1
Harry B. Rice, Ph.D.
VP, Regulatory and Scientific Affairs
Global Organization for EPA and DHA Omega-3s (GOED)
1075 Hollywood Avenue
Salt Lake City, UT 84105
RE: Petition for a Health Claim for Eicosapentaenoic Acid and Docosahexaenoic
Acid and Reduction of Blood Pressure in the General Population (Docket No.
FDA-2014-Q-1146)
Dear Dr. Rice:
This letter responds to the health claim petition submitted by the Global Organization for EPA
and DHA Omega-3s (GOED or you) pursuant to sections 403(r)(4) and 403(r)(5)(D) of the
Federal Food, Drug, and Cosmetic Act (the Act) (21 U.S.C. §§ 343(r)(4) and 343(r)(5)(D)). The
Food and Drug Administration (FDA or the agency) received your petition on April 24, 2014.
1
The petition requested that FDA authorize a health claim for the relationship between the
consumption of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) and reduction of
blood pressure (BP). Based on discussions with you after we received the petition,
2
we reviewed
it as a petition for a qualified health claim in accordance with the July 10, 2003 Task Force Final
Report on the Consumer Health Information for Better Nutrition Initiative.
3
The petition proposed the following model claims to be used on the labels or in the labeling of
conventional foods and dietary supplements containing EPA and DHA:
EPA and DHA help lower blood pressure in the general population.
EPA and DHA reduce BP, a risk factor for CHD (coronary heart disease).
EPA and DHA reduce the risk of CHD.
Research shows that EPA and DHA may be beneficial for moderating BP, a risk factor
for CHD.
Convincing scientific evidence indicates that EPA and DHA help lower blood pressure in
the general population, with comparable reductions to those achieved with other diet and
lifestyle interventions.
FDA filed the petition for comprehensive review on August 8, 2014 and posted the petition on
the FDA website for a 60-day comment period, consistent with FDA’s guidance on the
procedures for the submission of qualified health claim petitions (“qualified health claim
1
This petition is a revision of an earlier petition on the same subject received by FDA on December 5, 2013. GOED
submitted the revised petition after being notified by FDA that the original petition was incomplete and did not meet
the health claim petition requirements in 21 CFR 101.70.
2
See FDA Filing Letter for GOED Health Claim Petition, Docket No. FDA-2014-Q-1146-0002,
https://www.regulations.gov/document?D=FDA-2014-Q-1146-0002
.
3
See FDA, “Consumer Health Information for Better Nutrition Initiative: Task Force Final Report,” July 10, 2003
[
https://www.fda.gov/food/food-labeling-nutrition/consumer-health-information-better-nutrition-initiative-task-
force-final-report] (accessed May 17, 2019).
2
procedures guidance”).
4
The agency received a total of 22 comments in response to the petition.
Comments were from industry, academia, food and health organizations, and individual
consumers. FDA considered all 22 comments in its evaluation of the petition.
Of the 22 comments, six requested an extension of the 60-day comment period. In response,
FDA extended the comment period for 45 days. The remaining comments included nine
statements of support for the petition without additional information or substantiation. These
comments stated that EPA and DHA (EPA/DHA) are important nutrients that lower blood
pressure and that many people have low intakes of EPA/DHA. These comments also noted that
consumers should have information about the health effects of EPA/DHA and that EPA/DHA
supplements are safe and inexpensive. Two comments opposed to the petition were submitted,
also without additional information or supporting evidence. These comments stated that dietary
supplements should not bear health claims due to concerns of efficacy and safety.
Five comments provided additional information and evidence for use in FDA’s consideration of
the petition. One of these comments did not express support for or opposition to the petition but
did submit information on EPA/DHA intake levels to assist evaluation of the petition. Four
comments stated that the petitioner had satisfactorily demonstrated a relationship between the
consumption of EPA and DHA and reduction of blood pressure. These comments provided
additional scientific studies and data, which were used in our evaluation of the relationship
between EPA and DHA omega-3 fatty acids, blood pressure, and hypertension risk (see Section
II of this letter).
This letter sets forth the results of FDA’s scientific review of the evidence for the qualified
health claims requested in the petition. As explained in this letter, FDA has determined that the
current scientific evidence supports the use of certain qualified health claims in the labeling of
conventional foods and dietary supplements that contain EPA and DHA omega-3 fatty acids,
concerning the relationship between these substances and reducing the risk of hypertension and
coronary heart disease (CHD) by lowering blood pressure. Accordingly, this letter discusses the
factors that FDA intends to consider in the exercise of its enforcement discretion for the use of
qualified health claims, for both conventional foods and dietary supplements, about consumption
of EPA and DHA omega-3 fatty acids and reduced risk of hypertension and CHD through the
intermediate link of lowering blood pressure. For brevity, these claims will be referred to as “the
EPA/DHA qualified health claims(or “an EPA/DHA qualified health claim” when referring to
an individual claim) in this letter.
5
The “Conclusions” section of this letter contains the qualified
health claim language for which FDA intends to exercise enforcement discretion.
4
See FDA, “Interim Procedures for Qualified Health Claims in the Labeling of Conventional Human Food and
Human Dietary Supplements,” July 10, 2003
[
hhttps://www.fda.gov/regulatory-information/search-fda-guidance-documents/guidance-industry-interim-
procedures-qualified-health-claims-labeling-conventional-human-food-and] (accessed May 17, 2019).
5
As used in this letter, “EPA/DHA qualified health claim” is not intended to refer to any qualified health claim for
EPA and DHA other than the ones that are the subject of this letter.
3
I. Overview of Data and Eligibility for a Qualified Health Claim
A health claim characterizes the relationship between a substance and a disease or health-related
condition (21 CFR 101.14(a)(1)). The substance must be associated with a disease or health-
related condition for which the general U.S. population, or an identified U.S. population
subgroup is at risk (21 CFR 101.14(b)(1)). Health claims characterize the relationship between
the substance and reduced risk of contracting a particular disease or health-related condition.
6
In
a review of a qualified health claim, the agency first identifies the substance and the disease or
health-related condition that are the subject of the proposed claim, as well as the population to
which the claim is targeted.
7
FDA considers the data and information provided in the petition, in addition to other written data
and information available to the agency, to determine whether credible scientific evidence
supports a relationship between the substance and the disease or health-related condition.
8
At
this stage, the agency separates individual reports of human studies from other types of data and
information. FDA focuses its review on reports of human intervention and observational
studies.
9
In addition to individual reports of human studies, the agency also considers other types of data
and information in its review, such as meta-analyses,
10
review articles,
11
and animal and in vitro
studies. These other types of data and information may be useful to assist the agency in
understanding the scientific issues about the substance, the disease, or both, but cannot by
themselves support a health claim relationship. Reports that discuss a number of different
studies, such as meta-analyses and review articles, do not provide sufficient information on the
individual studies reviewed for FDA to determine critical elements, such as the study population
characteristics and the composition of the products used. Similarly, the lack of detailed
information on studies summarized in review articles and meta-analyses prevents FDA from
determining whether the studies are flawed in critical elements such as design, conduct of
studies, and data analysis. FDA must be able to review the critical elements of a study to
determine whether any scientific conclusions can be drawn from it. Therefore, FDA uses meta-
6
See Whitaker v. Thompson, 353 F.3d 947, 950-51 (D.C. Cir.) (upholding FDA’s interpretation of what constitutes a
health claim), cert. denied, 125 S. Ct. 310 (2004).
7
See FDA, “Guidance for Industry: Evidence-Based Review System for the Scientific Evaluation of Health Claims -
Final,” January 2009 (“guidance on scientific evaluation of health claims”) [
https://www.fda.gov/regulatory-
information/search-fda-guidance-documents/guidance-industry-evidence-based-review-system-scientific-evaluation-
health-claims] (accessed May 17, 2019).
8
For brevity, “disease” will be used as shorthand for “disease or health-related condition” in the rest of this letter
except when distinguishing between the two or quoting or paraphrasing a regulation that uses the longer term.
9
In an intervention study, subjects similar to each other are randomly assigned to either receive the intervention or
not to receive the intervention, whereas in an observational study, the subjects (or their medical records) are
observed for a certain outcome (e.g., disease). Intervention studies provide the strongest evidence for an effect. See
supra, note 7.
10
A meta-analysis is the process of systematically combining and evaluating the results of clinical trials that have
been completed or terminated (Spilker, 1991).
11
Review articles summarize the findings of individual studies.
4
analyses, review articles, and similar publications
12
to identify reports of additional studies that
may be useful to the health claim review and as background about the substance-disease
relationship.
13
If additional studies are identified, the agency evaluates them individually.
FDA uses animal and in vitro studies as background information regarding mechanisms of action
that might be involved in any relationship between the substance and the disease. The
physiology of animals is different than that of humans. In vitro studies are conducted in an
artificial environment and cannot account for a multitude of normal physiological processes,
such as digestion, absorption, distribution, and metabolism, which affect how humans respond to
the consumption of foods and dietary substances (Institute of Medicine (IOM), 2005a). Animal
and in vitro studies can be used to generate hypotheses or to explore a mechanism of action but
cannot adequately support a relationship between the substance and the disease.
FDA evaluates the individual reports of human studies to determine whether any scientific
conclusions can be drawn from each study. The absence of critical factors, such as a control
group or a statistical analysis, means that scientific conclusions cannot be drawn from the study
(Spilker, 1991; National Research Council (NRC), 2011). Studies from which FDA cannot draw
any scientific conclusions do not support the health claim relationship, and these are eliminated
from further review.
Because health claims involve reducing the risk of a disease in people who do not already have
that disease, FDA considers evidence from studies in individuals diagnosed with the disease that
is the subject of the health claim only if it is scientifically appropriate to extrapolate to
individuals who do not have the disease. That is, the available scientific evidence must
demonstrate that: (1) the mechanism(s) for the mitigation or treatment effects measured in the
diseased populations are the same as the mechanism(s) for risk reduction effects in non-diseased
populations; and (2) the substance affects these mechanisms in the same way in both diseased
people and healthy people. If such evidence is not available, the agency cannot draw any
scientific conclusions from studies that use diseased subjects to evaluate the substance-disease
relationship.
Next, FDA rates the remaining human intervention and observational studies for methodological
quality. This quality rating is based on several criteria related to study design (e.g., use of a
placebo control versus a non-placebo controlled group), data collection (e.g., type of dietary
assessment method), the quality of the statistical analysis, the type of outcome measured (e.g.,
disease incidence versus validated surrogate endpoint), and study population characteristics other
than relevance to the U.S. population (e.g., selection bias and whether important information
about the study subjects – e.g., age, smoker vs. non-smoker – was gathered and reported). For
example, if the scientific study adequately addressed all or most of the above criteria, it would
12
Other examples include book chapters, abstracts, letters to the editor, and committee reports.
13
Although FDA does not generally use meta-analyses in its health claim evaluations for the reasons discussed in
the text, the agency will include a meta-analysis in its scientific evaluation if the meta-analysis was conducted with
pooled data from all the publicly available studies from which scientific conclusions can be drawn (based on the
criteria in FDA’s guidance on scientific evaluation of health claims) and the statistical analyses were properly
conducted. See supra, note 7 [Section III.B, “Research Synthesis Studies”].
5
receive a high methodological quality rating. Moderate or low quality ratings would be given
based on the extent of the deficiencies or uncertainties in the quality criteria. Studies that are so
deficient that scientific conclusions cannot be drawn from them cannot be used to support the
health claim relationship, and these are eliminated from further review.
Finally, FDA evaluates the results of the remaining studies. The agency then rates the strength
of the total body of publicly available evidence.
14
The agency conducts this rating evaluation by
considering the study type (e.g., intervention, prospective cohort, case-control, cross-sectional),
the methodological quality rating previously assigned, the quantity of evidence (number of
studies of each type and study sample sizes), whether the body of scientific evidence supports a
health claim relationship for the U.S. population or target subgroup, whether study results
supporting the proposed claim have been replicated,
15
and the overall consistency
16
of the total
body of evidence.
17
Based on the totality of the scientific evidence, FDA determines whether
such evidence is credible to support a qualified health claim for the substance-disease
relationship, and, if so, considers what qualifying language should be included to convey the
limits on the level of scientific evidence supporting the relationship or to prevent the claim from
being misleading in other ways.
A. Substance
A health claim characterizes the relationship between a substance and a disease or health-related
condition (21 CFR 101.14(a)(1)). A substance means a specific food (defined to include both
conventional foods and dietary supplements) or component of food (21 CFR 101.14(a)(2)). The
petition identified EPA and DHA in combination as the substances that are the subject of the
proposed claim. EPA and DHA are omega-3 fatty acids that are components of some fatty fish
(primarily cold water fish), fish oils, other foods (e.g., seaweed), dietary supplements, and food
ingredients (e.g., algal oils). Accordingly, the agency concludes that EPA and DHA omega-3
fatty acids are components of food and therefore meet the definition of substance in the health
claim regulation (21 CFR 101.14(a)(2)).
B. Disease or Health-Related Condition
A disease or health-related condition means damage to an organ, part, structure, or system of the
body such that it does not function properly, or a state of health leading to such dysfunctioning
(21 CFR 101.14(a)(5)). The petition proposes model health claims about lowering blood
pressure to reduce the risk of hypertension and coronary heart disease. Hypertension is a disease
in which blood flows through blood vessels or arteries at higher than normal pressures.
14
See supra, note 7 [Section III.F].
15
Replication of scientific findings is important for evaluating the strength of scientific evidence (Wilson, 1990).
16
Consistency of findings among similar and different study designs is important for evaluating causation and the
strength of scientific evidence (Hill A.B., The environment and disease: association or causation? Proc R Soc Med
1965;58:295-300 ); See also Agency for Healthcare Research and Quality, “Systems to rate the scientific evidence”
(March 2002) [http://archive.ahrq.gov/clinic/epcsums/strengthsum.pdf
], defining “consistency” as “the extent to
which similar findings are reported using similar and different study designs.”
17
See supra, note 7 [Section III.F].
6
Hypertension is diagnosed when systolic blood pressure is 140 mmHg or diastolic blood
pressure is 90 mmHg when measured on more than one occasion. Pre-hypertension occurs
when systolic blood pressure is between 120-139 mmHg or diastolic blood pressure is between
80-89 mmHg.
18
Blood pressure is the force of blood pushing against the walls of the arteries and blood vessels as
the heart pumps blood. Hypertension, also called high blood pressure, is when this force against
the artery walls is too high.
19
Pre-hypertension, where blood pressure is elevated (higher than
normal) but not yet in the hypertensive range, increases the risk of developing hypertension in
the future.
20
Pre-hypertension is a risk factor for hypertension, meaning that it is also a state of
health leading to hypertension. Therefore, pre-hypertension is considered to be a health-related
condition. In addition, pre-hypertension and hypertension are risk factors for other diseases,
such as chronic kidney disease and cardiovascular disease (diseases of the heart and circulatory
system, including coronary heart disease and stroke).
21
Thus, hypertension is a state of health
leading to cardiovascular disease (CVD) and other diseases, as well as a disease in its own right.
Because hypertension and pre-hypertension are both states of health leading to CVD and other
diseases, they are also “health-related conditions” as defined in 21 CFR 101.14(a)(5).
Coronary heart disease (CHD) is one of the most common and serious forms of CVD and refers
to diseases of the heart muscle and supporting blood vessels. A number of risk factors, including
high blood total cholesterol, high low density lipoprotein (LDL) cholesterol levels, and high
blood pressure, are associated with increased risk of developing CHD.
22
CHD is a disease as
defined in 21 CFR 101.14(a)(5) because it involves damage to the heart and coronary arteries
such that they do not function properly.
23
C. Safety Review
Under 21 CFR 101.14(b)(3)(ii), if the substance that is the subject of a health claim is to be
consumed at other than decreased dietary levels, the substance must be a food, a food ingredient,
or a component of a food ingredient whose use at levels necessary to justify a claim has been
demonstrated by the proponent of the claim, to FDA's satisfaction, to be safe and lawful under
the applicable food safety provisions of the Act.
For conventional foods, evaluating whether a substance is “safe and lawful” involves considering
whether the substance (or, for substances that are inherent components of an ingredient, the
ingredient containing the substance) is generally recognized as safe (GRAS), approved as a food
18
See NHLBI, “Your Guide to Lowering Blood Pressure”
[https://www.nhlbi.nih.gov/files/docs/public/heart/hbp_low.pdf
], pp. 2-3 (accessed August 3, 2018).
19
See NHLBI, “High Blood Pressure[https://www.nhlbi.nih.gov/health-topics/high-blood-pressure] (accessed
August 3, 2018).
20
See supra, note 18.
21
See supra, notes 18-19.
22
See NHLBI, “Ischemic Heart Disease” [https://www.nhlbi.nih.gov/health-topics/ischemic-heart-disease]
(accessed May 17, 2019).
23
See supra, note 22.
7
additive, or authorized by a prior sanction issued by FDA (21 CFR 101.70(f)). Dietary
ingredients
24
in dietary supplements, however, are not subject to the food additive provisions of
the Act (see section 201(s)(6) of the Act (21 U.S.C. § 321(s)(6)). Rather, they are subject to the
adulteration provisions in section 402 of the Act (21 U.S.C. 342). The applicable adulteration
provisions of the Act require, for example, that the dietary ingredient not present a significant or
unreasonable risk of illness or injury under conditions of use recommended or suggested in
labeling or, if no conditions of use are suggested or recommended in the labeling, under ordinary
conditions of use (section 402(f)(1)(A) of the Act (21 U.S.C. 342(f)(1)(A))). Further, a dietary
supplement must not contain a poisonous or deleterious substance which may render the
supplement injurious to health under the conditions of use recommended or suggested in the
labeling (section 402(f)(1)(D) of the Act (21 U.S.C. 342(f)(1)(D))). Dietary ingredients that
were not marketed in the United States before October 15, 1994, are also subject to the new
dietary ingredient requirements in section 413 of the Act (21 U.S.C. 350b) and the corresponding
adulteration provision in section 402(f)(1)(B) of the Act (21 U.S.C. 342(f)(1)(B)).
EPA and DHA in Conventional Foods
FDA has affirmed menhaden oil as GRAS with specific limitations on use to ensure that the total
daily intake of EPA and DHA from conventional foods does not exceed 3 grams per person per
day (g/p/d) (62 FR 30751; June 5, 1997) (codified at 21 CFR 184.1472). EPA and DHA are the
major omega-3 fatty acids in fish oil and together comprise about 20 percent by weight of
menhaden oil. FDA established maximum use levels of menhaden oil in certain food categories
because of concerns over possible adverse effects of fish oil consumption on bleeding time,
glycemic control, and LDL cholesterol (62 FR 30751 at 30757; June 5, 1997). In 2005, FDA
published a final rule amending the GRAS affirmation regulation to reallocate the uses of
menhaden oil in conventional food, while maintaining the total daily intake of EPA and DHA
from menhaden oil at a level not exceeding 3 g/p/d (70 FR 14531; March 23, 2005) (codified at
21 CFR 184.1472). FDA placed specific limitations on the use of menhaden oil in food,
including level of use, food categories, and the functional use of the ingredient, to ensure that the
consumption of EPA and DHA from conventional food sources would not exceed 3 g/p/d. To
further ensure that total intake of EPA and DHA from conventional food sources does not exceed
3 g/p/d, FDA also added a requirement that menhaden oil not be used as a food ingredient in
combination with any other added oil that is a significant source of EPA and DHA.
FDA has received several GRAS notifications for food uses of other oils (e.g., algal oils and fish
oils from species other than menhaden) that are also sources of EPA and/or DHA (GRAS Notice
Nos. GRN000097, GRN000102, GRN000105, GRN000109, GRN000137, GRN000138,
GRN000146, GRN000193, GRN000200, GRN000279, GRN000319, GRN000355, and
GRN000553).
25
The GRAS notices for these oils proposed maximum use levels designed to
keep total intake levels of EPA and DHA within the 3 g/p/d limit established in the regulation
24
The term “dietary ingredient” is defined in section 201(ff)(1) of the Act (21 U.S.C. 321(ff)(1)) and includes
vitamins; minerals; herbs and other botanicals; dietary substances for use by man to supplement the diet by
increasing the total daily intake; and concentrates, metabolites, constituents, extracts, and combinations of the
preceding types of ingredients.
25
See FDA’s inventory of GRAS notices, available through links at https://www.fda.gov/food/generally-recognized-
safe-gras/gras-notice-inventory.
8
specifying conditions under which menhaden oil is GRAS for use as a direct human food
ingredient (21 CFR 184.1472). FDA responded without objection to these notifications.
Based on the data and information that FDA considered, which includes data and information
that FDA relied upon in reaching its conclusions about the safety of EPA and DHA omega-3
fatty acids in the menhaden oil rulemakings, current usual intakes of the American general
population, and the agency’s recent review of adverse event reports and other scientific
information for possible safety concerns, FDA concludes that the use of EPA and DHA, and
sources of these substances -- such as menhaden oil, other fish oils, and algal oils -- as
ingredients in conventional food is safe and lawful under 21 CFR 101.14(b)(3)(ii), provided that
such use is consistent with FDA’s GRAS regulation for menhaden oil (21 CFR 184.1472) and
the conditions of use specified in other relevant GRAS notifications to which FDA did not
object, including use limitations designed to ensure that daily intakes of EPA and DHA omega-3
fatty acids from conventional food sources do not exceed 3.0 g/day.
Current Dietary Intake Levels
Although some currently marketed dietary supplements provide a combined EPA and DHA
intake of over 8 g/ day when taken as recommended in their labeling, current intake data indicate
that for most of the population, total daily EPA and DHA intake is much lower. Data from
NHANES 2009-2014 indicate that the mean total usual intake of EPA and DHA from
conventional foods and dietary supplements for people age 4 years and older is 77 mg/day.
26
EPA and DHA in Dietary Supplements
Under the Dietary Supplement Health and Education Act of 1994, dietary supplements that are
not adulterated, misbranded, or otherwise in violation of applicable laws and regulations are
lawful for sale in the United States. There is no current regulatory limit on the amount of EPA
and DHA in dietary supplements.
In 2004, FDA considered the safety of EPA and DHA in dietary supplements as part of the
agency’s review of two petitions seeking qualified health claims about omega-3 fatty acids and
reduced risk of coronary heart disease. Based in large part on the scientific evaluation in the
1997 GRAS affirmation final rule for the use of menhaden oil as a conventional food ingredient,
the agency concluded that the use of EPA and DHA in dietary supplements is safe and lawful
under 21 CFR 101.14(b)(3)(ii), provided that total daily intake of EPA and DHA from
conventional food and dietary supplement sources does not exceed 3 g/day. As discussed earlier
in this section, the 1997 GRAS affirmation final rule for menhaden oil concluded, based on an
evaluation of the scientific literature available at that time, that intake of EPA and DHA over 3
g/day was not generally recognized as safe because of evidence that intake at such levels might
lengthen bleeding time, increase LDL cholesterol, and adversely affect glycemic control in
people with type 2 diabetes.
26
See “Memorandum to the File (Docket No. FDA-2014-Q-1146): Consumption estimates of EPA and DHA for
individuals aged 4 years and older in the United States.” (August 28, 2018).
9
Currently, dietary supplements are marketed with combined EPA and DHA content ranging from
less than a milligram to 8 g/day. To evaluate whether dietary supplements at higher levels of
EPA and DHA intake are safe and lawful, FDA has updated its safety review to examine more
recent evidence regarding the effects of consuming EPA and DHA at levels above 3 g/day on
glycemic control, blood cholesterol, and risk of excessive bleeding. After searching FDA’s
adverse event report database and evaluating relevant studies on EPA and DHA, we concluded
that EPA and DHA intake up to 10 g/day appears to have no adverse effect on glycemic control,
as measured by fasting blood glucose and HbA1c.
27
We further concluded that EPA and DHA
intake does not appear to raise total cholesterol.
28
There is some evidence that EPA and DHA
intake may raise LDL-cholesterol by approximately 2-4 mg/dL, but the studies are inconsistent,
and there is also supportive, but not conclusive, evidence that EPA and DHA may reduce the risk
of CHD by other mechanisms.
29
With regard to bleeding risk, we concluded that consumption of
less than or equal to 5 grams/day of EPA and DHA does not increase the risk of excessive
bleeding based on clinical trials that measured clinical outcomes such as bleeding episodes and
blood loss.
30
Therefore, we conclude that dietary supplements that provide no more than 5 g/day
EPA and DHA when used as recommended or suggested in their labeling are safe and lawful
under 21 CFR 101.14(b)(3)(ii).
Above the 5 g/day level, however, there is not enough scientific evidence to draw a conclusion.
