A homogeneity test (Higgins I2) and a visual funnel plot analysis were conducted to ascertain whether the effect sizes came from a homogeneous source (11), and a regression analysis was used to assess whether any relationship between sample size and results occurred.

Results

An initial search revealed 5,287 articles in the area, from which 571 were indexed as clinical trials. From subsequent hand-searching of the abstracts, 60 potential studies were revealed as being in the specific area of the systematic review. Analysis of the full texts revealed 40 studies that met the inclusion criteria (see Figure S1 in the online data supplement). Nine studies involved folic acid, methylfolate, folinic acid, or a combination of folic acid and vitamins B6 and B12; eight involved tryptophan (or 5-HTP); four involved S-adenosylmethionine (SAMe); eight involved omega-3 (a combination of eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA], EPA alone, or ethyl-EPA); two studies each were found on zinc, inositol, vitamin C, and vitamin D; and isolated studies were found for creatine, B12, and an amino acid combination. Common trial lengths were 4, 6, and 8 weeks (range 3–52 weeks), with a mean sample size of 63 participants (SD=75; range 20–475) and a mean age of 44 years (SD=11 years). The tables present results separately for one-carbon cycle nutraceuticals (Table 1), omega-3 (Table 2), tryptophan (Table 3), and other nutraceuticals (Table 4); the expanded tables in the online data supplement provide additional study features. Over two-thirds of the participants were female (69%). Of the 40 studies reviewed, 31 were randomized, double-blind, and placebo-controlled trials. Four studies had a 100% completion rate, with a mean overall completion rate of 85%.

TABLE 1. Studies of One-Carbon Cycle Nutraceuticals as Adjunctive Treatment for Depressiona

Intervention, First Author, and YearDaily DoseDesignStudy DurationNAntidepressantPrimary OutcomeResultb
S-Adenosylmethionine (SAMe)
Alpert (12) 20041600 mg (target dose)Open-label; treatment-resistantc6 weeks30Fluoxetine/paroxetine/citalopram ≥20 mg/day, escitalopram ≥10 mg/day, sertraline ≥50 mg/day, or venlafaxine ≥75 mg/dayHAM-D 17+
Bambling (13) 20151600 vs. 800 mgDosage-blind; open-label; RA; treatment-resistantd15 weeks36Any SSRIBDI+e
De Berardis (14) 2013800 mgOpen-label; treatment-resistantf; single-blind8 weeks25SSRI, SNRI, agomelatine, mirtazapine, or bupropion at adequate dose for at least 6 weeksHAM-D+
Papakostas (15) 20101600 mg (target dose) vs. placeboDouble-blind; RCT6 weeks73SSRI or SNRI at adequate and stable dose for at least 4 weeksHAM-D 17n.s.(+)g
Folic acid
Coppen (16) 20000.5 mg vs. placeboDouble-blind; RCT10 weeks127Fluoxetine 20 mg/dayHAM-D 17+h
Resler (17) 200810 mg vs. placeboDouble-blind; RCT6 weeks27Fluoxetine 20 mg/dayHAM-D 17+
Basoglu (18) 20092.5 mg vs. escitalopram onlyRA; open-label6 weeks42Escitalopram 10 mg/dayMADRSn.s.
Venkatasubramanian (19) 20131.5 vs. 5 mgDosage-blind; open-label, RA6 weeks42Fluoxetine 20 mg/dayHAM-D 17+
Bedson (20) 20145 mg vs. placeboDouble-blind; RCT12 weeks475Any antidepressant at adequate dose and durationBDI-IIn.s.
Folic acid (FA) and vitamins B12 and B6
Almeida (21) 2014FA 2 mg + B12 0.5 mg + B6 25 mg vs. placeboDouble-blind; RCT52 weeks153Citalopram 20–40 mg/dayMADRSn.s.(+)i
Methylfolate and folinic acid (ME: methylfolate; FO: folinic acid)
Godfrey (22) 1990ME 15 mg vs. placeboDouble-blind; RCT6 months24jUndefined antidepressant treatmentHAM-D 17+
Alpert (23) 2002FO 30 mg (target dose)Open-label; treatment-resistantc8 weeks22SSRI or venlafaxine for at least 4 weeksHAM-D 17+k
Papakostas (24) 2012ME 15 mg (target dose) vs. placebolDouble-blind; RCT; SSRI-resistantmFluoxetine/paroxetine/citalopram ≥20 mg/day, escitalopram ≥10 mg/day, or sertraline ≥50 mg/dayHAM-D 17
 Trial 160 days148n.s.
 Trial 260 days75+
Vitamin B12
Syed (25) 20131000 μg i.m. vs. antidepressant onlyOpen-label; RA; single-blind6 weeks73TCA equivalent to imipramine 100–200 mg/day or SSRI equivalent to fluoxetine 20–40 mg/dayHAM-D 20+

aAdditional study characteristics are shown in Table S1 in the data supplement that accompanies the online version of this article. BDI: Beck Depression Inventory; FA: folic acid; FO: folinic acid; HAM-D: Hamilton Depression Rating Scale; MADRS: Montgomery-Åsberg Depression Rating Scale; ME: methylfolate; RA: randomized allocation; RCT: randomized placebo-controlled trial; SNRI: serotonin-norepinephrine reuptake inhibitor; SSRI: selective serotonin reuptake inhibitor; TCA: tricyclic antidepressant.

b“+”indicates a statistically significant reduction in depression rating scores in the treatment group compared with the control group between baseline and endpoint or, if the study was open label, treatment significantly reduced depression symptoms on the depression rating scale over the course of the trial. “n.s.” indicates no significant difference in depression rating scores between the treatment and control conditions or, if the study was open label, the treatment did not significantly reduce depression symptoms on the depression rating scale over the duration of the trial.

cTreatment resistance was defined as at least one failed antidepressant trial for the current episode.

dTreatment resistance was defined as a history of suboptimal treatment response in the previous three episodes.

eSignificant reduction in symptoms from baseline to endpoint. However, there was no significant difference between the 1600-mg and 800-mg daily doses in the reduction of depression symptoms.

fTreatment resistance was defined as failures of two antidepressants of different classes.

gSuperior response and remission rates for SAMe group versus placebo (a priori primary outcome) but a nonsignificant between-groups reduction in HAM-D scores.

hFor females only; result not found in males.

iSuperior response and remission rates for FA/B12/B6 group versus placebo but nonsignificant reduction in MADRS scores for FA/B12/B6 versus placebo.

jPatients with depression and red cell folate levels below 200 μg/L.

kSignificant decrease in HAM-D scores over the course of the trial but modest response and remission rates (31% and 19%, respectively).

lStudy comprised two sequential double-blind randomized controlled trials. Trial 1 comprised a 60-day study divided into two 30-day periods. Participants were randomly assigned to one of three groups: 1) l-methylfolate for 60 days (7.5 mg/day for 30 days, then 15 mg/day for 30 days); 2) placebo for 30 days followed by l-methylfolate (7.5 mg/day) for 30 days; or 3) placebo for 60 days. Trial 2 was identical to Trial 1 except the dose of l-methylfolate was 15 mg/day for both 30-day periods.

mSSRI resistance was defined as a failure of one to two SSRI trials for the current episode.

