Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Nutritional Neuroscience

An International Journal on Nutrition, Diet and Nervous System

ISSN: 1028-415X (Print) 1476-8305 (Online) Journal homepage: http://www.tandfonline.com/loi/ynns20

Omega-3 fatty acids' supplementation in


Alzheimer's disease: A systematic review

Scheine Canhada, Kamila Castro, Ingrid Schweigert Perry & Vivian Cristine
Luft

To cite this article: Scheine Canhada, Kamila Castro, Ingrid Schweigert Perry & Vivian Cristine
Luft (2017): Omega-3 fatty acids' supplementation in Alzheimer's disease: A systematic review,
Nutritional Neuroscience, DOI: 10.1080/1028415X.2017.1321813

To link to this article: http://dx.doi.org/10.1080/1028415X.2017.1321813

Published online: 03 May 2017.

Submit your article to this journal

Article views: 115

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ynns20

Download by: [The UC San Diego Library] Date: 30 May 2017, At: 17:53
Omega-3 fatty acids’ supplementation in
Alzheimer’s disease: A systematic review
Scheine Canhada 1 , Kamila Castro2,3, Ingrid Schweigert Perry 3,4, Vivian Cristine
Luft1,3
1
Postgraduate Program in Epidemiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil,
2
Postgraduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto
Alegre, Brazil, 3Food and Nutrition Research Centre (CESAN), Hospital de Clínicas de Porto Alegre,
Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil, 4Postgraduate Program in Collective Health,
Academic Unit of Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil

Introduction: Alzheimer’s disease (AD) is a neurodegeneration disorder characterized by progressive


impairments of memory, language, reasoning, and other cognitive functions. Evidence suggests that
omega-3 fatty acids may act as a possible protection factor in AD.
Objective: To evaluate the results available in the literature involving omega-3 fatty acids supplementation
and its effect on cognitive function in AD patients.
Methods: A systematic review of MEDLINE (from PubMed), Excerpta Medica Database, and Cochrane
Library databases was conducted according to Preferred Reporting Items for Systematic Reviews and
Meta-Analyses guidelines. Inclusion criteria consisted in original intervention studies, controlled by
placebo, that assessed the impact of supplementation or dietary intake of omega-3 fatty acids on
cognitive function, in humans with AD, without limitation for prime date of publication.
Results: Initial search resulted in 361 articles. Seven studies fully met the inclusion criteria. Most studies did
not find statistically significant results for the omega-3 fatty acids supplementation compared to placebo, and
those who show some benefit do it only in a few cognitive assessment scales. However, the effects of omega-
3 fatty acids appear to be most effectively demonstrated in patients with very mild AD.
Conclusion: The effects of omega-3 fatty acids supplementation in mild AD corroborate epidemiological
observational studies showing that omega-3 fatty acids may be beneficial in disease onset, when there is
slight impairment of brain function. Although some studies have shown changes in scales of cognitive
function in more severe cases, they are not enough to support omega-3 fatty acids supplementation in the
treatment of AD.
Keywords: Omega-3 fatty acids, Eicosapentaenoic acid, Docosahexaenoic acid, Alzheimer’s disease, Cognition, Neuroinflammation

Introduction components may interfere directly in disease’s patho-


Alzheimer’s disease (AD) is a neurodegeneration dis- logic hallmarks – like Aβ’s excessive production or
order, with slow and continuous progression, character- deposition and neurodegeneration, – although mech-
ized by progressive impairments of memory, language, anisms still remain uncertain.9
reasoning, and other cognitive functions.1 AD results Evidence suggests that omega-3 fatty acids’ modu-
from nervous cell damage which is associated with the late numerous molecular and cellular processes,
accumulation of insoluble forms of amyloid-β (Aβ) in which include brain and visual development, inflam-
plaques and aggregation of the microtubule protein matory reactions, thrombosis, and carcinogenesis.10
tau in neurofibrillary tangles.1 AD prevalence is esti- Omega-3 fatty acids are considered essential for the
mated to quadruplicate until 2050.2 human body because we are unable to produce them,
There is a growing interest in lifestyle and dietary so they must be obtained from dietary sources.11,12
components as possible protection factors for AD, Among them, alpha-linolenic acid is present in flax-
including omega-3 fatty acids.3–8 Some dietary seed and canola oils, and eicosapentaenoic acid
(EPA) and docosahexaenoic acid (DHA) are found
Correspondence to: Scheine Canhada, Postgraduate Program in in fatty fish such as salmon, trout, and tuna, and in
Epidemiology, School of Medicine, Federal University of Rio Grande do
Sul, Ramiro Barcelos, 2400, Santa Cecília, Porto Alegre, RS, Brazil.
fish-oil supplements.12 Observational studies have
Email: scheinelc@gmail.com suggested that omega-3 fatty acids and its food

© 2017 Informa UK Limited, trading as Taylor & Francis Group


DOI 10.1080/1028415X.2017.1321813 Nutritional Neuroscience 2017 1
Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

