Accepted Manuscript

Nutraceuticals for Major Depressive Disorder- More is Not Merrier: an 8-week double-blind, randomized, controlled trial

Jerome Sarris PhDMHSc , Gerard J Byrne MBBSPhD , Con Stough PhD , Chad Bousman PhDMPH , David Mischoulon MDPhD , Jenifer Murphy BPsych (hons)PhD , Patricia MacDonald BPsych (Hons) , Laura Adams BPsychMCouns , Sonia Nazareth MPsych , Georgina Oliver MSc , Lachlan Cribb BSci (Hons) , Karen Savage BPsych (Hons) , Ranjit Menon MD , Suneel Chamoli MD , Michael Berk MDPhD , Chee Ng MBBSMD

PII: S0165-0327(18)31411-3 DOI: https://doi.org/10.1016/j.jad.2018.11.092 Reference: JAD 10310

To appear in: Journal of Affective Disorders

Received date: 29 June 2018 Revised date: 18 October 2018 Accepted date: 12 November 2018

Please cite this article as: Jerome Sarris PhDMHSc , Gerard J Byrne MBBSPhD , Con Stough PhD , Chad Bousman PhDMPH , David Mischoulon MDPhD , Jenifer Murphy BPsych (hons)PhD , Patricia MacDonald BPsych (Hons) , Laura Adams BPsychMCouns , Sonia Nazareth MPsych , Georgina Oliver MSc , Lachlan Cribb BSci (Hons) , Karen Savage BPsych (Hons) , Ranjit Menon MD , Suneel Chamoli MD , Michael Berk MDPhD , Chee Ng MBBSMD , Nutraceu- ticals for Major Depressive Disorder- More is Not Merrier: an 8-week double-blind, randomized, controlled trial, Journal of Affective Disorders (2018), doi: https://doi.org/10.1016/j.jad.2018.11.092

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. ACCEPTED MANUSCRIPT Highlights:  A combination nutraceutical treatment did not outperform placebo in treating symptoms of depression, and was in fact less effective  Genetic polymorphisms and BDNF levels were not related to treatment response  A very high placebo response rate may have warranted a placebo run-in design  Future research may focus on precision-based approaches tailoring treatment to deficiencies, neurochemical abnormalities, or pharmacogenetic differences

ACCEPTED MANUSCRIPT

1

ACCEPTED MANUSCRIPT Nutraceuticals for Major Depressive Disorder- More is Not Merrier:

an 8-week double-blind, randomized, controlled trial

Jerome Sarris PhD, MHSc1,2, Gerard J Byrne MBBS PhD3, Con Stough PhD4, Chad Bousman PhD, MPH 5,6,

David Mischoulon, MD, PhD7 Jenifer Murphy BPsych (hons), PhD2, Patricia MacDonald BPsych (Hons) 3,

Laura Adams BPsych, MCouns, Sonia Nazareth MPsych 3, Georgina Oliver MSc2, Lachlan Cribb BSci (Hons)2,

Karen Savage BPsych (Hons)2 Ranjit Menon MD6 Suneel Chamoli MD3, Michael Berk MD, PhD 6,8,9,10, Chee

Ng, MBBS, MD6

1 NICM Health Research Institute, Western Sydney University

2 The , Department of Psychiatry, The Melbourne Clinic, Professorial Unit

3 The , Department of Psychiatry

4 Swinburne University of Technology, Centre for Human Psychopharmacology

5 University of Calgary, Departments of Medical Genetics, Psychiatry, and Physiology & Pharmacology,

Calgary, AB, Canada

6 The University of Melbourne, Department of Psychiatry, Parkville

7 Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital,

Harvard Medical School

8 Deakin University, IMPACT SRC, School of Medicine, Barwon Health, Geelong 9 Florey ACCEPTEDInstitute of Neuroscience and Mental Health, MANUSCRIPT The University of Melbourne, Parkville, 10 Orygen, The Centre of Excellence in Youth Mental Health, The University of Melbourne, Parkville

Word Count: 4250

Figures and Tables: 6

2

ACCEPTED MANUSCRIPT

Correspondence:

Professor Jerome Sarris NICM Health Research Institute, Western Sydney University, Westmead, New South Wales [email protected]

ACCEPTED MANUSCRIPT

3

ACCEPTED MANUSCRIPT ABSTRACT

Background: One of the most pressing questions in “Nutritional Psychiatry” is whether using combinations of different nutraceuticals with putative antidepressant activity may provide an enhanced synergistic antidepressant effect.

Methods: A phase II/III, Australian multi-site, 8-week, double-blind, RCT involving 158 outpatients with a DSM-5 diagnosis of MDD. The intervention consisted of a nutraceutical combination: S-adenosyl methionine; Folinic acid; Omega-3 fatty acids; 5-HTP, Zinc picolinate, and relevant co-factors versus placebo. The primary outcome was change in MADRS scores, and hypothesis-driven analyses of potential moderators of response involving key SNPs, and BDNF.

Results: Placebo was superior to the nutraceutical combination in reducing MADRS score (differential reduction -1.75 points), however a mixed linear model revealed a non-significant Group X Time interaction (p = 0.33). Response rates were 40% for the active intervention and 51% for the placebo; remission rates were 34% and 43% for active and placebo groups, respectively. No significant differences were found between groups on any other secondary depression, anxiety, psychosocial, or sleep outcome measures. Key SNPs and BDNF did not significantly moderate response. No significant differences occurred between groups for total adverse effects, aside from more nausea in the active group.

Limitations: Very high placebo response rates suggest a placebo run-in design may have been valuable.

Interpretation: The adoption of a nutraceutical „shotgun‟ approach to treating MDD was not supported, and appeared to be less effective than adding placebo to treatment as usual.

Funding: NHMRC APP1048222

Trial Registration: ANZCTR 12613001300763

Key Words: Antidepressant, Nutraceutical, Nutrient, Depression, Clinical Trial, Pharmacogenomics RunningACCEPTED Header: Nutraceutical Combination Clinical MANUSCRIPT Trial in Major Depression

4

ACCEPTED MANUSCRIPT 1. Introduction

Widespread use of nutraceuticals (standardised, pharmaceutical-grade nutrient-based supplements) for diverse medical issues is normative. Indeed the nutraceutical market is almost a quarter the size of the global pharmaceutical market, expected to continue to grow markedly in the next 5 years. Yet the efficacy of most supplements and nutraceuticals is not established. Against this background, people with Major Depressive Disorder (MDD), a prevalent and highly disabling mental illness, causing marked occupational and social impairment and reduced quality of life, are frequent users of such agents

(Donohue and Pincus, 2007). This is understandable as current treatment outcomes are inadequate, with around two-thirds of those treated with first-line antidepressants not reaching remission (Rush,

2007). MDD is a chronic condition for many, often requiring multiple treatment attempts. One potential approach to improving non-response to antidepressants is the use of adjunctive nutraceuticals (J Sarris et al., 2015c, 2015a).