Specifically, we could not draw any conclusions about bleeding risk associated with EPA and
DHA intake at levels greater than 5 g/day because the published studies of the effects of EPA
and DHA on risk of excessive bleeding did not evaluate doses at such higher intake levels.
Because the current scientific evidence does not show that dietary supplements that provide more
than 5 g/day EPA and DHA are adulterated under the food safety provisions of the Federal Food,
Drug, and Cosmetic Act, such products are lawful. Due to the lack of scientific evidence about
risk of excessive bleeding at intakes of more than 5 g/day, however, we are unable to determine
whether these products are safe within the meaning of the “safe and lawful” requirement for
conventional foods and dietary supplement to bear a health claim (21 CFR 101.14(b)(3)(ii)).
Because of the uncertainty about possible adverse effects on bleeding risk from consuming more
than 5 grams EPA and DHA per day, FDA intends to consider, as a factor in the exercise of its
enforcement discretion for any EPA/DHA qualified health claim, that dietary supplements
bearing the qualified health claim provide no more than 5 g/day of EPA and DHA when used as
labeled. In section IV, FDA describes this and other factors it plans to consider in the exercise of
enforcement discretion for EPA/DHA qualified health claims.
27
See Memorandum to Docket No. FDA-2014-Q-1146: Survey of Comprehensive Reviews of the Effects of EPA
and DHA Intake on Glycemic Control and Blood Cholesterol (June 17, 2019).
28
Id.
29
Id.
30
See Memorandum to Docket No. FDA-2014-Q-1146: Review of Scientific Literature on EPA and DHA Intake
and Risk of Excessive Bleeding (June 17, 2019).
10
II. The Agency’s Consideration of a Qualified Health Claim
The evidence included in the petition was for lowering blood pressure and reduced risk of
hypertension. Lowering blood pressure is one of many mechanisms for lowering the risk of
CHD. FDA and NIH have identified diastolic blood pressure (DBP) and systolic blood pressure
(SBP) as surrogate endpoints for predicting risk of hypertension.
31
SBP and DBP are also
surrogate endpoints for risk of CHD.
32
Since either elevated SBP (≥ 140mmHg) or DBP (≥ 90
mmHg) can be used to diagnose hypertension, the reduction of either is beneficial in reducing the
risk of hypertension (James et al., 2014; NHLBI, 2003). For the purposes of this review, the
agency evaluated only studies that measured SBP, DBP, or incidence of hypertension. FDA did
not evaluate the totality of the evidence for effects of EPA and DHA intake on other surrogate
endpoints for CHD such as blood (serum or plasma) total cholesterol, blood (serum or plasma)
LDL cholesterol, or other mechanisms of CHD risk. Rather, FDA evaluated the totality of the
evidence for combined intake of EPA and DHA, which the petition describes as the substances
that are the subject of the claim, and reducing the risk of hypertension and CHD through
lowering SBP or DBP.
The petition cited 717 publications as evidence to substantiate the relationship for the proposed
claims. These publications consisted of: 21 government documents; 7 reports, 29 review articles,
15 meta-analyses, 8 letters, 47 abstracts, 1 book chapter; 46 articles written in a foreign language
without an English translation; 286 publications that evaluated the effect of intake of EPA, DHA,
or both on blood pressure or risk of hypertension; and 260 publications describing studies that
evaluated other substance-disease relationships or did not evaluate any substance-disease
relationship.
33
The 286 publications evaluating the effect of EPA and DHA intake on blood
pressure or risk of hypertension described a total of 277 studies, 245 intervention studies
(discussed in Section II.B) and 32 observational studies (discussed in Section II.C). Comments
on the petition cited 30 additional publications that were not already included in the petition. Of
these 30 publications, there were 3 meta-analyses, 14 review articles, 5 books or book chapters, 1
letter, 2 government documents, and 3 other publications that did not evaluate the substance-
disease relationship.
34
The remaining 2 publications (O'Sullivan et al., 2012; Skilton et al., 2013)
were reports of observational studies that evaluated the relationship between EPA and DHA
intake and blood pressure or risk of hypertension (discussed in Section II.C). In addition to the
studies cited in the petition or in comments, FDA identified thirty publications
35
describing
31
FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource [Internet].
Silver Spring (MD): Food and Drug Administration (US); 2016-. Validated Surrogate Endpoint. 2017 Sep.
[https://www.ncbi.nlm.nih.gov/books/NBK453484/] (accessed August 3, 2018). see supra, note 7 [Section III.C].
32
Id.
33
See “Memorandum to File (Docket No. FDA-2014-Q-1146): List of Publications Excluded from Review” (June
17, 2019).
34
Id.
35
Albert et al., 2015; Alfaddagh et al., 2017;Ansari et al., 2017; Augustine et al., 2014; Barbosa et al., 2017; Bosch
et al., 2012; Carvalho et al., 2014; Chan et al., 2016a; Chan et al., 2016b; Clark et al., 2016; Clark et al., 2018;
Elajami et al., 2017; Grieger et al., 2014; Harslof et al., 2014; Hlais et al., 2013; Kristensen et al., 2016; Kromhout et
al., 2010; Lind et al., 2015; Logan and Spriet et al., 2015; Minihane et al., 2016; Moriyama et al., 2018; Nodari et
11
twenty-eight relevant intervention studies through a literature search for studies evaluating the
relationship between EPA and DHA intake and blood pressure or risk of hypertension.
A. Assessment of Review Articles, Meta-Analysis, Book Chapters, Letters, and
Government Reports and Other Reports
Although useful for background information, review articles, meta-analyses, book chapters,
letters, and government reports do not contain sufficient information on the individual studies
reviewed and, therefore, FDA could not draw any scientific conclusions from these sources.
FDA could not determine factors such as the study population characteristics or the composition
of the products used (e.g., conventional food, dietary supplement). Similarly, the lack of detailed
information on studies summarized in review articles, meta-analyses, book chapters, letters, and
government reports prevents the agency from determining whether the studies are flawed in
critical elements such as design, conduct of studies, and data analysis. FDA must be able to
review the critical elements of a study to determine whether any scientific conclusions can be
drawn from it. As a result, the review articles, meta-analyses, book chapter, letters, and
government reports supplied by the comments and in the petition did not provide information
from which scientific conclusions can be drawn regarding the substance-disease relationship
claimed by the petitioner.
B. Assessment of Intervention Studies
FDA evaluated 284 publications reporting on 273 intervention studies that represented three
bodies of evidence related to the proposed health claims. These publications included studies
investigating the relationship between (1) EPA intake and blood pressure or risk of hypertension;
(2) DHA intake and blood pressure or risk of hypertension; and (3) intake of EPA and DHA
combined and blood pressure or risk of hypertension.
Of these 273 intervention studies, conclusions could not be drawn from 169 for the reasons
discussed below.
36
In 24 studies, subjects consumed either only EPA
37
(15 studies) or only DHA
38
(9 studies).
There were also three studies that contained both an EPA-only group and a DHA-only group, but
al., 2011; Pase et al., 2015; Roncaglioni et al., 2013; Root et al., 2013; Soares et al., 2014; Su et al., 2015; Tavazzi et
al., 2008; Witte et al., 2014; Wu et al., 2014.
36
This section contains a general discussion of major flaws in the reports of intervention studies from which
scientific conclusions could not be drawn. Such studies may have other flaws in addition to those specifically
mentioned.
37
Croset et al., 1990; Dokholyan et al., 2012; Hagiwara et al., 2011; Haiden et al., 2012; Hall et al., 2008; Hamazaki
et al., 1990; Harris et al., 2008; Iketani et al., 2013; Miyajima et al., 2001; Moriyama et al., 2013; Satoh et al., 2009;
Shimizu et al., 1995; Takaki et al., 2011; Yamakawa et al., 2012; Yoshimura et al., 1987.
38
Geppert et al., 2006; Harrison et al., 2004; Kelley et al., 2007; Neff et al., 2011; Sagara et al., 2011; Sanders et al.,
2006; Stark et al., 2004; Surai et al., 2000; Theobald et al., 2007.
12
no group that consumed EPA and DHA combined.
39
Scientific conclusions about whether
consuming EPA and DHA combined reduces the risk of hypertension cannot be drawn from
these 27 studies because they were designed to test the independent effects of EPA or DHA on
blood pressure, and not to test the effects of these two nutrients in combination. The
physiological effects of a substance when consumed as a single nutrient may be different than its
effects when consumed with other nutrients (IOM, 2005c; Sempos et al., 1999). Scientific
conclusions about the substance-disease relationship cannot be drawn from studies that did not
analyze whether intake of EPA and DHA combined is associated with risk of hypertension.
Therefore, the 27 studies that evaluated intake of EPA or DHA as a single nutrient were
eliminated from further review.
40
Fifty-nine studies did not include a control group or other appropriate control to distinguish the
effects of EPA and DHA on blood pressure from other factors that could affect blood pressure.
41
Without an appropriate control group, it cannot be determined if the changes in SBP or DBP
were due to EPA and DHA intake or uncontrolled extraneous factors. Thus, scientific
conclusions could not be drawn from these studies.
Twenty-nine studies
42
did not conduct statistical analysis to ensure an accurate comparison of
results between the control and treatment groups. Statistical analysis is a critical factor in
evaluating evidence for the substance-disease relationship because it provides the comparison
between subjects consuming EPA and DHA and those not consuming EPA and DHA (i.e.,
39
Three studies (Mori et al., 1999, Grimsgaard et al., 1998, and Rontoyanni et al., 2012) had both EPA-only and
DHA-only arms. The results of Mori et al., 1999, were also discussed in Woodman et al., 2002, Mori et al., 2003,
and Mas et al., 2010.
40
Thirteen of the 27 intervention studies that measured EPA only or DHA only contained other major flaws. For
example, 10 studies did not include a control group or other appropriate control (Dokholyan et al., 2012; Hagiwara
et al., 2011; Haiden et al., 2012; Hall et al., 2008; Iketani et al., 2013; Neff et al., 2011; Rontoyanni et al., 2012;
Surai et al., 2000; Yamakawa et al., 2012; Yoshimura et al., 1987). One study (Moriyama et al., 2013) did not report
a validated surrogate endpoint. Two studies (Croset et al., 1990; Hamazaki et al., 1990) did not conduct statistical
analysis to ensure an accurate comparison of results between the control and treatment groups.
41
Agouridis et al., 2012; Benito et al., 2006; Bhise et al., 2005; Bitzur et al., 2010; Caron-Dorval et al., 2008; Celik
et al., 2008; Chin and Dart, 1994; Cicero et al.,2010; Clark et al., 2016; Clark et al., 2018; Damsgaard et al., 2008;
Dangardt et al., 2010; De Caterina et al., 1993; de Jong et al., 2011; Deferne and Leeds, 1992; Delgado-Pando et al.,
2014; Dixit et al., 2004; Doenyas-Barak et al., 2012; Ernst et al., 1988; Ernst et al., 1991; Farrell et al., 1998; Fiedler
et al., 2005; Geppert et al., 2008; Grossman et al., 1993; Hackman et al., 2006; Hamazaki et al., 1984; Kappus et al.,
2011; Kasim et al., 1988; Kho et al., 1989; Laidlaw and Holub, 2003; Leng et al., 1998; Lok et al., 2012; Madden et
al., 2007; Manor et al., 2013; Olivieri et al., 1988; Parinyasiri et al., 2004; Rolf et al., 1990; Rose and Holub, 2006;
Russo et al., 1995; Sanders and Hinds, 1992; Sanders et al., 1981; Santos et al., 2000; Schmidt et al., 1990; Schmidt
et al., 1992; Simpson et al., 1991; Singer et al., 1983; Singer et al., 1984a; Singer et al., 1985a; Singer et al., 1985b;
Singer et al., 1985c; Singer et al., 1990a; Singer et al., 1991; Singer et al., 1993; Surai et al., 2000; Svensson et al.,
2004; Vakhapova et al., 2011;Valdivielso et al., 2009; Weisser et al., 1990; Yam et al, 2002; Yosefy et al., 1999.
42
Bach et al., 1989; Bennett et al., 1995; Chan et al., 2002; Davidson et al., 1989; Dusing et al., 1990; Ebrahimi et
al., 2009; Freund-Levi et al., 2006; Gans et al., 1990; Grundt et al., 1995; Haglund et al., 1990; Haines et al., 1986;
Hellsten et al., 1993; Kristensen et al., 1987; Mackness et al., 1994; Nodari et al., 2011; Oldendzki et al., 2011;
Passfall et al., 1993; Prisco et al., 1998; Sakamoto et al., 2000; Schaefer et al., 1996; Simao et al., 2010; Simao et al.,
2012; Singer et al., 1984b; Singer et al., 1986a; Singer et al., 1986b; Szabo de Edelenyi et al., 2012; Vericel et al.,
1999; Witte et al., 2014; Yosefy et al., 1996.
13
control group) to determine whether the study showed a reduction in blood pressure or risk of
hypertension. Hence, scientific conclusions could not be drawn from these studies.
Twenty-two studies
43
did not measure incidence of hypertension or a validated surrogate
endpoint of hypertension (SBP or DBP).
44
Because these studies did not measure incidence of
hypertension or a validated surrogate endpoint of hypertension, no scientific conclusions about
the relationship between EPA and DHA consumption and blood pressure or risk of hypertension
could be drawn from them.
For two studies, the baseline blood pressure values were very different between the fish oil and
control groups (Andreassen et al., 1997; Mills et al., 1989). When baseline values are
significantly different between groups in a parallel study, this difference must be accounted for
in the statistical analysis; otherwise, it is not possible to determine if differences between groups
at the end of the study were due to the intervention or to the differences that already existed at
the beginning of the study. Because this study did not properly adjust for the difference in
baseline values, an accurate comparison of the results between the two groups is not possible.
Thus, no scientific conclusions about a relationship between EPA and DHA and blood pressure
or risk of hypertension could be drawn from these studies.
Two studies exposed subjects to experimental conditions that do not normally occur in everyday
life and therefore are not relevant to the U.S. population (lower body negative pressure, cold
pressor test) (Mills et al., 1990; Hughes et al., 1991). Because these studies do not represent
real-world conditions, scientific conclusions about the relationship between EPA and DHA
consumption and blood pressure or risk of hypertension could not be drawn from them.
To determine the independent effects of EPA and DHA in combination on blood pressure,
intervention studies must control for the amount of sodium and potassium in the diet because
both of these nutrients influence blood pressure. Adding EPA and DHA to the usual diet in the
form of dietary supplements should not result in significant changes in the amount of sodium and
potassium that subjects consume; thus, we did not eliminate such intervention studies if they did
not report the levels of sodium and potassium consumed by the subjects. However, twenty-eight
intervention studies
45
had designs that could lead to significant changes in the amount of sodium
43
Alexopoulos et al., 2004; Augustine et al., 2014; Badalamenti et al., 1997; Badalamenti et al., 1995; Bilo et al.,
1988; Branten et al, 2002; Carvalho et al., 2014; de Fijter et al., 1995; Delarue et al., 2003; Lahoz et al., 1999;
Larnkjaer et al., 2006; Matsumura et al., 2012; McVeigh et al, 1993; Nishizawa et al., 2006; Shah et al., 2007; Su et
al., 2015; van der Heide et al., 1990; van der Heide et al., 1992; van der Heide et al., 1993; Ventura et al. 1993;
Walser et al., 2006; Walser et al., 2008.
44
See FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource [Internet].
Silver Spring (MD): Food and Drug Administration (US); 2016-. Validated Surrogate Endpoint. 2017 Sep.
[https://www.ncbi.nlm.nih.gov/books/NBK453484/] (accessed August 3, 2018). See also Rasnake CM, Trumbo PR,
Heinonen TM. Surrogate endpoints and emerging surrogate endpoints for risk reduction of cardiovascular disease.
Nutr Rev 2008;66(2):76-81. See also Desai M, Stockbridge N, Temple R. Blood pressure as an example of a
biomarker that functions as a surrogate. AAPS J 2006;8(1):E146-152.
45
Derosa et al., 2009; Derosa et al., 2012; Donadio et al., 1994; Donadio et al., 1999; Dyerberg et al., 2004;
Dyerberg et al., 2006 ;Erkkila et al., 2008; Fahs et al., 2010; Grieger et al., 2014; Gulseth et al., 2010; Gustafsson et
al., 1996; Hallund et al., 2010; Harslof et al., 2014; Jain et al., 2002; Lind et al., 2015; Lindqvist et al., 2009;
14
and potassium consumed
46
but did not properly control for these nutrients and/or did not report
the levels of these nutrients in the subjects’ diets. Therefore, these studies were eliminated from
further review because scientific conclusions about the relationship between EPA and DHA
consumption and blood pressure or risk of hypertension could not be drawn from them.
After the elimination of the studies discussed above, 104 intervention studies (112 publications)
remained from which scientific conclusions could be drawn about the relationship between EPA
and DHA intake and blood pressure or risk of hypertension. Those studies are discussed below
by category.
For the purposes of FDA’s review, studies that measured blood pressure were sorted into two
categories: 1) subjects with normal blood pressure or pre-hypertension (SBP≤139 mm Hg or
DBP ≤ 89 mm Hg) and 2) subjects with hypertension (SBP ≥ 140 mm Hg or DBP ≥ 90 mm Hg).
Because the mechanisms by which EPA and DHA may reduce blood pressure in normotensive
and hypertensive subjects are considered to be the same (Borghi and Cicero, 2006; Mozaffarian
et al., 2011), studies in hypertensive subjects were included in this review. Studies were further
sorted into three categories based on the type of intervention: 1) studies in which subjects
consumed EPA and DHA from fish oil supplements, prescription drugs, or foods enriched with
EPA and DHA, but otherwise followed their usual diets; 2) studies in which subjects consumed
EPA and DHA in fish oil supplements or foods enriched with EPA and DHA and made other
changes in their diets (e.g. sodium restriction, substitution of fish oil for other food items); and 3)
studies in which subjects consumed fatty fish as the source of EPA and DHA.
1. Studies that added prescription drugs containing highly purified fish oil to the diets of
normal or pre-hypertensive subjects (SBP 139 mmHg or DBP 89 mmHg)
Lungershausen et al. (1994)
47
was a moderate quality randomized, double-blind, placebo
controlled cross-over
48
study in which 42 Australian subjects consumed their usual diets plus
either 4 g/day corn oil (control) or 4 g/day of Omacor (1.9 g/day EPA and 1.5 g/day DHA; 3.4
g/day total) for six weeks each period. SBP and DBP were significantly lower (P=0.012 and
P=0.006, respectively)
49
with consumption of Omacor compared to the control.
Holman et al. (2009) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 732 British subjects with type II diabetes consumed 1) 2 g/day olive oil
Margolin et al., 1991; Moriyama et al., 2018; Murphy et al., 2007; Ness et al., 1999; Oh et al., 1991; Paniagua et al.,
2011; Rasmussen et al., 2006; Rossing et al., 1996; Sirtori et al., 1997; Soares et al., 2014; Von Houwelingen et al.,
1987; Wong et al., 2013.
46
The types of studies with designs that could lead to changes in levels of sodium and potassium consumed
included: 1) studies in which the subjects consumed EPA and DHA in fish oil supplements or EPA and DHA-
enriched foods and made other changes in their diets (e.g., sodium restriction, substitution of fish oil for other food
items); 2) studies in which the subjects consumed fatty fish as the source of EPA and DHA.
47
The results of this study were also discussed in Lungershausen and Howe, 1994.
48
A cross-over design study involves all subjects crossing over from the intervention group to the control group, and
vice versa, after a defined time period.
49
For the outcome of a study to demonstrate a statistically significant difference between groups, P (probability
value) must be < 0.05. See supra, note 7 [Section III.F].
15
(control) (n=178), 2) 2 g/day Omacor (920 mg/day EPA and 760 mg/day DHA; 1.7 g/day total)
(n=183), 3) 2 g/day olive oil plus atorvastatin (Lipitor) (control) (n=183), or 4) atorvastatin
(Lipitor) plus 2g/day Omacor (920 mg/day EPA and 760 m/day DHA; 1.7 g/day total) (n=188)
in addition to their usual diets for four months. There was no significant difference in SBP or
DBP between the olive oil control group and the Omacor group. Additionally, there was no
significant difference in SBP or DBP between the Lipitor plus olive oil control group and the
Omacor group.
Mori et al. (2009) was a high quality randomized, double-blind, placebo controlled parallel trial
in which 74 Australian subjects with chronic kidney disease consumed 1) 4 g/day olive oil
(control) (n=15), 2) 4 g/day Omacor (1.8 g/day EPA and 1.5 g/day DHA; 3.4 g/day total)
(n=20), 3) 4 g/day Omacor (1.8 g/day EPA and 1.5 g/day DHA; 3.4 g/day total) plus coenzyme
Q (200 mg/day) (n=18), or 4) coenzyme Q (200 mg/day) (control) (n=21) in addition to their
usual diets for eight weeks. There was no significant difference in SBP or DBP between the
olive oil control group and Omacor group. However, SBP and DBP were significantly lower
(P<0.001) in the Omacor plus coenzyme Q group compared to the coenzyme Q control group.
Skulas-Ray et al. (2012) was a moderate quality randomized, double-blind, placebo controlled
crossover trial in which 26 Australian subjects consumed 1) a corn oil control, 2) 1 g/day Lovaza
(465 mg/day EPA and 375 mg/day DHA, 850 mg/day total) or 3) 4 g/day Lovaza (1.8 g/day
EPA, 1.5 g/day DHA, 3.4 g/day total) for 8 weeks each period, in addition to their usual diets.
Blood pressure was measured at rest and during a task that required subjects to prepare for two
minutes and then deliver a 3-minute speech about a hypothetical situation. Subjects also
completed a foot cold pressor task in which they immersed one foot in 39 º F water for 2.5
minutes. There was no significant difference in SBP or DBP between the 850 mg/day EPA and
DHA period and the placebo period at rest and during the speech task. There was also no
significant difference in SBP or DBP between the 3.4 g/day total EPA and DHA period and the
placebo period during rest or during the speech task. FDA did not evaluate the data from the
foot cold pressor task because it does not reflect normal conditions that are relevant to the U.S.
population. For the same reason, we did not evaluate data with regard to overall treatment effect
because those data included blood pressure measurements from the foot cold pressor task.
Nordoy et al. (2001) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 42 Norwegian subjects consumed 2 g/day corn oil plus 10 mg/day
atorvastatin (n=20) (control) in addition to a low-fat diet or 2 g/day Omacor (900 mg/day EPA
and 780 mg/day DHA; 1.7 g/day total) plus 10 mg/day atorvastatin (n=22) in addition to a low-
fat diet (30 percent total fat) for 5 weeks. SBP was significantly lower (P=0.03) in the Omacor
group as compared to the control group. There was no significant difference in DBP between the
two groups.
Kim et al. (2011) was a moderate quality randomized, open label controlled parallel trial in
which 61 Korean subjects consumed 20 mg/day simvastatin (Zocor) (n=31) (control) or 4 g/day
Omacor (1.9 g/day EPA and 1.5 g/day DHA; 3.4 g/day total) plus 20 mg/day simvastatin (Zocor)
(n=30) for 6 weeks. All subjects also followed a low cholesterol diet. There was no significant
16
difference in SBP or DBP between the control group and the Omacor plus simvastatin (Zocor)
group.
Studies in subjects with CVD
Elajami et al. (2017)
50
was a moderate quality randomized, open-label controlled parallel trial in
which 262 American subjects with coronary artery disease (CAD) consumed their usual diet
(control group) (n=128) or 4 g/day Lovaza (1.8 g EPA and 1.5 g DHA, 3.3 g/day total) (n=134)
in addition to their usual diets for 1 year. There was no significant difference in SBP or DBP
between the control group and the Lovaza group.
Chan et al. (2016a)
51
was a moderate quality randomized, single-blind, open-label controlled
cross-over study in which 20 Australian subjects (4 subjects had CAD) on statins (with or
without ezetimibe) consumed a standardized diet (44 percent carbohydrates, 33 percent fat, 20
percent protein and 278 mg cholesterol) (control) or 4 g/day Omacor (1.8 g EPA and 1.52 g
DHA, 3.32 g/day total) in addition to the standardized diet for 8 weeks each period. SBP and
DBP were significantly lower (P=0.01) with consumption of Omacor compared to the control
period.
2. Studies that added prescription drugs containing highly purified fish oil to the diets of
subjects with hypertension (SBP 140 mmHg or DBP90 mmHg)
Pettersson et al. (1994) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 32 Swedish subjects with immunoglobulin A (IgA) nephropathy and
proteinuria consumed 6 g/day corn oil (control) (n=17) or 6 g/day Omacor (3.3 g/day EPA and
1.8 g/day DHA; 5.1 g/day total) (n=17) for 6 months. There was no significant difference in
SBP or DBP between the corn oil control group and the Omacor group.