TABLE 1. Studies of One-Carbon Cycle Nutraceuticals as Adjunctive Treatment for Depressiona

Enlarge table

TABLE 2. Studies of Omega-3 as Adjunctive Treatment for Depressiona

Intervention, First Author, and YearDaily DoseDesignStudy DurationNAntidepressantPrimary OutcomeResultb
EPA/DHA
Su (26) 20034.4 g EPA + 2.2 g DHA vs. placeboDouble-blind; RCT8 weeks (plus 1 week placebo run-in)28Any antidepressantscHAM-D 21+
Carney (27) 2009; coronary heart disease patientsd0.93 g EPA + 0.75 g DHA vs. placeboDouble-blind; RCT10 weeks (plus 2 week placebo run-in)122Sertraline 50 mg/dayBDI-IIn.s.
Gertsik (28) 20121.8 g EPA + 0.4 g DHA vs. placeboDouble-blind; RCT8 weeks (plus 1 week placebo run-in)42Citalopram 20–40 mg/day (titrated depending on response)HAM-D 21+
EPA vs. DHA
Mozaffari-Khosravi (29) 20131 g EPA vs. 1 g DHA vs. placeboDouble-blind; RCT12 weeks81Any antidepressantHAM-D 17+e
Ethyl-EPA (E-EPA)
Nemets (30) 20022 g vs. placeboDouble-blind; RCT4 weeks20Any antidepressantfHAM-D 24+
Peet (31) 20021 g vs. 2 g vs. 4 g vs. placeboDouble-blind; RCT12 weeks70Any antidepressantHAM-D 17+g
Jazayeri (32) 20081 g E-EPA + fluoxetine vs. 1 g E-EPA monotherapy vs. fluoxetine onlyDouble-blind; RCT; double dummy8 weeks60Fluoxetine 20 mg/dayHAM-D 17+h
Bot (33) 2010; diabetes patientsi1 g vs. placeboDouble-blind; RCT12 weeks25Any antidepressantMADRSn.s.

aAdditional study characteristics are shown in Table S2 in the data supplement that accompanies the online version of this article. BDI-II: Beck Depression Inventory; DHA: docosahexaenoic acid; EPA: eicosapentaenoic acid; E-EPA: ethyl-eicosapentaenoic acid; HAM-D: Hamilton Depression Rating Scale; MADRS: Montgomery-Åsberg Depression Rating Scale; RCT: randomized placebo-controlled trial.

b“+” indicates a statistically significant reduction in depression rating scores in the treatment group compared with the control group between baseline and endpoint or, if the study was open label, treatment significantly reduced depression symptoms on the depression rating scale over the course of the trial. “n.s.” indicates no significant difference in depression rating scores between the treatment and control conditions or, if the study was open label, the treatment did not significantly reduce depression symptoms on the depression rating scale over the duration of the trial.

cOne patient in each group was not taking an antidepressant (0.07% of the sample).

dAll patients were diagnosed with coronary heart disease defined as at least 50% stenosis in at least one major coronary artery, a history of revascularization, or a history of hospitalization for an acute coronary syndrome.

eEPA significantly reduced depression symptoms over both placebo and DHA.

fOne patient was not taking an antidepressant (5% of the sample).

gSignificant reduction in depression symptoms found for the group taking 1 g of E-EPA only, compared with placebo. Result not found for the 2-g and 4-g E-EPA groups.

hSignificant reduction in depression symptoms for E-EPA + fluoxetine in comparison to E-EPA only and fluoxetine only.

iAll patients were diagnosed with comorbid diabetes mellitus type 1 or 2.

TABLE 2. Studies of Omega-3 as Adjunctive Treatment for Depressiona

Enlarge table

TABLE 3. Studies of Tryptophan as Adjunctive Treatment for Depressiona

Intervention, First Author, and YearDaily DoseDesignStudy DurationNAntidepressantPrimary OutcomeResultb
Tryptophan
Levitan (34) 20004 g (target dose) vs. placeboDouble-blind; RCT8 weeks (plus 5-day placebo run-in)39Fluoxetine 20 mg/dayHAM-D 29n.s.
l-Tryptophan
Shaw (35) 1975Clomipramine only vs. clomipramine + 6 g l-tryptophan vs. clomipramine + desipramine vs. desipramine + 6 g l-tryptophanDouble-blind; RCT; triple dummy4 weeks54Clomipramine ≤175 mg/day or desipramine ≤225 mg/dayBDIn.s.
Thomson (36) 19823 g l-tryptophan only vs. amitriptyline only vs. 3 g l-tryptophan + amitriptyline vs. placebo onlyDouble-blind; RCT12 weeks (plus 1 week placebo run-in)115Amitriptyline 150 mg/dayHAM-D 18+c
dl-Tryptophan
Glassman (37) 196912, 15, or 18 g (depending on body weight) vs. placeboDouble-blind; CT3 weeks20Phenelzine 60 mg/dayHAM-D (modified)+
Ayuso Gutierrez (38) 19716 g vs. placeboDouble-blind; RCT3 weeks30Nialamide 500 mg/day i.m. (target dose)HAM-D+
Walinder (39) 19760.1 g/kg of body weight vs. placeboDouble-blind; RCT3.5 weeks26Clomipramine 150 mg/dayCronholm-Ottosson Depression Scale+
Walinder (40) 19810.1 g/kg of body weight vs. placeboDouble-blind; RCT3 weeks26Zimelidine 200 mg/dayCronholm-Ottosson Depression Scalen.s.
l-5-HTP
Nardini (41) 1983300 mg vs. placeboDouble-blind; RCT4 weeks26Chlorimipramine 50 mg/dayHAM-D+

aAdditional study characteristics are shown in Table S3 in the data supplement that accompanies the online version of this article. BDI: Beck Depression Inventory; CT: controlled trial; HAM-D: Hamilton Depression Rating Scale; N.A. not available; RCT: randomized placebo-controlled trial.

b“+” indicates a statistically significant reduction in depression rating scores in the treatment group compared with the control group between baseline and endpoint or, if the study was open label, treatment significantly reduced depression symptoms on the depression rating scale over the course of the trial. “n.s.” indicates no significant difference in depression rating scores between the treatment and control conditions or, if the study was open label, the treatment did not significantly reduce depression symptoms on the depression rating scale over the duration of the trial.

cAll active treatments were more effective than placebo. The combination (amitriptyline + l-tryptophan) was superior to the other active treatments alone (amitriptyline only and l-tryptophan only).

TABLE 3. Studies of Tryptophan as Adjunctive Treatment for Depressiona

Enlarge table

TABLE 4. Studies of Other Nutraceuticals as Adjunctive Treatment for Depressiona

Intervention, First Author, and YearDaily DoseDesignStudy DurationNAntidepressantPrimary Outcome MeasureResultb
Zinc
Siwek (42) 200925 mg vs. placeboDouble-blind; RCT12 weeks60Imipramine 100–200 mg/dayHAM-D 17n.s.c
Ranjbar (43) 201325 mg vs. placeboDouble-blind; RCT12 weeks44Citalopram 20–60 mg/day or fluoxetine 20–60 mg/dayBDI+
Vitamin C
Amr (44) 2013; pediatric patients1 g vs. placeboDouble-blind; RCT6 months27Fluoxetine 10–20 mg/dayCDRS+
Sahraian (45) 20151 g vs. placeboDouble-blind; RCT8 weeks43Citalopram 20 mg/dayHAM-D 21n.s.
Vitamin D3
Khoraminya (46) 20131500 IU vs. placeboDouble-blind; RCT8 weeks42Fluoxetine 20 mg/dayHAM-D 24+
Zanetidou (47) 2011; patients age >65300,000 IU vs. antidepressant onlyOpen-label; CT4 weeks24dAny antidepressantHAM-D+
Inositol
Levine (48) 199912 g vs. placeboDouble-blind; RCT4 weeks36SSRIHAM-Dn.s.
Nemets (49) 199912 g vs. placeboDouble-blind; RCT4 weeks42SSRIHAM-D 24n.s.
Amino acids
Ille (50) 2007Individualized amino acid mixture vs. placeboDouble-blind; RCT4 weeks40Mirtazapine (no dose specified)HAM-D+
Creatine
Lyoo (51) 20125 g (target dose) vs. placeboDouble-blind; RCT8 weeks52Escitalopram 20 mg/dayHAM-D 17+

aAdditional study characteristics are shown in Table S4 in the data supplement that accompanies the online version of this article. BDI: Beck Depression Inventory; CDRS: Children's Depression Rating Scale; CT: controlled trial; HAM-D: Hamilton Depression Rating Scale; RCT: randomized placebo controlled trial; SSRI: selective serotonin reuptake inhibitor.

b“+” indicates a statistically significant reduction in depression rating scores in the treatment group compared with the control group between baseline and endpoint or, if the study was open label, treatment significantly reduced depression symptoms on the depression rating scale over the course of the trial. “n.s.” indicates no significant difference in depression rating scores between the treatment and control conditions or, if the study was open label, the treatment did not significantly reduce depression symptoms on the depression rating scale over the duration of the trial.

cA significant treatment effect was found in a subgroup of patients who were considered treatment resistant, compared with placebo.

dTreatment group N=24; controls N=15.