The biopathophysiology of MDD involves a range of abnormalities such as monoaminergic system disturbance, neuro-endocrine changes, reduced brain-derived neurotropic factor (BDNF), and cytokine alterations (Antonijevic, 2006; Hindmarch, 2001; Plotsky et al., 1998; Raison et al., 2006; Ressler and

Nemeroff, 2000). Several of these key neurobiological mechanisms may be modulated by nutraceuticals such as S-adenosyl methionine (SAMe), 5-hydroxytryptophan (5-HTP), eicosapentaenoic acid (EPA), zinc, and folic acid (either as folinic acid or methylfolate); with emerging evidence for the use of some of these nutraceuticals as adjunctive antidepressants (Sarris et al., 2016). While the conventional paradigm mandates the study of individual agents to avoid the confound of multiple interventions, this approach may not be beneficial in studying nutraceuticals. Nutrients commonly work in concert (Jacobs and Tapsell, 2007) and, as outlined above, a range of nutraceuticals modulate a range of key pathways involved with the pathogenesis of depression (see Figure 1). ACCEPTED ………………………. FigureMANUSCRIPT 1 About Here ……………………

SAMe is an endogenous sulphur-containing compound that is a critical neurochemical component involved in the „one carbon‟ cycle responsible for the methylation of neurotransmitters that regulate

5

ACCEPTED MANUSCRIPT mood. SAMe may improve depressed mood via enhanced methylation of catecholamines and increased serotonin turnover, reuptake inhibition of norepinephrine, enhanced dopaminergic activity, decreased prolactin secretion, and increased phosphatidylcholine conversion (Bottiglieri and Hyland, 1994;

Papakostas et al., 2003). Our previous 12-week 3-arm double-blind RCT (n=144) using SAMe monotherapy (1600mg/day) versus selective serotonin reuptake inhibitor (SSRI) escitalopram (20mg) and placebo in adults with MDD found a significant difference between SAMe from baseline to week 12

(p=0.039) versus placebo in a subset of the parent study sample (Sarris et al., 2014). At week 12 endpoint, the remission rates on the Hamilton Depression Rating Scale (HAM-D< 7) were 34% for

SAMe, 23% for escitalopram and 6% for placebo, significantly in favour of SAMe (p=0.014). In addition, a 6-week double-blind RCT by colleagues Papakostas et al. (2010) involving 73 MDD patients non-responsive to SSRIs found response (HAM–D>50% reduction) and remission rates were significantly higher for patients treated with adjunctive SAMe (36.1% and 25.8% respectively) than adjunctive placebo (17.6% vs. 11.7% respectively).

5-HTP is an essential monoamine precursor derived from L-tryptophan required for the synthesis of serotonin, which has been studied extensively as an antidepressant. Eight controlled adjunctive studies using L-tryptophan or 5-HTP with antidepressants provide encouraging positive evidence, with augmentation effects shown with phenelzine sulphate, clomipramine, tranylcypromine, and fluoxetine

(Byerley et al., 1987).

Omega-3 fatty acids have a critical role in neurological activity and in depression, especially if inflammatory processes are implicated (Raison et al., 2006; Tassoni et al., 2008). The antidepressant activity of omega-3 fatty acids (in particular EPA) appears to occur via modulation of norepinephrine, dopamine and serotonin re-uptake, degradation, synthesis and receptor binding; anti-inflammatory effects; and the stabilization of the neuronal cell membrane (Mischoulon and Freeman, 2013). There is also evidence for the efficacy of EPA as an adjunct to SSRIs (Chalon, 2006; Tassoni et al., 2008;

WilliamsACCEPTED et al., 2006). MANUSCRIPT

Zinc is a divalent cation that is one of the most prevalent trace elements in the amygdala, hippocampus, and neocortex brain regions, and is involved in hippocampal neurogenesis via upregulation of BDNF, while also modifying N-methyl-D-aspartate (NMDA) and glutamate activity

6

ACCEPTED MANUSCRIPT (Szewczyk et al., 2011). Evidence suggests potential benefits of zinc as a stand-alone intervention or as an adjunct to antidepressants for depression (Lai et al., 2012).

Folate (folic acid being nutraceutical form) is involved with methylation pathways in the „one carbon‟ cycle, contributes to the metabolism and synthesis of various monoamines, and most notably is involved in the synthesis of SAMe from homocysteine (Papakostas et al., 2003). Several studies have assessed the antidepressant effect of folic acid with concomitant antidepressant use (Fava and

Mischoulon, 2009) with mostly positive results in enhancing either antidepressant response rates, or increasing the onset of response.

In view of the theoretical potential to impact neurobiological pathways associated with antidepressant treatments and favourable existing data for the antidepressant effect of individual nutraceuticals such as SAMe, 5-HTP, EPA, zinc and folinic acid, we used a double-blind RCT design to evaluate whether such an evidence-based nutraceutical combination (NC) would be an effective intervention in adults with MDD who were currently inadequately responsive to treatment as usual. The study also sought to assess the relationship between treatment response and changes in key biomarkers (brain-derived neurotrophic factor [BDNF], zinc, folate, B12, fatty acids and homocysteine), and evaluate whether treatment response was modified by single nucleotide polymorphisms (SNPs) or other polymorphisms in specific genes involving methylation, monoamine, and pharmacokinetic pathways.

2. Materials and methods

2.1 Overview

We report on the results of a phase II/III, multi-site, 8-week, double-blind, placebo controlled RCT studying a NC vs matching placebo in participants with current MDD who were not adequately responsive to their current antidepressant medication at an adequate therapeutic dose (SSRI, SNRI,

NaRI, tetracyclicACCEPTED or 5-HT2c antagonist) or undergoing MANUSCRIPT „treatment as usual‟ (standard visits to their GP, antidepressant treatment, general psychological care, self-care, or no care). The trial sites were at The

Melbourne Clinic (The University of Melbourne), Richmond, Melbourne, Australia; and The Royal

Brisbane and Women‟s Hospital (University of Queensland), Herston, Brisbane, Australia. Recruitment occurred from September 2013 until July 2017. The study had ethical clearance (TMC-HREC 220, UQ-

7

ACCEPTED MANUSCRIPT MREC 2013000199), and is registered on ANZCTR (protocol number: 12613001300763). The study was funded by an Australian National Health and Medical Research Council project grant (APP1048222), and was co-sponsored by FIT-BioCeuticals (who were not involved in any aspect of study design, statistical analysis or manuscript preparation). For further information on the project‟s aims and methods cf.

(Sarris et al., 2015).

2.2 Inclusion and exclusion criteria

Eligible participants were: aged 18 to 70 years; currently taking an SSRI, SNRI, NaRI, tetracyclic

(mirtazapine) or 5-HT2c antagonist (agomelatine) for a minimum of four weeks, (and on a stable dose for a minimum of two weeks), or on stable treatment as usual medical care (as detailed above); fulfilling the DSM-5 diagnostic criteria for MDD on the MINI; presenting with moderate to severe depression (Montgomery-Asberg Depression Rating Scale: MADRS ≥ 18) at time of study entry (14-25 on MADRS for participants not taking an antidepressant due to an ethics committee safety decision); meeting SAFER 2.0 criteria (Desseilles et al., 2013) for participation in a clinical trial; fluent in written and spoken English and had the capacity to consent to the study and follow its procedures.