Toft et al. (1995) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 78 Norwegian subjects consumed 4 g/day corn oil (control) (n=40) or 4 g/day
Omacor (2.0 g/day EPA and 1.4 g/day DHA, 3.4 g/day total) (n=38) in addition to their usual
diets for sixteen weeks. SBP was significantly lower (P=0.04) in the Omacor group compared to
the corn oil control group. There was no significant difference in DBP between groups.
Lungershausen et al. (1997) was a moderate quality randomized, double-blind, placebo
controlled parallel trial in which 32 Australian subjects with insulin-dependent diabetes mellitus
(IDDM) or noninsulin–dependent diabetes mellitus (NIDDM) consumed 4 g/day corn oil
(control) (n=16) or 4 g/day Omacor (2 g/day EPA and 1.4 g/day DHA; 3.4 g/day total) (n=16) in
addition to their usual diets for 12 weeks. SBP was significantly lower (P=0.039) in the Omacor
50
The results of this study were also discussed in Alfaddagh et al., 2017.
51
The results of this study were also discussed in Chan et al., 2016b.
17
group compared to the corn oil placebo control group. There was no significant difference in
DBP between the control and Omacor groups.
Bonaa et al. (1990) was a moderate quality randomized, double-blind, controlled parallel trial in
which 156 Norwegian subjects consumed 6 g/day corn oil control (n=78) or 6 g/day Omacor (3.3
g/day EPA and 1.8 g/day DHA; 5.1 g/day total) (n=78) in addition to their usual diets for 10
weeks. Sitting SBP was significantly lower (P=0.002) in the Omacor group compared to the
corn oil control group. Sitting DBP was also significantly lower (P=0.011) in the Omacor group
compared to the corn oil control group. Standing SBP and DBP were also significantly lower
(P=0.002 and P=0.0001) in the Omacor group compared to the control group.
Studies in subjects with CVD
Bosch et al. (2012) was a moderate quality randomized, double-blind, placebo controlled parallel
study in which 12,536 Canadian subjects who had or were at high risk for diabetes and at high
risk for CVD events (history of myocardial infarction (MI) , stroke, or revascularization)
consumed 1 g/day olive oil (n=6,281) or 1 g/day Omacor (465 mg EPA and 375 mg DHA, 840
mg/day total) (n=6,255) in addition to their usual diets for 6.2 years. There was no significant
difference in SBP or DBP between the control group and the Omacor group.
Holm et al. (2001) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 41 Norwegian subjects with a heart transplant consumed 4 g/day corn oil (control)
(n=20) or 4 g/day Omacor (1.86 g/day EPA and 1.5 g/day DHA; 3.4 g/day total) (n=21) in
addition to their usual diets for one year. The mean change in 24 hour SBP was significantly
different (P=0.02) in the fish oil group compared to the corn oil control group, resulting in lower
SBP in the EPA and DHA group compared to the control. However, there was no significant
difference in the mean change in 24 hour DBP between the fish oil group and the control group.
3. Studies that added fish oil supplements to the diets of normal or pre-hypertensive subjects
(SBP 139 mmHg or DBP 89 mmHg)
Ansari et al. (2017) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 43 Iranian subjects with type 2 diabetes consumed 3 capsules of paraffin
(control) (n=22) or 3.8 g/day fish oil (1,800 mg EPA and 900 mg DHA, 2.7 g/day total) (n=21)
in addition to their usual diets for 10 weeks. There was no significant difference in SBP with
consumption of fish oil as compared to the control. However, DBP was significantly lower
(P=0.040) in the fish oil group compared to the control.
Barbosa et al. (2017) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 80 Brazilian subjects consumed 3 g/day sunflower oil (control) (n=40) or 3
g/day fish oil (1.1 g/day EPA, 690 mg/day DHA, 1.8 g/day) (n=40) in addition to their usual diet
for 2 months. There was no significant difference in SBP or DBP between the control group and
the fish oil group.
18
Kristensen et al. (2016) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 114 Danish subjects with psoriatic arthritis consumed 3 g/day olive oil
(control) (n=56) or 3 g/day n-3 fatty acids from fish oil (~1.5 g EPA and ~1.5 g DHA, ~3 g/day)
(n=58) in addition to their usual diets for 24 weeks. There was no significant difference in SBP
or DBP between the control group and the fish oil group.
Albert et al. (2015) was a moderate quality randomized, double-blind, controlled cross-over
study in which 47 New Zealander subjects consumed 5 g/day canola oil (control) or 5 g/day
mixture of krill (88%) and salmon oil (12%) (230 mg EPA, 154 mg DHA, 384 mg/day total) in
addition to their usual diets for 8 weeks each period There was no significant difference in SBP
or DBP between the control group and the krill and salmon oil group.
Logan and Spriet (2015) was a moderate quality randomized, single-blind, placebo controlled
parallel trial in which 24 Canadian women consumed either a standard diet (50 percent
carbohydrates, 30 percent fat, 20 percent protein) and 3 g/day olive oil (control) (n=12) or a
standard diet and 5 g/day fish oil (2 g EPA, 1 g DHA, 3 g/day total) (n=12) for 12 weeks. There
was no significant difference in SBP or DBP between the control group and the fish oil group.
Pase et al. (2015) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 72 Australian subjects consumed 6 g/day sunola oil (control) (n=32) or 6 g/day fish
oil (480 mg EPA, 480 DHA, 960 mg/day total) (n=38) in addition to their usual diet. There was
no significant difference in SBP or DBP between the control group and any of the fish oil group.
Wu et al. (2014) was a moderate quality randomized, double-blind, placebo controlled cross-over
study in which 84 British subjects at moderate risk of CVD consumed 3 capsules of corn oil
(control) or 3 capsules of fish oil (900 mg EPA, 600 mg DHA, 1.5 g/day) in addition to their
usual diet for 8 weeks each period. Subjects were genotyped for the Asp298 endothelial nitric
oxide synthase (eNOS) polymorphism that has been associated with increased risk of CVD.
Subjects were stratified by gene variants GG (carriers of Glu/Glu) (n=40) and GT/TT (carriers of
Glu/Asp or Asp/Asp) (n=44). There was no significant difference in SBP or DBP between the
control group and the fish oil groups in carriers of the GG or the GT/TT genotype. There was
also no significant difference in SBP or DBP between the control group and the fish oil group in
all subjects.
Hlais et al. (2013) was a moderate quality randomized, single-blind, controlled parallel trial in
which 98 Lebanese subjects consumed 1) 8 g/day sunflower oil (control) (n=20), 2) 2 g/day fish
oil (990 mg EPA, 392 mg DHA, 1.382 g/day total) (n=22), 3) 1 g/day fish oil (495 mg EPA, 196
mg DHA, 691 mg/day) and 8 g/day sunflower oil (n=17), 4) 2 g/day fish oil (990 mg EPA, 392
mg DHA, 1.382 g/day total) 8 g/day sunflower oil (n=18),or 5) 4 g/day fish oil (1.98 g/ EPA, 784
mg DHA, 2.76 g/day total) and 8 g/day sunflower oil (n=21) in addition to their usual diets for
12 weeks. There was no significant difference in SBP or DBP between the control group and any
of the fish oil groups.
19
Minihane et al. (2016) was a randomized, double-blind, placebo-controlled, crossover study in
which 312 British subjects consumed 1) 3.2 g/day of a control oil (80/20 mixture palm oil and
soybean oil), 2) 3.2 g/day fish oil (1.8 g/day EPA and DHA), or 3) 50/50 corn oil/fish oil with
700 mg/day EPA and DHA for 8 weeks each period. In the analysis of data for all subjects
combined, there was no significant difference in SBP and DBP compared to control when
subjects consumed either 3.2 g/day EPA and DHA or 700 mg/day EPA and DHA. The study also
analyzed the data based on baseline hypertension status, dividing the subjects into two groups:
17 subjects who were described as having dual hypertension” (SBP ≥140 mmHg) and DBP ≥90
mmHg) and 31 subjects who had isolated systolic hypertension (SBP ≥140 mmHg but DBP ≤90
mmHg). In the 31 subjects with isolated systolic hypertension, SBP was significantly lower
(P=0.046) compared to control when the subjects consumed 1.8 g/day EPA and DHA or 700
mg/day EPA and DHA. In the 17 subjects with dual hypertension, however, there was no
significant difference in SBP among the three diets. There was also no significant difference in
DBP among the three diets in subjects with isolated systolic hypertension or dual hypertension.
Armstrong et al. (2012) was a moderate quality randomized, double-blind, placebo controlled,
parallel study in which 98 American subjects of African American, Black or African ancestry
consumed 5 g/day of corn oil/ soybean oil placebo (control) (n=46) or 5 g/day fish oil (2 g/day
EPA and 1 g/day DHA; 3 g/day total) (n=49) in addition to their usual diets for six weeks.
Subjects were stratified in both control and EPA and DHA groups based on ALOX5
(Arachidonate 5-lipoxygenase) gene variants that are common in people of African ancestry and
are associated with a differential CVD risk. There was no significant difference in SBP or DBP
between the two groups for all subjects or within genotypes.
Atar et al. (2012) was a moderate quality randomized, double-blind, placebo controlled parallel
study in which 73 Iranian women with type II diabetes consumed 2 g/day cornstarch (control)
(n=34) or 2 g/day fish oil (720 mg/day EPA and 480 mg/day DHA; 1.2 g/day total) (n=39) in
addition to their usual diets for 8 weeks. There was no significant difference in SBP or DBP
between the EPA and DHA group and the cornstarch control group.
Carter et al. (2012) was a moderate quality randomized, double blind, placebo controlled parallel
trial in which 67 normotensive or pre-hypertensive American subjects consumed 9 g/day olive
oil (control) (n=33) or 9 g/day fish oil (1.6 g/day EPA and 1.1 g/day DHA; 2.7 g/day total)
(n=34) for 8 weeks. There was no significant difference in SBP and DBP between the fish oil
group (n=19) and the control group (n=19) in normotensive subjects. Additionally, there was no
significant difference in SBP or DBP between the fish oil group (n=15) and the olive oil control
group (n=14) in the pre-hypertensive subjects.
Faghihi et al. (2012) was a moderate quality randomized, double-blind placebo controlled
parallel trial in which 41 Iranian subjects (with schizophrenia, bipolar, or schizoaffective
disorder on olanzapine plus either sodium valproate or lithium) consumed a placebo control
(n=21) or fish oil (n=20) in addition to their usual diets for 6 weeks. The dose of fish oil was 1
g/day (300 mg/day total EPA and DHA) for week one, 2 g/day (600 mg/day EPA and DHA for
week 2, 3 g/day (900 mg/day EPA and DHA) for week three and through the end of the study.
20
There was no significant difference in SBP between the EPA and DHA group and the control
group. DBP was not reported.
Ginty et al. (2012) was a moderate quality randomized, double-blind, placebo controlled parallel
study in which 34 British subjects consumed corn oil (control) (n=14) or 1 g/day EPA and 400
mg/day DHA (1.4 g/day total) (n=20) in addition to their usual diets for three weeks. All
subjects also completed a psychological stress task (mental arithmetic) at baseline and at 21
days. The stress task included a two-minute pre-resting period, six-minute resting period, six-
minute mental arithmetic period, and a six-minute recovery period. Blood pressure was measured
every two minutes during the psychological stress task. There was no significant difference in
SBP or DBP between the fish oil group and the corn oil control group at rest or during the
psychological stress task.
Nilsson et al. (2012) was a moderate quality randomized, placebo controlled cross-over study in
which 44 British subjects consumed a placebo control (366 mg/day dicalcium phosphate, 150
mg/day microcrystalline cellulose, and 4 mg/day magnesium salts of fatty acids) or 5 g/day fish
oil (1.5 g/day EPA and 1.1 g/day DHA; 2.6 g/day total) for 5 weeks each period, in addition to
their usual diets. The reduction in SBP was significantly greater (P<0.05) with fish oil
consumption compared to the control. There was no significant difference in the change in DBP
with consumption of fish oil compared to the control.
Noreen and Brandauer (2012) was a moderate quality randomized, double-blind, placebo
controlled parallel trial in which 40 American subjects consumed 4 g/day safflower oil (control)
(n=20) or 4 g/day fish oil (1.6 g/day EPA and 800 mg/day DHA; 2.4 g/day total) (n=20) in
addition to their usual diets for 6 weeks. SBP was significantly lower (P=0.004) in the EPA and
DHA group compared to the control group. There was no significant difference in DBP between
the fish oil and control groups.
Dewell et al. (2011) was a moderate quality randomized, double-blind, placebo controlled
parallel trial, in which 60 American subjects consumed 1) 4 or 6 g/day soybean oil (control)
(n=20), 2) 4 g/day fish oil (700 mg/day EPA and 500 mg/day DHA; 1.2 g/day total) (n=20), or 3)
6 g/day fish oil (2.1 g/day EPA and 1.5 g/day DHA; 3.6 g/day total) (n=20) in addition to their
usual diets for 8 weeks. There was no significant difference in SBP and DBP between the 4
g/day fish oil (1.2 g/day EPA and DHA) and control groups. SBP and DBP, however, were
significantly lower (P<0.05) in the 6 g/day fish oil group (3.6 g/day EPA and DHA) compared to
the control group.
Sanders et al. (2011) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 310 British subjects consumed 1) 3 g/day olive oil (control) (n=71), 2) 3
g/day fish oil (0.45 g/day EPA and DHA) (n=80), 3) 3 g/day fish oil (0.9 g/day EPA and DHA)
(n=79), or 4) 3 g/day fish oil (1.8 g/day EPA and DHA) (n=80) in addition to their usual diets for
12 months. There was no significant difference in SBP or DBP between the control group and
either of the fish oil groups.
21
Sjoberg et al. (2010) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 67 Australian subjects consumed 1) 6 g/day sunola oil (control) (n=17), 2)
2 g/day fish oil (112 mg/day EPA and 524 mg/day DHA; 636 mg/day total) (n=16), 3) 4 g/day
fish oil (224 mg/day EPA and 1.04 g/day DHA; 1.3 g/day total) (n=17), or 4) 6 g/day fish oil
(336 mg/day EPA and 1.6 g/day DHA; 2 g/day total) (n=17) in addition to their usual diets for 12
weeks. There was no significant difference in SBP or DBP between the control group and any of
the fish oil groups.
Wong et al. (2010) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 97 Chinese subjects with NIDDM consumed 4 g/day olive oil (control) (n=48) or 4
g/day fish oil (1.68 g/day EPA and 1 g/day DHA; 2.7 g/day total) (n=49) in addition to their
usual diets for 12 weeks. There was no significant difference in SBP or DBP between the fish
oil group and olive oil control group.
Buckley et al. (2009) was a moderate quality randomized, double blind, placebo controlled
parallel trial in which 25 Australian subjects consumed 6 g/day sunflower oil (control) (n=13) or
6 g/day fish oil (1.5 g/day DHA and 0.36 g/day EPA; 1.9 g/day total) (n=12) in addition to their
usual diets during five weeks of exercise training. DBP was significantly lower (P=0.04) in the
EPA and DHA group compared to the control group. There was no significant difference in SBP
between the fish oil group and the control group.
Ferraro et al. (2009) was a moderate quality randomized, controlled parallel trial in which 30
Italian subjects with proteinuric IgA nephropathy consumed Renin-Angiotensin System Blockers
(RASB) therapy only (ramipril 10 mg/day and irbesartan 300 mg/day) (n=15) (control) or RASB
therapy plus 3 g/day fish oil (2.5 g/day total EPA and DHA) (n=15) in addition to their usual
diets for 6 months. There was no significant difference in SBP or DBP between the two groups.
Maki et al. (2009) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 76 American subjects consumed 1) 2 g/day olive oil (control) (n=25), 2) 2 g/day
krill oil (216 mg/day EPA and 90 mg/day DHA; 306 mg/day total) (n=25), or 3) 2 g/day
menhaden oil (212 mg/day EPA and 178 mg/day DHA; 390 mg/day total) (n=25) in addition to
their usual diets for 4 weeks. There was no significant difference in SBP or DBP between the
krill oil group and the control group. SBP was significantly lower (P=0.032) in the menhaden oil
group compared to the control group. There was no significant difference in DBP between the
menhaden oil group and the control group.
Meyer et al. (2009) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 48 Australian subjects consumed 1) 5 g/day sunola oil (control) (n=17), 2)5 g/day
fish oil (210 mg/day EPA and 810 mg/day DHA; 1 g/day total) (n=15), or 3) 5 g/day seal oil
(340 mg/day EPA and 450 mg/day DHA; 790 mg/day total) (n=16) in addition to their usual
diets for 6 weeks. SBP was significantly lower (P<0.05) in the fish oil group compared to the
sunola control group. SBP was also significantly lower (P<0.01) in the seal oil group compared
to the sunola control group. There was no significant difference in DBP between the control
group and the fish oil group or seal oil group.
22
Micallef and Garg (2009) was a moderate quality randomized, double-blind placebo controlled
parallel trial in which 60 Australian subjects consumed 1) 4 g/day sunola oil (control) (n=15), 2)
4 g/day sunola oil and 2 g/day plant sterol (control) (n=15), 3) 4 g/day fish oil (320 mg/day EPA
and 1.12 g/day DHA;1.4 g/day total) (n=15), or 4) 4 g/day fish oil (320 mg/day EPA and 1.12
g/day DHA; 1.4 g/day total) plus 2 g/day plant sterol (n=15) in addition to their usual diets for
three weeks. There was no significant difference in SBP or DBP between the fish oil only group
and the sunola control group. There was also no significant difference in SBP or DBP between
the fish oil plus plant sterol group and the plant sterol control group.
Rizza et al. (2009) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 50 Italian subjects consumed 2 g/day olive oil (control) (n=24) or 2 g/day fish oil
(1.7 g/day EPA and DHA) (n=26) in addition to their usual diets for 12 weeks. There was no
significant difference in SBP or DBP between the fish oil group and the olive oil control group.
Barcelo-Coblijn et al. (2008) was a moderate quality randomized, double-blind, placebo
controlled parallel trial in which 30 Canadian subjects consumed 1) 1 g/day sunflower oil
(control) (n=9), 2) 600 mg/day fish oil (252 mg/day EPA and 3 mg/day DHA; 255 mg/day total)
(n=11), or 3) 1.2 g/day fish oil (504 mg/day EPA and 6 mg/day DHA; 510 mg/day total) (n=10)
in addition to their usual diets for 12 weeks. There was no significant difference in SBP or DBP
between the sunflower oil control group and either of the fish oil groups.
Vernaglione et al. (2008) was a moderate quality single-blind, placebo controlled sequential
intervention
52
in which 24 Italian subjects on hemodialysis for end stage renal disease consumed
1) 2 g/day olive oil (control), 2) 2 g/day fish oil (900 mg/day EPA and 150 mg/day DHA; 1.05
g/day total), and 3) 2 g/day olive oil control again for 4 months each period. SBP and DBP were
significantly lower (P≤0.05) with fish oil consumption compared to the olive oil control.
Browning et al. (2007) was a moderate quality randomized placebo controlled cross-over study
in which 30 British women consumed 5 g/day of an oleic and linoleic oil mixture (control) or 5
g/day fish oil (2.9 g/day DHA and 1.3 g/day EPA; 4.2 g/day total) for 12 week periods each, in
addition to their usual diets. Subjects who were in the highest tertile for baseline serum sialic
acid, a marker of inflammation, were assigned to the raised inflammatory status group (n=12)
and subjects in the lowest tertile for baseline sialic acid were assigned to the reference group
(n=18). In subjects with raised inflammatory status, there was no significant difference in SBP
or DBP with consumption of fish oil as compared to the oleic and linoleic oil control. There was
also no significant difference in SBP or DBP in the reference group with fish oil consumption as
compared to the control.
Cazzola et al. (2007) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 155 Italian men were assigned to 1) 9 g/day corn oil (control) (n=40),
2) 6 g/day corn oil and 3 g/day fish oil (1.4 g/day EPA and 270 mg/day DHA; 1.6 g/day total)
(n=39), 3) 3 g/day corn oil and 6 g/day fish oil (2.7 g/day EPA and 510 mg/day DHA, 3.2g/day
total) (n=38) or 4) 9 g/day fish oil (4.1 g/day EPA and 810 mg/day DHA; 4.9 g/day total) (n=38)
52
In a sequential intervention, subjects receive a control and a treatment in a fixed sequence.
23
in addition to their usual diets for 12 weeks. There was no significant difference in SBP or DBP
between any of the fish oil groups and the control group.
Hill et al. (2007) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 65 Australian subjects 1) consumed 6 g/day sunflower oil (control) (n=18), 2)
consumed 7 g/day sunflower oil and followed a prescribed exercise regimen (control) (n=14), 3)
consumed 6 g/day fish oil (360 mg/day EPA and 1.6 g/day DHA; 1.9 g/day total) (n=17), or 4)
consumed 6 g/day fish oil (360 mg/day EPA and 1.6 g/day DHA; 1.9 g/day total) and followed
the prescribed exercise regimen (n=16) in addition to their usual diets for twelve months. There
was no significant difference in SBP or DBP between the fish oil groups and the sunflower oil
control groups.
Barden et al. (2006) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 83 pregnant Australian women with allergic disease consumed 4 g/day
olive oil (n=43) or 4 g/day fish oil (1.1 g/day EPA and 2.2 g/day DHA; 3.3 g/day total) (n=40) in
addition to their usual diets from the 20
th
week of pregnancy until delivery at 36 weeks. There
was no significant difference in SBP or DBP between the control and fish oil groups during
pregnancy at 20, 30 or 36 weeks. There was also no significant difference in SBP or DBP
between groups at 6 weeks post-delivery.
Damsgaard et al. (2006) was a moderate quality randomized, controlled parallel trial in which 83
Danish infants consumed standard infant formula or cow’s milk (control) (n=43) or standard
infant formula or cow’s milk plus 5 ml of fish oil (~554 mg/day EPA and ~369 mg/day DHA;
~924 mg/day) (n=40) for 3 months. SBP was significantly lower (P=0.02) in the fish oil group
compared to the control group. There was no significant difference in DBP between the two
groups.
Monahan et al. (2004) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 18 American subjects consumed 10 g/day olive oil (control) (n=9) or 10
g/day fish oil (3 g/day EPA and 2 g/day DHA; 5 g/day total) (n=9) in addition to their usual diets
for one month. There was no significant difference in SBP or DBP between the fish oil group
and the olive oil control group.
Finnegan et al. (2003) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 91 British subjects consumed a diet that incorporated 1) 25 g/day
margarine made from sunflower and safflower oils (control) (n=30), 2) 25 g/day margarine made
from fish oil (500 mg/day EPA and DHA) plus an estimated 200 mg/day EPA and DHA from
the subjects’ usual diets for a target intake of 700 mg/day EPA and DHA (n=30), or 3) 25 g/day
margarine made from fish oil (500 mg/day EPA and DHA) plus 3 g/day fish oil capsules (800
mg/day EPA and DHA) plus an estimated 200 mg/day EPA and DHA from the subjects’ usual
diets for a target intake of 1.5 g/day EPA and DHA (n=31) for 6 months. Actual intakes of EPA
and DHA were 800 mg/day and 1.7 g/day in the two treatment groups. Subjects used the
margarine provided by the study in place of the margarine they usually ate and otherwise
maintained their usual diets. There was no significant difference in SBP or DBP between the
control group and either of the fish oil groups.
24
Geelen et al. (2003) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 74 subjects from the Netherlands consumed 3.5 g/day high oleic
sunflower oil (control) (n=35) or 3.5 g/day fish oil (700 mg/day EPA and 560 mg/day DHA; 1.3
g/day total) (n=39) in addition to their usual diets for 12 weeks. There was no significant
difference in SBP or DBP between the fish oil and control groups.
Nestel et al. (2002) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 38 Australian subjects consumed 1) 4 g/day olive oil (control) (n=14), 2) 4 g/day
fish oil (3.04 g/day EPA and 68 mg/day DHA; 3.1 g/day total) (n=12), or 3) 4 g/day fish oil (2.84
g/day DHA and 164 mg/day EPA; 3.0 g/day total) (n=12) in addition to their usual diets for 7
weeks. There was no significant difference in SBP or DBP between the control group and either
of the fish oil groups.
Conquer et al. (1999) was a moderate quality randomized double-blind, controlled parallel trial
in which 19 Canadian men consumed 20 g/day vegetable oil (control) (n=9) or 20 g/day seal oil
(1.3 g/day EPA, 1.7 g/day DHA; 3.0 g/day total) (n=10) in addition to their usual diets for 42
days. There was no significant difference in SBP or DBP between the fish oil group and the
vegetable oil control group.