TABLE 4. Studies of Other Nutraceuticals as Adjunctive Treatment for Depressiona

Enlarge table

Most studies used the DSM-IV criteria for a diagnosis of major depressive disorder, with the HAM-D being used in 29 studies (most of the others used the Beck Depression Inventory or the Montgomery-Åsberg Depression Rating Scale). A variety of antidepressant pharmacotherapies were used in the studies, which primarily used open inclusion of all SSRIs or commonly specified prescription of fluoxetine, citalopram, or escitalopram at adequate doses and durations. A positive effect of the adjunctive intervention was revealed in 68% of the clinical trials (including six out of eight omega-3 studies).

One-Carbon Cycle Nutraceuticals

Fifteen data sets in 14 studies were located concerning adjunctive use of nutrients involved in the one-carbon cycle: SAMe, folic acid (or related forms: folinic acid, methylfolate), B6, and B12 (Table 1). Eight of these were double-blind randomized controlled trials, with seven studying open-label augmentation after nonresponse to antidepressant medication. Eight studies used DSM-IV criteria, two DSM-III, three ICD-10, and the study by Syed et al. (25) made diagnoses according to depression level. Sample size ranged from 22 to 475, with the majority of studies having a primarily female sample (two folic acid studies had samples that were 85% [17] and 100% [19] female). Study lengths ranged from 6 to 52 weeks. Dosage variance was found among studies using oral SAMe (800 or 1600 mg/day) and methylfolate (15 or 30 mg/day). The HAM-D was used in all but four studies as the primary outcome measure. A range of antidepressants were used by participants, with fluoxetine being used solely in three studies. Ten out of the 15 trials revealed an effect in favor of this class of nutraceuticals, either over placebo or beyond baseline in nonresponsive depression.

A sufficient number of folic acid studies were available to perform a meta-analysis. Four sets of data on folic acid (0.5 to 10 mg) were included in the meta-analysis. Two out of the four studies revealed a benefit in favor of folic acid; however, the largest study (Bedson et al. [20]), with a robust sample of 475 subjects, revealed no significant difference from placebo. The pooled data in a random-effects meta-analytic model revealed a nonsignificant difference between folic acid and placebo (p=0.23; z=1.19, 95% confidence interval [CI], –0.31 to 1.29), with an inconsequential effect size (g) of 0.49 (Figure 1). Sensitivity analyses revealed that when the disproportionately large study (20) (N=475) was removed, the nonsignificant effect was maintained (p=0.29). When a fixed-effects model was adopted in place of a random-effects model, the significance of the results was also not altered (p=0.78). A Higgins test revealed substantial heterogeneity among folic acid depression studies (I2=93%, p<0.001), with the funnel plot showing one marked outlier study (17) (figure not shown); this reflects potential study bias. A regression analysis showed no relationship between sample size and the level of effect size (figure not shown). While heterogeneity may be produced when folic acid and methylfolate studies are combined, when the only methylfolate randomized controlled trial with available baseline and end-point data (Papakostas et al. [24] trial 1/phase 1: 7.5 mg; trial 2/phase 1: 15 mg) was added to the meta-analysis, a similar nonsignificant effect was revealed (p=0.25; z=1.15, 95% CI, –0.22 to 0.83).

FIGURE 1.

FIGURE 1. Meta-Analyses for Folic Acid and Omega-3 as Adjunctive Treatment for Depression

Omega-3

We located eight studies that met the inclusion criteria. All studies were double-blind randomized controlled trials. The trials involved EPA and DHA combinations (N=3), ethyl-EPA (N=4), and EPA versus DHA (N=1) (Table 2). The range of EPA varied between 930 mg and 4.4 g (commonly 1 to 2 g per day, which is a therapeutic dose). The sample sizes of the study arms were generally modest (full sample size range, 20–122), and all but one study (27) had a majority of female participants. Study lengths ranged from 4 to 12 weeks, with a variety of antidepressants used by the participants. All studies diagnosed the depression by means of DSM-IV criteria, except the study by Peet et al. (31), who studied physician-diagnosed depression treated with antidepressants.

Eleven sets of data were included in the meta-analysis (including the four-arm Peet et al. study [31] and the three-arm Jazayeri et al. study [32]). Seven out of the 11 data sets revealed a benefit in favor of omega-3, but only six were statistically significant. The pooled data in a random-effects meta-analytic model revealed an effect size (Hedges g) of 0.61 (z=2.63, 95% CI, 0.15 to 1.06), which was highly statistically significant (p=0.009; Figure 1). Sensitivity analyses revealed that when meta-analysis of the data was restricted to EPA-inclusive studies (removing the DHA-only arm of the Mozaffari-Khosravi et al. study [29]), this effect was slightly strengthened (g=0.69, p=0.007). When a fixed-effects model was adopted in place of a random-effects model, the significance of the results was p<0.001. Aside from the methodological limitations of the generally small samples and the samples defined by diabetes and coronary heart disease in the studies by Bot et al. (33) and Carney et al. (27), respectively, most of the other study elements were reasonably consistent. A Higgins test, however, revealed substantial data heterogeneity among the omega-3 depression studies (I2=82%, p<0.001), with the funnel plot showing three outlier studies (figure not shown); this reflects potential study bias. A regression analysis showed no relationship between sample size and the level of effect size (data not shown).

Tryptophan

Eight studies investigating the various forms of tryptophan (including one 5-HTP study) were reviewed (Table 3). Many of the earlier studies (six studies before 1990) used dl-tryptophan, which is potentially less effective than l-tryptophan, as the D isomer is inactive. One study used l-5-HTP, the active precursor of serotonin and a derivative of tryptophan, in conjunction with a tricyclic antidepressant. The majority of studies (N=7) were published before 1985 and used the diagnosis of “endogenous depression” (N=4) or “affective disorder” (N=2). The HAM-D was used by most studies to asses depression symptoms (N=5), and two studies used the Cronholm-Ottosson Depression Scale. Because most of the studies reviewed were conducted between 1969 and 1983, predominately tricyclic antidepressants (N=4) and monoamine oxidase inhibitors (N=2) were prescribed in conjunction with tryptophan (or 5-HTP). Four out of the seven tryptophan studies and the one L-5-HTP study found a positive effect of the adjunctive treatment, relative to placebo and/or a control condition. We considered undertaking a meta-analysis of tryptophan but were unable to perform one because of missing raw data in many of the early studies.

Other Nutraceuticals

Ten other studies involving a range of nutraceuticals were located, but there were too few on specific nutraceuticals for us to perform a meta-analysis (Table 4). Two studies each tested zinc, vitamin C, vitamin D, and inositol; one study used a mixture of amino acids, and one tested creatine. All studies except one (47) were double-blind randomized controlled trials ranging from 4 to 26 weeks. All studies used DSM-IV to establish the depression diagnosis, and all but two studies used the HAM-D for assessment. A range of antidepressants were coprescribed, with the most common being fluoxetine (N=3). In all but one study (44), the majority of participants were female. The individual study findings demonstrated positive and significant results for vitamin D (a separate negative community-based, nonclinical trial notwithstanding [52]), creatine, and an amino acid combination; mixed results for zinc and vitamin C; and no significant benefit over placebo for inositol.

Safety

All the nutraceuticals were generally well tolerated; gastrointestinal symptoms (e.g., constipation, stomach upset, and diarrhea) were the most commonly reported adverse events across all nutraceutical groups (omega-3, one-carbon cycle, and tryptophan). Across all studies, the rates of dropouts due to side effects were very low (less than 2% of the samples), further supporting tolerability.