Exclusion criteria were: currently taking MAOIs (reversible or non-reversible) or tricyclic antidepressants (due to greater safety concerns with older agents), or specified nutraceuticals (e.g. St

John‟s wort, SAMe, 5-HTP at any dose, or folic acid >500mcg per day, omega-3 >180mg EPA per day, zinc > 10mg per day- in such cases a one to two week washout (depending on nutraceutical half-life) was employed before rescreening and potential inclusion); presenting with suicidal ideation

(> 3 on MADRS suicidal thoughts domain) at the time of study entry; three or more failed trials of pharmacotherapy or somatic therapy (e.g. electroconvulsive therapy) for the current major depressive episode; Meeting DSM-IV diagnostic criteria for bipolar disorder I/II or presence of psychosis on structured interview (MINI 6.0); a clinical diagnosis of a substance/alcohol use disorder within the last 12 monthsACCEPTED on structured interview (MINI 6.0); recentlyMANUSCRIPT commenced psychotherapy (more than four weeks of stable treatment was acceptable); taking warfarin or phenytoin; known or suspected clinically unstable systemic medical disorder (including cancer, organ failure, or serious cardio/cerebrovascular disease); pregnancy or breastfeeding; not currently using medically approved contraception (including abstinence) if female and of childbearing age; or having an allergy or intolerance to seafood or to any of the investigated nutraceuticals.

8

ACCEPTED MANUSCRIPT

2.3 Outcome measures

Participants meeting inclusion criteria were screened firstly with the SAFER 2.0 criteria (for assessing suitability for study entry to establish a stable episode of depression) and the MINI version 6.0 (for assessing MDD and other comorbid psychiatric disorders diagnosis). Research assistants with psychology or mental health qualifications were trained on the MINI and primary assessment measures by a clinical psychologist.

The primary outcome scale measuring depression levels was the MADRS. Secondary outcome scales included: Beck Depression Inventory (BDI-II); Hamilton Anxiety Rating Scale (HAMA); Short Form

Survey-12 (SF-12); Leeds Sleep Evaluation Questionnaire (LSEQ); Arizona Sexual Experience Scale

(ASEX); the CORE Assessment of Psychomotor Change; and the Clinical Global Impression (CGI) severity (CGI-S) and improvement (CGI-I) scales. The Systematic Assessment for Treatment Emergent

Effects (SAFTEE) was used to assess for adverse events (AEs), while Serotonin Syndrome assessments were completed at each follow-up assessment session, due to the inclusion of 5-HTP in the formula, a known serotonergic agent. This was assessed via the Sternbach and Hunter Serotonin Toxicity Criteria.

Inter-rater reliability was assessed between 6 and 12 months to ensure inter-site validity of the MADRS ratings. A Krippendorff‟s alpha (Hayes and Krippendorff, 2007) of 0.788 was calculated indicating sound agreement between raters. SNPs in specific genes involved in methylation or monoamine pathways, or the pharmacokinetics of the nutrients, were assessed via blood sample at baseline (Table 1). Serum levels of BDNF were also collected at baseline and week 8.

………………………. Table 1 About Here ……………………

2.4 Randomisation, blinding, and treatment intervention

Participants were randomly assigned via computerized number generation to either the NC group or placebo. An independent researcher conducted the randomization, while trial researchers and investigatorsACCEPTED were blinded as to which groups MANUSCRIPT the participants were assigned to (as were the participants themselves). All participants were required to take 2 tablets and 2 capsules twice per day for 8 weeks.

The NC formula was provided by BioCeuticals, who were responsible for authentication of the purity of the nutrients and the preparation via Pharmaceutical Good Manufacturing Practice in a Therapeutic

Goods Administration certified facility. The products provided consisted of soft-gel capsules and tablets,

9

ACCEPTED MANUSCRIPT and stored at room temperature 15 to 25 degrees Celsius). The tablets consisted of a per day dose of

SAMe (800mg/day) folinic acid (500mcg/day); co-factor vitamin B12 (200mcg/day), while the capsules consisted of a per day dose of omega-3 fatty acid concentrate (EPA-esters 1000mg/day, DHA-esters

656mg/day) 5-HTP (200mg/day) zinc picolinate (30 mg elemental/day); and co-factors vitamin B6

(100mg/day), vitamin C (60mg/day), and magnesium (amino acid chelate, elemental 40mg/day) to assist in the conversion of 5-HTP to serotonin. In addition, a cofactor vitamin E (40IU/day), was added to prevent oxidation of the omega-3. The placebo capsules and tablets were matched internally and externally in terms of colour, shape, and size, with non-active excipients (e.g. microcellulose) added and 13mg of fish oil added to the placebo capsules to assist in blinding for any potential fishy aftertaste experienced. Stability testing was undertaken with the capsules, at the 2.5 year time point and the active constituents were within a-priori acceptable parameters. Serum nutrient levels, genotyping, and

BDNF were analysed by Australian Clinical Labs scientists. In respect to the polymorphisms, these were assayed from DNA extracted from whole blood using Qiagen, QIAmp mini-columns according to the manufacturer‟s instruction. The genotyping was then performed by single base extension assays and analysed on the Sequenom Massarray. Serum BDNF levels for each participant in the study were obtained through ELISA kits performed by Australian Clinical Labs scientists. Whole blood samples were collected, from which serum BDNF was extracted and tested. Extracts were run in triplicate to enhance confidence in the results, with the mean taken from the three results.

2.5 Procedure Participants were recruited through advertisements in local newspapers, radio, social media, websites, and posters and brochures displayed in public places. Participants were required to attend five visits at the study site at weeks 0 (baseline), 2, 4, 6 and 8, as well as a safety assessment over the phone or in person at week 1. At the baseline visit, participants were asked to complete consent forms, screening assessments, and mood, anxiety, sleep, and health questionnaires. All eligible participants were randomlyACCEPTED allocated to a treatment arm, and corresponding MANUSCRIPT treatment was provided. All subsequent visits followed the same outline of the baseline session excluding consent forms and screening assessments and including a safety assessment. In addition, participants were required to provide a blood sample at baseline (prior to commencing the intervention) and week 8 visit (shortly before ceasing the intervention). To compensate for their time and travel expenses, participants were given

10

ACCEPTED MANUSCRIPT AUS $50 at the week 2 visit and another AUS $50 at the final week 8 visit. On completion they were also provided two months‟ supply of SAMe tablets.

2.6 Statistical analyses

The study was powered to detect a potential small to moderate statistical difference between groups

(using all data via intention-to-treat analysis). Based on a two-tailed analysis with α=0.05, β=0.80, and a critical F1,157 of 3.90, 156 participants were required to detect a small effect size difference (F=0.175) on the MADRS score between groups. Primary analysis of the data was conducted with blinding to group allocations. Linear mixed models (LMM) were used to determine differences in MADRS symptom severity scores over the study period by group allocation. Unadjusted models included fixed effects of

Time, Group, and Time x Group interactions, as well as random effects of subject (Participant*Site) and

Time. A random intercept was also included if the resulting Wald test was significant. Potential confounders and other covariates (such as recruitment site, age and gender) were entered into adjusted models including covariate x Time, covariate x Group and covariate x Group x Time interaction terms. An autoregressive covariance structure was used in each model as it best represented the visual structure of our data, produced the smallest Bayesian Information Criterion (BIC) and was supported by residual covariance significance tests. All tests of treatment effects were conducted using a two- sided alpha level of 0.05.