McVeigh et al. (1994) was a moderate quality randomized, double-blind, placebo controlled
cross-over study in which 20 Irish subjects with NIDDM consumed 10 g/day of olive oil
(control) or 10 g/day of fish oil (1.8 g/day EPA and 1.2 g/day DHA; 3.0 g/day total) for 6 weeks
each period, in addition to their usual diets. There was no significant difference in SBP and DBP
between the fish oil group and the olive oil control group.
Chin et al. (1993) was a moderate quality randomized, single-blind, placebo controlled parallel
study in which 29 Australian men consumed 1) 20 g/day mixed oil (palm, safflower, olive oil)
(control) (n=9), 2) 5 g/day fish oil (890 mg/day EPA and 580 mg/day DHA; 1.5 g/day total)
(n=8), 3) 10 g/day fish oil (1.8 g/day EPA and 1.2 g/day DHA; 2.9 g/day total) (n=6), or 4) 20
g/day fish oil (3.6 g/day EPA, 2.3 g/day DHA; 5.9 g/day total) (n=6) in addition to their usual
diets for 28 days. There was no significant difference in SBP and DBP between the mixed oil
control group and any of the fish oil groups.
Demke et al (1988) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 31 American subjects consumed 5 g/day safflower oil (control) (n=18) or 5 g/day
fish oil (930 mg/day EPA and 790 mg/day DHA; 1.7 g/day total) (n=13) in addition to their
usual diets for 28 days. There was no significant difference in SBP or DBP between the fish oil
group and the safflower oil control group.
Deslypere et al. (1992) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 58 Belgian subjects consumed 1) 9 g/day olive oil and palm oil (control)
(n=14), 2) 3 g/day fish oil (809 mg/day EPA and 165 mg/day DHA; 974 mg/day total) plus 6
g/day olive oil (n=15), 3) 6 g/day fish oil (1.6 g/day EPA and 329 mg/day DHA; 1.9 g/day total)
plus 3 g/day olive oil (n=15), or 4) 9 g/day fish oil (2.4 g/day EPA and 495 mg/day DHA; 2.9
25
g/day total) (n=14) in addition to their usual diets for 1 year. There was no significant difference
in SBP or DBP between the olive oil control group and any of the fish oil groups.
Flaten et al. (1990) was a moderate quality randomized, double-blind, placebo controlled parallel
study in which 56 Norwegian men consumed 14 g/day olive oil (control) (n=29) or 14 g/day fish
oil (3.6 g/day EPA and 2.9 g/day DHA; 6.5 g/day total) (n=27) in addition to their usual diets for
six weeks. There was no significant difference in SBP or DBP between the fish oil group and
olive oil control group.
Mortensen et al. (1983) was a high quality double-blind, placebo controlled cross-over study in
which 20 Danish men consumed 10 g/day vegetable oil (control) or 10 g/day fish oil (1.98 g/day
EPA and 1.25 g/day DHA; 3.23 g/day total) for 4 weeks each period, in addition to their usual
diets. SBP was significantly lower (P<0.05) with fish oil consumption as compared to the
vegetable oil control. However, there was no significant difference in DBP with fish oil
consumption as compared to the control.
Ryu et al. (1990) was a moderate quality randomized, placebo controlled parallel trial in which
20 American men consumed 6 g/day wheat germ oil (control) (n=10) or 6 g/day marine oil (2.1
g/day EPA and 900 mg/day DHA; 3 g/day total) (n= 10) in addition to their usual diets for 4
weeks. There was no significant difference in SBP or DBP between the two groups.
Sacks et al. (1994a)
53
(Trials of Hypertension Prevention) was a moderate quality randomized,
double-blind, placebo controlled parallel trial in which 350 American subjects consumed 1) 6
capsules/day olive oil (control) (n=86), 2) 3 tablets of cellulose (control) (n=89), or 3) 6 g/day
fish oil (1.44 g/day EPA and 960 mg/day DHA; 2.4 g/day total) (n=175) in addition to their usual
diets for 6 months. There was no significant difference in SBP or DBP between the fish oil
group and either control group.
Bonnema et al. (1995) was a moderate quality randomized, double-blind, controlled parallel trial
in which 28 Danish subjects with diabetes (IDDM or NIDDM) and incipient nephropathy
consumed 6 g/day olive oil (control) (n=14) or 6 g/day fish oil (2 g/day EPA and 1.3 g/day DHA;
3.3 g/day total) (n=14) in addition to a conventional diabetes diet for six months. There was no
significant difference in SBP or DBP between the fish oil group and olive oil control group.
Donnelly et al. (1992) was a moderate quality randomized, double-blind, placebo controlled
cross-over study in which 13 Canadian subjects with end stage renal disease consumed 12 g/day
olive oil (control) or 12 g/day fish oil (2.16 g/day EPA and 1.4 g/day DHA; 3.6 g/day total) for 4
weeks each period, in addition to their usual diets. There was no significant difference in SBP or
DBP with consumption of fish oil as compared to the olive oil control.
Bruckner et al. (1987) was a moderate quality randomized, single-blinded, placebo controlled
trial in which 21 American men consumed 1.5 g/10 kg body weight/day of olive oil (control)
(n=10) or 1.5 g/10 kg body weight/day fish oil (18 percent EPA and 12.64 percent DHA) (n=11)
53
The results of this study were also discussed in Sacks et al., 1994b and Cutler et al., 1992.
26
in addition to their usual diets for 3 weeks. There was no significant difference in SBP or DBP
between the fish oil group and the olive oil control group.
Blonk et al. (1990) was a moderate quality randomized, controlled parallel trial in which 45
Dutch subjects consumed 1) no capsules (control) (n=10), 2) 3 g/day fish oil (900 mg/day EPA
and 600 mg/day DHA; 1.5 g/day total) (n=11), 3) 6 g/day fish oil (1.8 g/day EPA and 1.2 g/day
DHA; 3 g/day) (n= 10), or 4) 12 g/day fish oil (3.6 g/day EPA and 2.4 g/day DHA, 6 g/day)
(n=14) in addition to their usual diets for 12 weeks. There was no significant difference in SBP
or DBP between the control group and any of the fish oil groups.
Dart et al. (1989) was a moderate quality double-blind, placebo controlled cross-over trial in
which 21 British subjects consumed 20 g/day olive oil (control) or 20 g/day fish oil (3.52 g/day
EPA and 2.5 g/day DHA; 6 g/day total) for 2 months each period, in addition to a low-fat diet
(30 percent total fat). SBP was significantly lower (P<0.05) with consumption of fish oil
compared to the olive oil control. There was no significant difference, however, in DBP with
fish oil as compared to the control.
DiGiacomo et al. (1989) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 32 subjects with Raynaud's phenomenon consumed 12 capsules/day of
olive oil (control) (n=16) or 12 capsules/day fish oil (3.96 g/day EPA and 2.64 g/day DHA; 6.6
g/day total) (n=16) in addition to their usual diets for 12 weeks. Blood pressure (SBP only) was
measured by using a digital blood pressure cuff on left index finger. Readings were taken at
room temperature and after the subjects’ hands were immersed in water (hand immersion test) at
104ºF, 77ºF, 59ºF, and 50ºF until there was an absence of blood flow or pulse into the finger or
until measurements were obtained at all water temperatures. There was no significant difference
in digital SBP between the two groups at room temperature. FDA did not consider the data on
digital SBP in subjects exposed to different water temperatures during the hand immersion test
portion of the trial because these experimental conditions were designed to evaluate whether fish
oil improves tolerance to cold temperatures and delays the onset of vasospasms associated with
Raynaud’s phenomenon by increasing blood flow and blood pressure. This portion of the trial
was not designed to evaluate whether fish oil intake lowers blood pressure.
Studies in subjects with CVD
Tavazzi et al. (2008) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 6, 975 Italian subjects with heart failure consumed 1 g/day placebo
(control) (n=3,481) or 1 g/day fish oil (~ 386 mg to 401 mg EPA, ~464 to 481 mg DHA, ~850-
882 mg/day total) (n=3,494) in addition to their usual diets for 3.9 years. There was no
significant difference in SBP or DBP between the control group and the fish oil group.
Axelrod et al. (1994) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 18 American subjects with NIDDM (4 subjects also had CAD) consumed
5 g/day safflower oil (control) (n=9) or 5 g/day fish oil (1.5 g/day EPA and 1.0 g/day DHA; 2.5
g/day total) (n=9) in addition to their usual diets for 6 weeks. SBP was significantly lower
(P=0.043) in the fish oil group compared to the safflower control group at 4 weeks. However, at
27
6 weeks SBP was not significantly lower between the two groups. Additionally, there was no
significant difference in DBP between the two groups at 4 or 6 weeks.
Bairati et al. (1991) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 125 Canadian subjects with CAD consumed 15 g/day olive oil (control)
(n=59) or 15 g/day fish oil (2.7 g/day EPA and 1.8 g/day DHA; 4.5 g day total) (n=66) in
addition to their usual diets for six months. SBP was significantly lower (P<0.05) in the fish oil
group compared to the olive oil control group. There was, however, no significant difference in
DBP between the two groups.
Rogers et al. (1987) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 60 British subjects with CVD consumed olive oil (control) (n=30) or fish
oil (n=30). The length of the study varied from 17 to 42 days with a mean length of 32 days.
Subjects consumed 16 mL/day of olive oil or fish oil supplements (2.9 g/day EPA and 1.9 g/day
DHA; 4.8 g/day total) for one week. For the rest of the trial, subjects consumed 10 mL/day of
olive oil or fish oil supplements (1.6 g/day EPA and 1.2 g/day DHA; 2.8 g/day total). There was
no significant difference in mean SBP or DBP between the two groups. The percent attributable
reduction in DBP, however, was significantly greater in the fish oil group (P<0.05) compared to
the olive oil control group.
Mehta et al. (1988) was a moderate quality randomized, double-blind, placebo controlled cross-
over trial in which 8 American men with CAD consumed a placebo control or 18 capsules/day
fish oil (3.2 g/day EPA and 2.2 g/day DHA; 5.4 g/day total) for 4 weeks each period, in addition
to their usual diets. SBP was significantly lower (P<0.05) with fish oil consumption compared to
the control when blood pressure was taken in a supine (lying down) or erect (standing) position.
SBP was also significantly lower (P<0.05) after a 3-minute exercise test and a 6-minute exercise
test. There was no significant difference in DBP between the fish oil period and the control
period when blood pressure was taken in a supine or erect position or after a 3 or 6-minute
exercise test.
O'Keefe et al. (2006) was a moderate quality randomized, double-blind, placebo controlled
cross-over trial in which 18 American men with a history of MI consumed a 50:50 mixture of
corn and olive oil (control) or 1.5 g/day fish oil (225 mg/day EPA and 585 mg/day DHA; 810
mg/day total) for 4 months each period. At the end of each test period, subjects completed an
exercise stress test. Blood pressure was measured at baseline (before the test) and during the
exercise stress test at peak heart rate. There was no significant difference in SBP with
consumption of fish oil as compared to the control. However, DBP was significantly higher
(P<0.05) with fish oil consumption compared to the control.
Radaelli et al. (2006) was a moderate quality randomized, placebo controlled parallel trial in
which 25 Italian subjects with post-MI systolic heart failure consumed a placebo control (n=10)
or 2 g/day fish oil (630 mg/day EPA and 1.1 g/day DHA, 1.7 g/day total) (n=15) in addition to
their usual diets for four months. There was no significant difference in SBP or DBP between
the two groups.
28
4. Studies that combined EPA and DHA supplementation with other dietary interventions in
normal or pre-hypertensive subjects (SBP 139 mmHg or DBP 89 mmHg)
Cobiac et al. (1991a) was a high quality randomized, double-blind, placebo controlled cross-over
trial in which 50 Australian subjects consumed diets that provided 1) 8 g/day sunflower oil/low
sodium (1,610 mg/day) (low sodium control diet), 2) 8 g/day sunflower oil/normal sodium
(3,450 mg/day) (normal sodium control diet), 3) 8 g/day fish oil (2.5 g/day EPA and 1.7 g/day
DHA; 4.2 g/day total)/low sodium (fish oil/low sodium diet), or 4) 8 g/day fish oil (2.5 g/day
EPA and 1.7 g/day DHA; 4.2 g/day total)/normal sodium (fish oil/normal sodium diet) for 4
weeks each period. SBP and DBP were significantly lower (P<0.05) with consumption of the
fish oil/low sodium diet as compared to the low sodium control diet. There was no significant
difference in SBP or DBP with consumption of the fish oil/normal sodium diet as compared to
the normal sodium control diet.
Cobiac et al. (1992) was a high quality randomized, double-blind, placebo controlled cross-over
trial in which 106 Australian subjects consumed diets that provided 1) 8 g/day sunflower oil/low
sodium (1,610 mg/day) (low sodium control diet), 2) 8 g/day sunflower oil/normal sodium
(3,450 mg/day) (normal sodium control diet), 3) 8 g/day fish oil (2.5 g/day EPA and 1.7 /day
DHA; 4.2 g/day total)/low sodium (fish oil/low sodium diet), or 4) 8 g/day fish oil (2.5 g/day
EPA and 1.7 g/day DHA; 4.2 g/day total)/normal sodium (fish oil/normal sodium diet) for 4
weeks each period. DBP was significantly lower (P<0.02) with consumption of the fish oil/low
sodium diet as compared to the sunflower oil/low sodium control diet. There was no significant
difference in SBP with consumption of the fish oil/low sodium diet as compared to the low
sodium control diet. Also, there was no significant difference in SBP or DBP with the
consumption of the fish oil/normal sodium diet as compared to the normal sodium control diet.
Kestin et al. (1990) was a high quality randomized, double-blind, placebo controlled parallel trial
in which 22 Australian men consumed 188 g/day linoleic acid supplements (14.3 g/day linoleic
acid) (control) (n=11) or 188 g/day fish oil supplements (2.1 g/day EPA and 1.3 g/day DHA; 3.4
g/day total) (n=11) for 6 weeks. The fish oil supplements were an emulsion of low-fat milk and
fish oil. The control supplements were an emulsion of low fat milk and safflower oil (linoleic
acid). A mixture of vegetable oils (olive and palm oil) and pure cholesterol were also added to
both supplements to balance the amount of saturated, monounsaturated, and polyunsaturated fats
and cholesterol in the test and control supplements. Subjects were instructed to consume a diet
with 40 percent of energy from total fat, with 10 percent of energy from total fat coming from
the fish oil or linoleic acid supplements. SBP was significantly lower (P=0.01) in the fish oil
group compared to the linoleic acid control group. There was no significant difference in DBP
between the two groups.
Kenny et al. (1992) was a high quality randomized, double-blind, placebo controlled cross-over
study in which 8 American men were fed a typical American diet providing 9 g/day safflower oil
(control) or 9 g/day fish oil (2.5 g/day EPA and 1.08 g/day DHA; 3.6 g/day total) for 7 days each
period. There was no significant difference in SBP or DBP with the fish oil diet as compared to
the control diet.
29
5. Studies of EPA and DHA from fatty fish in normal or pre-hypertensive subjects (SBP
139 mmHg or DBP 89 mmHg)
Cobiac et al. (1991b) was a high quality randomized, controlled parallel trial in which 31
Australian men were divided into three groups: 1) control diet (n=6), 2) fish diet that included
salmon (1 kg raw weight/wk) and sardines in salad oil (150 g/week) (4.5 g/day EPA and DHA)
(n=12), or 3) 15 g/day fish oil (4.5 g/day EPA and DHA) (n=13) for 5 weeks. Subjects were
instructed to limit fat intake to 30 percent of energy. The control group was provided lean steak,
chicken breast and ham. They were also provided a milk based supplement that was a mixture of
palm oil, safflower oil, olive oil, skim milk powder, cholesterol, sodium and potassium. The fish
diet group was provided Atlantic salmon, Norwegian sardines, and a liquid supplement that
contained less oil, some added cholesterol, no skim milk powder, and added sugar. The fish oil
group consumed the same diet as the control but also consumed fish oil supplements. The fish
oil supplement contained no added cholesterol, a small amount of palm and olive oil and no
added cholesterol. The foods and supplements consumed by all three groups provided 10
percent of energy as fat and were matched for total polyunsaturated fat, monounsaturated fat,
saturated fat, cholesterol, protein, fat, carbohydrate, sodium and potassium. There was no
significant difference in SBP or DBP between the control group and either the fish diet group or
the fish oil supplement group.
Ramel et al (2010) was a high quality randomized, single-blind, controlled parallel trial in which
278 subjects in Spain, Ireland, and Iceland consumed 1) no seafood and 6 capsules/day
sunflower oil (control), 2) 150 g/day cod 3 times/week (~300 mg/day EPA and DHA), 3) 150 g
salmon 3 times/week (~2.1 g/day EPA and DHA), or 4) 6 capsules/day fish oil (~1.3g/day EPA
and DHA) for eight weeks. In addition, all subjects were instructed to decrease caloric intake by
~30 percent of estimated energy expenditure. To minimize the differences among diets, subjects
were also instructed to consume ~30 percent of calories as total fat, ~50 percent of calories as
carbohydrate (including ~20-25 g/day dietary fiber), and ~20 percent of calories as protein.
There was no significant difference in SBP and DBP between the control group and either the
fish (cod or salmon) group or the fish oil group.
Lara et al. (2007) was a moderate quality controlled cross-over study in which 41 British subjects
consumed a standard diet (50 percent carbohydrates, 35 percent fat, 15 percent protein)
containing 125 g/day salmon (800 mg/day EPA and 1.6 g/day DHA; 2.4 g/day total) or a
standard diet containing no fish or fish oil supplements (control) for 4 weeks each. SBP and DBP
were significantly lower (P=0.001 and P=0.009, respectively) with consumption of salmon as
compared to the control.
Vandongen et al. (1993)
54
was a high quality randomized, controlled parallel trial in 120
Australian men. Five groups received 40 percent of calories from fat plus 1) olive/palm/
safflower oil placebo (1:4.5:4.5 mixture) (control) (n=18), 2) fish diet that provided 1.3 g/day of
EPA (DHA content not specified) plus olive/palm/safflower oil placebo (n=17), 3) 6 g/day fish
54
The results of this study were also discussed in Beilin et al., 1993.
30
oil (1.3 g/day EPA and 900 mg/day DHA; 2.2 g/day total) (n=17), 4) fish diet (1.3 g/day EPA;
DHA content not specified) plus 6 g/day fish oil (1.3 g/day EPA and 860 mg DHA; 2.2 g/day
total) (n=17), or 5) 12 g/day fish oil (2.6 g/day EPA and 1.7 g/day DHA; 4.3 g/day total) (n=16)
for 12 weeks. Two groups received 30 percent calories from fat plus olive/palm/safflower oil
placebo (n=17) (control) or fish diet (1.3 g/day EPA; DHA content not specified) plus
olive/palm/safflower oil placebo (n=18) for 12 weeks. Subjects were provided fish to consume
during one meal per day. There was no significant difference in SBP or DBP between any of the
fish or fish oil groups and the control groups.
6. Studies that added fish oil supplements to the diets of subjects with hypertension (SBP
140 mmHg or DBP 90 mmHg)
Kromhout et al. (2010) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 2,428 Dutch subjects with history of MI consumed a diet that
incorporated ~18 g/day margarine (control) (n=1,236) or ~18 g/day margarine made with fish oil
(~ 226 mg EPA, ~150 mg DHA, ~400 mg/day total) (n=1,192) for 40 months. Subjects used the
margarine provided by the study in place of the margarine they usually ate and otherwise
maintained their usual diets. There was no significant difference in SBP or DBP between the
control group and the fish oil group.
Root et al. (2013) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 51 American subjects consumed 3 g/day safflower oil (control) (n=27) or 3 g/day
fish oil (1.05 g EPA, 690 mg DHA, 1.74 g/day total) (n=30) in addition to their usual diets for 4
weeks. There was no significant difference in SBP or DBP between the control group and the
fish oil group.
Landmark et al. (1993)
55
was a moderate quality randomized, double-blind, placebo controlled
cross-over study in which 18 Norwegian men consumed their usual diets plus 1) 10 capsules of
olive oil (control), 2) 10 capsules of olive oil plus 40 mg/day nifedipine (blood pressure
medication) (control), 3) 10 capsules of fish oil (1.8 g/day EPA and 2.8 g/day DHA; 4.6 g/day
total), or 4) 10 capsules of fish oil (1.8 g/day EPA and 2.8 g/day DHA; 4.55 g/day total) plus 40
mg day nifedipine for 4 weeks each period. There was no significant difference in supine (lying
down) or standing SBP or DBP with fish oil as compared to the olive oil control. There was also
no significant difference in supine or standing SBP or DBP with fish oil plus 40 mg nifedipine as
compared to olive oil plus 40 mg day nifedipine.
Levinson et al. (1990) was a high quality randomized, double-blind, placebo controlled parallel
trial in which 16 American subjects consumed 50 g/day corn oil/palm oil (control) (n=8) or 50
g/day fish oil (9 g/day EPA and 6 g/day DHA; 15 g/day total) (n=8) in addition to their usual diet
for 6 weeks. There was no significant difference in SBP between the two groups. However,
DBP was significantly lower (p0.05) in the fish oil group compared to the corn oil/palm oil
control group.
55
The results of this study were also discussed in Mundal et al., 1993.
31
Meland et al. (1989) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 40 Norwegian men consumed 20 g/day olive oil and corn oil (control)
(n=20) or 20 g/day fish oil (3.6 g/day EPA and 2.4 g/day DHA; 6.0 g/day total) (n=20) in
addition to their usual diet for six weeks. There was no significant difference in SBP or DBP
between the fish oil group and the olive oil and corn oil control group.
Radack et al. (1991) was a moderate quality randomized, double-blind, placebo controlled cross-
over study in which 33 American subjects consumed 6 g/day safflower oil (control) or 6 g/day
fish oil (1.2 g/day EPA and 840 mg/day DHA; 2.0 g/day total) for 12 weeks each period, in
addition to their usual diets. DBP was significantly lower (p<0.05) in the fish oil group
compared to the control group. There was no significant difference in SBP with consumption of
fish oil compared to control.
Steiner et al. (1989) was a moderate quality randomized, double-blind, placebo controlled cross-
over study in which 28 Swiss subjects consumed 1) 4 capsules/day of salad oil (control),
2) 4 capsules of fish oil (560 mg/day EPA and 240 mg/day DHA; 800 mg/day total), or
3) 8 capsules of fish oil (1.1 g/day EPA and 480 mg/day DHA; 1.6 g/day total) for 4 weeks each
period, in addition to their usual diets. SBP and DBP were significantly lower (P<0.05)
compared to control when consuming 800 mg/day EPA and DHA or 1.6 g/day EPA and DHA.
Urakaze et al. (1989a)
56
was a moderate quality randomized, controlled parallel study in which
30 Japanese renal allograft recipients followed their usual diets (control) (n=16) or consumed 5.4
g/day fish oil (1.5 g/day EPA and 700 mg/day DHA; 2.2 g/day total) in addition to their usual
diets (n=14) for 6 months. There was no significant difference in SBP or DBP between the two
groups.
Wang et al. (2008) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 52 Chinese subjects consumed 3 placebo capsules (control) (n=26) or 3 g/day fish
oil (540 mg/day EPA and 360 mg/day DHA; 900 mg/day total) (n=26) for 8 weeks. There was
no significant difference in SBP or DBP between the control and fish oil groups.
Schmitz et al. (2002) was a moderate quality randomized, double-blind, placebo controlled
parallel trial in which 24 American subjects with chronic kidney disease who had a newly
constructed vascular graft consumed 4 g/day corn oil (control) (n=12) or 4 g/day fish oil (1.8
g/day EPA and 960 mg/day DHA; 2.7 g/day total) (n=12) in addition to their usual diets for 12
months. SBP and DBP were significantly lower (P<0.05) in the fish oil group compared to the
control group.
Wing et al. (1990) was a moderate quality randomized, double-blind, placebo controlled cross-
over trial in which 20 Australian subjects consumed 15 g/day olive oil (control) or 15 g/day fish
oil (2.7 g/day EPA and 1.8 mg/day DHA; 4.5 g/day total) for 8 weeks each period, in addition to
56
The results of this study were also discussed in Urakaze et al., 1989b.
32
their usual diets. There was no significant difference in SBP and DBP between the fish oil
period and the control period.
Gray et al. (1996) was a moderate quality randomized, double-blind, placebo controlled parallel
trial in which 19 American subjects consumed 18 g/day corn oil (n=10) (control) or 18 g/day fish
oil (2.16 g/day EPA and 1.28 g/day DHA; 3.44 g/day total) (n=9) in addition to their usual diets
for 8 weeks. There was no significant difference in SBP or DBP between the fish oil group and
the corn oil control group.