Despite the mild nature of the adverse effects reported in the included studies, nutraceuticals are not without risk or serious safety concerns when used at high doses, over long periods of time, and/or when combined with certain medications. Potential carcinogenicity should be considered regarding supplements containing folic acid, as higher folate blood levels are linked to increased risk of prostate cancer (53, 54). While meta-analyses have not revealed definitive evidence for the association between folic acid supplementation and a range of cancers (55), and adequate folate consumption from vegetables and whole grains has potential cancer-protective properties, high dosages of folic acid may not be advised in people with cancer, since it increases cell proliferation (56). Folic acid has also been implicated in accelerating cognitive decline with age and reducing the efficacy of particular antifolate drugs, such as immunosuppressants (57).

Omega-3 also should be prescribed with caution in people with prostate cancer, since a meta-analysis showed that certain fatty acids may have an association with prostate cancer risk (58). It should be noted that this association was weak and further larger epidemiological studies are required to assess this comprehensively. Higher-dose omega-3 supplementation has also been suspected to increase bleeding, impair immune function, increase lipid peroxidation, and impair lipid and glucose metabolism (59). There is also evidence that omega-3 likely increases LDL cholesterol concentrations, but only when dosages of DHA and combined EPA/DHA are over 2 g/day (59).

SAMe has been associated with an increased risk of hypomania or manic switching in depressed patients. However, the switching has primarily been reported in patients with a diagnosed bipolar disorder and with intravenous or intramuscular administrations of SAMe (60, 61). The phenomenon has not been observed in clinical trials of oral administration of SAMe (62). Tryptophan and 5-HTP, when used in conjunction with other serotonergic agents (such as antidepressants, opioid pain medications), may cause a toxic state, serotonin syndrome, due to an excess of serotonin in the brain (63). Hypercalcemia and vascular calcification may occur when vitamin D is used at high doses (≥275 µg/day) (64), while high doses of zinc (>25 mg/day) can lead to copper deficiency (65). At doses above 1000 mg/day, vitamin C has been linked to an increased risk of kidney stones in men due to its conversion to oxalate and excretion in urine (66). Caution is warranted when reviewing the safety data on nutraceuticals, and many of the suggested potential risks have not been revealed in randomized controlled trials or meta-analyses.

Discussion

The overall findings revealed a substantive number of human clinical trials testing adjunctive nutraceuticals to augment antidepressant activity in depression. Primarily positive results in replicated studies were found for SAMe, methylfolate, omega-3 (EPA or ethyl-EPA specifically), and vitamin D. Due to positive isolated studies, tentative consideration may also be extended to creatine and an amino acid combination. Further research is needed to clarify whether zinc, vitamin C, or tryptophan (or more specifically 5-HTP, the active precursor of serotonin) may be of value, while inositol is unlikely to have any utility as an adjunctive antidepressant agent. It should be noted, however, that previous findings suggest that inositol may have an antidepressant effect as a monotherapy agent rather than an adjunctive agent for depression (67, 68).

The finding from the omega-3 meta-analysis demonstrates that this augmentation approach significantly reduces depressive symptoms beyond placebo and thus has potential clinical and public health significance. As detailed in a recent general meta-analysis, it is advised that EPA or ethyl-EPA dominant formulas be used, as DHA may not be effective (69). In summation, EPA-rich omega-3 fish oil may be recommended for the adjunctive treatment of major depressive disorder. In respect to folic acid and methylfolate, results are less clear. As the meta-analysis revealed, folic acid cannot be firmly recommended; however, the “active” forms of methylfolate and folinic acid can be tentatively recommended. It should be noted that while the review included various formulations (folic acid, folinic acid, methylfolate), the latter is a patented derivative that has been the subject of several large-scale commercially sponsored clinical trials. There is a potential concern that commercially sponsored trials may be biased toward positive results. For example, a review by Perlis et al. (70) found that industry-funded randomized controlled trials with a reported conflict of interest were 4.9 times more likely to report positive results. Regardless, the vast majority of efficacy-focused biological medicine studies have commercial sponsorship, and while there is the potential for inherent bias, the methodology of these studies was rigorous.

All of the nutraceuticals reviewed in this article have mechanistic antidepressant activity underpinning their use. The one-carbon cycle agents (SAMe, folic acid/methylfolate, B6/B12) are critical in the methylation processes of monoamines (71). In particular, SAMe may improve depressed mood through enhanced methylation of catecholamines and increased serotonin turnover, reuptake inhibition of norepinephrine, enhanced dopaminergic activity, decreased prolactin secretion, and increased phosphatidylcholine conversion (72). Omega-3 (in particular, EPA) exerts antidepressant activity potentially through modulation of norepinephrine, dopamine, and serotonin reuptake, degradation, synthesis, and receptor binding; through enhancement of glutathione antioxidant capacity (73); and through enhancement of cell membrane fluidity (74).

Another possible explanation for the antidepressant efficacy of these compounds may reside with their anti-inflammatory properties, which are well demonstrated with SAMe (75) and EPA (76, 77). A recent double-blind monotherapy randomized controlled trial (N=155) showed that patients with major depressive disorder who have biomarkers of increased inflammation (e.g., interleukins or C-reactive protein) may benefit from EPA over both placebo and DHA, compared with those with low levels of inflammation biomarkers (78). Zinc is one of the most prevalent trace elements in the amygdala, hippocampus, and neocortex, and aside from having anti-inflammatory and immunological-modulating properties, it is involved with hippocampal neurogenesis through up-regulation of brain-derived neurotrophic factor (BDNF), while also modifying N-methyl-d-aspartate (NMDA) and glutamate activity (79, 80).

Amino acids are essential precursors of all proteins involved in the manufacture of neurochemicals. In particular, tryptophan (and particularly its derivative 5-HTP) is an essential monoamine precursor required for the synthesis of serotonin (81). Creatine plays a pivotal role in brain energy homeostasis, and altered cerebral energy metabolism at a cellular level may be involved in the pathophysiology of depression (51). Oral creatine supplementation may modify brain high-energy phosphate metabolism in people with depression. Inositol is incorporated into neuronal cell membranes as inositol phospholipids, and it is the key metabolic precursor in the phosphoinositide (PI) intracellular secondary messenger cycle (82). The PI cycle is involved with a broad range of neurotransmitter systems, including adrenergic, serotonergic, dopaminergic, glutaminergic, and cholinergic receptor types (83). The potential exists that both inositol and SSRIs converge on the same mechanism of action, as 5-HT2 receptors are linked to the PI cycle signal transduction pathway (84). Regardless of this preclinical activity, as reviewed above, this effect was not manifested in adjunctive treatment studies. However, as discussed above, inositol may have a greater therapeutic effect as a monotherapy agent for depression (67, 68) or for panic disorder (85).

Vitamin D can be considered a neurosteroid, with vitamin D receptors being identified in areas involved with depression, such as the prefrontal cortex, hypothalamus, and substantia nigra (86). Vitamin D has been revealed to increase the expression of genes encoding for tyrosine hydroxylase (precursor of dopamine and norepinephrine) (86). Further, a major dopamine metabolite in the striatum and accumbens has been found in methamphetamine-treated animals administered vitamin D (87). Vitamin C is an essential vitamin involved in various neuroendocrine activities and is needed for the production of neurotransmitters, such as serotonin (88). An animal study showed that the coadministration of vitamin C (1 mg/kg p.o.) potentiated the action of subeffective doses of fluoxetine (1 mg/kg), thus providing a synergistic antidepressant-like effect with an SSRI (89).