LMMs were again used, in the active group only, to investigate genotypic influences on treatment response. Models included Polymorphism, Time, and Polymorphism x Time interaction terms, with polymorphisms coded dominant, additive or recessive based on inspection of BIC. Finally, LMMs including Group, Polymorphism and Group x Polymorphism interaction terms were used to assess for differences in baseline severity of psychiatric scales by group and genotype. The serotonin-transporter- linked polymorphic region known as the 5HTTLPR polymorphism was analysed including its AG internal ACCEPTED SNP (rs25531), as performed by Nguyen MANUSCRIPT et al., (2015). The LG allele of the rs25531 SNP behaves similarly to the short allele (Wendland et al., 2006). As such, Individuals were coded as S/S if they were homozygous for the short allele, or were S/LG. Individuals with the long allele subtype of

LG/LG were also coded as S/S. LG/LA and S/LA were coded as S/L while LA homozygotes were coded as

L/L. All polymorphisms were tested for deviation from Hardy Weinberg equilibrium (using an online tool available from: http://www.oege.org/software/hwe-mr-calc.shtml (Rodriguez et al., 2009)) with p >

11

ACCEPTED MANUSCRIPT 0.01 as the cut off for inclusion. Correction for multiple comparisons was performed in genetic models using a Benjamini-Hochberg correction for all independent tests (Benjamini and Hochberg, 1995).

Modelling was performed using the Statistical Package for Social Sciences software (SPSS, version 23.0,

IBM, Chicago).

………………………. Figure 2 About Here ……………………

3. Results

3.1 Sample and participant characteristics

After screening and baseline exclusions, a total sample of 158 was available for analysis in the NC group versus placebo. The CONSORT flowchart is summarised in Figure 2. Treatment groups were largely matched on key sociodemographic, health, clinical and treatment factors (Table 2). However, mean age was higher in the placebo group (p = 0.022). Furthermore, baseline illness severity, per the

CGI-S, was higher in the placebo group than the NC group (p = 0.039). Similarly, the placebo group demonstrated greater severity on the baseline HAMA and MADRS, although neither reached statistical significance. Of the 158 who were randomised into the study, 72% of participants completed the entire 8 weeks, with 56 (69%) completers in the treatment group, and 57 (74%) in the placebo group. There were no significant differences in dropout rates between the two groups. Over 78% (n =

124) of the sample were at least 70% adherent based on returned capsule data.

…………………… Table 2 and 3 About Here …………………

3.2 Primary and secondary outcomes

A greater relative reduction from baseline to week 8 on the primary outcome (MADRS) of 11.7 points

(SD = 7.82) was observed for the placebo group compared to 9.95 points (SD = 8.57) in the NC group (see Table 2). A linear mixed methods model however revealed a non-significant Group X Time interaction (F(1,126) = 0.944, p = 0.33); see Figure 3. Overall improvement was noted, irrespective of group,ACCEPTED with a significant effect for Time (F(1,126) MANUSCRIPT = 151, p < 0.001) observed. Response (MADRS decrease of ≥50%) rates were 40% in NC group and 51% in placebo group; remission (final MADRS score <10) rates were 34% for NC and 43% for placebo; neither of which differed significantly between treatment groups, χ²(1,138) = 1.83, p = 0.23 and χ²(1,139) = 1.29, p = 0.30, respectively.

……………………. Figure 3 About Here …………………….

12

ACCEPTED MANUSCRIPT Recruitment site, age, gender, presence of comorbid psychiatric illness, concurrent antidepressant treatment and baseline severity of depression were individually entered as covariates into the model, none of which altered this relationship or showed any evidence of a Group X Time interaction (all results NS). There was a significant interaction between the presence of a comorbid anxiety disorder and Time, indicating a steeper response gradient for those without comorbid anxiety (F(1,332) =

7.52, p = 0.006). Furthermore, a significant Group x Antidepressant treatment interaction indicated greater depression severity at baseline in those taking antidepressants. There was, however, no interaction between Antidepressant treatment and Time, or Antidepressant treatment by Time and

Group, indicating that antidepressant use did not influence treatment response in either condition.

Splitting the sample into those receiving antidepressants (n = 110; F(1,225) = 1.40, p = 0.24), and those medication-free (n = 48; F(1,100) = 0.00, p = 0.99) demonstrated similar non-significant Group

X Time interactions.

On the BDI-II a more pronounced effect in favour of placebo occurred, however the Group x Time effect similarly did not reach significance, F(1, 193) = 2.31, p = 0.13. Similarly, no significant effect was found on other secondary outcomes: HAM-A: F(1, 157) = 0.59, p = 0.445; LSEQ: F(1, 349) =

1.18, p = 0.28; CGI-S: F(1,335) = 2.52, p = 0.13; or CGI-I: F(1,432) = 1.47, p = 0.28). On the SF-

12, only item 11 „Feeling downhearted or blue‟ showed a significant improvement with time (F[1,517]

= 10.8, p = 0.001), however no Group X Time interactions were present for any item. No appreciable changes to response to the NC or placebo were observed after removing participants with mild depression (MADRS 14-18). Finally, including only study completers in the analysis did not markedly alter the results, although a significant treatment effect in favour of placebo was evident on the BDI,

F(1,142) = 4.49, p = 0.036.

3.3 Genomic moderators of response A total 21ACCEPTED polymorphisms from 15 key genes was MANUSCRIPTavailable for analysis in 112 (71%) participants (see Table 1). The G allele of rs953413 (ELOVL2 gene) was associated with a greater depression severity at baseline, measured on both the MADRS and BDI-II, F(1, 133) = 4.98, p = 0.028 and F(1,105) =

9.62, p = 0.002, respectively. A Group X genotype interaction was found between A779C (TPH1) and

HAMA F(2,101) = 3.39, p = 0.038, as well as between rs953413 (ELOVL2) and CGI-S, and F(2, 206)

= 5.07, p = 0.007, respectively. After applying correction for multiple comparisons, significance was

13

ACCEPTED MANUSCRIPT only upheld for the association between the rs953413 G allele and greater baseline depression on the

BDI-II.

Of the investigated SNPs (in the active NC group; n=59), none demonstrated a significant Genotype X

Time interaction, indicating no significant influence on treatment response: Serotonin transporter

(HTTLPR; p = 0.18, Stin2; p = 0.64, cystathionine b-synthase (CBS69bpDup; p = 0.81), BDNF

(Val66Met; p = 0.96), P-glycoprotein (C3435T; p = 0.47), SLC39A8 (A391T; p = 0.75), Tryptophan hydroxylase (rs1386494; p = 0.26, A779C; p = 0.77, A218C; p = 0.96), FADS (rs174537; p = 0.80, rs3834458; p = 0.88), MTHFR (A1298C; p = 0.71, C677T; p = 0.73), TCN2 (Pro259Arg; p = 0.87),

MTR (rs1805087; p = 0.94), CBS (C699T; p = 0.54), BHMT (716G4A; p = 0.93), COMT (Val158Met; p

= 0.36), SLC39A3 (rs4806874; p = 0.98), ELOVL2 (rs953413; p = 1.0).