Cussons et al. (2009) was a moderate quality randomized, double-blind, placebo controlled
cross-over study in which 25 Australian women with polycystic ovary syndrome consumed 4
g/day olive oil (control) or 4 g/day fish oil (1.08 g/day EPA and 2.24 g/day DHA; 3.3 g/day
total) for 8 weeks each period, in addition to their usual diets. SBP was significantly lower
(P=0.018) with fish oil as compared to the control. Similarly, DBP was significantly lower
(P=0.005) with fish oil as compared to the control.
Morris et al. (1993) was a moderate quality randomized, double-blind, placebo controlled cross-
over trial in which 18 American subjects consumed 1) an olive oil placebo (control), 2) 6 g/day
fish oil (2.08 g/day EPA and 684 mg/day DHA; 2.8 g/day total), or 3) 12 g/day fish oil (4.1 g/day
EPA and 1.4 g/day DHA; 5.5 g/day total) for 12 weeks each period. During the placebo period,
half of the subjects were given 6 g/day of olive oil and half were given 12 g/day of olive oil to
ensure blinding. There was no significant difference in SBP or DBP with 6 g/day fish oil as
compared to the control. Similarly, there was no significant difference in SBP or DBP with
12 g/day fish oil as compared to the control.
duPlooy et al. (1992) was a moderate quality randomized, single-blind, placebo controlled
parallel trial in which 26 South African subjects consumed 0.5 mL olive oil (control) (n=12) for
24 weeks or cumulative doses of fish oil (n=14). Subjects in the fish oil group consumed 1) 0.05
mL fish oil (10 mg/day EPA and 3 mg/day DHA; 13 mg/day total), 2) 0.5 mL fish oil (100
mg/day EPA and 33 mg/day DHA; 133 mg/day total), 3) 5 mL fish oil (1.0 g/day EPA and 333
mg/day DHA; 1.3 g/day total), and 4) 50 mL fish oil (10 g/day EPA and 3.3 g/day DHA; 13.3
g/day total) for six weeks each (n=14) for a total of 24 weeks, in addition to their usual diets.
There was no significant difference in SBP or DBP between any of the EPA and DHA doses and
the control group.
Norris et al. (1986) was a moderate quality randomized, double-blind, placebo controlled cross-
over trial in which 16 British subjects consumed 16.5 g/day of a placebo or 16.5 g/day fish oil
(3.2 g/day EPA and 2.1 g/day DHA; 5.2 g/day total) for six weeks each, in addition to their usual
diets. When blood pressure was taken with subjects lying down, SBP was significantly lower
(P<0.02) with fish oil as compared to the control, but DBP was not significantly different. When
blood pressure was taken with subjects standing, DBP was significantly lower (P<0.05) with fish
oil as compared to the control, but SBP was not.
Lofgren et al. (1993) was a moderate quality randomized, double-blind, placebo controlled cross-
over study in which 38 American men consumed 20 g/day safflower oil (control) and 20 g/day
33
fish oil (3.6 g/day EPA and 2.4 g/day DHA; 6.0 g/day total) for 12 weeks each, in addition to an
American Heart Association step 1 diet. There was no significant difference in SBP or DBP
between the control and fish oil diets.
Study in subjects with CVD
Roncaglioni et al. (2013) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 12,505 Italian subjects with multiple CVD risk factors or atherosclerotic
vascular disease consumed 1 g/day olive oil (control) (n=6,266) or 1 g/day fish oil (850 mg/day
EPA and DHA total) (n=6,239) in addition to their usual diets for 5 years. There was no
significant difference in SBP or DBP between the control group and the fish oil group.
Hendra et al. (1990) was a moderate quality randomized, double-blind, placebo controlled
parallel study in which 80 British subjects with NIDDM (17 subjects also had CAD and 7 had
nephropathy) consumed 10 g/day olive oil (control) (n=40) or 10 g/day fish oil (1.8 g/day EPA
and 1.2 g/day DHA; 3 g/day total) (n=40) for six weeks each, in addition to their usual diets.
There was no significant difference in SBP or DBP with consumption of fish oil as compared to
control.
7. Studies that combined EPA and DHA with other dietary interventions in subjects with
hypertension (SBP 140 mmHg or DBP90 mmHg)
Howe et al. (1994) was a high quality randomized, double-blind, placebo controlled parallel trial
in which 56 Australian subjects were assigned to one of four groups: 1) normal sodium and 8
g/day olive oil (n=14), 2) normal sodium and 8 g/day fish oil (5 g n-3
57
fatty acids) (n=14) ,
3) low sodium and 8 g/day olive oil (n=14) , 4) low sodium and 8 g/day fish oil (5 g/day n-3 fatty
acids) (n=14) for six weeks. In the two normal sodium groups, subjects were given delayed-
release sodium tablets that provided an additional 1,840 mg/day (80 mmol/day) sodium to restore
sodium intake to between 3,450 and 3,634 mg/day (150 to 158 mmol/day). In the two low
sodium groups, the subjects were given placebo tablets in lieu of sodium supplements to
maintain blinding. The sodium intake in the low sodium groups was around 1,610 mg/day to
1,840 mg/day (70 to 80 mmol/day). There was no significant difference in SBP or DBP between
the normal sodium/fish oil group and the normal sodium/olive oil control group. Additionally,
there was no significant difference in SBP or DBP between the low sodium/fish oil group and the
low sodium/olive oil control group.
Singer et al. (1990b)
58
was a high quality, single-blind cross-over study in which 16 German
men consumed 80 mg/day propranolol and 9 g/day fish oil (1.8 g/day EPA and 1.1 g/day DHA;
57
“N-3” is a common scientific abbreviation for “omega-3.”
58
This study also included a subgroup (n=15) that consumed 9 g/day fish oil (1.8 g/day EPA and 1.1 g/day DHA;
2.9 g/day total) for 36 weeks followed by 9 g/day olive oil (control) for 4 weeks. The statistics comparing these two
groups were not reported. Additionally, the olive oil period was an inappropriate control since the duration was only
four weeks compared to the 36 week duration of the fish oil period. Since the duration of the study for the control
and treatment periods did not match and the statistics between the groups were not reported, scientific conclusions
could not be drawn by comparing SBP and DBP readings with consumption of fish oil to SBP and DBP readings
34
2.9 g/day total) for 12 weeks, followed by 80 mg/day propranolol and 9 g/day olive oil placebo
for 12 weeks, and finally 80 mg/day propranolol for 12 weeks. Subjects were instructed to
consume an isocaloric diet that substituted the polyunsaturated fat in the fish oil supplement for
an equal amount of saturated fat in other foods from their usual diet (e.g. cold cuts, meat, sausage
or cheese). With propranolol and fish oil consumption, SBP and DBP were significantly lower
(P<0.01) compared to propranolol alone. SBP and DBP were also significantly lower (P<0.01)
with propranolol and fish oil compared to propranolol and olive oil.
Hughes et al. (1990) was a high quality randomized, double-blind, placebo controlled cross-over
study in which 13 normotensive American men and 13 hypertensive American men consumed
10 capsules/day of wheat germ oil (77 mg of oil per capsule) (control) or 10 g/day fish oil (3.5
g/day EPA and 1.5 g/day DHA; 5.0 g/day total) for thirty days each period. Subjects were also
instructed to consume an isocaloric diet with 35-50 percent of calories from fat. There was no
significant difference in SBP and DBP in normotensive subjects between the control group and
the EPA and DHA group. Similarly, there was no significant difference in SBP and DBP in
hypertensive subjects between the control group and the EPA and DHA group.
Knapp et al. (1989) was a moderate quality randomized, placebo controlled parallel trial in which
30 American men consumed 1) 50 mL/day safflower oil (control) (n=8), 2) 50 ml/day of mixed
oils (n=8) (control), 3) 50 mL/day of fish oil (9.0 g/day EPA and 6 g/day DHA; 15 g/day total)
(n=8), or 4) 10 mL/day fish oil (1.8 g/day EPA and 1.2 g/day DHA; 3 g/day total) (n=8) for four
weeks. Subjects reduced the intake of fat in their usual diets to account for fat intake from the
fish oil supplements. There was no significant difference in SBP or DBP between the safflower
oil control group and the 10 mL or 50 mL fish oil groups. There was no significant difference in
SBP or DBP between the mixed oil control group and the 10mL fish oil group. SBP and DBP,
however, were significantly lower (P<0.015 and P< 0.038, respectively) in the 50-mL fish oil
group compared to the mixed oil control group.
C. Assessment of the Relevant Observational Studies
FDA reviewed 34 observational studies that evaluated the effect of EPA and DHA intake on risk
of hypertension or blood pressure. For the reasons discussed below, no scientific conclusions
about the relationship between EPA and DHA intake and blood pressure or risk of hypertension
could be drawn from any of the observational studies.
59
Eighteen observational studies estimated EPA and DHA intake from foods in the diet.
60
In
observational studies that calculate nutrient intake from conventional foods, these calculations
with consumption of olive oil. The high quality rating applies only to the part of the study described in the main text,
not the part described in this footnote.
59
This section contains a general discussion of major flaws in the reports of observational studies from which
scientific conclusions could not be drawn. Such studies may have other flaws in addition to those specifically
mentioned.
60
Asserhoj et al., 2009; Beegom et al., 1997; Ebbesson et al., 2007; Jarvinen et al., 2006; Jolly et al., 2011; Leary et
al., 2005; Mirmiran et al., 2012; Monge-Rojas et al., 2005; Morris et al, 1995; Oken et al., 2007; O'Sullivan et al.,
2012; Rytter et al., 2012; Skilton et al., 2013; Titova et al., 2013; Ueshima et al., 2007; Ulbak et al., 2004; van
35
are based on recorded dietary intake methods such as food frequency questionnaires, diet recall
interviews, or diet records, in which the types and amounts of foods consumed are estimated. A
common weakness of observational studies is the limited ability to ascertain the actual food or
nutrient intake for the population studied as a result of poor memory, over- or underestimation of
portion sizes, and recall bias (Flegal et al., 1999). Furthermore, the nutrient content of foods can
vary (e.g., due to type of fish, soil composition, food processing and cooking procedures, or
storage conditions such as duration and temperature). Thus, FDA cannot ascertain an accurate
amount of EPA and DHA consumed based on reported dietary intake of foods.
Intakes of EPA and DHA have been shown to correlate well with the percent EPA and DHA in
erythrocytes (r=0.95), plasma phospholipids (r=0.60), and whole blood (r=0.97) (Patterson et al.,
2015). Therefore, FDA considers these biomarkers to be valid and reliable measures of EPA and
DHA intake. Biomarkers that are reliable estimates of nutrient intake are not affected by the
limitations of estimating food intake in observational studies as discussed above.
Sixteen observational studies
61
measured a valid biomarker of intake for EPA and DHA.
However, none of these observational studies controlled for sodium and potassium intake or
smoking, all of which are known to affect blood pressure (IOM, 2005b; FDA, 2009). Therefore,
no scientific conclusions about the association between EPA and DHA intake and blood pressure
or hypertension risk could be drawn from these studies.
III. Strength of the Scientific Evidence
Below, the agency rates the strength of the total body of publicly available evidence. The agency
conducts this rating evaluation by considering the study type (e.g., intervention, prospective
cohort, case-control, cross-sectional), the methodological quality rating previously assigned, the
number of studies and number of subjects per group, whether the body of scientific evidence
supports a health claim relationship for the U.S. population or a target subgroup, whether study
results supporting the proposed claim have been replicated,
62
and the overall consistency
63
of the
total body of evidence.
64
Based on the totality of the scientific evidence, FDA determines
whether such evidence is credible to support a qualified health claim for the substance/disease
relationship and, if so, considers what qualifying language should be included to convey the
limits on the level of scientific evidence supporting the relationship or to prevent the claim from
being misleading in other ways.
Woudenbergh et al., 2009; Xun et al., 2011. Three of the 18 studies (Ulbak et al.2004; Asserhoj et al., 2009; and
Rytter et al., 2012) were observational follow-up studies to an intervention study that did not evaluate the
relationship between EPA and DHA and blood pressure or hypertension risk.
61
Arnarson et al., 2012; Ayer et al., 2009; Dewailly et al., 2001; Godin et al., 2003; Huang et al., 2010; Huang et al.,
2012; Kagawa et al., 1982; Leng et al., 1994; Liu et al., 2011; Mayneris-Perxachs et al., 2010; Novgorodtseva et al.,
2011; Oda et al., 2005; Park et al., 2010; Van Rossem et al., 2012; Wennberg et al., 2007; Zhang et al.,2012.
62
See supra, note 15.
63
See supra, note 16.
64
See supra, note 7 [Section III. F].
36
As discussed in Section II, the totality of scientific evidence about a possible relationship
between EPA and DHA and risk of high blood pressure includes 112 publications
65
reporting on
104 intervention studies from which scientific conclusions can be drawn. Seventy-five of these
studies were conducted in subjects with normal blood pressure or pre-hypertension (SBP≤139
mmHg or DBP ≤ 89 mm Hg), and 29 studies were conducted in subjects with hypertension (SBP
≥ 140 mm Hg or DBP ≥ 90 mm Hg). Of those 104 studies that looked at the relationship between
blood pressure and combined intake of EPA and DHA from conventional foods, dietary
supplements or prescription drugs, only 36
66
showed a statistically significant benefit. The
duration of these moderate to high quality studies showing a benefit ranged from four weeks to
one year, and the combined dose of EPA and DHA ranged from 390 mg/day to 15 g/day. The
remaining 68 intervention studies that showed no benefit were also moderate to high quality
randomized controlled trials. The duration of these studies ranged from 7 days to one year, with
EPA and DHA intake ranging from 13 mg/day to 13 g/day. One study (O'Keefe et al., 2006)
showed a statistically significant increase in DBP with EPA and DHA consumption. None of the
other 67 studies reported a statistically significant effect of EPA and DHA intake from
conventional foods, dietary supplements, or prescription drugs.
Based on the findings of these 104 intervention studies, FDA concludes there is some credible
evidence suggesting a relationship between the combined intake of EPA and DHA from
conventional foods, dietary supplements, or prescription drugs and blood pressure reduction.
65
Albert et al., 2015; Alfaddagh et al., 2017; Ansari et al., 2017; Armstrong et al., 2012; Atar et al., 2012; Axelrod
et al., 1994; Bairati et al., 1991; Barbosa et al., 2017; Barcelo-Coblijn et al., 2008; Beilin et al., 1993; Bosch et al.,
2012; Barden et al., 2006; Blonk et al., 1990; Bonaa et al., 1990; Bonnema et al., 1995 ;Browning et al., 2007;
Bruckner et al., 1987; Buckley et al., 2009; Carter et al., 2012; Cazzola et al., 2007; Chan et al., 2016a; Chan et al.,
2016b; Chin et al., 1993;Cobiac et al., 1991a; Cobiac et al., 1991b; Cobiac et al., 1992; Conquer et al., 1999;
Cussons et al., 2009; Cutler et al., 1992; Damsgaard et al., 2006; Dart et al., 1989; Demke et al., 1988; Deslypere et
al., 1992; Dewell et al., 2011; DiGiacomo et al., 1989; Donnelly et al., 1992; duPlooy et al., 1992; Elajami et al.,
2017; Faghihi et al., 2012; Ferraro et al., 2009; Finnegan et al., 2003; Flaten et al., 1990; Geelen et al., 2003; Ginty
et al., 2012; Gray et al., 1996; Hendra et al., 1990; Hill et al., 2007; Hlais et al., 2013; Holm et al., 2001; Holman et
al., 2009; Howe et al., 1994; Hughes et al., 1990; Kenny et al., 1992; Kestin et al., 1990; Kim et al., 2011; Knapp et
al., 1989; Kristensen et al., 2016; Kromhout et al., 2010; Landmark et al., 1993; Lara et al., 2007; Levinson et al.,
1990; Lofgren et al., 1993; Logan and Spriet, 2015; Lungershausen and Howe, 1994; Lungershausen et al., 1994;
Lungershausen et al., 1997; Maki et al., 2009; McVeigh et al., 1994; Mehta et al., 1988; Meland et al., 1989; Meyer
et al., 2009; Micallef and Garg, 2009; Minihane et al., 2016; Monahan et al., 2004; Mori et al., 2009; Morris et al.,
1993; Mortensen et al., 1983; Mundal et al., 1993; Nestel et al., 2002; Nilsson et al., 2012; Nordoy et al., 2001;
Noreen and Brandauer, 2012; Norris et al., 1986; O'Keefe et al., 2006; Pase et al., 2015; Pettersson et al., 1994;
Radack et al., 1991; Radaelli et al., 2006; Ramel et al., 2010; Rizza et al., 2009; Rogers et al., 1987; Roncaglioni et
al., 2013; Root et al., 2013; Ryu et al., 1990; Sacks et al., 1994a; Sacks et al., 1994b; Sanders et al., 2011; Schmitz et
al., 2002; Singer et al., 1990b; Sjoberg et al., 2010; Skulas-Ray et al., 2012; Steiner et al., 1989; Tavazzi et al., 2008;
Toft et al., 1995; Urakaze et al., 1989a; Urakaze et al., 1989b;Wu et al., 2014; Vandongen et al., 1993; Vernaglione
et al., 2008; Wang et al., 2008; Wing et al., 1990; Wong et al., 2010.
66
Ansari et al., 2017; Axelrod et al., 1994; Bairati et al., 1991; Bonaa et al., 1990; Buckley et al., 2009; Chan et al.,
2016a; Cobiac et al., 1991a; Cobiac et al., 1992; Cussons et al., 2009; Damsgaard et al., 2006; Dart et al., 1989;
Dewell et al., 2011; Holm et al., 2001; Kestin et al., 1990; Knapp et al., 1989; Lara et al., 2007; Levinson et al.,
1990; Lungershausen et al., 1994; Lungershausen et al., 1997; Maki et al., 2009; Mehta et al., 1988; Meyer et al.,
2009; Minihane et al., 2016; Mori et al., 2009; Mortensen et al., 1983; Nilsson et al., 2012; Nordoy et al., 2001;
Noreen and Brandauer, 2012; Norris et al., 1986; Radack et al., 1991; Rogers et al., 1987; Schmitz et al., 2002;
Singer et al., 1990; Steiner et al., 1989; Toft et al., 1995; Vernaglione et al., 2008.
37
However, this evidence is highly inconsistent. The findings of the minority of intervention
studies that found a statistically significant lowering of blood pressure from combined intake of
EPA and DHA are undermined by the larger body of evidence that found no effect on blood
pressure (or, in one study, a statistically significant rise in DBP). Study results were inconsistent
regardless of baseline SBP or DBP or the study duration (7 days to five years). Results were also
inconsistent in studies conducted in subjects with preexisting CVD (including CAD and other
forms of CHD). In addition, results were inconsistent across the whole range of EPA and DHA
intakes studied (13 mg/day to 15 g/day). Consistency of the findings among similar and different
study designs is important for evaluating causation and the strength of the evidence. Lack of
consistency among studies evaluating the same substance-disease relationship weakens the
strength of the evidence. Therefore, FDA concludes that while there is some credible evidence
suggesting that combined intake of EPA and DHA from conventional foods and dietary
supplements may reduce the risk of hypertension by lowering blood pressure, this evidence is
inconclusive and highly inconsistent.
IV. General Requirements for Health Claims and Enforcement Discretion Factors
A qualified health claim about reducing the risk of hypertension or CHD on the label or in the
labeling of conventional foods and dietary supplements containing EPA and DHA in
combination is required to meet all applicable statutory and regulatory requirements under the
Federal Food, Drug, and Cosmetic Act, with the exception of the requirement that the health
claim meet the significant scientific agreement standard and the requirement that the claim be
made in accordance with an authorizing regulation. Other exceptions to the general requirements
for health claims that FDA intends to consider in the exercise of its enforcement discretion for
qualified health claims are discussed below, as well as enforcement discretion factors specific to
qualified health claims describing how consuming EPA and DHA in combination may reduce
the risk of hypertension or CHD by lowering blood pressure.
A. Daily Dietary Intake Needed to Achieve the Claimed Effect
The general requirements for health claims provide that, if the claim is about the effects of
consuming the substance at other than decreased dietary levels, the level of the substance must
be sufficiently high and in an appropriate form to justify the claim. Where no definition for
“high” has been established, the claim must specify the daily dietary intake necessary to achieve
the claimed effect (21 CFR 101.14(d)(2)(vii)). However, the agency finds that this provision
cannot be applied to qualified health claims for combined EPA and DHA and reduced risk of
hypertension because, as discussed in Section III, the scientific evidence for this relationship,
which is inconclusive and highly inconsistent, does not support the establishment of a
recommended daily dietary intake level for the general U.S. population.
B. Level of EPA and DHA Intake From Dietary Supplements When Used as Labeled
For a substance to be eligible for a health claim, its use at the levels necessary to justify a claim
must be demonstrated to be safe and lawful (21 CFR 101.14(b)(3)(ii)). In Section I.C of this
letter (“Safety Review”), we discussed the limited conditions of use under which EPA- and
38
DHA-containing ingredients (e.g., menhaden oil, other fish oils, algal oils) are GRAS for use in
conventional foods. The limitations on conditions of use are designed to ensure that total daily
intake of EPA and DHA from conventional foods does not exceed 3 g/day. There is, however, no
current limit on the amounts of EPA and DHA that may be used in dietary supplements.
67
In the absence of a limit on the amounts of EPA and DHA that may be used in dietary
supplements, we evaluated evidence on risks related to the use of EPA and DHA to determine
whether we should consider the level of EPA and DHA in a dietary supplement as a factor in the
exercise of our enforcement discretion for use of the qualified health claim. Because the FD&C
Act directs us to evaluate the safety of a dietary supplement in the context of the conditions of
use recommended or suggested in its labeling (see 21 U.S.C. 342(f)(1)), we focused on the total
daily intake of EPA and DHA that a dietary supplement provides when used as labeled, rather
than on the EPA and DHA content of a single capsule or dose of the product.
Health claims in the labeling of conventional foods and dietary supplements are used as a
marketing tool to encourage consumers to buy and consume the products that bear them. Health
claims have been shown to increase sales of the products on which they appear (Roe et al.,
1999). Thus, FDA’s exercise of enforcement discretion for EPA/DHA qualified health claims
may result in increased intake of EPA and DHA. Because of this potential for greater
consumption of EPA and DHA, we evaluated information on possible adverse effects on
glycemic control, blood cholesterol, and risk of excessive bleeding at levels of EPA and DHA
intake currently available in dietary supplements on the U.S. market (i.e., up to 8 g/day of EPA
and DHA combined).
68
Recent comprehensive reviews suggest that intake of EPA and DHA has
no adverse effects on glycemic control in subjects with normal glucose levels or in subjects with
type II diabetes. These reviews also suggest that intake of EPA and DHA does not raise total
cholesterol. Based on our evaluation of several comprehensive reviews, the evidence as to
whether EPA and DHA intake increases low density lipoprotein cholesterol (LDL-C) is
inconsistent. While some reviews suggest that there is a small but statistically significant
increase in LDL-C of approximately 2 to 4 mg/dL, another review suggests that long-term
consumption of EPA and DHA does not lead to an adverse effect on LDL-C. Additionally, there
is some evidence that despite any potential increase in LDL-C, EPA and DHA intake may reduce
the risk of CHD through other mechanisms.
69
67
Dietary ingredients in dietary supplements are not required to be GRAS for their intended use, although FDA can
set a regulatory limit on the level of a dietary ingredient to prevent adulteration. See 21 U.S.C. §§ 321(s)(6), 342(f),
371(a). FDA has not set any regulatory limit on the level of EPA, DHA, or other omega-3 fatty acids in dietary
supplements.
68
See Memoranda to Docket No. FDA-2014-Q-1146: Review of Scientific Literature on EPA and DHA Intake and
Risk of Excessive Bleeding (June 17, 2019) and Survey of Comprehensive Reviews of the Effects of EPA and DHA
Intake on Glycemic Control and Blood Cholesterol (June 17, 2019).
69
See, e.g., “Letter Responding to Health Claim Petition dated November 3, 2003 (Martek Petition): Omega-3 Fatty
Acids and Reduced Risk of Coronary Heart Disease” (September 2004). [
http://wayback.archive-
it.org/7993/20171114183727/https://www.fda.gov/Food/IngredientsPackagingLabeling/LabelingNutrition/ucm0729
32.htm].