The methodology of this review has several limitations, and general cautions regarding the findings need to be considered. First, our search criteria were modestly restrictive, and several excluded studies (i.e., case studies and trials with non-English-speaking patients or those with general depressive symptoms) were not included, and thus a more expansive perspective might have been reached with their inclusion. Because much of the early literature on SAMe is in Italian, it is not covered by this review. Readers are advised to additionally consult the Italian literature on this nutraceutical. Caution also needs to be extended to the findings of some of the nutraceuticals (aside from omega-3, SAMe, and folic acid/methylfolate) due to a limited number of studies and the small sample sizes of the studies reviewed. Potential publication bias was also revealed in the folic acid and omega-3 meta-analyses (from funnel plot analysis), as well as heterogeneity among studies (in respect to I2 analysis and marked differences in medications used, study lengths, nutraceutical dosages, and participant characteristics). Several studies of treatment-resistant depression were also open label, and while any effect beyond baseline is of merit, the lack of placebo cautions interpretation of the results. Lastly, the older studies did not provide comprehensive details of the methodology or the raw statistics, and thus a more thorough assessment could not be achieved. Caveats regarding the prescription of the nutraceuticals relate to the quality of nutrient products (especially with SAMe, though current formulations have potentially more stable shelf-lives) and the correct formulation and dosage (especially with omega-3). Expense may also be an issue in the case of SAMe and methylfolate.

Nutraceutical applications in psychiatry are advancing, as reflected in recent international collaborative consensus and position statements discussing the potential of nutraceutical use in psychiatry (10, 90) As noted, much more work is needed, and while an evolving body of research is strengthening the potential of nutraceuticals (and dietary considerations) as an important element in modern psychiatric practice, we are only beginning to study their potential applications. A major barrier to this field is the often unpatentable nature of these compounds, and large-scale randomized controlled trials may be unfeasible due to a potential lack of financial incentive. Despite the challenges of soliciting non-industry-sponsored funding, randomized controlled trials with robust sample sizes and the application of pharmacogenomic and neuroimaging technologies to determine biomarkers of response are now required. Finally, while the current studies (whether as first-line therapy or in treatment-resistant depression) were pooled in the present article, as the database of this research expands, future subanalyses potentially can be conducted assessing these individual nutraceuticals as first-line agents.

Another area of potential interest is the use of combination nutraceuticals. Our research group is currently testing an adjunctive nutraceutical formulation in the treatment of major depressive disorder not responsive to stable antidepressant medication (7). Nutrients commonly work in concert, and as detailed above, a range of nutraceuticals modulate several key pathways involved with the pathogenesis of depression. The formulation currently being tested (involving SAMe, ethyl-EPA, zinc, folinic acid, and 5-HTP and relevant cofactors) may provide an array of antidepressant action beyond that of isolated approaches. In conclusion, as detailed above, several nutraceuticals may hold a potential clinical application to enhance the antidepressant effect of medications. Further, the integration of nutraceuticals into clinical practice guidelines should be considered, especially in circumstances where the risk-benefit ratio may not justify the use of pharmacological treatment (such as in mild symptom presentations and in the treatment of children, adolescents, and pregnant women [if the nutraceuticals are established as safe]). While future research needs to determine how this is applied to the individual (and whether it is more pertinent in cases of nutrient deficiencies), many nutraceuticals are low-cost options that are worthy of clinical consideration.

From the ARCADIA Mental Health Research Group, Professorial Unit, Department of Psychiatry, The Melbourne Clinic, University of Melbourne, Melbourne, Australia; the Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, Australia; the Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston; the IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Australia; Orygen, the National Centre of Excellence in Youth Mental Health and the Centre of Youth Mental Health, Melbourne, Australia; and the Florey Institute for Neuroscience and Mental Health, Melbourne, Australia.
Address correspondence to Dr. Sarris ().

Dr. Sarris is supported by a C.R. Roper Fellowship at the University of Melbourne. Dr. Berk is supported by NHMRC Senior Principal Research Fellowship 1059660.

All authors except Dr. Murphy disclose having been provided previous or current research funding to study pharmaceuticals and nutraceuticals for mood disorders, including but not restricted to omega 3, folinic acid, 5-MTHF, tryptophan, and S-adenosylmethionine. Dr. Sarris reports no specific direct conflict of interest; general disclosures involve presentation honoraria, travel support, clinical trial grants, or book royalties from Integria Healthcare, MediHerb, Pfizer, Taki Mai, Pepsico, BioCeuticals, Blackmores, Soho-Flordis, Elsevier, HealthEd, and the National Health and Medical Research Council. Dr. Mischoulon has received research support from FisherWallace, Nordic Naturals, Methylation Sciences, and PharmoRx Therapeutics; he has received honoraria for consulting, speaking, and writing from the Massachusetts General Hospital Psychiatry Academy; and he has received royalties from Lippincott Williams & Wilkins for the book Natural Medications for Psychiatric Disorders: Considering the Alternatives. Dr. Papakostas has served as a consultant for Abbott, AstraZeneca, Avanir, Axsome (on behalf of Massachusetts General Hospital [MGH]), Brainsway, Bristol-Myers Squibb, Cephalon, Dey, Eli Lilly, Genentech (on behalf of MGH), GlaxoSmithKline, Evotec, H. Lundbeck, Inflabloc, Janssen Global Services (on behalf of MGH), Jazz Pharmaceuticals, Johnson & Johnson (on behalf of MGH), Methylation Sciences, Novartis, One Carbon Therapeutics (on behalf of MGH), Osmotica (on behalf of MGH), Otsuka, PAMLAB, Pfizer, Pierre Fabre Laboratories, Ridge Diagnostics (formerly known as Precision Human Biolaboratories), Shire, Sunovion, Taisho, Takeda, Theracos, and Wyeth; he has received honoraria (for lectures or consultancy) from Abbott, AstraZeneca, Avanir, Bristol-Myers Squibb, Brainsway, Cephalon, Dey, Eli Lilly, Evotec, Forest, GlaxoSmithKline, Inflabloc, Jazz, H. Lundbeck, Medichem, Meiji Seika, Novartis, Otsuka, PAMLAB, Pfizer, Pierre Fabre Laboratories, Ridge Diagnostics, Shire, Sunovion, Takeda, Theracos, Titan, and Wyeth; he has received research support (paid to hospital) from AstraZeneca, Bristol-Myers Squibb, Forest, the National Institute of Mental Health, Neuralstem, PAMLAB, Pfizer, Ridge Diagnostics (formerly known as Precision Human Biolaboratories), Sunovion, Tal Medical, and Theracos; he has also served (not currently) on the speaker’s bureau for Bristol-Myers Squibb and Pfizer. Dr. Fava has received research support from and/or served as an adviser or consultant to Acadia, Alkermes, AstraZeneca, Avanir, AXSOME Therapeutics, Biogen, Bristol-Myers Squibb, Cerecor, Dinippon Sumitomo, Eli Lilly, EnVivo, Euthymics Bioscience, Forest, FORUM, GenOmind, GlaxoSmithKline, Intracellular, Janssen R&D, Johnson & Johnson Pharmaceutical Research and Development, Lundbeck, Merck, Methylation Sciences, MSI Methylation Sciences, the National Center for Complementary and Alternative Medicine, the National Coordinating Center for Integrative Medicine, NIDA, NIMH, Naurex, Nestle Health Sciences, Neuralstem, Novartis, Nutrition 21, Osmotica, Otsuka, PamLab, Pfizer, PharmoRx Therapeutics, Photothera, PPD, Puretech Ventures, PsychoGenics, RCT Logic (formerly Clinical Trials Solutions), Reckitt Benckiser, Ridge Diagnostics, Roche, Sanofi-Aventis, Servier Laboratories, the Stanley Medical Research Institute, Sunovion, Taisho, Takeda, Tal Medical, and VistaGen; he has had speaking or publishing roles for the American Society of Clinical Psychopharmacology, Belvoir Media Group, CME Institute/Physicians Postgraduate Press, and MGH Psychiatry Academy; he has equity holdings in Compellis and PsyBrain; he is named on patents for sequential parallel comparison design, licensed by MGH to Pharmaceutical Product Development, and a patent application for a combination of ketamine plus scopolamine in major depressive disorder, licensed by MGH to Biohaven; and he is a copyright holder for the MGH Cognitive and Physical Functioning Questionnaire, the Sexual Functioning Inventory, the Antidepressant Treatment Response Questionnaire, Discontinuation-Emergent Signs and Symptoms, the Symptoms of Depression Questionnaire, and SAFER and has publications with Lippincott Williams & Wilkins, Wolkers Kluwer, and World Scientific Publishing. Dr. Berk has received grant/research support from Stanley Medical Research Foundation, MBF, NHMRC, NHMRC Senior Principal Research Fellowship (1059660), Cooperative Research Centre, Simons Autism Foundation, Cancer Council of Victoria, Rotary Health, Meat and Livestock Board, Woolworths, BeyondBlue, Geelong Medical Research Foundation, Bristol-Myers Squibb, Eli Lilly, GlaxoSmithKline, Organon, Novartis, Mayne Pharma, and Servier; he has been a speaker for AstraZeneca, Bristol-Myers Squibb, Eli Lilly, GlaxoSmithKline, Lundbeck, Pfizer, Sanofi Synthelabo, Servier, Solvay, and Wyeth; he has been a consultant to AstraZeneca, Bristol-Myers Squibb, Eli Lilly, Bioadvantex, Merck, Glaxo SmithKline, Lundbeck, Janssen Cilag, and Servier; and he is a co-inventor of two provisional patents regarding the use of NAC and related compounds for psychiatric indications, which, while assigned to the Mental Health Research Institute, could lead to personal remuneration upon a commercialization event. Dr. Ng has served on advisory boards for Servier, Wyeth, and Eli Lilly; has received research grant support from Wyeth and Lundbeck; and has received honoraria from Servier, Bristol-Myers Squibb, Organon, Eli Lilly, GlaxoSmithKline, Janssen-Cilag, Astra-Zeneca, Wyeth, and Pfizer. Dr. Murphy reports no financial relationships with commercial interests.