3.4 Brain-derived neurotrophic factor (BDNF)

Analyses revealed decreases in BDNF levels from baseline to week 8 of 2.29 ng/ml for the NC group and 0.601ng/ml for the placebo group, which were not significantly different (t(67) = -0.860, p =

0.32). In non-medicated participants, BDNF levels increased in the NC group (0.281ng/ml ± 4.97) while decreasing in the placebo group (-2.28ng/ml ± 3.33), although the difference was similarly not significant, t(17) = 1.35, p = 0.20. A reverse finding was found in those taking antidepressants (-

3.08ng/ml ± 9.96 in the NC group vs 0.169ng/ml ± 8.24 in placebo), which also did not reach significance, t(48) = -1.25, p = 0.22). Although BDNF levels were slightly higher in Val homozygotes

(23.9 ng/ml ± 8.01) than in the combined Met/Met or Val/Met genotypes (21.5 ng/ml ± 7.26), the

Val66Met SNP did not significantly predict baseline BDNF levels (F(1,98) = 2.28, p = 0.13). Nor did the Val66Met SNP predict change in BDNF in the active combination group (F(1,28) = 0.00, p = 0.98).

3.5 Safety and adverse reactions Overall, ACCEPTEDthe NC and placebo were very well tolerated, MANUSCRIPT with similar numbers of adverse events between the groups, χ²(1,158) = 0.042, p = 0.52. There was a greater frequency of early termination secondary to adverse events in the NC group (n = 9) compared to the placebo group (n = 3) although the difference did not quite reach significance, χ²(1,158) = 2.93, p = 0.09. Four serious adverse events occurred in the study, with three in the placebo group and one in the NC group. These adverse events were evaluated as unrelated to the study medication. Serotonergic symptoms were noted in two

14

ACCEPTED MANUSCRIPT participants who were receiving the NC (one participant was taking antidepressant medication) but these did not meet the diagnostic criteria of serotonin syndrome. Upon completion of the study 43% of the NC group and 46% of the placebo group believed they were receiving the active therapy, indicating no clear subjective distinction in the adverse events experienced between groups, X(2,108) = 0.43, p =

0.81.

The following specific adverse events (presented as a summation of all instances) were most frequently reported across the study: diarrhoea (NC: 23, placebo: 30), sweating (NC: 16, placebo:

19), headache (NC: 11, placebo: 14), fatigue (NC: 12, placebo: 11), concentration issues, confusion or forgetfulness (NC: 14, placebo: 9), nausea (NC: 15, placebo: 5; p = 0.023), twitching/shaking (NC: 8, placebo: 12), irritability (NC: 8, placebo: 11), muscle or stomach cramps (NC: 6, placebo: 11) and hot flushes (NC: 7, placebo: 5). Aside from nausea being significantly more frequent in the NC

(potentially due to the zinc or omega-3), there were no other significant between-group differences.

4. Discussion

Contrary to what had been anticipated, as was advocated in Nutritional Medicine as Mainstream in

Psychiatry cf. J Sarris et al., (2015b) the nutraceutical combination was found to be inferior to placebo in reducing depressive symptoms in MDD. Thus this study showed no support for the use of a „shot- gun‟ approach, and the NC intervention may potentially be completely ineffective, even iatrogenic.

However the combination may not the best combination or dose/ratio of the nutrients studied.

It should be recognised however that the NC did have a modest effect on reducing depressive symptoms, which the high placebo response may have somewhat obscured. High placebo response rates bedevil psychiatric research in general, and trials with high placebo response rates are much less likely to show expected separation (Chi et al., 2016; Sonawalla and Rosenbaum, 2002).

In terms of the genomic outcomes, none of the investigated polymorphisms appeared to meaningfully influenceACCEPTED response to treatment in the NC group, MANUSCRIPTproviding no support for a pharmacogenomic related response. Of the investigated polymorphisms, one was associated with baseline severity of depression; rs953413 (greater baseline severity measured on MADRS and BDI-II in G allele carriers). It is recognised that for genomic outcomes, particularly regarding SNPs with low minor allelic frequencies, the study was only sufficiently powered to detect a relatively large separation by genotype, which is uncommon in depression studies. Regarding the BDNF results, particularly considered in light of the 15

ACCEPTED MANUSCRIPT large standard deviations at each time point, „natural‟ or non-treatment based fluctuations appeared to play a greater role than treatment. Interestingly, despite an overall improvement across time noted in the study, this was not reflected in an increase in BDNF levels.

The present study has several strengths, including its double-blind, randomised, placebo-controlled design, robust sample size, rigorous inclusion/exclusion criteria, use of a slight amount of fish oil in the placebo group to maintain blinding, and homogeneity of baseline depression severity. Limitations are however recognised. Firstly, as identified above, certain factors, such as the lack of a placebo run- in, may have increased the placebo-response. Expectancy may have been one such factor. It was noted that many participants in the study felt unsatisfied with conventional antidepressant treatment and believed that nutrient imbalances may have been the „missing-link‟ underpinning their depression.

As such, many participants felt open-minded towards nutraceutical therapy, suggesting an expectancy which may have contributed to observed placebo response rates. One reason for this inflated expectancy, is our use of Facebook and Google advertising which account for 42% of our enquiries.

Facebook and Google targets via algorithms users with specific interest in certain areas (in this case supplements and mental health. „Grade inflation‟ at baseline is not uncommon in studies with severity based inclusion criteria and may also amplify placebo response rates, while response rates can be unpredictable in participants with mild (MADRS < 18) or drug treatment naïve depression (Fava et al.,

2003; Robinson and Rickels, 2000). Although the effect size of the placebo response in this study

(Cohen‟s d=1.75) did not greatly exceed those reported in a recent meta-analysis of antidepressant trials (d=1.69) (Rief et al., 2009), the placebo effect may be expected to be lower in nutraceutical intervention studies (Freeman et al., 2010). We adopted the SAFER 2.0 screening tool to mitigate against this risk, and the researchers administering the MADRS were consistently trained during the participant sessions to avoid any unintended therapeutic effect. This appeared to have not prevented a strong placebo-response rate, and there is the consideration that a placebo run-in phase may have been a ACCEPTEDsagacious design option. Secondly, the MANUSCRIPT inclusion of participants undergoing a range of treatment as usual medical care interventions including use of different antidepressant classes, adds a confounding element to the study. Lastly, the effect of several nutraceuticals combined together is unknown, both in terms of pharmacokinetics and pharmacodynamics, and we cannot rule out that such a combination may create a less effective antidepressant effect. Regardless, the novelty of

16

ACCEPTED MANUSCRIPT exploring such a combination of evidence-based nutrients has rarely been subject to a rigorous clinical trial, in addition to exploring biomarkers as moderators of response.