39
Our review of the evidence on bleeding risk found that adverse events were rarely experienced in
the studies we evaluated, which looked at intakes of EPA and DHA up to 5.4 g/day. The average
dose seen in the studies was around 5 g/day. Based on the data from these studies, we conclude
that ingestion of less than or equal to 5 g/day of EPA and DHA from dietary supplements is
unlikely to cause excessive bleeding. With little or no data on intakes over 5 g/day, the potential
effects on bleeding risk at those levels of intake are less clear. Therefore, to ensure that
consumption of high-potency EPA and DHA supplements based on the presence of EPA/DHA
qualified health claims in product labeling does not increase the potential for adverse effects, we
intend to consider, as a factor in the exercise of our enforcement discretion, that dietary
supplements bearing the claim not provide more than 5 gr/day of EPA and DHA combined when
used according to their labeling.
70
C. Minimum Content of EPA and DHA to Prevent Consumer Deception
As a factor in the exercise of our enforcement discretion, we are specifying a minimum amount
of combined EPA and DHA that a conventional food or dietary supplement bearing an
EPA/DHA qualified health claim should contain. The purpose of this minimum content
enforcement discretion factor is to prevent consumer deception from health claims on the labels
of products that contain only trivial or insignificant amounts of EPA and DHA. To identify an
appropriate minimum content amount, FDA considered the two lowest levels of combined EPA
and DHA evaluated in the studies that demonstrated a benefit in lowering blood pressure. As
discussed in sections III and IV.A, the scientific evidence for such a benefit does not support the
establishment of a recommended daily dietary intake level because it is inconclusive and highly
inconsistent; however, limiting our consideration of enforcement discretion to products that
contain EPA and DHA in amounts that have been observed to lower blood pressure in at least
some well-conducted scientific studies will help ensure that consumers do not see the claim on
products that contain so little EPA and DHA that they are very unlikely to provide any such
benefit.
The lowest level of intake among the studies that showed a benefit in lowering blood pressure
was 0.39 g/day EPA and DHA (Maki et al., 2009). The next lowest level of intake among studies
that showed a benefit in lowering blood pressure was 0.8 g/day EPA and DHA (Meyer et al.,
2009; Steiner et al., 1989). Although the lowest intake (0.39 g/day) was in the study by Maki et
al. (2009), the studies by Steiner et al. (1989) and Meyer et al. (2009) with the next lowest level
of EPA and DHA (0.8 g/day) provide a more scientifically appropriate basis for a minimum
content amount. The Maki et al. (2009) study was conducted in U.S. subjects and looked at
intake of EPA and DHA from two different sources, krill oil and menhaden oil, compared to a
control. There was no significant difference in SBP or DBP when the krill oil was used as a
70
We considered whether the level of 5 g/day from dietary supplements should be adjusted downward to allow for
intake of EPA and DHA from conventional foods and decided no adjustment was necessary. The studies we
evaluated provided EPA and DHA supplements as an addition to subjects’ usual diets, which (with the possible
exception of a few subjects with eating patterns outside the norm) would already contain some EPA and DHA.
Therefore, the results of the studies on bleeding risk with intakes of up to 5 g/day EPA and DHA from the test
product reflect the subjects’ intake of EPA and DHA from all sources, including the EPA and DHA that was in the
subjects’ usual diet.
40
source of EPA and DHA (0.3 g/day). However, there was a benefit in lowering SBP, but not
DBP, when menhaden oil was provided as the source of EPA and DHA (0.39 g/day). The
authors of Maki et al. (2009) noted that because blood pressure was not lowered when krill oil
was the source of EPA and DHA, and only SBP but not DBP was significantly lowered when
menhaden oil was the source of EPA and DHA, the benefit in lowering SBP from menhaden oil
could be a chance finding. For this reason, we believe the 0.39 g/day amount from the study by
Maki et al. should not be the basis for the minimum amount of EPA and DHA content in a
conventional food or dietary supplement that bears an EPA/DHA qualified health claim.
The Steiner et al. (1989) study was conducted in Swiss subjects with hypertension and evaluated
two levels of EPA and DHA, 0.8 g/day and 1.6 g/day. In this study, both levels of intake
significantly lowered SBP and DBP. The third study, Meyer et al. (2009), provided 0.8 g/day of
EPA and DHA from seal oil and was conducted in Australian subjects with normal blood
pressure or pre-hypertension. In this study, SBP, but not DBP. was significantly lowered after
consumption of EPA and DHA from seal oil. Since these two studies showed a benefit in
lowering blood pressure among subjects with a wide range of blood pressure levels (normal, pre-
hypertensive, and hypertensive), we conclude that they provide the most scientifically
appropriate basis for a minimum amount of EPA and DHA in conventional foods and dietary
supplements that bear an EPA/DHA qualified health claim.
As discussed in the Safety Review section, usual intake of EPA and DHA in the United States is
very low (mean total intake of 77 mg/day from all sources for people age 4 years and older).
Because of the low level of intake and the number of foods containing EPA and DHA being very
limited (primarily fish and seafood products), we do not find it necessary to divide the 0.8 g/day
minimum amount among multiple eating occasions throughout the day. Unlike some nutrients
that can be found in many types of foods that may be consumed throughout the day, we do not
expect the typical U.S. diet to include EPA- and DHA-containing foods on multiple eating
occasions during the day. Therefore, we are specifying 0.8 g EPA and DHA (combined total) as
the minimum amount per serving for a conventional food or dietary supplement to bear an
EPA/DHA qualified health claim.
We are uncertain how much consumers will increase their intake of EPA and DHA from
conventional foods and dietary supplements due to the use of qualified health claims about the
relationship between intake of EPA and DHA in combination and reduced risk of hypertension
and CHD through the mechanism of blood pressure reduction. To help consumers gauge their
total intake of EPA and DHA, FDA intends to consider, as a factor in the exercise of its
enforcement discretion, that conventional foods and dietary supplements that bear an EPA/DHA
qualified health claim state the amount of EPA and DHA per serving in the claim. Alternatively,
dietary supplements may declare the amount of EPA and DHA per serving in “Supplement
Facts,” instead of in the claim (see 21 CFR 101.36(b)(3)). Information on the content of EPA
and DHA allows consumers to understand how their food and dietary supplement choices fit into
the context of their total daily diet, as required by 21 CFR 101.14(d)(2)(v).
41
D. Total Fat, Saturated Fat, and Cholesterol Enforcement Discretion Factors for CHD-
related Health Claims
In regulations authorizing health claims about reducing the risk of CHD through a cholesterol-
lowering mechanism, FDA has generally required, with a few exceptions, that foods bearing
such claims meet the "low fat" criterion defined by 21 CFR 101.62(b)(2), the "low saturated fat"
criterion defined by 21 CFR 101.62(c)(2), and the "low cholesterol" criterion defined by 21 CFR
101.62(d)(2) (see authorized claims in 21 CFR 101.75, 101.77, 101.81, 101.82, and 101.83). We
discuss below how the agency intends to consider these criteria as factors in deciding whether to
exercise enforcement discretion for EPA/DHA qualified health claims about reducing the risk of
hypertension and CHD by lowering blood pressure.
“Low fat” criterion
Several of the authorized health claims about reduced risk of CHD require foods bearing the
claim to meet the definition of "low fat" in 21 CFR 101.62(b)(2) (see authorized claims in 21
CFR 101.75, 101.77, 101.82, and 101.83). The "low fat" requirement was first introduced in the
regulation authorizing a health claim about reducing the risk of CHD by lowering intake of
saturated fat and cholesterol (21 CFR 101.75). In the proposed rule for that claim (56 FR 60727
at 60739; November 27, 1991), FDA stated that, although total fat is not directly related to
increased risk for CHD, it may have significant indirect effects on CHD risk. The agency
explained that low fat diets facilitate reducing the intake of saturated fat and cholesterol to
recommended levels. Furthermore, the agency noted that obesity is a major risk factor for CHD,
and that dietary fats, which have more than twice as many calories per gram as proteins and
carbohydrates, are major contributors to total calorie intakes.
FDA has made several exceptions to the “low fat” requirement in CHD health claims, and its
views on dietary fat and health have evolved with changes in nutrition science. For example,
instead of the "low fat" criterion, fish and game meat are required to meet the "extra lean"
criterion in the authorizing regulation for the health claim about saturated fat and cholesterol and
reduced risk of CHD (21 CFR 101.75(c)(2)(ii)), as these foods are appropriately included in a
diet low in harmful fats and cholesterol. Additionally, a food that bears the plant sterol/stanol
esters and CHD health claim is not required to be “low fat," but the food’s total fat content may
not exceed the total fat disqualifying level in 21 CFR 101.14(a)(4) (see 21 CFR
101.83(c)(2)(iii)(C)). In explaining the exception from the "low fat" criterion for foods that bear
the plant sterol/stanol esters claim, FDA noted that the 2000 Dietary Guidelines for Americans
changed the earlier recommendation for a diet low in total fat to a recommendation for a diet
moderate in total fat, replacing the previous focus on limiting total fat with a focus on keeping
intake of saturated fat and cholesterol low (65 FR 54686 at 54708; Sept. 8, 2000). The current
Dietary Guidelines for Americans (2015-2020 Dietary Guidelines) no longer recommend
limiting total fat intake (USDA, 2015). Instead, the 2015-2020 Dietary Guidelines prioritize
increasing intakes of polyunsaturated and monounsaturated fats and decreasing intakes of
saturated fat and trans fat.
42
FDA agrees that increasing consumption of heart-healthy fats and decreasing consumption of
unhealthy fats is more important in reducing CHD risk than consuming diets low in total fat.
Therefore, FDA has decided not to consider as an enforcement discretion factor whether a
dietary supplement or conventional food that bears an EPA/DHA qualified health claim meets
the "low fat" criterion.
Low saturated fat” criterion
For individual foods other than fish (discussed below under “Extra lean criterion for fish”) and
dietary supplements, FDA intends to consider, as a factor in the exercise of its enforcement
discretion for the use of an EPA/DHA qualified health claim, that the food meets all "low
saturated fat" criteria in 21 CFR 101.62(c)(2)). This food category includes primarily foods that
do not normally contain EPA and DHA but have been enriched with EPA- and DHA-containing
ingredients. For meal products as defined in 21 CFR 101.13(l) and main dishes as defined in 21
CFR 101.13(m), FDA intends to consider, as a factor in the exercise of its enforcement
discretion, that such foods meet all "low saturated fat" criteria in 21 CFR 101.62(c)(3).
For dietary supplements, FDA intends to consider, as a factor in the exercise of its enforcement
discretion for the use of an EPA/DHA qualified health claim, that the dietary supplement meets
the first low saturated fat criterion in 21 CFR 101.62(c)(2)(i) by containing no more than 1 g
saturated fat per reference amount customarily consumed (RACC).
71
However, FDA does not
intend to consider as an enforcement discretion factor for dietary supplements the additional
“low saturated fat” criterion in 21 CFR 101.62(c)(2) that the product contain no more than 15
percent of calories from saturated fat. Among dietary supplements, products that contain 100%
fish oil as the source of EPA and DHA would contain the highest amounts of saturated fat. In a
fish oil, 20 - 30 percent of calories come from saturated fat (USDA, 2018). Because 100 percent
fish oil dietary supplements have no other source of calories than fish oil and reformulation to
reduce the percent of calories from saturated fat is not possible, such supplements would never
be able to meet the 15 percent criterion in 21 CFR 101.62(c)(2). Therefore, FDA intends to
consider, as a factor in the exercise of its enforcement discretion, that dietary supplements that
bear an EPA/DHA qualified health claim meet the "equal to or less than 1 g of saturated fat per
RACC" criterion in 21 CFR 101.62(c)(2), but does not intend to consider the “no more than 15
percent of calories from saturated fatcriterion as a factor in the exercise of its enforcement
discretion.
“Low cholesterol” criterion
For individual foods other than fish (discussed below under “Extra lean criterion for fish”) and
dietary supplements, FDA intends to consider, as a factor in the exercise of its enforcement
discretion for an EPA/DHA qualified health claim, that the food meets the low cholesterol
71
The reference amount customarily consumed (RACC) for a food represents the amount of that food customarily
consumed per “eating occasion” (meal or snack) in the United States. FDA sets the RACCs for various foods and
food categories in its food labeling regulations (see 21 CFR 101.12). The serving sizes declared on food labels
(“label serving sizes”) are based on, but not necessarily identical to, the RACC (see 21 CFR 101.9(b)).
43
criterion of no more than 20 mg per RACC or per 50 grams (21 CFR 101.62(d)(2)). As with
saturated fat, supplements that contain 100% fish oil as the source of EPA and DHA would
contain the highest amounts of cholesterol. Fish oil dietary supplements in liquid form usually
have a serving size of one teaspoon (containing 4.5 g of total fat). One teaspoon of fish oil
contains about 22 – 34 mg of cholesterol (USDA, 2018), which would exceed the “low
cholesterolcriterion per RACC and per 50 g. Because fish oil dietary supplements in liquid
form usually have no other components and reformulation to reduce the cholesterol is not
possible, such liquid supplements would never be able to meet the “low cholesterol” criterion per
RACC or per 50g. Therefore, FDA does not intend to consider, as a factor in the exercise of its
enforcement discretion, that dietary supplements in liquid form that bear an EPA/DHA qualified
health claim meet the “low cholesterol” criterion per RACC or per 50 g.
With regard to combined EPA and DHA dietary supplements in other forms (e.g., softgel or
gummy), FDA intends to consider, as a factor in its discretion for the use of an EPA/DHA
qualified health claim, that the dietary supplement meet the low cholesterol criterion of no more
than 20 mg per RACC. Supplements that contain 100% fish oil, such as single-ingredient fish oil
softgels, would contain the highest amounts of cholesterol. Fish oil softgels usually have a
serving size of 1 - 2 softgels. FDA estimates that 1 - 2 softgels weigh about 1 - 3 g and contain
about 0.5 - 2 g of fish oil. This small amount of fish oil would not contain enough cholesterol to
exceed the criterion of 20 mg per RACC, but the cholesterol content of 100% fish oil softgels per
50 g would far exceed the 20 mg criterion. FDA estimates that 50 g of fish oil would contain
about 240 to 380 mg of cholesterol (USDA, 2018). However, since it is highly unlikely that
individuals would consume 50 g per day of dietary supplements containing combined EPA and
DHA, FDA has decided that it is not necessary to consider, as a factor in the exercise of its
enforcement discretion, that dietary supplements containing combined EPA and DHA contain no
more than 20 mg of cholesterol on a 50g basis.
“Extra lean” criterion for fish
FDA has defined fish in 21 CFR 123.3(d) as “fresh or saltwater finfish, crustaceans, other forms
of aquatic animal life (including, but not limited to, alligator, frog, aquatic turtle, jellyfish, sea
cucumber, and sea urchin and the roe of such animals) other than birds or mammals, and all
mollusks, where such animal life is intended for human consumption.” For the purpose of
EPA/DHA qualified health claims in the labeling of products that are essentially all fish, FDA
intends to consider certain factors in the exercise of its enforcement discretion. We consider
“products that are essentially all fish” to mean fish without any added ingredients and fish with
an “insignificant amount”
72
of added fat or carbohydrate, as defined in 21 CFR 101.9(f)(1).
Raw fish, boiled fish, and broiled fish are examples of products that are essentially all fish.
The regulation authorizing a health claim about diets low in saturated fat and cholesterol and
reduced risk of CHD excepts fish from the "low saturated fat" criterion and "the low cholesterol"
72
“An ‘insignificant amount’ shall be defined as that amount that allows a declaration of zero in nutrition labeling,
except that for total carbohydrate, dietary fiber, and protein, it shall be an amount that allows a declaration of ‘less
than 1 gram.’” 21 CFR 101.9(f)(1).
44
criterion (21 CFR 101.75(c)(2)(ii)). Instead, fish is required to be "extra lean" as defined in 21
CFR 101.62(e)(4) (contains less than 5 g total fat, less than 2 g saturated fat, and less than 95 mg
cholesterol per reference amount customarily consumed and per 100 g).
Most oily fish that are rich in EPA and DHA omega-3 fatty acids exceed the "extra lean"
criterion for saturated fat (2 g of saturated fat per RACC) but do not exceed the saturated fat
disqualifying level (4 g of saturated fat per RACC). One of the ways that FDA determines
whether to consider a nutrient content criterion as a factor in the exercise of its enforcement
discretion is whether there are risk reduction data among healthy individuals that would suggest
that there may be a benefit from consumption of the food, even though the food does not meet
the nutrient content criterion. Such data, for purposes of this review, would include data
showing an association with a lower risk of hypertension or CHD in observational studies
conducted in apparently healthy individuals. Because we have concluded that the findings of
some studies show that intake of EPA and DHA in combination may reduce the risk of
hypertension and CHD by lowering blood pressure, FDA has decided that it is not necessary to
consider, as a factor in the exercise of its enforcement discretion for products that are essentially
all fish, that such products meet the "extra lean" criterion for saturated fat. However, FDA has
decided to consider, as a factor in the exercise of its enforcement discretion for products that are
essentially all fish, that such products meet the "extra lean" criterion for cholesterol (95 mg of
cholesterol per RACC). Most fish that are rich sources of EPA and DHA do not exceed the
"extra lean" criterion for cholesterol; thus, this approach should not disqualify many products
that are essentially all fish. As discussed earlier, FDA now considers limiting intake of total fat
unimportant; therefore, FDA does not intend to consider the "extra lean" criterion for total fat as
a factor in deciding whether to exercise its enforcement discretion. This approach is consistent
with the agency’s decision not to consider the "low fat" criterion as a factor in the exercise of its
enforcement discretion for use of the EPA/DHA qualified health claims in the labeling of
products that are essentially all fish.
E. Disqualifying Nutrient Levels
Under the general requirements that apply to all health claims, a food may not bear a health
claim if the total fat, saturated fat, cholesterol, or sodium content of the food exceeds the
disqualifying level for that nutrient (21 CFR 101.14(e)(3)). For individual foods, the
disqualifying nutrient levels are 13 g of total fat, 4 g of saturated fat, 60 mg of cholesterol and
480 mg of sodium per RACC, per label serving size, and per 50 g if the RACC is 30 g or less or
2 tablespoons or less (21 CFR 101.14(a)(4)).
Dietary supplements containing EPA and DHA in combination generally meet the disqualifying
nutrient levels per RACC and per labeled serving size, as they have small RACCs and serving
sizes. However, given the fact that most of these products are composed almost entirely of oil,
supplements containing EPA and DHA typically exceed the disqualifying nutrient levels per 50
g. Considering the small serving sizes of supplements containing EPA and DHA and the daily
intake amounts typically recommended in the labeling of these products, however, it is unlikely
that consumers would consume 50 g of dietary supplements containing combined EPA and DHA
under normal circumstances. Thus, FDA intends to exercise enforcement discretion for such
45
supplements with respect to the requirement for foods with a small RACC (30 g or less or two
tablespoons or less) to meet the disqualifying nutrient level for fat per 50 g as a condition of
bearing an EPA/DHA qualified health claim.
F. 10 Percent Minimum Nutrient Content Requirement
Under the general requirements for health claims, a conventional food may not bear a health
claim unless it contains, prior to any nutrient addition, at least 10 percent of the Daily Value for
vitamin A, vitamin C, iron, calcium, protein, or dietary fiber per RACC (see 21 CFR
101.14(e)(6)). The purpose of this provision is to prevent the use of health claims on foods of
minimal nutritional value.
Conventional Foods. The 10 percent minimum content requirement applies to conventional
foods that bear an EPA/DHA qualified health claim. The 10% minimum nutrient content
requirement per RACC for protein is 5 grams. Products that are essentially all fish almost
always contain more than 5 grams of protein per RACC. Thus, such products would generally
meet the minimum nutrient requirement because of their protein content. Other conventional
foods may or may not meet the requirement, depending on their composition. FDA does not
intend to exercise enforcement discretion for the use of EPA/DHA qualified health claims in the
labeling of conventional foods that do not meet the 10 percent minimum content requirement.
Dietary Supplements. The 10 percent minimum nutrient content requirement does not apply to
dietary supplements (21 CFR 101.14(e)(6)).
V. Conclusions
Based on FDA’s consideration of the scientific evidence submitted with the petition and other
pertinent scientific evidence, FDA concludes that the current scientific evidence is appropriate
for consideration of qualified health claims regarding intake of EPA and DHA in combination
and lowering blood pressure to reduce the risk of hypertension and CHD, provided that the
qualified health claims are appropriately worded to avoid misleading consumers.
The petition proposed the following model claims to be used on the labels or in the labeling of
conventional foods and dietary supplements that contain EPA and DHA in combination:
EPA and DHA help lower blood pressure in the general population.
EPA and DHA reduce BP, a risk factor for CHD (coronary heart disease).
EPA and DHA reduce the risk of CHD.
Research shows that EPA and DHA may be beneficial for moderating BP, a risk factor
for CHD.
46
Convincing scientific evidence indicates that EPA and DHA help lower blood pressure in
the general population, with comparable reductions to those achieved with other diet and
lifestyle interventions.
The first and last proposed claims discuss the possible blood pressure lowering effects of
combined intake of EPA and DHA without specifying the disease or health-related condition that
is the subject of the claim. Health claims are defined as claims that characterize the relationship
between a substance and a disease or health-related condition (21 CFR 101.14(a)(1)). Blood
pressure is neither a disease nor a health-related condition; rather, it is a biomarker that is a
diagnostic criterion for certain diseases and health-related conditions, such as hypertension,
CHD, and pre-hypertension. Blood pressure levels also affect the risk of certain diseases and
health-related conditions, including hypertension and CHD (see Section I.B, “Disease or Health-
Related Condition”). To meet the definition of a health claim, the proposed claims that mention
only blood pressure-lowering must be modified to describe the relationship between combined
intake of EPA and DHA and reducing the risk of hypertension or CHD.
The third proposed claim discusses EPA and DHA and reduced risk of CHD. As noted in
section II, the evidence included in the petition was for lowering blood pressure and reduced risk
of hypertension. Lowering blood pressure is one of many mechanisms for lowering the risk of
CHD. FDA did not evaluate the totality of the evidence for effects of combined EPA and DHA
intake on other surrogate endpoints for CHD such as blood (serum or plasma) total cholesterol,
blood (serum or plasma) LDL cholesterol, or other mechanisms of CHD risk. Rather, FDA
evaluated the totality of the evidence for effects of combined EPA and DHA intake and reducing
the risk of hypertension and CHD by lowering SBP and/or DBP. Therefore, to make the third
proposed claim complete and enable the public to comprehend the information provided, as
required by 21 CFR 101.14(d), we are modifying it to clarify that the mechanism for the possible
reduction in CHD risk is lowering blood pressure (see claim 3a, below). Alternatively, the same
scientific concept could be conveyed by saying that consuming EPA and DHA in combination
may reduce the risk of CHD by reducing the risk of hypertension (see claim 3b, below).
All of the proposed claims lack qualifying language disclosing the limited scientific evidence
supporting the claim. This information is necessary to prevent the claims from misleading
consumers. Adding it will inform consumers about the level of science supporting the claim and
prevent them from being misled about the strength of the supporting evidence. FDA also
believes that information on the amount of EPA and DHA in a product is necessary to enable
consumers to understand the claim and its significance in the context of their daily diet, as
required by 21 CFR 101.14(d)(2)(v). As discussed above in section IV.A, FDA intends to
consider, as a factor in the exercise of its enforcement discretion, that the amount of EPA and
DHA in the product be stated, either as a combined total or separately for each nutrient, on the
labels of conventional foods and dietary supplements that bear an EPA/DHA qualified health
claim. Because the nutrition labeling regulations do not permit the amount of EPA and DHA in
a food to be declared in the Nutrition Facts label, consumers will not be aware of how much EPA
and DHA is in a conventional food bearing the claim unless that information is included in the
claim itself. For dietary supplements, however, the amount of EPA and DHA per serving need
not be stated in the claim as long as it is declared in “Supplement Facts” in accordance with 21
47
CFR 101.36. Information on EPA and DHA content allows consumers to understand how their
food and dietary supplement choices and intake fit into the context of their total daily diet (see 21
CFR 101.14(d)(2)(v)).
The fifth and final proposed claim states,Convincing scientific evidence indicates that EPA and
DHA help lower blood pressure with comparable reductions to those achieved with other diet
and lifestyle interventions(emphasis added). Describing the level of evidence supporting the
claim that EPA and DHA help lower blood pressure as “convincing” is not accurate. That term
overstates and therefore mischaracterizes the strength of the evidence for a relationship between
EPA and DHA intake and reduced risk of hypertension through blood pressure reduction. As
discussed in section III of this letter, the evidence suggesting that intake of EPA and DHA in
combination from conventional foods and dietary supplements may reduce the risk of
hypertension by lowering blood pressure is inconclusive and highly inconsistent. Only a minority
of the studies (36 out of 104) showed a beneficial effect on blood pressure, and results were
inconsistent among studies across the entire range of study durations and levels of combined
EPA and DHA intake. Further, the proposed claim’s assertion that EPA and DHA produce
“comparable reductions [in blood pressure] to those achieved with other diet and lifestyle
interventionsis not supported by any credible scientific evidence. The studies from which
scientific conclusions could be drawn did not evaluate how the effects of consuming combined
EPA and DHA compare with the effects of other diet and lifestyle interventions on blood
pressure. Because the scientific evidence does not support portions of this proposed claim and
modifying the claim to exclude the unsubstantiated portions results in a claim nearly identical to
the first proposed claim, FDA is including only the remaining four proposed claims in its
consideration of enforcement discretion.