References

1 Shelton RC: What are the comparative benefits and harms of augmentation treatments in major depression? J Clin Psychiatry 2015; 76:e531–e533Crossref, MedlineGoogle Scholar

2 Pietrzak RH, Kinley J, Afifi TO, et al.: Subsyndromal depression in the United States: prevalence, course, and risk for incident psychiatric outcomes. Psychol Med 2013; 43:1401–1414Crossref, MedlineGoogle Scholar

3 Papakostas GI, Ionescu DF: Towards new mechanisms: an update on therapeutics for treatment-resistant major depressive disorder. Mol Psychiatry 2015; 20:1142–1150Crossref, MedlineGoogle Scholar

4 Zhou X, Ravindran AV, Qin B, et al.: Comparative efficacy, acceptability, and tolerability of augmentation agents in treatment-resistant depression: systematic review and network meta-analysis. J Clin Psychiatry 2015; 76:e487–e498Crossref, MedlineGoogle Scholar

5 Sarris J, Kavanagh DJ, Byrne G: Adjuvant use of nutritional and herbal medicines with antidepressants, mood stabilizers and benzodiazepines. J Psychiatr Res 2010; 44:32–41Crossref, MedlineGoogle Scholar

6 Maes M, Leonard B, Fernandez A, et al.: (Neuro)inflammation and neuroprogression as new pathways and drug targets in depression: from antioxidants to kinase inhibitors. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:659–663Crossref, MedlineGoogle Scholar

7 Sarris J, Stough C, Bousman C, et al.: An adjunctive antidepressant nutraceutical combination in treating major depression: study protocol, and clinical considerations. Adv Integr Med 2015; 2:49–55CrossrefGoogle Scholar

8 Belmaker RH, Agam G: Major depressive disorder. N Engl J Med 2008; 358:55–68Crossref, MedlineGoogle Scholar

9 Berk M, Williams LJ, Jacka FN, et al.: So depression is an inflammatory disease, but where does the inflammation come from? BMC Med 2013; 11:200Crossref, MedlineGoogle Scholar

10 Sarris J, Logan AC, Akbaraly TN, et al.: Nutritional medicine as mainstream in psychiatry. Lancet Psychiatry 2015; 2:271–274Crossref, MedlineGoogle Scholar

11 Higgins JP, Thompson SG: Quantifying heterogeneity in a meta-analysis. Stat Med 2002; 21:1539–1558Crossref, MedlineGoogle Scholar

12 Alpert JE, Papakostas G, Mischoulon D, et al.: S-adenosyl-L-methionine (SAMe) as an adjunct for resistant major depressive disorder: an open trial following partial or nonresponse to selective serotonin reuptake inhibitors or venlafaxine. J Clin Psychopharmacol 2004; 24:661–664Crossref, MedlineGoogle Scholar

13 Bambling M, Parham SC, Coulson S, et al.: S-adenosylmethionine (SAMe) and magnesium orotate as adjunctives to SSRIs in sub-optimal treatment response of depression in adults: a pilot study. Adv Integr Med 2015; 2:56–62CrossrefGoogle Scholar

14 De Berardis D, Marini S, Serroni N, et al.: S-Adenosyl-L-methionine augmentation in patients with stage II treatment-resistant major depressive disorder: an open label, fixed dose, single-blind study. Scientific World Journal 2013; 2013:204649CrossrefGoogle Scholar

15 Papakostas GI, Mischoulon D, Shyu I, et al.: S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: a double-blind, randomized clinical trial. Am J Psychiatry 2010; 167:942–948LinkGoogle Scholar

16 Coppen A, Bailey J: Enhancement of the antidepressant action of fluoxetine by folic acid: a randomised, placebo controlled trial. J Affect Disord 2000; 60:121–130Crossref, MedlineGoogle Scholar

17 Resler G, Lavie R, Campos J, et al.: Effect of folic acid combined with fluoxetine in patients with major depression on plasma homocysteine and vitamin B12, and serotonin levels in lymphocytes. Neuroimmunomodulation 2008; 15:145–152Crossref, MedlineGoogle Scholar

18 Basoglu C, Ates MA, Algul A, et al.: Adjuvant folate with escitalopram treatment and homocystein, folate, vitamin B-12 levels in patients with major depressive disorder. Klin Psikofarmakol Bul. 2009; 19:135–142Google Scholar

19 Venkatasubramanian R, Kumar CN, Pandey RS: A randomized double-blind comparison of fluoxetine augmentation by high and low dosage folic acid in patients with depressive episodes. J Affect Disord 2013; 150:644–648Crossref, MedlineGoogle Scholar

20 Bedson E, Bell D, Carr D, et al.: Folate augmentation of treatment–evaluation for depression (FolATED): randomised trial and economic evaluation. Health Technology Assessment. 2014; 18(48):vii–viiiMedlineGoogle Scholar

21 Almeida OP, Ford AH, Hirani V, et al.: B vitamins to enhance treatment response to antidepressants in middle-aged and older adults: results from the B-VITAGE randomised, double-blind, placebo-controlled trial. Br J Psychiatry 2014; 205:450–457Crossref, MedlineGoogle Scholar

22 Godfrey PS, Toone BK, Carney MW, et al.: Enhancement of recovery from psychiatric illness by methylfolate. Lancet 1990; 336:392–395Crossref, MedlineGoogle Scholar

23 Alpert JE, Mischoulon D, Rubenstein GE, et al.: Folinic acid (Leucovorin) as an adjunctive treatment for SSRI-refractory depression. Ann Clin Psychiatry 2002; 14:33–38Crossref, MedlineGoogle Scholar

24 Papakostas GI, Shelton RC, Zajecka JM, et al.: L-methylfolate as adjunctive therapy for SSRI-resistant major depression: results of two randomized, double-blind, parallel-sequential trials. Am J Psychiatry 2012; 169:1267–1274LinkGoogle Scholar

25 Syed EU, Wasay M, Awan S: Vitamin B12 supplementation in treating major depressive disorder: a randomized controlled trial. Open Neurol J 2013; 7:44–48Crossref, MedlineGoogle Scholar