In conclusion, the adoption of a nutraceutical „shotgun‟ approach to treating MDD was not supported, and was in fact is less effective than adding placebo to an antidepressant. It is still possible that select nutrients may be of benefit in a targeted manner, and the benefits of enhancing dietary quality as an important element in improving or sustaining mental and physical health. Future research should focus on precision-based approaches tailoring supplementation based on nutrient deficiencies, neurochemical abnormalities, or pharmacogenetic differences. This may better reflect individual diversity in clinical practice, which is moving towards more precision-based medicine (Fernandes et al., 2017).

ACCEPTED MANUSCRIPT

17

ACCEPTED MANUSCRIPT Role of funding source

This grant is funded by an Australian National Health and Medical Research Council project grant (APP1048222), and is co-sponsored by FIT-BioCeuticals (who provided the trial product and placebo). Jerome Sarris is supported by an NHMRC Fellowship (APP1125000). Michael Berk is supported by a NHMRC Senior Principal Research Fellowship (1059660 and 1156072). BioCeuticals was not involved in any aspect of study design, analysis, manuscript preparation or the decision to submit this manuscript for publication.

Conflicts of Interest

No specific significant conflicts identified. JS has received honoraria, research support, royalties, or consultancy or travel grant funding from Integria Healthcare & MediHerb, Pfizer, Scius Health, Key Pharmaceuticals, Taki Mai, FIT-BioCeuticals (untied consultancy funds paid to the university) & Blackmores, SPRIM, Soho-Flordis, Healthworld, HealthEd, HealthMasters, Elsevier, Chaminade University, International Society for Affective Disorders, Complementary Medicines Australia, Terry White Chemists, ANS, Society for Medicinal Plant and Natural Product Research, Sanofi-Aventis, Omega-3 Centre, the National Health and Medical Research Council, CR Roper Fellowship. DM has received research support from the Bowman Family Foundation, FisherWallace, Ganeden, Nordic Naturals, Methylation Sciences, Inc. (MSI), and PharmoRx Therapeutics. He has received honoraria for consulting, speaking, and writing from the Massachusetts General Hospital Psychiatry Academy. He has received royalties from Lippincott Williams & Wilkins for published book “Natural Medications for Psychiatric Disorders: Considering the Alternatives.” CS has received research grants from Government: Australian Research Council; NHMRC; NDLERF; NIDA; VDLERF; ARC; Vicpolice; Vicroads; Nestle; SFI; Barry Callebaut; Blackmores; Flordis; Zeller; Clover Corporation; Horphag; GSK; Bayer; Pharmalink; Siemens; Securatek; Efamol; Cognis; Integria; Medvet, Cooper Nutrition, and paid strategic consulting advice for SFI, Blackmores, Bayer; ARU, Carlton FC, GSK India

Acknowledgments

We would like to thank the study participants who contributed to this project.

ACCEPTED MANUSCRIPT

References

Antonijevic, I.A., 2006. Depressive disorders -- is it time to endorse different pathophysiologies? Psychoneuroendocrinology 31, 1–15. https://doi.org/S0306-4530(05)00085-5 [pii]10.1016/j.psyneuen.2005.04.004 [doi] Benjamini, Y., Hochberg, Y., 1995. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300. https://doi.org/10.2307/2346101

18

ACCEPTED MANUSCRIPT Bottiglieri, T., Hyland, K., 1994. S-adenosylmethionine levels in psychiatric and neurological disorders: a review. Acta Neurol Scand Suppl 154, 19–26. Byerley, W.F., Judd, L.L., Reimherr, F.W., Grosser, B.I., 1987. 5-Hydroxytryptophan: a review of its antidepressant efficacy and adverse effects. J Clin Psychopharmacol 7, 127–137. Chalon, S., 2006. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins Leukot. Essent. Fat. Acids 75, 259–269. https://doi.org/10.1016/j.plefa.2006.07.005 Chi, G.Y.H., Li, Y., Liu, Y., Lewin, D., Lim, P., 2016. On clinical trials with a high placebo response rate. Contemp. Clin. Trials Commun. 2, 34–53. https://doi.org/10.1016/j.conctc.2015.10.002 Desseilles, M., Witte, J., Chang, T.E., Iovieno, N., Dording, C., Ashih, H., Nyer, M., Freeman, M.P., Fava, M., Mischoulon, D., 2013. Massachusetts General Hospital SAFER criteria for clinical trials and research. Harv Rev Psychiatry 21, 269–274. https://doi.org/10.1097/HRP.0b013e3182a75cc7 Donohue, J., Pincus, H., 2007. Reducing the Societal Burden of Depression: A Review of Economic Costs, Quality of Care and Effects of Treatment. Pharmacoeconomics 25, 7–24. Fava, M., Evins, A.E., Dorer, D.J., Schoenfeld, D.A., 2003. The problem of the placebo response in clinical trials for psychiatric disorders: culprits, possible remedies, and a novel study design approach. Psychother Psychosom 72, 115–127. https://doi.org/69738 Fava, M., Mischoulon, D., 2009. Folate in depression: efficacy, safety, differences in formulations, and clinical issues. J Clin Psychiatry 70 Suppl 5, 12–17. https://doi.org/10.4088/JCP.8157su1c.03 Fernandes, B.S., Williams, L.M., Steiner, J., Leboyer, M., Carvalho, A.F., Berk, M., 2017. The new field of „precision psychiatry.‟ BMC Med. 15, 80. https://doi.org/10.1186/s12916-017-0849-x Freeman, M.P., Mischoulon, D., Tedeschini, E., Goodness, T., Cohen, L.S., Fava, M., Papakostas, G.I., 2010. Complementary and alternative medicine for major depressive disorder: A meta-analysis of patient characteristics, placebo-response rates, and treatment outcomes relative to standard antidepressants. J. Clin. Psychiatry 71, 682–688. https://doi.org/10.4088/JCP.10r05976blu Hayes, A.F., Krippendorff, K., 2007. Answering the Call for a Standard Reliability Measure for Coding Data. Commun. Methods Meas. https://doi.org/10.1080/19312450709336664 Hindmarch, I., 2001. Expanding the horizons of depression: beyond the monoamine hypothesis. Hum Psychopharmacol 16, 203–218. https://doi.org/10.1002/hup.288 [doi] Jacobs, D.R.J., Tapsell, L.C., 2007. Food, not nutrients, is the fundamental unit in nutrition. Nutr Rev 65, 439– 450. Lai, J., Moxey, A., Nowak, G., Vashum, K., Bailey, K., McEvoy, M., 2012. The efficacy of zinc supplementation in depression: systematic review of randomised controlled trials. J Affect Disord 136, e31-9. https://doi.org/S0165-0327(11)00353-3 [pii]10.1016/j.jad.2011.06.022 Mischoulon, D., Freeman, M.P., 2013. Omega-3 Fatty Acids in Psychiatry. Psychiatr. Clin. North Am. https://doi.org/10.1016/j.psc.2012.12.002 Nguyen, T.B., Gunn, J.M., Potiriadis, M., Everall, I.P., Bousman, C. a., 2015. Serotonin transporter polymorphism ( 5HTTLPR ), severe childhood abuse and depressive symptom trajectories in adulthood. Br. J. Psychiatry OpenACCEPTED 1, 104–109. https://doi.org/10.1192/bjpo.bp.115.000380 MANUSCRIPT Papakostas, G.I., Alpert, J.E., Fava, M., 2003. S-adenosyl-methionine in depression: a comprehensive review of the literature. Curr Psychiatry Rep 5, 460–466. Papakostas, G.I., Mischoulon, D., Shyu, I., Alpert, J.E., Fava, M., 2010. 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 167, 942–948. https://doi.org/appi.ajp.2009.09081198 [pii]10.1176/appi.ajp.2009.09081198 Plotsky, P.M., Owens, M.J., Nemeroff, C.B., 1998. Psychoneuroendocrinology of depression. Hypothalamic-