In light of the above considerations, FDA intends to consider the exercise of its enforcement
discretion for the following qualified health claims:
1. Consuming EPA and DHA combined may help lower blood pressure in the general
population and reduce the risk of hypertension. However, FDA has concluded that the evidence
is inconsistent and inconclusive. One serving of [name of the food or dietary supplement]
provides [ ] gram(s) of EPA and DHA.
2. Consuming EPA and DHA combined may reduce blood pressure and reduce the risk of
hypertension, a risk factor for CHD (coronary heart disease). However, FDA has concluded that
the evidence is inconsistent and inconclusive. One serving of [name of the food or dietary
supplement] provides [ ] gram(s) of EPA and DHA.
3a. Consuming EPA and DHA combined may reduce the risk of CHD (coronary heart disease)
by lowering blood pressure. However, FDA has concluded that the evidence is inconsistent and
inconclusive. One serving of [name of the food or dietary supplement] provides [ ] gram(s) of
EPA and DHA.
3b. Consuming EPA and DHA combined may reduce the risk of CHD (coronary heart disease)
by reducing the risk of hypertension. However, FDA has concluded that the evidence is
48
inconsistent and inconclusive. One serving of [name of the food or dietary supplement] provides
[ ] gram(s) of EPA and DHA.
4. Research shows that consuming EPA and DHA combined may be beneficial for moderating
blood pressure, a risk factor for CHD (coronary heart disease). However, FDA has concluded
that the evidence is inconsistent and inconclusive. One serving of [name of the food or dietary
supplement] provides [ ] gram(s) of EPA and DHA.
Dietary supplements need not declare the amount of EPA and DHA per serving in the claim if it
is declared in “Supplement Facts” in accordance with 21 CFR 101.36.
FDA intends to consider exercising its enforcement discretion for the above qualified health
claims when all enforcement discretion factors identified in Section IV of this letter are met.
Please note that scientific information is subject to change, as are consumer consumption
patterns. FDA intends to evaluate new information that becomes available to determine whether
it necessitates a change in this decision. For example, scientific evidence may become available
that will support significant scientific agreement, that will support a qualified health claim for
claims that were denied, that will no longer support the use of the above qualified health claims,
or that may raise safety concerns about the substances that are the subject of the claims.
Sincerely,
Douglas Balentine, Ph.D.
Director
Office of Nutrition and Food Labeling
Center for Food Safety
and Applied Nutrition
49
References
Agency for Healthcare Research and Quality, “Systems to rate the scientific evidence” (March
2002) [http://archive.ahrq.gov/clinic/epcsums/strengthsum.pdf] (accessed August 3, 2018).
Albert BB, Derraik JG, Brennan CM, Biggs JB, Garg ML, Cameron-Smith D, Hofman PL,
Cutfield WS. Supplementation with a blend of krill and salmon oil is associated with increased
metabolic risk in overweight men. Am J Clin Nutr 2015;102(1):49-57.
Alfaddagh A, Elajami TK, Ashfaque H, Saleh M, Bistrian BR, Welty FK. Effect of
Eicosapentaenoic and Docosahexaenoic Acids Added to Statin Therapy on Coronary Artery
Plaque in Patients with Coronary Artery Disease: A Randomized Clinical Trial. J Am Heart
Assoc 2017;6(12).
Ansari S, Djalali M, Mohammadzadeh Honarvar N, Mazaherioun M, Zarei M, Agh F,
Gholampour Z, Javanbakht MH. The Effect of n-3 Polyunsaturated Fatty Acids Supplementation
on Serum Irisin in Patients with Type 2 Diabetes: A Randomized, Double-Blind, Placebo-
Controlled Trial. Int J Endocrinol Metab 2017;15(1):e40614.
Augustine AH, Lowenstein LM, Harris WS, Shearer GC, Block RC. Treatment with omega-3
fatty acid ethyl-ester alters fatty acid composition of lipoproteins in overweight or obese adults
with insulin resistance. Prostaglandins Leukot Essent Fatty Acids 2014;90(2-3):69-75.
Barbosa MM, Melo AL, Damasceno NR. The benefits of omega-3 supplementation depend on
adiponectin basal level and adiponectin increase after the supplementation: A randomized
clinical trial. Nutrition 2017;34:7-13.
Borghi C, Cicero A. Omega-3-polyunsaturated fatty acids: Their potential role in blood
pressure prevention and management. Heart Int 2006;2:98-105.
Bosch J, Gerstein HC, Dagenais GR, Diaz R, Dyal L, Jung H, Maggiono AP, Probstfield J,
Ramachandran A, Riddle MC, Ryden LE, Yusuf S. n-3 fatty acids and cardiovascular outcomes
in patients with dysglycemia. N Engl J Med 2012;367(4):309-318.
Carvalho AP, Kimi Uehara S, Nogueria Netto JF, Rosa G. Hypocaloric diet associated with the
consumption of jam enriched with microencapsulated fish oil decreases insulin resistance. Nutr
Hosp 2014;29(5):1103-1108.
Chan DC, Pang J, Barrett PH, Sullivan DR, Burnett JR, van Bockxmeer FM, Watts GF. omega-3
Fatty Acid Ethyl Esters Diminish Postprandial Lipemia in Familial Hypercholesterolemia. J Clin
Endocrinol Metab 2016a;101(10):3732-3739.
50
Chan DC, Pang J, Barrett PH, Sullivan DR, Mori TA, Burnett JR, van Bockxmeer FM, Watts
GF. Effect of omega-3 fatty acid supplementation on arterial elasticity in patients with familial
hypercholesterolaemia on statin therapy. Nutr Metab Cardiovasc Dis 2016b;26(12):1140-1145.
Clark CM, Monahan KD, Drew RC. Omega-3 polyunsaturated fatty acid supplementation
attenuates blood pressure increase at onset of isometric handgrip exercise in healthy young and
older humans. Physiol Rep 2016;4(14).
Clark CM, Monahan KD, Drew RC. Omega-3 polyunsaturated fatty acid supplementation
reduces blood pressure but not renal vasoconstrictor response to orthostatic stress in healthy
older adults. Physiol Rep 2018;6(8):e13674.
Desai M, Stockbridge N, Temple R. Blood pressure as an example of a biomarker that functions
as a surrogate. AAPS J 2006;8(1):E146-152.
Elajami TK, Alfaddagh A, Lakshminarayan D, Soliman M, Chandnani M, Welty FK.
Eicosapentaenoic and Docosahexaenoic Acids Attenuate Progression of Albuminuria in Patients
with Type 2 Diabetes Mellitus and Coronary Artery Disease. J Am Heart Assoc 2017;6(7).
FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools)
Resource [Internet]. Silver Spring (MD): Food and Drug Administration (US); 2016-. Validated
Surrogate Endpoint. 2017 Sep. [https://www.ncbi.nlm.nih.gov/books/NBK453484/] (accessed
August 3, 2018).
Flegal K.M. "Evaluating Epidemiologic Evidence of the Effects of Food and Nutrient
Exposures". Am J Clin Nutr 1999;69:1339S-1344S.
Grieger JA, Miller MD, Cobiac L. Investigation of the effects of a high fish diet on inflammatory
cytokines, blood pressure, and lipids in healthy older Australians. Food Nutr Res 2014;58.
Harslof LB, Damsgaard CT, Hellgren LI, Andersen AD, Vogel U, Lauritzen L. Effects on
metabolic markers are modified by PPARG2 and COX2 polymorphisms in infants randomized to
fish oil. Genes Nutr. 2014;9(3):396.
Hill AB. The environment and disease: Association or causation? Proc R Soc Med 1965; 58:
295-300.
Hlais S, El-Bistami D, El Rahi B, Mattar MA, Obeid OA. Combined fish oil and high oleic
sunflower oil supplements neutralize their individual effects on the lipid profile of healthy men.
Lipids 2013;48(9):853-861.
IOM (Institute of Medicine, National Academy of Sciences). 2005a. Dietary Supplements: A
Framework for Evaluating Safety. Chapter 7, Categories of Scientific Evidence – In Vitro Data.
The National Academies Press, Washington, D.C.
51
IOM. 2005b. Dietary Reference Intakes for Water, Potassium, Sodium, Chloride, and Sulfate.
The National Academies Press, Washington, D.C.
IOM. 2005c. Dietary Supplements: A Framework for Evaluating Safety. Chapter 8, Interactions.
The National Academies Press, Washington, D.C.
James PA, Oparil S, Carter BL, Cushman WC, Dennison-Himmelfarb C, Handler J, Lackland D,
LeFevre M, MacKenzie T, Ogedegbe O, Smith S, Svetkey L, Taler S, Townsend R, Wright J,
Narva A, Ortiz E. 2014 Evidence-based guidelines for the management of high blood pressure
in adults. Report from the panel members appointed to the Eighth Joint National Committee
(JNC 8). J Am Med Assoc 2014;311:507-520.
Kristensen S, Schmidt EB, Schlemmer A, Rasmussen C, Lindgreen E, Johansen MB,
Christensen JH. The effect of marine n-3 polyunsaturated fatty acids on cardiac autonomic and
hemodynamic function in patients with psoriatic arthritis: a randomised, double-blind, placebo-
controlled trial. Lipids Health Dis 2016;15(1):216.
Kromhout D, Giltay EJ, Geleijnse JM, Alpha Omega Trial G. n-3 fatty acids and cardiovascular
events after myocardial infarction. N Engl J Med 2010;363(21):2015-2026.
Lind MV, Martino D, Harslof LB, Kyjovska ZO, Kristensen M, Lauritzen L. Genome-wide
identification of mononuclear cell DNA methylation sites potentially affected by fish oil
supplementation in young infants: A pilot study. Prostaglandins Leukot Essent Fatty Acids
2015;101:1-7.
Logan SL, Spriet LL. Omega-3 Fatty Acid Supplementation for 12 Weeks Increases Resting and
Exercise Metabolic Rate in Healthy Community-Dwelling Older Females. PLoS One
2015;10(12):e0144828.
Minihane AM, Armah CK, Miles EA, Madden JM, Clark AB, Caslake MJ, Packard CJ, Kofler
BM, Lietz G, Curtis PJ, Mathers JC, Williams CM, Calder PC. Consumption of Fish Oil
Providing Amounts of Eicosapentaenoic Acid and Docosahexaenoic Acid That Can Be Obtained
from the Diet Reduces Blood Pressure in Adults with Systolic Hypertension: A Retrospective
Analysis. J Nut 2016;146(3):516-523.
Moriyama T, Kumon S, Kamiyama T, Karasawa K, Uchida K, Nitta K. The Renoprotective
Effects of Docosahexaenoic Acid as an Add-on Therapy in Patients Receiving Eicosapentaenoic
Acid as Treatment for IgA Nephropathy: A Pilot Uncontrolled Trial. Intern Med 2018;57(2):173-
179.
Mozaffarian D, Wu J. Omega-3-fatty acids and cardiovascular disease: Effects on risk factors,
molecular pathways, and clinical events. J Am Coll Cardiol 2011;58:2047-2067.
52
NHLBI and the Seventh Report of the Joint National Committee on Prevention, Detection,
Evaluation and treatment of high blood pressure, 2003.
[http://www.nhlbi.nih.gov/guidelines/hypertension/express.pdf](accessed August 3, 2018).
Nodari S, Triggiani M, Campia U, Manerba A, Milesi G, Cesana BM, Gheorghiade M, Dei Cas
L. n-3 polyunsaturated fatty acids in the prevention of atrial fibrillation recurrences after
electrical cardioversion: a prospective, randomized study. Circulation 2011;124(10):1100-1106.
National Research Council. 2011. Reference Manual on Scientific Evidence: Third Edition. The
National Academies Press, Washington, DC, page 220.
O'Sullivan TA, Bremner AP, Beilin LJ, Ambrosini GL, Mori TA, Huang RC, Oddy WH.
Polyunsaturated fatty acid intake and blood pressure in adolescents. J Hum Hypertens
2012;26(3):178‐187.
Pase MP, Grima N, Cockerell R, Stough C, Scholey A, Sali A, Pipingas A. The effects of long-
chain omega-3 fish oils and multivitamins on cognitive and cardiovascular function: a
randomized, controlled clinical trial. J Am Coll Nutr 2015;34(1):21-31.
Patterson AC, Chalil A, Henao JJ, Streit IT, Stark KD. Omega-3-polyunsaturated fatty acid
blood biomarkers increase linearly in men and women after tightly controlled intakes of 0.25, 0.5
and 1 g/d of EPA and DHA. Nutr Res 2015;35:1040-1051.
Rasnake CM, Trumbo PR, Heinonen TM. Surrogate endpoints and emerging surrogate endpoints
for risk reduction of cardiovascular disease. Nutr Rev 2008;66(2):76-81.
Roe, Brian & S. Levy, Alan & M. Derby, Brenda. The Impact of Health Claims on Consumer
Search and Product Evaluation Outcomes: Results from FDA Experimental Data. Journal of
Public Policy and Marketing 1999;18: 89-105.
Roncaglioni MC, Tombesi M, Avanzini F, Barlera S, Caimi V, Longoni P, Marzona I, Milani V,
Silletta MG, Tognoni G, Marchioli R. n-3 fatty acids in patients with multiple cardiovascular risk
factors. N Engl J Med 2013;368(19):1800-1808.
Root M, Collier SR, Zwetsloot KA, West KL, McGinn MC. A randomized trial of fish oil
omega-3 fatty acids on arterial health, inflammation, and metabolic syndrome in a young healthy
population. Nutr J 2013;12:40.
Sempos CT, Liu K, Ernst ND. Food and nutrient exposures: what to consider when evaluating
epidemiologic evidence. Am J Clin Nutr 1999;69(suppl):1330S-8S.
Skilton MR, Raitakari OT, Celermajer DS. High intake of dietary long‐chain omega‐3 fatty acids
is associated with lower blood pressure in children born with low birth weight: NHANES 2003‐
2008. Hypertension 2013;61(5):972‐976.
53
Soares TS, Piovesan CH, Gustavo Ada S, Macagnan FE, Bodanese LC, Feoli AM. Alimentary
habits, physical activity, and Framingham global risk score in metabolic syndrome. Arq Bras
Cardiol 2014;102(4):374-382.
Spilker B. 1991. Guide to Clinical Studies. Raven Press, New York, New York, pp. 59-64,
pp.497-501 and pp. 793-800.
Su HY, Lee HC, Cheng WY, Huang SY. A calorie-restriction diet supplemented with fish oil
and high-protein powder is associated with reduced severity of metabolic syndrome in obese
women. Eur J Clin Nutr 2015;69(3):322-328.
Tavazzi L, Maggioni AP, Marchioli R, Barlera S, Franzosi MG, Latini R, Lucci D, Nicolosi GL,
Porcu M, Tognoni G, Gissi HFI. Effect of n-3 polyunsaturated fatty acids in patients with
chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial.
Lancet 2008;372(9645):1223-1230.
U.S. Department of Health and Human Services and U.S. Department of Agriculture. 2015–2020
Dietary Guidelines for Americans. 8th Edition. December 2015. Available at
http://health.gov/dietaryguidelines/2015/guidelines/.
U.S. Food and Drug Administration. "Guidance for Industry: Evidence-Based Review System
for the Scientific Evaluation of Health Claims," January 2009. Retrieved from:
https://www.fda.gov/regulatory-information/search-fda-guidance-documents/guidance-industry-
evidence-based-review-system-scientific-evaluation-health-claims.
U.S. Food and Drug Administration. “Memorandum to the File: List of Publications Excluded
from Review”, 2019.
U.S. Food and Drug Administration. “Memorandum to the File: Consumption estimates of EPA
and DHA for individuals 4 years and old in the United States”, 2018.
U.S. Food and Drug Administration. “Memorandum to Docket No. FDA-2014-Q-1146: Survey
of Comprehensive Reviews of the Effects of EPA and DHA Intake on Glycemic Control and
Blood Cholesterol”, 2019.
U.S. Food and Drug Administration. “Memorandum to Docket No. FDA-2014-Q-1146: Review
of Scientific Literature on EPA and DHA Intake and Risk of Excessive Bleeding”, 2019.
U.S. Department of Agriculture (USDA), Agricultural Research Service, Nutrient Data
Laboratory. USDA National Nutrient Database for Standard Reference, Legacy. Version
Current: April 2018. Internet: http://www.ars.usda.gov/nutrientdata.
Wilson EB. 1990. An Introduction to Scientific Research. Dover Publications, New York. pp.
46-48.
54
Witte AV, Kerti L, Hermannstadter HM, Fiebach JB, Schreiber SJ, Schuchardt JP, Hahn A, Floel
A. Long-chain omega-3 fatty acids improve brain function and structure in older adults. Cereb
Cortex 2014;24(11):3059-3068.
Wu SY, Mayneris-Perxachs J, Lovegrove JA, Todd S, Yaqoob P. Fish-oil supplementation alters
numbers of circulating endothelial progenitor cells and microparticles independently of eNOS
genotype. Am J Clin Nutr 2014;100(5):1232-1243.
Please See Docket No. FDA-2014-Q-1146 for the following publications:
Agouridis AP, Kostapanos MS, Tsimihodimos V, Kostara C, Mikhailidis DP, Bairaktari ET,
Tselepis AD, Elisaf MS. Effect of rosuvastatin monotherapy or in combination with fenofibrate
or (omega)-3 fatty acids on lipoprotein subfraction profile in patients with mixed dyslipidaemia
and metabolic syndrome. Int J Clin Prac 2012;66(9):843-853.
Alexopoulos E, Stangou M, Pantzaki A, Kirmizis D, Memmos D. Treatment of severe IgA
nephropathy with omega-3 fatty acids: The effect of a "very low dose" regimen. Ren Fail
2004;26(4):453-459.
Andreassen AK, Hartmann A, Offstad J, Geiran O, Kvernebo K, Simonsen S. Hypertension
prophylaxis with omega-3 fatty acids in heart transplant recipients. J AmColl Cardiol
1997;29(6):1324-1331.
Armstrong P, Kelley DS, Newman JW, Staggers FE, Sr., Hartiala J, Allayee H, Stephensen CB.
Arachidonate 5-lipoxygenase gene variants affect response to fish oil supplementation by healthy
African Americans. J Nutr 2012;142:1417-1428.
Arnarson A, Geirsdottir OG, Ramel A, Jonsson PV, Steingrimsdottir L, Thorsdottir I. [Dietary
habits and their association with blood pressure among elderly Icelandic people]. Laeknabladid
2012;98:515-520.
Asserhoj M, Nehammer S, Matthiessen J, Michaelsen KF, Lauritzen L. Maternal fish oil
supplementation during lactation may adversely affect long-term blood pressure, energy intake,
and physical activity of 7-year-old boys. J Nutr 2009;139(2):298-304.
Atar MJH, Hajianfar H, Bahonar A. The effects of omega-3 on blood pressure and the
relationship between serum visfatin level and blood pressure in patients with type II diabetes.
ARYA Atheroscler 2012;8:27-31.
Axelrod L, Camuso J, Williams E, Kleinman K, Briones E, Schoenfeld D .Effects of a small
quantity of omega-3 fatty acids on cardiovascular risk factors in NIDDM. A randomized,
prospective, double-blind, controlled study. Diabetes Care 1994;17:37-44.
55
Ayer JG1, Harmer JA, Xuan W, Toelle B, Webb K, Almqvist C, Marks GB, Celermajer DS.
Dietary supplementation with n-3 polyunsaturated fatty acids in early childhood: effects on blood
pressure and arterial structure and function at age 8 y. Am J Clin Nutr 2009;90:438-446.
Bach R, Schmidt U, Jung F, Kiesewetter H, Hennen B, Wenzel E, Schieffer H, Bette L, Heyden
S. Effects of fish oil capsules in two dosages on blood pressure, platelet functions,
haemorheological and clinical chemistry parameters in apparently healthy subjects. Ann Nutr
Metab 1989; 33:359-367.
Badalamenti S, Salerno F, Lorenzano E, Paone G, Como G, Finazzi S, Sacchetta AC, Rimola A,
Graziani G, Galmarini D, Ponticelli C. Renal effects of dietary supplementation with fish oil in
cyclosporine- treated liver transplant recipients. Hepatology 1995;22(6):1695-1701.
Badalamenti S, Salerno F, Salmeron JM, Lorenzano E, Rimola A, Gines P, Jimenez W, Graziani
G, Arroyo V, Rodes J, Ponticelli C. Lack of renal effects of fish oil administration in patients
with advanced cirrhosis and impaired glomerular filtration. Hepatology 1997;25(2):313-316.
Bairati I, Roy L, Meyer F. Effects of a fish oil supplement on blood pressure and serum lipids in
patients treated for coronary artery disease. Can J Cardiol 1992;8(1):41-46.
Barcelo-Coblijn G, Murphy EJ, Othman R, Moghadasian MH, Kashour T, Friel JK. Flaxseed oil
and fish-oil capsule consumption alters human red blood cell n-3 fatty acid composition: a
multiple-dosing trial comparing 2 sources of n-3 fatty acid. Am J Clin Nutr 2008;88:801-809.
Barden AE, Dunstan JA, Beilin LJ, Prescott SL, Mori TA. n -- 3 fatty acid supplementation
during pregnancy in women with allergic disease: effects on blood pressure, and maternal and
fetal lipids. Clin Sci (Colch) 2006;111(4):289-294.
Beegom R, Singh RB. Association of higher saturated fat intake with higher risk of hypertension
in an urban population of Trivandrum in South India. Int J Cardiol 1997;58(1):63-70.
Beilin LJ, Mori TA, Vandongen R, Morris J, Burke V, Ritchie J. The effects of omega-3 fatty
acids on blood pressure and serum lipids in men at increased risk of cardiovascular disease. J
Hypertens Suppl 1993;11(5):S318-319.
Benito P, Caballero J, Moreno J, Gutierrez-Alcantara C, Munoz C, Rojo G, Garcia S, Soriguer
FC. Effects of milk enriched with (omega)-3 fatty acid, oleic acid and folic acid in patients with
metabolic syndrome. Clinical Nutrition 2006;25(4):581-587.
Bennett WM, Carpenter CB, Shapiro ME, Strom TB, Hefty D, Tillman M, Abrams J, Ryan D,
Kelley VR. Delayed omega-3 fatty acid supplements in renal transplantation. A double-blind,
placebo-controlled study. Transplantation 1995;59(3):352-356.
56
Bhise A, Krishnan PV, Aggarwal R, Gaiha M, Bhattacharjee J. Effect of low-dose omega-3 fatty
acids substitution on blood pressure, hyperinsulinemia and dyslipidemia in Indians with essential
hypertension: A pilot study. Indian J Clin Biochem 2005;20(2):4-9.
Bilo HJ, Schaap GH, Slagt ME, Popp-Snijders C, Oe PL, Donker AJ. Protein intake variation
and omega-3 polyunsaturated fatty acids do not influence carbohydrate metabolism in patients
with chronic renal insufficiency. Current Thera Res 1988;44:292-303.
Bitzur R, Cohen H, Cohen T, Dror TW, Herzog Y, Lifshitz Y, Lubish T, Harats D, Rubinstein A.
The metabolic effects of omega-3 plant sterol esters in mixed hyperlipidemic subjects.
Cardiovasc Drugs Ther 2010;24(5-6):429-437.
Blonk MC, Bilo HJ, Nauta JJ, Popp-Snijders C, Mulder C, Donker AJ. Dose-response effects of
fish-oil supplementation in healthy volunteers. Am J Clin Nutr 1990;52(1):120-127.
Bønaa KH, Bjerve KS, Straume B, Gram IT, Thelle D.. Effect of eicosapentaenoic and
docosahexaenoic acids on blood pressure in hypertension. A population-based intervention trial
from the Tromsø study. N Engl J Med 1990;795-801.
Bonnema SJ, Jespersen LT, Marving J, Gregersen G. Supplementation with olive oil rather than
fish oil increases small arterial compliance in diabetic patients. Diabetes, Nutrition and
Metabolism Clinical and Experimental 1995;8:81-87.
Branten AJW, Klasen IS, Wetzels JFM. Short-term effects of fish oil treatment on urinary
excretion of high- and low-molecular weight proteins in patients with IgA nephropathy.