sources are associated with lower incidences of AD deviation or confidence interval. Results are not
and dementias in general.4,6–8 However, omega-3 described when not available in the original articles.
fatty acids’ role in AD treatment, when the disease
already exists, is still controversial in the literature. Results
The present study systematically reviews the effects The search strategy resulted in a total of 361 articles.
of interventions using omega-3 fatty acids’ supplemen- After eligibility analyses, a total of seven papers ful-
tation on cognitive function in individuals with AD. filled all criteria. The selection process is shown in
Figure 1. Publication year varied from 2006 to 2015.
Methods All articles were available in English.
This is a systematic review of articles found in The questionnaires that were used in these studies
MEDLINE (US National Library of Medicine and were Alzheimer’s Disease Assessment Scale –
National Institute of Health) – using PubMed –, Cognitive section (ADAS-cog), Clinicians Global
Cochrane Library, and EMBASE (Excerpta Medica Impression of Change (CIBIC-plus), Mini-Mental
Database). Furthermore, the reference lists of the State Examination (MMSE), Hamilton Depression
selected relevant papers were screened by hand for Scale (HDRS), Montgomery Asberg Depression
potentially additional papers in the subject. The Rating Scale (MADRS), Clinical Dementia Rating
search strategy combined the terms “Fatty Acids, Scale (CDR), Alzheimer’s Disease Cooperative
Omega-3”[Mesh] AND “Alzheimer Disease”[Mesh]. Study – Activities of Daily Living (ADCS-ADL),
Guidelines established from Preferred Reporting Alzheimer’s Disease Cooperative Study – Instrumental
Items for Systematic Reviews and Meta-Analyses Activities of Daily Living (ADCS-IADL), Caregivers
(PRISMA) were followed.13 Burden Scale (CGB), Disability Assessment for
Inclusion criteria consisted in original intervention Dementia Scale (DAD), Neuropsychiatric Inventory
studies, controlled by placebo, that assessed omega-3 (NPI), Neuropsychological Test Battery (NTB), and
fatty acids’ impact on cognitive function markers, in Bristol’s Activities of Daily Living Scale (BADLS).
humans with AD, published in any language until The most frequent questionnaires were ADAS-cog
March 2017, without limitation for the initial date of and MMSE, both present in four studies.14–17 The
publication. Non-original articles, such as reviews, main scales are summarized in Table 1.
editorials, and letters, were excluded, as well as Sample size varied from 19 to 402 men and women,
studies in animals or in vitro. We searched for interven- summing a total of 429 individuals randomized for
tions using omega-3 fatty acids as supplementation (in omega-3 fatty acids’ intervention and 366 for
capsules, or in any other form than in food itself ) or as placebo. Patients were 55–90 years old and presented
increased dietary intake (throughout its food sources, mild and moderate AD, according to MMSE ques-
such as fish or fish oils). tionnaire, used in all seven studies, and additionally
We first applied eligibility criteria in titles’ analysis, CDR questionnaire, used in one study. Information
followed by abstracts’ analysis and full texts’ reading. about these studies are shown in Table 2.
Search, selection, and information extraction were per- In six studies, omega-3 fatty acids’ supplementation
formed independently by two reviewers. In order to combined both DHA and EPA14–16,24–26 and one
favor reliability, data were collected independently in study supplemented only DHA.17 Placebo groups
a table including first author, year, journal, study received olive oil capsules, soy oil, corn oil, and/or
design, population characteristics, initial and final an isocaloric product. In four studies, both groups,
sample size, type and dosage of supplementation, intervention and placebo, received other nutrients
exposure period, outcome measures, and results. A additionally, such as tocopherols and hydro-
third investigator solved eventual disagreements. quinones.14,16,24–26 None of the studies supplemented
One of the researchers evaluated bias risk for each omega-3 fatty acids by increasing intake of its food
study included in this review, after data extraction, sources. The intervention period varied from 4
through the application of a tool available on months to 18 months.
Cochrane Handbook for Systematic Reviews of Concomitant pharmacological treatment for AD
Interventions and with the support of the software was not allowed in some studies.16,24 In studies that
Review Manager. This information was reported in allowed the adjuvant treatment with medi-
the discussion section, regarding validity of effects cines,14,15,17,25,26 the stable use for 3 or 4 months
found for each study. prior to the beginning of intervention was required.
Results from the original papers are presented The first randomized clinical trial controlled by
comparing scores of cognitive function derived from placebo that evaluated omega-3 fatty acids’ impact
questionnaires, between intervention and placebo in AD was published in 2006. Freund-Levi et al. 14
groups, as in P values and, when statistically significant assessed omega-3 fatty acids’ supplementation in 204
(P < 0.05), additionally as means and its standard subjects with mild-to-moderate AD (mean age: 74 ±

2 Nutritional Neuroscience 2017


Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

Figure 1 Flow diagram of the systematic review of the impact of omega-3 fatty acids’ supplementation on cognitive function in
AD patients.

9 years). The intervention group received omega-3 (P > 0.05). In a subgroup with very mild AD
fatty acids in the dosages of 1720 mg DHA and (MMSE > 27 and CDR 0.5–1), there was a significant
600 mg EPA and the placebo group received difference in MMSE between intervention and placebo
4000 mg of corn oil (containing 2400 mg of linoleic in the first 6 months (placebo group had a decline of
acid) for 6 months, followed by more 6 months of −2.6 points and intervention group had −0.5 points;
omega-3 fatty acids’ supplementation for both P = 0.01).
groups. Medication for AD treatment was allowed. As In a second paper published in 2008, Freund-Levi
a result, there was no significant statistical difference et al. 25, using the exact same sample from 2006,
at 6 months and 12 months between groups in assessed other questionnaires. No significant statistical
ADAS-cog (P > 0.05), MMSE (P > 0.05) e CDR difference was found at 6 months and 12 months

Table 1 Summary of main scales used for cognitive assessment in AD.

Main scales Description

ADAS-cog18 Is a sensitive and reliable method for the assessment of cognitive function in dementias. It consists of a
psychometric scale of 11 items, and scores range from 0 (no impairment) to 70 (very severe impairment).
It is considered to have a good sensitivity to evaluate changes over time
ADCS-ADL; ADCS- It measures the functional ability to perform activities of daily life. ADL assess basic living skills such as
IADL19 bathing and eating, whereas IADL measure more complex tasks such as using the telephone or
preparing meals. A higher ADL or IADL score indicates a worsening functionality. Scores range from 0 to
27 for ADL and 0 to 14 for IADL
CDR20 It is a global measure that assesses memory, orientation, judgment, and other features. Is based on
caregiver interview. Classifies dementia into questionable, mild, moderate, and severe
CIBIC-plus21 It is a global measure capable of detecting changes in cognition, functionality, and behavior, thus
assessing dementia’s severity and progression. Requires separate interviews with patients and
caregivers. It is a 7-point Likert-type scale, in which 1 represents markedly improved; 4, no change; and
7, markedly worse
MMSE22 Evaluates cognitive function in the areas of orientation, memory, attention, calculation, language, and visual
construction. It is widely translated and used in clinical practice. Patients score between 0 and 30 points,
and cutoffs of 23/24 have typically been used to show significant cognitive impairment
NTB23 This scale assesses changes in cognitive function and is seen as a promising method for mild AD. NTB has
shown to be able to detect changes in memory performance

Nutritional Neuroscience 2017 3


4

Canhada et al.
Nutritional Neuroscience

Omega-3 fatty acids’ supplementation in Alzheimer’s disease


Table 2 Clinical trials assessing the impact of omega-3 fatty acid’s supplementation on cognitive function in Alzheimer’s disease patients.