19

ACCEPTED MANUSCRIPT pituitary-adrenal axis. Psychiatr Clin North Am 21, 293–307. Raison, C.L., Capuron, L., Miller, A.H., 2006. Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends Immunol. https://doi.org/10.1016/j.it.2005.11.006 Ressler, K.J., Nemeroff, C.B., 2000. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depress Anxiety 12 Suppl 1, 2–19. https://doi.org/10.1002/1520- 6394(2000)12:1+<2::AID-DA2>3.0.CO;2-4 [doi] Rief, W., Nestoriuc, Y., Weiss, S., Welzel, E., Barsky, A.J., Hofmann, S.G., 2009. Meta-analysis of the placebo response in antidepressant trials. J. Affect. Disord. https://doi.org/10.1016/j.jad.2009.01.029 Robinson, D.S., Rickels, K., 2000. Concerns about clinical drug trials. J. Clin. Psychopharmacol. https://doi.org/10.1097/00004714-200012000-00001 Rodriguez, S., Gaunt, T.R., Day, I.N.M., 2009. Hardy-Weinberg equilibrium testing of biological ascertainment for Mendelian randomization studies. Am. J. Epidemiol. 169, 505–514. https://doi.org/10.1093/aje/kwn359 Rush, A.J., 2007. Limitations in efficacy of antidepressant monotherapy. J Clin Psychiatry 68 Suppl 1, 8–10. Sarris, J., Logan, A.C., Akbaraly, T.N., Amminger, G.P., Balanza-Martinez, V., Freeman, M.P., Hibbeln, J., Matsuoka, Y., Mischoulon, D., Mizoue, T., Nanri, A., Nishi, D., Ramsey, D., Rucklidge, J.J., Sanchez- Villegas, A., Scholey, A., Su, K.P., Jacka, F.N., International Society for Nutritional Psychiatry, R., 2015a. Nutritional medicine as mainstream in psychiatry. Lancet Psychiatry 2, 271–274. https://doi.org/10.1016/S2215-0366(14)00051-0 Sarris, J., Logan, A.C., Akbaraly, T.N., Amminger, G.P., Balanza-Martinez, V., Freeman, M.P., Hibbeln, J., Matsuoka, Y., Mischoulon, D., Mizoue, T., Nanri, A., Nishi, D., Ramsey, D., Rucklidge, J.J., Sanchez- Villegas, A., Scholey, A., Su, K.P., Jacka, F.N., International Society for Nutritional Psychiatry, R., 2015b. Nutritional medicine as mainstream in psychiatry. Lancet Psychiatry 2, 271–274. https://doi.org/10.1016/S2215-0366(14)00051-0 Sarris, J., Logan, A.C., Akbaraly, T.N., Paul Amminger, G., Balanza-Martinez, V., Freeman, M.P., Hibbeln, J., Matsuoka, Y., Mischoulon, D., Mizoue, T., Nanri, A., Nishi, D., Parletta, N., Ramsey, D., Rucklidge, J.J., Sanchez-Villegas, A., Scholey, A., Su, K.P., Jacka, F.N., 2015c. International Society for Nutritional Psychiatry Research consensus position statement: nutritional medicine in modern psychiatry. World Psychiatry 14, 370–371. https://doi.org/10.1002/wps.20223 Sarris, J., Murphy, J., Mischoulon, D., Fava, M., Berk, M., Ng, C., 2016. Adjunctive Nutrient Nutraceuticals for Depression: A Systematic Review and Meta-analyses. Am. J. Psychiatry 173, 575–587. Sarris, J., Papakostas, G.I., Vitolo, O., Fava, M., Mischoulon, D., 2014. S-Adenosyl Methionine (SAMe) versus Escitalopram and Placebo in Major Depression: Efficacy and Effects of Histamine and Carnitine as Moderators of Response. J. Affect. Disord. 164, 76–81. Sarris, J., Stough, C., Bousman, C., Murphy, J., Savage, K., Smith, D.J., Menon, R., Chamoli, S., Oliver, G., Berk, M., Byrne, G.J., Ng, C., Mischoulon, D., 2015. An adjunctive antidepressant nutraceutical combination in treating major depression: Study protocol, and clinical considerations. Adv. Integr. Med. 2, 49–55. https://doi.org/10.1016/j.aimed.2015.02.001 Sonawalla,ACCEPTED S.B., Rosenbaum, J.F., 2002. Placebo response MANUSCRIPT in depression. Dialogues Clin. Neurosci. https://doi.org/10.1192/bjp.179.1.79 Szewczyk, B., Kubera, M., Nowak, G., 2011. The role of zinc in neurodegenerative inflammatory pathways in depression. Prog Neuropsychopharmacol Biol Psychiatry 35, 693–701. https://doi.org/S0278- 5846(10)00057-6 [pii]10.1016/j.pnpbp.2010.02.010 Tassoni, D., Kaur, G., Weisinger, R.S., Sinclair, A.J., 2008. The role of eicosanoids in the brain. Asia Pac. J. Clin. Nutr. https://doi.org/10.1002/9783527613625.ch8 Wendland, J.R., Martin, B.J., Kruse, M.R., Lesch, K.P., Murphy, D.L., 2006. Simultaneous genotyping of four

20

ACCEPTED MANUSCRIPT functional loci of human SLC6A4, with a reappraisal of 5-HTTLPR and rs25531. Mol. Psychiatry. https://doi.org/10.1038/sj.mp.4001789 Williams, A.L., Katz, D., Ali, A., Girard, C., Goodman, J., Bell, I., 2006. Do essential fatty acids have a role in the treatment of depression? J Affect Disord 93, 117–123. https://doi.org/S0165-0327(06)00134-0 [pii]10.1016/j.jad.2006.02.023 [doi]