26 Su K-P, Huang S-Y, Chiu C-C, et al.: Omega-3 fatty acids in major depressive disorder: a preliminary double-blind, placebo-controlled trial. Eur Neuropsychopharmacol 2003; 13:267–271Crossref, MedlineGoogle Scholar

27 Carney RM, Freedland KE, Rubin EH, et al.: Omega-3 augmentation of sertraline in treatment of depression in patients with coronary heart disease: a randomized controlled trial. JAMA 2009; 302:1651–1657Crossref, MedlineGoogle Scholar

28 Gertsik L, Poland RE, Bresee C, et al.: Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder. J Clin Psychopharmacol 2012; 32:61–64Crossref, MedlineGoogle Scholar

29 Mozaffari-Khosravi H, Yassini-Ardakani M, Karamati M, et al.: Eicosapentaenoic acid versus docosahexaenoic acid in mild-to-moderate depression: a randomized, double-blind, placebo-controlled trial. Eur Neuropsychopharmacol 2013; 23:636–644Crossref, MedlineGoogle Scholar

30 Nemets B, Stahl Z, Belmaker RH: Addition of omega-3 fatty acid to maintenance medication treatment for recurrent unipolar depressive disorder. Am J Psychiatry 2002; 159:477–479LinkGoogle Scholar

31 Peet M, Horrobin DF: A dose-ranging study of the effects of ethyl-eicosapentaenoate in patients with ongoing depression despite apparently adequate treatment with standard drugs. Arch Gen Psychiatry 2002; 59:913–919Crossref, MedlineGoogle Scholar

32 Jazayeri S, Tehrani-Doost M, Keshavarz SA, et al.: Comparison of therapeutic effects of omega-3 fatty acid eicosapentaenoic acid and fluoxetine, separately and in combination, in major depressive disorder. Aust N Z J Psychiatry 2008; 42:192–198Crossref, MedlineGoogle Scholar

33 Bot M, Pouwer F, Assies J, et al.: Eicosapentaenoic acid as an add-on to antidepressant medication for co-morbid major depression in patients with diabetes mellitus: a randomized, double-blind placebo-controlled study. J Affect Disord 2010; 126:282–286Crossref, MedlineGoogle Scholar

34 Levitan RD, Shen JH, Jindal R, et al.: Preliminary randomized double-blind placebo-controlled trial of tryptophan combined with fluoxetine to treat major depressive disorder: antidepressant and hypnotic effects. J Psychiatry Neurosci 2000; 25:337–346MedlineGoogle Scholar

35 Shaw DM, Macsweeney DA, Hewland R, et al.: Tricyclic antidepressants and tryptophan in unipolar depression. Psychol Med 1975; 5:276–278Crossref, MedlineGoogle Scholar

36 Thomson J, Rankin H, Ashcroft GW, et al.: The treatment of depression in general practice: a comparison of L-tryptophan, amitriptyline, and a combination of L-tryptophan and amitriptyline with placebo. Psychol Med 1982; 12:741–751Crossref, MedlineGoogle Scholar

37 Glassman AH, Platman SR: Potentiation of a monoamine oxidase inhibitor by tryptophan. J Psychiatr Res 1969; 7:83–88Crossref, MedlineGoogle Scholar

38 Ayuso Gutierrez JL, Aliño JJ: Tryptophan and an MAOI (nialamide) in the treatment of depression: a double-blind study. Int Pharmacopsychiatry 1971; 6:92–97Crossref, MedlineGoogle Scholar

39 Walinder J, Skott A, Carlsson A, et al.: Potentiation of the antidepressant action of clomipramine by tryptophan. Arch Gen Psychiatry 1976; 33:1384–1389Crossref, MedlineGoogle Scholar

40 Wålinder J, Carlsson A, Persson R: 5-HT reuptake inhibitors plus tryptophan in endogenous depression. Acta Psychiatr Scand Suppl 1981; 290:179–190Crossref, MedlineGoogle Scholar

41 Nardini M, De Stefano R, Iannuccelli M, et al.: Treatment of depression with L-5-hydroxytryptophan combined with chlorimipramine, a double-blind study. Int J Clin Pharmacol Res 1983; 3:239–250MedlineGoogle Scholar

42 Siwek M, Dudek D, Paul IA, et al.: Zinc supplementation augments efficacy of imipramine in treatment resistant patients: a double blind, placebo-controlled study. J Affect Disord 2009; 118:187–195Crossref, MedlineGoogle Scholar

43 Ranjbar E, Kasaei MS, Mohammad-Shirazi M, et al.: Effects of zinc supplementation in patients with major depression: a randomized clinical trial. Iran J Psychiatry 2013; 8:73–79MedlineGoogle Scholar

44 Amr M, El-Mogy A, Shams T, et al.: Efficacy of vitamin C as an adjunct to fluoxetine therapy in pediatric major depressive disorder: a randomized, double-blind, placebo-controlled pilot study. Nutr J 2013; 12:31Crossref, MedlineGoogle Scholar

45 Sahraian A, Ghanizadeh A, Kazemeini F: Vitamin C as an adjuvant for treating major depressive disorder and suicidal behavior, a randomized placebo-controlled clinical trial. Trials 2015; 16:94Crossref, MedlineGoogle Scholar

46 Khoraminya N, Tehrani-Doost M, Jazayeri S, et al.: Therapeutic effects of vitamin D as adjunctive therapy to fluoxetine in patients with major depressive disorder. Aust N Z J Psychiatry 2013; 47:271–275Crossref, MedlineGoogle Scholar

47 Zanetidou S, Belvederi Murri M, Buffa A, et al.: Vitamin D supplements in geriatric major depression. Int J Geriatr Psychiatry 2011; 26:1209–1210Crossref, MedlineGoogle Scholar

48 Levine J, Mishori A, Susnosky M, et al.: Combination of inositol and serotonin reuptake inhibitors in the treatment of depression. Biol Psychiatry 1999; 45:270–273Crossref, MedlineGoogle Scholar

49 Nemets B, Mishory A, Levine J, Belmaker RH: Inositol addition does not improve depression in SSRI treatment failures. J Neur Transm 1999; 106:795-798Crossref, MedlineGoogle Scholar

50 Ille R, Spona J, Zickl M, et al.: “Add-on”-therapy with an individualized preparation consisting of free amino acids for patients with a major depression. Eur Arch Psychiatry Clin Neurosci 2007; 257:222–229Crossref, MedlineGoogle Scholar

51 Lyoo IK, Yoon S, Kim TS, et al.: A randomized, double-blind placebo-controlled trial of oral creatine monohydrate augmentation for enhanced response to a selective serotonin reuptake inhibitor in women with major depressive disorder. Am J Psychiatry 2012; 169:937–945LinkGoogle Scholar

52 Sanders KM, Stuart AL, Williamson EJ, et al.: Annual high-dose vitamin D3 and mental well-being: randomised controlled trial. Br J Psychiatry 2011; 198:357–364Crossref, MedlineGoogle Scholar

53 Wang R, Zheng Y, Huang JY, et al.: Folate intake, serum folate levels, and prostate cancer risk: a meta-analysis of prospective studies. BMC Public Health 2014; 14:1326Crossref, MedlineGoogle Scholar

54 Tio M, Andrici J, Cox MR, et al.: Folate intake and the risk of prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2014; 17:213–219Crossref, MedlineGoogle Scholar

55 Qin X, Cui Y, Shen L, et al.: Folic acid supplementation and cancer risk: a meta-analysis of randomized controlled trials. Int J Cancer 2013; 133:1033–1041Crossref, MedlineGoogle Scholar

56 Baggott JE, Oster RA, Tamura T: Meta-analysis of cancer risk in folic acid supplementation trials. Cancer Epidemiol 2012; 36:78–81Crossref, MedlineGoogle Scholar

57 European Food Safety Authority: ESCO Report on Analysis of Risks and Benefits of Fortification of Food With Folic Acid 2009. http://www.efsa.europa.eu/en/supporting/pub/3eGoogle Scholar