Authors Sample Methods Results

Freund-Levi Initial sample: 204 Design: randomized, double-blind, placebo-controlled study Group w3/w3: 89 individuals, 72.6±9.0 years, 57%
2017

et al.14 Final sample: 174 patients completed the protocol (85%) Follow-up: 6 months of placebo-controlled intervention, women,
Recruitment: patients from a specialist memory clinic followed by 6 months of treatment for both groups Group placebo/w3: 85 individuals, 72.9±8.6 years, 46%
Local: Stockholm, Sweden Group w3/w3: 1720 mg DHA + 600 mg EPA, daily (from w3 women
Inclusion criteria: AD (DSM-IV); MMSE- 15 score between 15 fatty acid concentrate in triglyceride form) Outcomes: MMSE, ADAS-cog, and CDR: There was no
and 30 points; patient living in his or her own home; Group placebo/w3: isocaloric placebo oil (4000 mg of corn statistically significant difference during 6 and 12 months
Treatment with a stable dose of acetylcholine esterase oil) daily for 6 months, followed by 6 months of treatment with between groups on the ADAS-cog (P > 0.05), MMSE
inhibitors for at least 3 months; and plan to continue w3 fatty acid supplementation (P > 0.05), or CDR (P > 0.05).
acetylcholine esterase inhibitors for the duration of the study Both groups received 24 mg of vitamin E (tocopherol) added In a subgroup analyses (n = 32), with subjects with very
Exclusion criteria: treatment with nonsteroidal anti- in their capsules mild cognitive dysfunction (MMSE > 27 e CDR 0.5–1), a
inflammatory agents (low-dose acetylsalicylic acid was Assessments: Baseline, 6 and 12 months: global function significant (−0.5 points vs. −2.6 points; P = 0.01)
accepted), w3 preparations, or anticoagulants; alcohol (CDR), cognitive function (MMSE), and the modified cognitive reduction in decline rate was observed in the omega-3
abuse; concomitant serious disease; did not have a caregiver portion (ADAS-COG) fatty acids’ group when compared with placebo between
baseline and 6 months.
Chiu et al. 16 Initial sample: 46 Design: randomized, double-blind, placebo-controlled study Group 1 (omega-3 fatty acids): 20 individuals, 74.0 years
Final sample: 29 completed the protocol (63.04%) Follow-up: 24 weeks (70.1–77.8), 8 AD and 12 with MCI, 65% women (at
Recruitment: newspaper advertisements Group w3: 1080 mg EPA + 720 mg DHA, daily (the source of baseline)
Local: Tapei, Taiwan the omega-3 fatty acids was menhaden fish body oil Group 2 (placebo): 15 individuals, 76.5 (71.8–81.1), 9 AD
Inclusion criteria: AD (DSM-IV), mild-or-moderate severity concentrate) and 6 with MCI, 46.7% women (at baseline)
(MMSE score between 10 and 26, and a CDR score of 1 or Group placebo: olive oil esters (dosage not informed) Outcomes: ADAS, MMSE, HDRS: there were no
2), or amnesic MCI, age range between 55 and 90 years old Both groups receive antioxidants in their capsules (1.2 mg of differences between groups (P > 0.05, P = 0.87 and,
Exclusion criteria: inadequate motor or sensory capacity; hydroquinone and 12 mg of tocopherols, daily) P = 0.76, respectively).
ischemic lesion on brain computed tomography (CT); a Assessments: baseline and at weeks 6, 12, 18, and 24: In subgroup analyses, the omega-3 fatty acids’ fatty acids
modified Hachinski Ischemic Scale scoreN4; a 17-item HDRS cognitive portion of the ADAS (ADAS-cog), CIBIC-plus scale, group showed significant improvement in ADAS-cog
(Hamilton, 1960) scoreN13; abnormal levels of folic acid, and HDRS compared to the placebo group in participants with mild
vitamin B12, or thyroid function; severe comorbidity (another Baseline, weeks 12, 18, and 24: Erythrocyte membrane fatty cognitive impairment (−3.23 ± 3.82 vs. −0.37 ± 1.4,
neurodegenerative diseases, chronic debilitating neurological acid compositions P = 0.03), which was not observed in those with AD.
illness, brain trauma, tumors, severe pulmonary, renal, liver Baseline and week 24: plasma fatty acids There was a relative improvement in the CIBIC-plus score
disease, cardiac disease, or autoimmune disease, or in omega-3 fatty acids’ fatty acid group of −0.35 (−0.61 to
conditions expected to cause death within 1 year, alcoholism, −0.09, P = 0.008) compared to placebo group every 6
schizophrenia, and bipolar disorder, receiving cholinesterase weeks
agents during the screen or taking NSAID on a long-term
basis)
Continued
Table 2 Continued