ACCEPTED MANUSCRIPT

21

ACCEPTED MANUSCRIPT

Table 1. List of SNPs and polymorphisms dbSNP ID Alternative ID Gene Symbol Gene Name rs4680 Val158Met COMT catechol-o-methyltransferase rs1801133 C677T MTHFR methylenetetrahydrofolate reductase rs1801131 A1298C MTHFR methylenetetrahydrofolate reductase rs1799913 A779C TPH1 tryptophan hydroxylase 1 rs1800532 A218C TPH1 tryptophan hydroxylase 1 rs1386494 -- TPH2 tryptophan hydroxylase 2 rs25531 -- SLC6A4 serotonin transporter rs13107325 A391T SLC39A8 zinc transporter rs4806874 -- SLC39A3 zinc transporter rs174537 -- FADS1 fatty acid desaturase 1 rs3834458 -- FADS2 fatty acid desaturase 2 rs953413 ELOVL2 Elongation of very long chain fatty acids protein 2 rs234706 C699T CBS cystathionine b-synthase rs3733890 716G4A BHMT betaine homocysteine methyltransferase rs1805087 Asp919Gly MTR methionine synthase rs1801198 Pro259Arg TCN2 transcobalamin-II rs1045642 C3435T ABCB1 P-glycoprotein rs6265 Val66Met BDNF brain-derived neurotrophic factor

Other polymorphisms

5HTTLPR SLC6A4 serotonin transporter

STin2 VNTR SLC6A4 serotonin transporter

845ins68 68bp dup CBS cystathionine b-synthase

ACCEPTED MANUSCRIPT

22

ACCEPTED MANUSCRIPT Table 2. Demographic and clinical characteristics

Characteristics Treatment (n=81) Placebo (n=77)

Age, mean (SD) 40.2 (12.3) 44.7 (12.6)

Female gender, % (n) 75.3 (61) 79.2 (61)

Years of Education, mean (SD) 14.1 (2.30) 14.2 (2.23)

Married/defacto, % (n) 60.8 (48) 53.3 (40)

Employment status, % (n)

Full/part time 73.1 (57) 67.1 (49)

Student 15.4 (12) 11.7 (9)

Unemployed 7.70 (6) 6.70 (6)

Other 7.4 (6) 16.9 (13)

Health status

Cardiovascular disease, % (n) 4.90 (4) 2.60 (2)

Endocrine disorders, % (n) 12.3 (10) 10.4 (8)

Immune disease, % (n) 16.0 (13) 11.7 (9)

Current smoker, % (n) 11.1 (8) 12.5 (9)

Alcohol consumption, mean (SD)/week 3.47 (4.53) 3.49 (6.17)

General health*, mean (SD) 2.09 (.990) 2.01 (.914)

Comorbid psychiatric illness (current), % (n)

Generalised anxiety disorder 35.8 (29) 33.3 (27)

Panic disorder 11.0 (9) 6.58 (5)

Social phobia 11.0 (9) 5.19 (4)

Clinical features

Length of episode (weeks), mean (SD) 47.0 (43.9) 58.4 (78.2)

Number of lifetime episodes, mean (SD) 6.06 (5.03) 5.83 (5.87)

Age of onset, mean (SD) 23.6 (11.5) 25.1 (13.6)

Taking antidepressant medication, % (n) 71.6 (58) 67.5 (52)

Illness severity, mean (SD)

Baseline MADRS 24.0 (4.69) 24.6 (5.23)

Baseline HAMA 14.0 (4.55) 15.6 (5.52) BaselineACCEPTED CGI-S MANUSCRIPT3.81 (.622) 4.00 (.513) Other treatment, % (n)

History of psychotherapy

Current treatment 40.3 (29) 37.5 (27)

Previous treatment 41.7 (30) 48.6 (35)

* Self-reported on the SF-12

23

ACCEPTED MANUSCRIPT Table 3. Changes in primary and secondary endpoints across the trial (Baseline-Week 8)*

Measure Treatment (n=56) Placebo (n=57) Group X Time interaction

MADRS -9.95 (8.57) -11.7 (7.82) F(1,125) = 0.944, p = 0.33

BDI-II -11.7 (10.0) -16.1 (9.76) F(1,133) = 2.31, p = 0.13

HAMA -5.15 (5.49) -5.94 (6.79) F(1, 157) = 0.586, p = 0.45

CGI-S -0.962 (1.14) -1.13 (1.05) F(1,335) = 2.52, p = 0.13

CGI-I -0.333 (1.62) -0.592 (1.54) F(1,432) = 1.47, p = 0.28

LSEQ 7.91 (19.2) 12.1 (17.3) F(1, 349) = 1.18, p= 0.28

*Baseline to endpoint data in study completers only. MADRS= Montgomery-Asberg Depression Rating Scale; BDI-II= Beck Depression Rating Scale-II; HAMA= Hamilton Anxiety Rating Scale; CGI-S= Clinical Global Impressions-Severity; CGI-I= Clinical Global Impressions-Improvement; LSEQ= Leeds Sleep Evaluation Questionnaire

ACCEPTED MANUSCRIPT

24

ACCEPTED MANUSCRIPT

Figure 1. Neuropathology of depression and potential for nutraceutical intervention*

* Modified from Sarris et al. 20154. IL-1/6= interleukin 1/6; TNF-a= tumour necrosis factor alpha; SE= serotonin; NE= noradrenaline; DA = dopamine; BDNF= brain derived neurotropic factor; 5-HTP= 5- hydroxytryptophan; SAMe= S-adenosyl methionine; EPA= eicosapentaenoic acid; HPA= hypothalamic– pituitary–adrenal

ACCEPTED MANUSCRIPT

25

ACCEPTED MANUSCRIPT Figure 2. CONSORT Flow Diagram displaying participant flow from enrolment to trial completion.

Expressed interest in study (n=2637)

1.1 Enrolment

Assessed for eligibility (baseline visit; n=227)

Excluded (n=69)  Not meeting inclusion criteria (n=69)  Declined to participate (n=0)

Randomised (n=158)

1.2 Allocation Allocated to active nutrient combination (n=81) Allocated to placebo (n=77)  Commenced allocated intervention (n=78)  Commenced allocated intervention (n=74)  Did not commence allocated intervention  Did not commence allocated intervention

(n=3) (n=3)  Withdrawal of consent (n=2)  Withdrawal of consent (n=2)  Medical issues (n=1)  Change to medication (n=1)

1.3 Follow-Up

Discontinued intervention (n=22) Discontinued intervention (n=17)  Adverse events (n=9)  Adverse events (n=3)  Investigator decision (n=2)  Investigator decision (n=5)  Participant withdrew consent (n=7)  Participant withdrew consent (n=7)  Protocol deviation (n=1)  Non-compliance (n=1)

 Non-compliance (n=1)  Lost to follow up (n=1)  Lost to follow up (n=2)

ACCEPTED MANUSCRIPT 1.4 Analysis Trial completers: 56 Trial completers: 57

Analysed (n=81) Analysed (n=77)  Excluded from analysis (n=0)  Excluded from analysis (n=0)

26

ACCEPTED MANUSCRIPT Figure 3. Results on the primary outcome.

MADRS 30

25

SD

± 20

15 NC

10 Placebo

Mean MADRS score score MADRS Mean 5

0 Baseline W2 W4 W6 W8 Week

MADRS= Montgomery-Asberg Depression Rating Scale; NC= Nutraceutical Combination;

Group X Time interaction p =0.33; Error bars represent standard deviation of the mean.

ACCEPTED MANUSCRIPT

27