58 Fu YQ, Zheng JS, Yang B, et al.: Effect of individual omega-3 fatty acids on the risk of prostate cancer: a systematic review and dose-response meta-analysis of prospective cohort studies. J Epidemiology 2015; 25:261–274Crossref, MedlineGoogle Scholar

59 European Food Safety Authority Panel on Dietetic Products, Nutrition and Allergies: Scientific opinion on the tolerable upper intake level of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA). EFSA Journal 2012; 10:2815CrossrefGoogle Scholar

60 Carney MW, Martin R, Bottiglieri T, et al.: Switch mechanism in affective illness and S-adenosylmethionine. Lancet 1983; 1:820–821Crossref, MedlineGoogle Scholar

61 Lipinski JF, Cohen BM, Frankenburg F, et al.: Open trial of S-adenosylmethionine for treatment of depression. Am J Psychiatry 1984; 141:448–450LinkGoogle Scholar

62 Murphy BL, Babb SM, Ravichandran C, et al.: Oral SAMe in persistent treatment-refractory bipolar depression: a double-blind, randomized clinical trial. J Clin Psychopharmacol 2014; 34:413–416Crossref, MedlineGoogle Scholar

63 Ciprian-Ollivier J, Cetkovich-Bakmas M, Albin J, et al.: SSRI, L-tryptophan and serotonin syndrome: experience with 75 patients. Eur Neuropsychopharmacol 1996; 6(6, suppl 3):143–143CrossrefGoogle Scholar

64 European Food Safety Authority Panel on Dietetic Products, Nutrition and Allergies: Scientific opinion on the tolerable upper intake level of vitamin D. EFSA Journal 2012; 10:2813CrossrefGoogle Scholar

65 European Food Safety Authority Panel on Dietetic Products, Nutrition and Allergies: Scientific opinion on dietary reference values for zinc. EFSA Journal 2014; 12:3844CrossrefGoogle Scholar

66 Ferraro PM, Curhan GC, Gambaro G, et al.: Total, dietary, and supplemental vitamin C intake and risk of incident kidney stones. Am J Kidney Dis (Epub ahead of print, Oct 10, 2015)MedlineGoogle Scholar

67 Mukai T, Kishi T, Matsuda Y, et al.: A meta-analysis of inositol for depression and anxiety disorders. Hum Psychopharmacol 2014; 29:55–63Crossref, MedlineGoogle Scholar

68 Levine J, Barak Y, Gonzalves M, et al.: Double-blind, controlled trial of inositol treatment of depression. Am J Psychiatry 1995; 152:792–794LinkGoogle Scholar

69 Lin PY, Mischoulon D, Freeman MP, et al.: Are omega-3 fatty acids antidepressants or just mood-improving agents? The effect depends upon diagnosis, supplement preparation, and severity of depression (letter). Mol Psychiatry 2012; 17:1161–1163; author reply 1163–1167Crossref, MedlineGoogle Scholar

70 Perlis RH, Perlis CS, Wu Y, et al.: Industry sponsorship and financial conflict of interest in the reporting of clinical trials in psychiatry. Am J Psychiatry 2005; 162:1957–1960LinkGoogle Scholar

71 King WD, Ho V, Dodds L, et al.: Relationships among biomarkers of one-carbon metabolism. Mol Biol Rep 2012; 39:7805–7812Crossref, MedlineGoogle Scholar

72 Papakostas GI: Evidence for S-adenosyl-L-methionine (SAM-e) for the treatment of major depressive disorder. J Clin Psychiatry 2009; 70(suppl 5):18–22Crossref, MedlineGoogle Scholar

73 Smesny S, Milleit B, Schaefer MR, et al.: Effects of omega-3 PUFA on the vitamin E and glutathione antioxidant defense system in individuals at ultra-high risk of psychosis. Prostaglandins Leukot Essent Fatty Acids 2015; 101:15–21Crossref, MedlineGoogle Scholar

74 Sarris J, Mischoulon D, Schweitzer I: Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J Clin Psychiatry 2012; 73:81–86Crossref, MedlineGoogle Scholar

75 Pfalzer AC, Choi SW, Tammen SA, et al.: S-adenosylmethionine mediates inhibition of inflammatory response and changes in DNA methylation in human macrophages. Physiol Genomics 2014; 46:617–623Crossref, MedlineGoogle Scholar

76 Tsunoda F, Lamon-Fava S, Asztalos BF, et al.: Effects of oral eicosapentaenoic acid versus docosahexaenoic acid on human peripheral blood mononuclear cell gene expression. Atherosclerosis 2015; 241:400–408Crossref, MedlineGoogle Scholar

77 Honda KL, Lamon-Fava S, Matthan NR, et al.: EPA and DHA exposure alters the inflammatory response but not the surface expression of Toll-like receptor 4 in macrophages. Lipids 2015; 50:121–129Crossref, MedlineGoogle Scholar

78 Rapaport MH, Nierenberg AA, Schettler PJ, et al.: Inflammation as a predictive biomarker for response to omega-3 fatty acids in major depressive disorder: a proof-of-concept study. Mol Psychiatry 2016; 21:71–79Crossref, MedlineGoogle Scholar

79 Szewczyk B, Kubera M, Nowak G: The role of zinc in neurodegenerative inflammatory pathways in depression. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:693–701Crossref, MedlineGoogle Scholar

80 Swardfager W, Herrmann N, McIntyre RS, et al.: Potential roles of zinc in the pathophysiology and treatment of major depressive disorder. Neurosci Biobehav Rev 2013; 37:911–929Crossref, MedlineGoogle Scholar

81 Byerley WF, Judd LL, Reimherr FW, et al.: 5-Hydroxytryptophan: a review of its antidepressant efficacy and adverse effects. J Clin Psychopharmacol 1987; 7:127–137Crossref, MedlineGoogle Scholar

82 Kim H, McGrath BM, Silverstone PH: A review of the possible relevance of inositol and the phosphatidylinositol second messenger system (PI-cycle) to psychiatric disorders—focus on magnetic resonance spectroscopy (MRS) studies. Hum Psychopharmacol 2005; 20:309–326Crossref, MedlineGoogle Scholar

83 Fisher SK, Heacock AM, Agranoff BW: Inositol lipids and signal transduction in the nervous system: an update. J Neurochem 1992; 58:18–38Crossref, MedlineGoogle Scholar

84 Einat H, Belmaker RH: The effects of inositol treatment in animal models of psychiatric disorders. J Affect Disord 2001; 62:113–121Crossref, MedlineGoogle Scholar

85 Palatnik A, Frolov K, Fux M, et al.: Double-blind, controlled, crossover trial of inositol versus fluvoxamine for the treatment of panic disorder. J Clin Psychopharmacol 2001; 21:335–339Crossref, MedlineGoogle Scholar

86 Bertone-Johnson ER: Vitamin D and the occurrence of depression: causal association or circumstantial evidence? Nutr Rev 2009; 67:481–492Crossref, MedlineGoogle Scholar

87 Cass WA, Smith MP, Peters LE: Calcitriol protects against the dopamine- and serotonin-depleting effects of neurotoxic doses of methamphetamine. Ann N Y Acad Sci 2006; 1074:261–271Crossref, MedlineGoogle Scholar

88 Chambial S, Dwivedi S, Shukla KK, et al.: Vitamin C in disease prevention and cure: an overview. Indian J Clin Biochem 2013; 28:314–328Crossref, MedlineGoogle Scholar

89 Binfaré RW, Rosa AO, Lobato KR, et al.: Ascorbic acid administration produces an antidepressant-like effect: evidence for the involvement of monoaminergic neurotransmission. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:530–540Crossref, MedlineGoogle Scholar

90 Sarris J, Logan AC, Akbaraly TN, et al.: International Society for Nutritional Psychiatry Research consensus position statement: nutritional medicine in modern psychiatry. World Psychiatry 2015; 14:370–371Crossref, MedlineGoogle Scholar