Authors Sample Methods Results

Quinn Initial sample: 402 Design: randomized, double-blind, placebo-controlled trial Group DHA: 238 individuals, 76 (9.3) years, 47.1% female
et al. 17 Final sample: 295 completed the protocol (73.4%) Follow-up: 18 months sex (at baseline)
Recruitment: Fifty-one US clinical research sites of the Group DHA: 2000 mg of DHA daily (algal-derived) Group placebo: 164 individuals, 76 (7.8) years, 59.8
Alzheimer’s Disease Cooperative Study Group placebo: corn or soy oil (dosage not informed) female sex (at baseline)
Local: US Assessments: baseline, 6 months, 12 months: cognitive Outcomes: There was no statistically significant difference
Inclusion criteria: probable AD if MMSE score was between subscale (ADAS-cog) and CDR sum of boxes. Brain atrophy between groups on the ADAS-cog (P = 0.41), CDR
14 and 26, they were medically stable, they consumed on [volumetric magnetic resonance imaging (MRI)] in a (P = 0.68), ADCS-ADL (P = 0.38), NPI (P = 0.11), or MMSE
average no more than 200 mg/d of DHA, and they were not subsample of participants (n = 102). ADCS-ADL, NPI, and the (P = 0.88) on the 18-month follow-up.
taking DHA or omega-3 fatty acids’ fatty acid supplements Quality of Life AD scale. Brain atrophy decline difference was not statistically
Exclusion criteria: taking drugs with central anticholinergic Baseline and 18 months: MMSE significant between groups during 18 months (P = 0.79)
effects or sedatives or receiving any investigational treatment
for AD. Stable use (>3 months) of cholinesterase inhibitors or
mean time was permitted
Scheltens Initial sample: 259 Design: randomized, controlled, double-blind, parallel-group, Active: 130 individuals, 74.4 (6.9) years, 52.3% male (at
et al. 24 Final sample: 238 completed the protocol (92%) multi-country trial baseline)
Recruitment: from 27 AD Center Follow-up: 24 weeks Control: 129 individuals, 73.2 (8.4) years, 49.6% male (at
Local: The Netherlands, Germany, Belgium, Spain, Italy, and Group active: 1200 mg DHA + 300 mg EPA + phospholipids, baseline)
France choline, UMP, vitamin B12, B6, and folate, vitamins C and E, Outcomes: NTB memory domain: Z-score was
Inclusion criteria: age ≥50 years, AD (NINCDS-ADRDA), and selenium from Souvenaid® significantly increased in the active vs. the control group
MMSE score of ≥20, recent MRI or computed tomography over the 24-week intervention period (P = 0.023; Cohen’s

Canhada et al.
Group control: isocaloric control product
(CT) scan with no evidence of any other potential causes of Both study products were available as a 125 ml drink with an d = 0.21; 95% confidence interval [−0.06]–[0.49]); a trend
dementia identical taste and appearance, taken once daily for an effect was observed on the NTB total composite z-
Exclusion criteria: other neurological diseases; a Geriatric Assessments: baseline, 12, and 24 weeks: memory function score (P = 0·053); no statistically significant difference
Depression Scale (15-item) score of >6; use of a domain score (NTB); executive function (WMS-r Digit Span, between groups was found on the NTB executive
cholinesterase inhibitor or NMDA-receptor antagonist within 3 Trail Making Tests parts A and B), category Fluency function domain (P = 0.686)

Omega-3 fatty acids’ supplementation in Alzheimer’s disease


months prior to baseline; use of omega-3 fatty acids (Controlled Oral Word Association Test), functional ability EEG measures of functional connectivity in the delta band
containing supplements or regular consumption of oily fish (DAD) were significantly different between study groups during
(>twice/week) within 2 months prior to baseline; use of Nutritional blood parameters (plasma vitamin E, erythrocyte 24 weeks in favor of the active group (P = 0.011)
atropine, scopolamine, tolterodine, hyoscyamine, biperiden, DHA and EPA, and homocysteine), and EEG to assess eyes-
benztropine, trihexyphenidyl, oxybutynin, antipsychotics, closed resting-state oscillatory brain activity and functional
vitamins B, C, and/or E (>200% of the recommended daily brain connectivity in a subset of patients
intake), consumption of high-energy and/or high-protein
Nutritional Neuroscience

nutritional supplements, a change in dose of lipid-lowering


medications, antidepressants, antihypertensives, or the use of
other investigational products within 1 month prior to
baseline; excessive alcohol intake or drug abuse; nursing
home institutionalization; or investigator’s uncertainty
regarding the willingness or ability of the patient to comply
with the protocol
Continued
2017
5
6

Canhada et al.
Nutritional Neuroscience

Table 2 Continued

Omega-3 fatty acids’ supplementation in Alzheimer’s disease


Authors Sample Methods Results

Shinto Initial sample: 39 Design: 3-arm, parallel group, randomized, double-blind, Placebo: 13 individuals, 75.2 (10.8) years, 54% female (at
et al. 15 Final sample: 34 completed the protocol (87%) placebo-controlled pilot clinical trial baseline)
Recruitment: - Follow-up: 12 months Omega-3 fatty acids: 13 individuals, 75.9 (8.1) years, 39%
Local: US Group w3: omega-3 fatty acids’ fish oil concentrate containing female (at baseline)
Inclusion criteria: age ≥55 years, AD (NINCDS/ADRDA), a daily dose of 675 mg DHA and 975 mg EPA Omega-3 fatty acids + LA: 13 individuals, 76.7 (10.6)
MMSE score 15–26, CDR 0.5 to 1.0, not depressed (CESD Group w3/LA: omega-3 fatty acids’ (fish oil concentrate years, 39% female (at baseline)
2017

<4.0), general health status that would not interfere with containing a daily dose of 675 mg DHA and 975 mg EPA) plus Outcomes: ADAS-cog: The ω-3 + LA and ω-3 were not
patient’s ability to participate and complete the study, and LA (600 mg/day) significantly different than the placebo group (P = 0.98,
caregiver/informant able to accompany participant to study Group placebo: soy oil P = 0.86)
visits Assessments: Baseline to 12 months: cognitive subscale ADL: The ω-3 + LA and ω-3 were not significantly different
Exclusion criteria: non-AD dementia, residence in a long-term (ADAS-cog), cognitive function (MMSE), Activities of Daily than the placebo group (P = 0.15, P = 0.82).
care facility at screening visit, history of stroke, health Living/Instrumental Activities of Daily Living (ADL/IADL) MMSE: The mean change over 12 months between
conditions such as cancer (prostate cancer gleason grade placebo group and intervention group with ω-3 was not
<3 and non-metastatic cancers were acceptable), liver statistically significant (−4.6 points (±1.4) for the placebo
disease, history of ventricular fibrillation or ventricular group vs. −4.3 points (±1.3) for the ω-3 group; P = 0.80);
tachycardia, major psychiatric disorder, major central nervous but there was a significant difference between placebo
system diseases, taking lipid-lowering medication, and ω-3+LA (−4.6 points (±1.4) for placebo vs. −1.0
hyperlipidemic, fish oil or cod liver oil supplementation within points (±0.7) for ω-3+LA; P < 0.01), suggesting that the
30 days of enrollment, greater than one 6 ounce serving per combination therapy decreased the rate of decline in
week of fish or seafood within 30 days of enrollment, lipoic MMSE over 12 months
acid supplementation within 30 days of enrollment, taking IADL: The mean change over 12 months between
systemic corticosteroids, neuroleptics, antiparkinsonian placebo group and ω-3 group was statistically significant
agents, and narcotic analgesics. The following medications (4.2 points (±0.9) vs. 0.7 points (±1.0); P < 0.01) and
and supplements were allowed if stable for 4 months prior to between placebo and ω-3+LA (4.2 points (±0.9) vs. 0.9
study enrollment, acetylcholinesterase inhibitors, memantine, points (±1.1); P = 0.01)
vitamin E, and ginkgo biloba
Freud-Levi Initial sample: 204 Design: randomized, double-blind placebo-controlled clinical Group w3/w3: 89 individuals, 72.6 ± 9.0 years. 57%
et al. 25 Final sample: 174 completed the protocol (85%) trial women
Recruitment: patients from a specialist memory clinic Follow-up: 12 months Group placebo/w3: 85 individuals, 46% women,
Local: Stockholm, Sweden Group w3: 430 mg DHA and 150 mg EPA in four capsules 72.9 ± 8.6 years
Inclusion criteria: AD (DSM-IV), MMSE score inclusive of 15 daily Outcomes: NPI: no significant difference over 6 and 12
and 30, living in their own homes; Group placebo: isocaloric placebo oil (containing 1 g of corn months was observed between the w3/w3 and the
Treatment with a stable dose of acetylcholine esterase oil, including 0.6 g of linoleic acid) for 6 months, followed by 6 placebo/w3 groups (P = 0.52)
inhibitors for at least 3 months before the study start months’ open treatment with omega-3 fatty acids for all No significant differences in depressive symptoms
Exclusion criteria: use of NSAID (low-dose acetylsalicylic acid patients occurred in either the v3/v3 or the placebo/v3 groups at
was accepted), ω3 preparations, anticoagulation agents; 4 mg vitamin E (tocopherol) was added to each EPAX1050TG 0–6 and 6–12 months, whether assessed using the
alcohol abuse; concomitant serious disease; or did not have and placebo capsule depressive domain of NPI (P = 0.84) or by MADRS
a caregiver Assessments: Baseline, 6 and 12 months: Neuropsychiatric (P = 0.49)
Exclusion criteria: treatment with nonsteroidal anti- symptoms (NPI); Depressive symptoms (MADRS); Burden of No significant differences in DAD score change or in the
inflammatory agents (low-dose acetylsalicylic acid was the caregiver was evaluated using three items from CGB. three items of care givers burden (CGB) between the v3/
accepted), w3 preparations, or anticoagulants; alcohol Activities of daily living were rated (DAD) v3 and placebo/v3 at 0 and 6 months were found
abuse; concomitant serious disease; did not have a caregiver
Continued
Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

between intervention and placebo in NPI (P = 0.52),

Outcomes: There was no statistically significant difference


53.8% female sex (at baseline – except for the number of

(P = 0.576), BADLS (P = 0.595) in the 4 months of follow-


between groups on the MMSES7 (P = 0.711), MMSEWB
MADRS (P = 0.49), DAD (P > 0.05) e CGB (P >

Group omega-3 fatty acids: Eight individuals, 71.1 (4.8)


years, 56.8% female sex (at baseline – except for the
number of individuals, data refer to 37 CNID and AD

Group placebo: Eleven individuals, 71.1 (9.5) years,


0.05).

individuals, data refer to 39 CNID and AD patients)


Chiu et al. 16 studied 46 subjects with mild-to-mod-
erate AD or mild cognitive impairment (mean age:
55–90 years). During 24 weeks, intervention group
received omega-3 fatty acids in the form of 1080 mg
Results

of EPA and 720 mg of DHA, while placebo group


received olive oil. Medication for AD treatment was
not allowed. There was no significant statistical differ-
ence in ADAS-cog (P > 0.05), MMSE (P = 0.87), and
HDRS (P = 0.76) between the two groups in 24 weeks.
A significant improvement was observed in CIBIC-
plus in the intervention group of −0.35 (IC95%
patients)

−0.61 a −0.09, P = 0.008) every 6 weeks compared


to placebo group. In a subgroup analysis, participants
up

with mild cognitive impairment, but not with AD,


showed a significant additional delay in ADAS-cog
Both groups also received 20 mg mixed tocopherols, daily, in
Group omega-3 fatty acids: 625 mg DHA and 600 mg EPA,

Assessments: Baseline, 1 and 4 months: cognitive function

decline compared to placebo group (−3.23 ± 3.82 vs.


(MMSE: MMSES7 and MMSEWB), activities of daily living

−0.37 ± 1.4, P = 0.03).


Quinn et al. 17 assessed 402 subjects with mild-to-
moderate AD (age: 76 ± 8.7 years). The intervention
Group placebo: olive oil (dosage not informed)
Design: randomized, double-blind, placebo-

group received 2000 mg of DHA and the placebo


group received corn or soy oil for 18 months.
Methods

Medication for AD treatment was allowed. A sub-


sample received magnetic resonance imaging (MRI)
assessments. There was no significant effect from the
intervention in cognitive decline rhythm in ADAS-
cog (P = 0.41), CDR (P = 0.68), ADCS-ADL (P =
Follow-up: 4 months

(BADLS) and others

0.38), NPI (P = 0.11), or in MMSE (P = 0.88).


controlled study

their capsules

There was also no change in cerebral volume at 18


months of trial (P = 0.79).
Scheltens et al. 24 assessed 259 subjects with mild AD
daily

(age of 55 years or higher). The intervention group


received 1200 mg of DHA and 300 mg of EPA with
needs that would prevent the study’s requirements from being
of other psychiatric or neurological conditions; current drug or
minimum of 3 months prior to entering the study; MMSE score
have been on a stable dose of a cholinesterase inhibitor for a

Exclusion criteria: if another dementia was suspected; history


Inclusion criteria: AD (diagnosis based on NINCDS-ADRDA);
Final sample: 19 completed the protocol (100%). There were

the addition of other nutrients in the form of the


accompany them to research visits and who lived with them
alcohol addiction; disabilities, disorders or special dietary

product Souvenaid®, whereas the placebo group


supplements; did not have a relative or friend that could
undertaken; already taking omega-3 fatty acids’ PUFA

received isocaloric control product for 24 weeks.


Medication for AD treatment was not allowed. A sub-
Recruitment: through specialist memory clinics
no dropouts but twodid not complete all visits

sample was assessed through EEG. In the NTB


memory domain, a significant increase was found in
or visited them at least 2 times a week

the intervention group compared to control group at


24 weeks (P = 0.023, additional alteration of 0.21,
Sample

IC95% −0.06–0.49). There was a trend for effect in


the NTB total composite in 24 weeks (P = 0.053). No
significant effect from the intervention was found in
the NTB execution function domain (P = 0.686).
between 16 and 30
Local: Bristol, UK
Initial sample: 19

EEG measures of functional connectivity were signifi-


cantly different in one of the bands – the delta band –
in favor of the intervention group (P = 0.011).
Table 2 Continued

Shinto et al. 15 studied 39 subjects with more than 55


years with probable AD in a randomized placebo-con-
trolled pilot with three arms. Two groups received
et al. 26

omega-3 fatty acids’ supplementation, one only


Authors

Phillips

omega-3 fatty acids (975 mg EPA and 675 mg


DHA), and the other with the addition of alpha

Nutritional Neuroscience 2017 7


Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

lipoic acid (975 mg EPA, 675 mg DHA, and 600 mg The CIBIC-plus21 is a scale that provides a global
ALA); placebo group received soy oil. The interven- measure designed to detect changes not only in cogni-
tion lasted 12 months and medication for AD was tion, but also in overall function and behavior, thus
allowed. There were no differences in ADAS-cog and assessing dementia’s severity and progression. Only
ADL between placebo and omega-3 fatty acids (P = Chiu et al. 16 used this scale to assess the impact of
0.86; P = 0.82) or between placebo and omega-3 omega-3 fatty acids’ supplementation, finding statisti-
fatty acids + ALA (P = 0.98; P = 0.15). In MMSE, cally significant improvements with the intervention
the mean alteration in 12 months between placebo compared to placebo. However, as this same study
group and intervention group with only omega-3 did not find benefits in other two scales that specifi-
fatty acids was not significant (−4.6 ± 1.4 vs. −4.3 ± cally measure the cognitive capacity – ADAS-cog
1.3 points; P = 0.80), whereas the difference between and MMSE – it is possible that the positive results in
placebo and omega-3 fatty acids + ALA was signifi- CIBIC-plus are the reflection of omega-3 fatty acid’s
cant (−4.6 ± 1.4 points for placebo vs. −1.0 ± 0.7 action in other systems, such as cardiovascular or
points for omega-3 fatty acids + ALA; P < 0.01). immune systems – in which the omega-3 fatty acids’
The mean alteration in IADL in 12 months was sig- beneficial effects have already been reported.
nificant between placebo group and omega-3 fatty From the studies that used MMSE scale as an
acids group (4.2 ± 0.9 points for placebo group vs. assessment method,14–17,26 only one demonstrated
0.7 ± 1.0 points for omega-3 fatty acids group; P < statistically significant effects in favor of interven-
0.01) and between placebo and omega-3 fatty tion,15 but only when in combination with ALA.
acids + ALA (4.2 ± 0.9 points for placebo group vs. MMSE22 assess cognitive function in the orientation,
0.9 ± 1.1 points for omega-3 fatty acids + ALA memory, attention, calculation, language, and visual
group; P = 0.01). construction areas. The mechanism that resulted in
Phillips et al. 26 assessed omega-3 fatty acids ‘sup- the delay of cognitive decline progression remains
plementation in 19 subjects with AD (mean age: uncertain, so it is not possible to assert if the effects
71.1 ± 4.8 years – all data refer to 76 patients, which were the result of the combination or only of ALA,
includes 57 with cognitive impairment and 19 with because there was no exclusive intervention with the
AD). The intervention group received daily omega-3 latter in that study.
fatty acids in the dosages of 625 mg DHA and ADCS-ADL scale19 was used in two studies.15,17
600 mg EPA and the placebo group received olive oil This scale measures the subject’s functional ability in
for 4 months. Medication for AD treatment was performing daily living activities. While Quinn
allowed. As a result, there was no significant statistical et al. 17 used the full scale and did not find statistical
difference at 4 months between groups in MMSES7 significance, Shinto et al. 15 used a modified version
(P = 0.711), MMSEWB (P = 0.576), or BADLS to measure ADL and IADL. ADL assessed basic
(P = 0.595). daily functions, such as bathing and eating, whereas
IADL assessed more complex skills, such as using
Discussion the phone and cooking meals. In this study, despite
Recent literature reveals weak associations between the fact that placebo group has shown an apparent
omega-3 fatty acids’ supplementation and cognitive superior decline, no significant difference was found
outcomes in AD. Most studies do not find statistically in ADL between placebo and omega-3 fatty acids or
significant results when supplementation is compared between placebo and omega-3 fatty acids + ALA.
to placebo. On the other hand, some benefit was Regarding IADL, both groups that received omega-3
found in just a few evaluated scales.15,16,24 Omega-3 fatty acids obtained delay in functional decline when
fatty acids’ supplementation seems more effective in compared with placebo. This scale measures more
very mild AD.14 complex daily functions, and maybe for this reason it
Some studies showed improvements with omega-3 was more sensitive to Alzheimer’s progression than
fatty acids ‘supplementation in overall analyses, and ADL.15
not only in subgroups. Chiu et al. 16 found better From the seven studies included in this review, only
results for the intervention group, compared to one used NTB scale for assessing cognitive function24
placebo, in the CIBIC-plus scale. Shinto et al. 15 to detect changes in early stages of AD.23 The authors
found better results in the MMSE scale with omega- found a significant increase in the NTB memory
3 fatty acids in addition to ALA, and in the IADL domain in the intervention group with the product
scale in both groups that received omega-3 fatty Souvenaid® compared to placebo, but not in the
acid’s supplementation, compared to placebo. executive function and total composite score domains.
Scheltens et al. 24 also found better results with The effect observed in a subgroup of subjects whose
omega-3 fatty acids in the memory domain of NTB MMSE scores were the highest and cognitive decline
scale. was the smaller14 corroborates findings from

8 Nutritional Neuroscience 2017


Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

epidemiological observational studies. Several cohort plasma levels after 1.8 g/day of oil fish – smaller
studies suggest that fish consumption, omega-3 fatty than in other study, in which increases of 2.4- to 3.6-
acids’ source, would have a role in Alzheimer’s preven- folds were observed after 2.4 g/day of fish oil.14,25 In
tion, but not in the treatment when the disease is addition, the study of Chiu et al. 16 was performed in
already manifested.4,6–8 Subgroup analysis of subjects Taiwan, a country with a high fish consumption,27
with mild AD also allowed the conclusion that so perhaps a higher omega-3 fatty acids’ dosage is
improvements were originated in the memory necessary to reach a significant contrast compared to
domain, one of the disease’s key symptoms that control group. Shinto et al. 15 used 975 mg EPA +
marks the beginning of the symptomatology. 675 mg DHA, a quite similar dosage to the one used
Some limitations in the studies may explain part of by Chiu et al. 16, but with a DHA:EPA relation
their results and, as so, deserve consideration. First, much smaller than other studies.14,17,25 Phillips
in at least three studies15,16,26 the sample size was too et al. 26 used practically the same amounts for DHA
small, which can limit statistical power. Second, loss (625 mg) and EPA (600 mg). Freund-Levi et al. 14,25,
during the treatment period was frequent – in one of on the other hand, supplemented fish oil (DHA
the studies only 63%16 of the subjects completed the 1.7 g/day and EPA 0.6 g/day) in higher doses than
follow-up. Two authors tested if dropout was different studies that revealed benefits in cardiovascular dis-
between groups,16,17 not finding statistically significant eases and AD prevention. The authors chose an elev-
differences. Reasons for losing participants during the ated DHA:EPA relation (2.8-folds more DHA than
follow-up included lack of adhesion from caregivers, EPA) based on studies that show DHA deficiency in
withdrawal of consent, adverse gastrointestinal brains affected by AD and on studies that associate
effects, other diseases, and death. Third, the high het- the DHA ingestion with Aβ reduction in hippocampus
erogeneity of methods used to assess aspects of and parietal cortex in rats in a dose-dependent way.
patient’s cognition, along with the heterogeneity in These areas are the first affected by AD in the brain,
interventions composition and dosages, makes it diffi- and they alter the verbal episodic memory. Scheltens
cult to establish greater comparative analysis. et al. 24 also administered a higher DHA quantity
Other study characteristics can also contribute to throughout the use of the product Souvenaid®
the absence of significant results of omega-3 fatty (1200 mg DHA + 300 mg EPA), which contains
acids in AD in some of these papers. Primarily, in additional nutrients – potentially responsible for part
two of the studies16,26 the placebo included olive oil, of the observed benefits. Quinn et al. 17, on the other
which is a source of monounsaturated fatty acids, hand, did not observe significant impact with DHA
already shown by other authors as inversely associated exclusive use (2 g/day). The high heterogeneity in
with cognitive decline. In this case, the placebo would the dosages supplemented made unfeasible to
not be totally inert and, therefore, would limit the conduct a meta-analysis.
differences compared to the intervention group. To our knowledge, this is the most recent systematic
Second, in five studies14,15,17,25,26 participants were review focusing exclusively in intervention studies,
allowed to receive medications for AD treatment with randomized clinical trials, controlled by
along with omega-3 fatty acids, which could contrib- placebo, assessing omega-3 fatty acids’ supplemen-
ute to good outcomes also in the control group. tation in AD, regarding cognitive function outcomes.
However, in these five studies there was no apparent These findings support other reviews, that previously
difference in the use of medications between groups, described omega 3’s supplementation in other popu-
and in two of them15,17 tests were applied to detect lations and other outcomes,28–31 as also still inconclu-
differences and no statistical significance was found. sive, as for the treatment of AD.
Third, the trials observed in this review had, as Although few studies have shown alterations in
exposure period to treatment, a variation between some of the cognitive function scales, it is not
4 and 18 months, a time considered appropriate from enough to support, in a systematic way, omega-3
the ethical point of view in this population, but that fatty acids’ supplementation in AD treatment. The
can be too short since Alzheimer’s is a chronic effects found in mild AD corroborate observational
disease. Fourth, omega-3 fatty acids’ dosages could epidemiological studies that show omega-3 fatty
have been insufficient to provide significant benefits. acids’ benefits in the early stages of the disease, when
Indeed, there is no consensus among AD studies there is still a mild cerebral functional impairment.
about dosages of EPA e DHA. In Chiu et al. 16, In this sense, more studies are needed, with sufficient
1080 mg of EPA and 720 mg of DHA was sup- sample size and longer intervention period, for differ-
plemented daily. In this study, there was an increase ent stages of the disease, with the use of standardized
in the DHA levels and total omega-3 fatty acids in cognitive function scales, that also assess the depen-
plasma and in membranes of erythrocytes, but not dency level in daily activities, in which the effect of
in EPA levels. There was an increase of 1.5-fold in each of the omega-3 fatty acids is evaluated, in

Nutritional Neuroscience 2017 9


Canhada et al. Omega-3 fatty acids’ supplementation in Alzheimer’s disease

sufficient doses, so that we can have a clearer con- pilot trial of omega-3 fatty acids and alpha lipoic acid in
Alzheimer’s disease. J Alzheimers Dis 2014;38(1):111–20.
clusion of the real benefits of supplementation regard- 16 Chiu C-C, Su K-P, Cheng T-C, Liu H-C, Chang C-J, Dewey
ing clinically relevant outcomes. ME, et al. The effects of omega-3 fatty acids monotherapy
in Alzheimer’s disease and mild cognitive impairment: a
preliminary randomized double-blind placebo-controlled study.
Disclaimer statements Prog Neuropsychopharmacol Biol Psychiatry 2008;32(6):
Contributors None. 1538–44.
17 Quinn JF, Raman R, Thomas RG., Yurko-Mauro K, Nelson
EB., Van Dyck C, et al. Docosahexaenoic acid supplementation
Funding None. and cognitive decline in Alzheimer disease: a randomized trial.
JAMA 2010;304(17):1903–11.
Conflicts of interest None. 18 Rosen WG, Mohs R, Davis K. A new rating scale for
Alzheimer’s disease. Am J Psychiatry 1984;141(11):1356–64.
Ethics approval None. 19 Galasko D, Bennett D, Sano M, Ernesto C, Thomas R,
Grundman M, et al. An inventory to assess activities of daily
living for clinical trials in Alzheimer’s disease. The Alzheimer’s
References disease cooperative study. Alzheimer Dis Assoc Disord 1997;11
1 Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, (Suppl. 2):33–9.
Cummings JL. Alzheimer’s disease. Nat Rev Dis Prim 2016;1 20 Morris JC. The Clinical Dementia Rating (CDR):
(15056):1–18. current version and scoring rules. Neurology 1993;43(11):
2 Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. 2412–4.
Forecasting the global burden of Alzheimer’s disease. 21 Schneider LS, Olin JT, Doody RS, Clark CM, Morris JC,
Alzheimer’s Dement 2007;3(3):186–91. Reisberg B, et al. Validity and reliability of the Alzheimer’s
3 Luchsinger JA, Gustafson DR. Adiposity and Alzheimer’s disease cooperative study-clinical global impression of change.
disease. Curr Opin Clin NutrMetab Care 2009;12(1):15–21. The Alzheimer’s disease cooperative study. Alzheimer Dis
4 Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Assoc Dis 1997;11(Suppl. 2):22–32.
Wilson RS, et al. Consumption of fish and n-3 fatty acids and 22 Folstein MF, Folstein SE, Mchugh PR. “Mini-Mental State”: a
risk of incident Alzheimer disease. Arch Neurol 2003;60(7): practical method for grading the cognitive state of patients for
940–6. the clinician. J Psychiatr Res 1975;12(3):189–98.
5 Morris MC, et al. The role of nutrition in Alzheimer’s disease: 23 Harrison J, Minassian SL, Jenkins L, Black RS, Koller M,
epidemiological evidence. Eur J Neurol 2009;16(Suppl. 1):1–7. Grundman M. A neuropsychological test battery for use in
6 Huang T-L, Zandi PP, Tucker KL, Fitzpatrick AL, Kuller LH, Alzheimer disease clinical trials. Arch Neurol 2007;64(9):1323–9.
Fried LP, et al. Benefits of fatty fish on dementia risk are stronger 24 Scheltens P, Twisk JW, Blesa R, Scarpini E, von Arnim CA,
for those without APOE e4. Neurology 2005;65(9):1409–14. Bongers A, et al. Efficacy of Souvenaid in mild Alzheimer’s
7 Barberger-Gateau P, Raffaitin C, Letenneur L, Berr C, Tzourio disease: results from a randomized, controlled trial. J
C, Dartigues JF, et al. Dietary patterns and risk of dementia: the Alzheimers Dis 2012;31(1):225–36.
three-city cohort study. Neurology 2007;69(20):1921–30. 25 Freund-Levi Y, Basun H, Cederholm T, Faxén-Irving G,
8 Kalmijn S, Launer LJ, Ott A, Witteman JC, Hofman A, Breteler Garlind A, Grut M, et al. Omega-3 supplementation in mild
MM. Dietary fat intake and the risk of incident dementia in the to moderate Alzheimer’s disease: effects on neuropsychiatric
Rotterdam study. Ann Neurol 1997;42(5):776–82. symptoms. Int J Geriatr Psych 2008;23(2):161–9.
9 Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer 26 Phillips M, Childs C, Calder P, Rogers P. No effect of omega-3
disease. Nat Rev Neurol 2011;7(3):137–52. fatty acid supplementation on cognition and mood in individuals
10 Surette M. The science behind dietary omega-3 fatty acids. Can with cognitive impairment and probable Alzheimer’s disease:
Med Assoc J 2008;178(2):177–80. a randomised controlled trial. Int J Mol Sci 2015;16(10):
11 Whelan J, Rust C. Innovative dietary sources of n-3 fatty acids. 24600–13.
Annu Rev Nutr 2006;26:75–103. 27 Hibbeln JR. Fish consumption and major depression. Lancet
12 Taber L, Chiu C-H, Whelan J. Assessment of the arachidonic 1998;351(9110):1213.
acid content in foods commonly consumed in the American 28 Mazereeuw G, Lanctôt KL, Chau SA, Swardfager W, Herrmann
diet. Lipids 1998;33:1151–7. N. Effects of omega-3 fatty acids on cognitive performance: a
13 Moher D, Liberati A, Tetzlaff J, Altman DG, the PRISMA meta-analysis. Neurobiol Aging 2012;33(7):1482.e17–29.
Group. Preferred reporting items for systematic reviews and 29 Burckhardt M, Herke M, Wustmann T, Watzke S, Langer G,
meta-analyses: the PRISMA statement. Ann Intern Med Fink A. Omega-3 fatty acids for the treatment of dementia.
2009;151(4):264–9. Cochrane Database Syst Rev 2016;4:1–67.
14 Freund-Levi Y, Eriksdotter-Jönhagen M, Cederholm T, Basun 30 Thomas J, Thomas CJ, Radcliffe J, Itsiopoulos C. Omega-3
H, Faxén-Irving G, Garlind A, et al. Omega-3 fatty acid treat- fatty acids in early prevention of inflammatory neurodegenera-
ment in 174 patients with mild to moderate Alzheimer disease: tive disease: a focus on Alzheimer’s disease. Biomed Res Int
OmegAD study: a randomized double-blind trial. Arch Neurol 2015;2015:1–13. 172801.
2006;63(10):1402–8. 31 Pusceddu M, Kelly P, Stanton C, Cryan JF, Dinan TG. N-3 poly-
15 Shinto L, Quinn J, Montine T, Dodge HH, Woodward W, unsaturated fatty acids through the lifespan: implication for psy-
Baldauf-Wagner S, et al. A randomized placebo-controlled chopathology. Int J Neuropsychopharmacol 2016;19(12):1–23.

10 Nutritional Neuroscience 2017

You might also like