ClinNephrol 2002;58(4):267-274.
Browning LM, Krebs JD, Moore CS, Mishra GD, O'Connell MA, Jebb SA. The impact of long
chain n-3 polyunsaturated fatty acid supplementation on inflammation, insulin sensitivity and
CVD risk in a group of overweight women with an inflammatory phenotype. Diabetes Obes
Metab 2007; 9:70-80.
Bruckner G, Webb P, Greenwell L, Chow C, Richardson D. Fish oil increases peripheral
capillary blood cell velocity in humans. Atherosclerosis 1987;66:237-245.
Buckley JD, Burgess S, Murphy KJ, Howe PR. DHA-rich fish oil lowers heart rate during
submaximal exercise in elite Australian Rules footballers. J Sci Med Sport 2009;12:503-507.
Caron-Dorval D, Paquet P, Paradis AM, Rudkowska I, Lemieux S, Couture P, Vohl MC. Effect
of the PPAR-Alpha L162V polymorphism on the cardiovascular disease risk factor in response
to n-3 polyunsaturated fatty acids. J Nutrigenet Nutrigenomics 2008;1(4):205-212.
Carter JR, Schwartz CE, Yang H, Joyner MJ. Fish oil and neurovascular control in humans. Am
J Physiol Heart Circ Physiol 2012;303:H450-456.
57
Cazzola R, Russo-Volpe S, Miles EA, Rees D, Banerjee T, Roynette CE, Wells SJ, Goua M,
Wahle KW, Calder PC, Cestaro B. Age- and dose-dependent effects of an eicosapentaenoic acid-
rich oil on cardiovascular risk factors in healthy male subjects. Atherosclerosis 2007;193:159-
167.
Celik S, Doesch A, Erbel C, Blessing E, Ammon K, Koch A, Katus HA, Dengler TJ. Beneficial
effect of omega-3 fatty acids on sirolimus- or everolimus-induced hypertriglyceridemia in heart
transplant recipients. Transplantation 2008;86(2):245-250.
Chan DC, Watts GF, Barrett PHR, Beilin LJ, Redgrave TG, Mori TA. Regulatory effects of
HMG CoA reductase inhibitor and fish oils on apolipoprotein B-100 kinetics in insulin-resistant
obese male subjects with dyslipidemia. Diabetes 2002;51(8):2377-2386.
Chin JP, Dart AM. HBPRCA Astra Award. Therapeutic restoration of endothelial function in
hypercholesterolaemic subjects: effect of fish oils. Clin Exp Pharmacol Physiol
1994;21(10):749-755.
Chin JP, Gust AP, Nestel PJ, Dart AM. Marine oils dose-dependently inhibit vasoconstriction of
forearm resistance vessels in humans. Hypertension 1993; 21:22-28.
Cicero AF1, Derosa G, Di Gregori V, Bove M, Gaddi AV, Borghi C. Omega 3 polyunsaturated
fatty acids supplementation and blood pressure levels in hypertriglyceridemic patients with
untreated normal-high blood pressure and with or without metabolic syndrome: a retrospective
study. Clin Exp Hypertens 2010;32:137-144.
Cobiac L, Clifton PM, Abbey M, Belling GB, Nestel PJ. Lipid, lipoprotein, and hemostatic
effects of fish vs fish-oil n-3 fatty acids in mildly hyperlipidemic males. Am J Clin Nutr
1991b;53:1210-1216.
Cobiac L, Nestel PJ, Wing LM, Howe PR. A low-sodium diet supplemented with fish oil lowers
blood pressure in the elderly. J Hypertens 1992;10:87-92.
Cobiac L, Nestel PJ, Wing LM, Howe PR. Effects of dietary sodium restriction and fish oil
supplements on blood pressure in the elderly. Clin Exp Pharmacol Physiol 1991a;18:265-268.
Conquer JA, Cheryk LA, Chan E, Gentry PA, Holub BJ. Effect of supplementation with dietary
seal oil on selected cardiovascular risk factors and hemostatic variables in healthy male subjects.
Thromb Res 1999; 96:239-250.
Croset M, Vericel E, Rigaud M, Hanss M, Courpron P, Dechavanne M, Lagarde M. Functions
and tocopherol content of blood platelets from elderly people after low intake of purified
eicosapentaenoic acid. Thromb Res 1990;57:1-12.
58
Cussons AJ, Watts GF, Mori TA, Stuckey BGA. Omega-3 fatty acid supplementation decreases
liver fat content in polycystic ovary syndrome: a randomized controlled trial employing proton
magnetic resonance spectroscopy. J Clin Endocrinol Metab 2009;94(10):3842-3848.
Cutler JA, Whelton PK, Appel L, The Trials of Hypertension Prevention Collaborative Research
Group. The effects of nonpharmacologic interventions on blood pressure of persons with high
normal levels: Results of the trials of hypertension prevention, phase I. J Am Med Assoc
1992;267(9):1213-1220.
Damsgaard CT, Frokiaer H, Andersen AD, Lauritzen L. Fish oil in combination with high or low
intakes of linoleic acid lowers plasma triacylglycerols but does not affect other cardiovascular
risk markers in healthy men. J Nutr 2008;138(6):1061-1066.
Damsgaard CT, Schack-Nielsen L, Michaelsen KF, Fruekilde MB, Hels O, Lauritzen L. Fish oil
affects blood pressure and the plasma lipid profile in healthy Danish infants. J Nutr 2006(1):94-
99.
Dangardt F, Osika W, Chen Y, Nilsson U, Gan LM, Gronowitz E, Strandvik B, Friberg P.
Omega-3 fatty acid supplementation improves vascular function and reduces inflammation in
obese adolescents. Atherosclerosis 2010;212(2):580-585.
Dart AM, Riemersma RA, Oliver MF. Effects of Maxepa on serum lipids in
hypercholesterolaemic subjects. Atherosclerosis 1989;80:119-124.
Davidson MH, Bagdade JD, Liebson PR, Subbaiah P, Messer JV, Schoenberger JA.
Comparative effects of marine and olive oil dietary supplementation on coronary risk factors. J
Appl Cardiol 1989;4:145-151.
De Caterina R, Caprioli R, Giannessi D, Sicari R, Galli C, Lazzerini G, Bernini W, Carr L, Rindi
P. n-3 fatty acids reduce proteinuria in patients with chronic glomerular disease. Kidney
Int1993;44(4):843-850.
de Fijter CW, Popp-Snijders C, Oe LP, Tran DD, van der Meulen J, Donker AJ. Does additional
treatment with fish oil mitigate the side effects of recombinant human erythropoietin in dialysis
patients? Haematologica 1995;80(4):332-334.
de Jong C, Boehm G, Kikkert HK, Hadders-Algra M. The Groningen LCPUFA study: No effect
of short-term postnatal long-chain polyunsaturated fatty acids in healthy term infants on
cardiovascular and anthropometric development at 9 years. Pediatr Res 2011(4):411-416.
Deferne JL, Leeds AR. The antihypertensive effect of dietary supplementation with a 6-
desaturated essential fatty acid concentrate as compared with sunflower seed oil. J Hum
Hypertens1992;6(2):113-119.
59
Delarue J, Matzinger O, Binnert C, Schneiter P, Chiolero R, Tappy L. Fish oil prevents the
adrenal activation elicited by mental stress in healthy men. Diabetes Metab 2003;29:289-295.
Delgado-Pando G, Celada P, Sanchez-Muniz FJ, Jiménez-Colmenero F, Olmedilla-Alonso B.
Effects of improved fat content of frankfurters and pates on lipid and lipoprotein profile of
volunteers at increased cardiovascular risk: a placebo-controlled study. Eur J Nutr 2014;53:83-
93.
Demke DM, Peters GR, Linet OI, Metzler CM, Klott KA. Effects of a fish oil concentrate in
patients with hypercholesterolemia. Atherosclerosis 1988;70:73-80.
Derosa G, Cicero AF, Fogari E, D'Angelo A, Bonaventura A, Romano D, Maffioli P. Effects of
n-3 PUFAs on postprandial variation of metalloproteinases, and inflammatory and insulin
resistance parameters in dyslipidemic patients: Evaluation with euglycemic clamp and oral fat
load. J Clin Lipid 2012;6:553-564.
Derosa G, Maffioli P, D'Angelo A, Salvadeo SA, Ferrari I, Fogari E, Gravina A, Mereu R,
Randazzo S, Cicero AF. Effects of long chain omega-3 fatty acids on metalloproteinases and
their inhibitors in combined dyslipidemia patients. Expert Opin Pharmacother 2009;10:1239-
1247.
Deslypere JP. Influence of supplementation with N-3 fatty acids on different coronary risk
factors in men--a placebo controlled study. Verh K Acad Geneeskd Belg 1992;54:189-216.
Dewailly E E, Blanchet C, Gingras S, Lemieux S, Sauvé L, Bergeron J, Holub BJ. Relations
between n-3 fatty acid status and cardiovascular disease risk factors among Quebecers. Am J
Clin Nutr 2001;74:603-611.
Dewell A, Marvasti FF, Harris WS, Tsao P, Gardner CD. Low- and high-dose plant and marine
(n-3) fatty acids do not affect plasma inflammatory markers in adults with metabolic syndrome. J
Nutr 2011;141:2166-2171.
DiGiacomo RA, Kremer JM, Shah DM. Fish-oil dietary supplementation in patients with
Raynaud's phenomenon: a double-blind, controlled, prospective study. Am J Med
1989;86(2):158-164.
Dixit MP, Dixit NM, Scott K. Managing Henoch-Schonlein purpura in children with fish oil and
ACE inhibitor therapy. Nephrology 2004;9(6):381-386.
Doenyas-Barak K, Berman S, Abu-Hamad R, Golik A, Rahimi-Levene N, Efrati S. N-3 fatty
acid supplementation to routine statin treatment inhibits platelet function, decreases patients'
daytime blood pressure, and improves inflammatory status. Eur J Clin Pharmacol
2012;68(8):1139-1146.
60
Dokholyan RS, Albert CM, Appel LJ, Cook NR, Whelton P, Hennekens CH. A trial of omega-3
fatty acids for prevention of hypertension. Am J Cardiol 2004;93(8):1041-1043.
Donadio Jr JV, Bergstralh EJ, Offord KP, Spencer DC, Holley KE. A controlled trial of fish oil
in IgA nephropathy. N Engl J Med 1994;331(18):1194-1199.
Donadio JV, Jr., Grande JP, Bergstralh EJ, Dart RA, Larson TS, Spencer DC. The long-term
outcome of patients with IgA nephropathy treated with fish oil in a controlled trial. Mayo
Nephrology Collaborative Group. J Am Soc Nephrol 1999;10(8):1772-1777.
Donnelly SM, Ali MA, Churchill DN. Effect of n-3 fatty acids from fish oil on hemostasis, blood
pressure, and lipid profile of dialysis patients. J Am Soc Nephrol 1992;2(11):1634-1639.
Du Plooy WJ, Venter CP, Muntingh GM, Venter HL, Glatthaar II, Smith KA. The cumulative
dose response effect of eicosapentaenoic and docosahexaenoic acid on blood pressure, plasma
lipid profile and diet pattern in mild to moderate essential hypertensive black patients.
Prostaglandins LeukotEssent Fatty Acids 1992;46:315-321.
Dusing R, Struck A, Gobel BO, Weisser B, Vetter H. Effects of n-3 fatty acids on renal function
and renal prostaglandin E metabolism. Kidney Intl 1990;38(2):315-319.
Dyerberg J, Christensen JH, Eskesen D, Astrup A, Stender S. Trans, and n-3 polyunsaturated
fatty acids and vascular function-a yin yang situation? Atherosclerosis Suppl 2006; 7:33-35.
Dyerberg J, Eskesen DC, Andersen PW, Astrup A, Buemann B, Christensen JH, Clausen P,
Rasmussen BF, Schmidt EB, Tholstrup T, Toft E, Toubro S, Stender S. Effects of trans- and n-3
unsaturated fatty acids on cardiovascular risk markers in healthy males. An 8 weeks dietary
intervention study. Eur J Clin Nutr 2004;58:1062-1070.
Ebbesson SO, Tejero ME, Nobmann ED, Lopez-Alvarenga JC, Ebbesson L, Romenesko T,
Carter EA, Resnick HE, Devereux RB, MacCluer JW, Dyke B, Laston SL, Wenger CR, Fabsitz
RR, Comuzzie AG, Howard BV. Fatty acid consumption and metabolic syndrome components:
the GOCADAN study. J Cardiometab Syndr 2007;2:244-249.
Ebrahimi M, Ghayour-Mobarhan M, Rezaiean S, Hoseini M, Parizade SM, Farhoudi F,
Hosseininezhad SJ, Tavallaei S, Vejdani A, Azimi-Nezhad M, Shakeri MT, Rad MA, Mobarra
N, Kazemi-Bajestani SM, Ferns GA. Omega-3 fatty acid supplements improve the
cardiovascular risk profile of subjects with metabolic syndrome, including markers of
inflammation and auto-immunity. Acta Cardiol 2009;64(3):321-327.
Erkkila AT, Schwab US, de Mello VDF, Lappalainen T, Mussalo H, Lehto S, Kemi V, Lamberg-
Allardt C, Uusitupa MI. Effects of fatty and lean fish intake on blood pressure in subjects with
coronary heart disease using multiple medications. Eur J Nutr 2008;47(6):319-328.
61
Ernst E, Saradeth T, Achhammer G. n-3 fatty acids and acute-phase proteins. Eur J Clin Invest
1991;21(1):77-82.
Ernst E, Matrai A. The effect of omega-3-fatty acids on blood rheology in hyperlipoproteinemias
- A pilot study. Med Sci Res 1988;16:69-70.
Faghihi T, Jahed A, Mahmoudi-Gharaei J, Sharifi V, Akhondzadeh S, Ghaeli P. Role of Omega-
3 fatty acids in preventing metabolic disturbances in patients on olanzapine plus either sodium
valproate or lithium: A randomized double-blind placebo-controlled trial. Daru 2012;20:43.
Fahs CA, Yan H, Ranadive S, Rossow LM, Agiovlasitis S, Wilund KR, Fernhall B. The effect of
acute fish-oil supplementation on endothelial function and arterial stiffness following a high-fat
meal. Appl Physiol Nutr Metab 2010;35(3):294-302.
Farrell DJ. Enrichment of hen eggs with n-3 long-chain fatty acids and evaluation of enriched
eggs in humans. Am J Clin Nutr 1998;68(3):538-544.
Ferraro PM, Ferraccioli GF, Gambaro G, Fulignati P, Costanzi S. Combined treatment with
renin-angiotensin system blockers and polyunsaturated fatty acids in proteinuric IgA
nephropathy: a randomized controlled trial. Nephrol Dial Transplant 2009;24(1):156-160.
Fiedler R, Mall M, Wand C, Osten B. Short-term administration of omega-3 fatty acids in
hemodialysis patients with balanced lipid metabolism. J Ren Nutr 2005;15(2):253-256.
Finnegan YE, Minihane AM, Leigh-Firbank EC, Kew S, Meijer GW, Muggli R, Calder PC,
Williams CM. Plant- and marine-derived n-3 polyunsaturated fatty acids have differential effects
on fasting and postprandial blood lipid concentrations and on the susceptibility of LDL to
oxidative modification in moderately hyperlipidemic subjects. Am J Clin Nutr 2003;77:783-795.
Flaten H, Hostmark AT, Kierulf P, Lystad E, Trygg K, Bjerkedal T, Osland A. Fish-oil
concentrate: effects on variables related to cardiovascular disease. Am J Clin Nutr 1990;52:300-
306.
Freund-Levi Y, Eriksdotter-Jonhagen M, Cederholm T, Basun H, Faxen-Irving G, Garlind A,
Vedin I, Vessby B, Wahlund LO, Palmblad J. Omega-3 fatty acid treatment in 174 patients with
mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch
Neurol 2006;63:1402-1408.
Gans RO, Bilo HJ, Weersink EG, Rauwerda JA, Fonk T, Popp-Snijders C, Donker AJ. Fish oil
supplementation in patients with stable claudication. Am J Surg 1990;160(5):490-495.
Geelen A, Zock PL, Swenne CA, Brouwer IA, Schouten EG, Katan MB. Effect of n-3 fatty acids
on heart rate variability and baroreflex sensitivity in middle-aged subjects. Am Heart J
2003;146:344.
62
Geppert J, Demmelmair H, Hornstra G, Koletzko B. Co-supplementation of healthy women with
fish oil and evening primrose oil increases plasma docosahexaenoic acid, gamma-linolenic acid
and dihomo-gamma-linolenic acid levels without reducing arachidonic acid concentrations. Br J
Nutr 2008;99(2):360-369.
Geppert J, Kraft V, Demmelmair H, Koletzko B. Microalgal docosahexaenoic acid decreases
plasm triacylglycerol in normolipidaemic vegetarians: a randomised trial. Br J Nutr
2006;95(4):779-786.
Ginty AT, Conklin SM. Preliminary evidence that acute long-chain omega-3 supplementation
reduces cardiovascular reactivity to mental stress: A randomized and placebo controlled trial.
Biol Psychol 2012;89:269-272.
Godin C, Shatenstein B, Paradis G, Kosatsky T. Absence of cardiovascular benefits and sportfish
consumption among St. Lawrence River anglers. Environ Res 2003;93(3):241-247.
Gray DR, Gozzip CG, Eastham JH, Kashyap ML. Fish oil as an adjuvant in the treatment of
hypertension. Pharmacotherapy 1996;16(2):295-300.
Grimsgaard S, Bonaa KH, Hansen JB, Myhre ES. Effects of highly purified eicosapentaenoic
acid and docosahexaenoic acid on hemodynamics in humans. Am J Clin Nutr 1998 68:52-59.
Grossman E, Peleg E, Shiff E, Rosenthal T. Hemodynamic and neurohumoral effects of fish oil
in hypertensive patients. Am J Hypertens 1993;6(12):1040-1045.
Grundt H, Nilsen DW, Hetland O, Aarsland T, Baksaas I, Grande T, Woie L. Improvement of
serum lipids and blood pressure during intervention with n-3 fatty acids was not associated with
changes in insulin levels in subjects with combined hyperlipidaemia. J Intern Med 1995
237(3):249-259.
Gulseth HL, Gjelstad IM, Tierney AC, Shaw DI, Helal O, Hees AM, Delgado-Lista J,
Leszczynska-Golabek I, Karlstrom B, Lovegrove J, Defoort C, Blaak EE, Lopez-Miranda J,
Dembinska-Kiec A, Riserus U, Roche HM, Birkeland KI, Drevon CA. Dietary fat modifications
and blood pressure in subjects with the metabolic syndrome in the LIPGENE dietary intervention
study. Br J Nutr 2010;104(2):160-163.
Gustafsson IB, Ohrvall M, Ekstrand B, Vessby B. Moderate amounts of n-3 fatty acid enriched
seafood products are effective in lowering serum triglycerides and blood pressure in healthy
subjects. J Hum Nutr Diet 1996;135-45.
Hackman RM, Havel PJ, Schwartz HJ, Rutledge JC, Watnik MR, Noceti EM, Stohs SJ, Stern JS,
Keen CL. Multinutrient supplement containing ephedra and caffeine causes weight loss and
improves metabolic risk factors in obese women: a randomized controlled trial. Int J Obes
2006(10):1545-1556.
63
Hagiwara H, Nishiyama Y, Katayama Y. Effects of eicosapentaenoic acid on asymmetric
dimethylarginine in patients in the chronic phase of cerebral infarction: a preliminary study. J
Stroke Cerebrovasc Dis 2011;20(5):474-478.
Haglund O, Wallin R, Luostarinen R, Saldeen T. Effects of a new fluid fish oil concentrate,
ESKIMO-3, on triglycerides, cholesterol, fibrinogen and blood pressure. J Intern Med
1990;227:347-353.
Haiden M, Miyasaka Y, Kimura Y, Tsujimoto S, Maeba H, Suwa Y, Iwasaka T, Shiojima I.
Effect of eicosapentaenoic acid on regional arterial stiffness: Assessment by tissue Doppler
imaging. World J Cardiol 2012;4(8):256-259.
Haines AP, Sanders TA, Imeson JD, Mahler RF, Martin J, Mistry M, Vickers M, Wallace PG.
Effects of a fish oil supplement on platelet function, haemostatic variables and albuminuria in
insulin-dependent diabetics. Thromb Res1986;43:643-655.
Hall WL, Sanders KA, Sanders TAB, Chowienczyk PJ. A high-fat meal enriched with
eicosapentaenoic acid reduces postprandial arterial stiffness measured by digital volume pulse
analysis in healthy men. J Nutr 2008;138(2):287-291.
Hallund J, Madsen BO, Bugel SH, Jacobsen C, Jakobsen J, Krarup H, Holm J, Nielsen HH,
Lauritzen L. The effect of farmed trout on cardiovascular risk markers in healthy men. Br J Nutr
2010;104:1528-1536.
Hamazaki T, Nakazawa R, Tateno S, Shishido H, Isoda K, Hattori Y, Yoshida T, Fujita T, Yano
S, Kumagai A. Effects of fish oil rich in eicosapentaenoic acid on serum lipid in hyperlipidemic
hemodialysis patients. Kidney Intl 1984;26(1):81-84.
Hamazaki T, Takazakura E, Osawa K, Urakaze M, Yano S. Reduction in microalbuminuria in
diabetics by eicosapentaenoic acid ethyl ester. Lipids 1990;25:541-545.
Harris WS, Lemke SL, Hansen SN, Goldstein DA, DiRienzo MA, Su H, Nemeth MA, Taylor
ML, Ahmed G, George C. Stearidonic acid-enriched soybean oil increased the omega-3 index,
an emerging cardiovascular risk marker. Lipids 2008;43:805-811.
Harrison RA, Sagara M, Rajpura A, Armitage L, Birt N, Birt CA, Yamori Y. Can foods with
added soya-protein or fish-oil reduce risk factors for coronary disease? A factorial randomised
controlled trial. Nutr Metab Cardiovas 2004;14:344-350.
Hellsten G, Boman K, Saarem K, Hallmans G, Nilsson TK. Effects on fibrinolytic activity of
corn oil and a fish oil preparation enriched with omega-3-polyunsaturated fatty acids in a long-
term study. Curr Med Res Opin 1993;13:133-139.
64
Hendra TJ, Britton ME, Roper DR, Wagaine-Twabwe D, Jeremy JY, Dandona P, Haines AP,
Yudkin JS. Effects of fish oil supplements in NIDDM subjects. Controlled study. Diabetes Care
1990;13:821-829.
Hill AM, Buckley JD, Murphy KJ, Howe PR. Combining fish-oil supplements with regular
aerobic exercise improves body composition and cardiovascular disease risk factors. Am J Clin
Nutr 2007;85:1267-1274.
Holm T, Andreassen AK, Aukrust P, Andersen K, Geiran OR, Kjekshus J, Simonsen S,
Gullestad L. Omega-3 fatty acids improve blood pressure control and preserve renal function in
hypertensive heart transplant recipients. Eur Heart J 2001;22(5):428-436.
Holman RR, Paul S, Farmer A, Tucker L, Stratton IM, Neil HA. Atorvastatin in Factorial with
Omega-3 EE90 Risk Reduction in Diabetes (AFORRD): a randomised controlled trial.
Diabetologia 2009;52:50-59.
Howe PR, Lungershausen YK, Cobiac L, Dandy G, Nestel PJ. Effect of sodium restriction and
fish oil supplementation on BP and thrombotic risk factors in patients treated with ACE
inhibitors. J Hum Hypertens1994;8(1):43-49.
Huang T, Bhulaidok S, Cai Z, Xu T, Xu F, Wahlqvist ML, Li D. Plasma phospholipids n-3
polyunsaturated fatty acid is associated with metabolic syndrome. Mol Nutr Food Res
2010;54(11):1628-1635.
Huang T, Shou T, Cai N, Wahlqvist ML, Li D. Associations of plasma n-3 polyunsaturated fatty
acids with blood pressure and cardiovascular risk factors among Chinese. Int J Food Sci Nutr
2012;63(6):667-673.
Hughes GS, Jr., Ringer TV, Francom SF, Caswell KC, DeLoof MJ, Spillers CR. Effects of fish
oil and endorphins on the cold pressor test in hypertension. Clin Pharmacol Ther 1991;50:538-
546.
Hughes GS, Ringer TV, Watts KC, DeLoof MJ, Francom SF, Spillers CR. Fish oil produces an
atherogenic lipid profile in hypertensive men. Atherosclerosis 1990;84:229-237.
Iketani T, Takazawa K, Yamashina A. Effect of eicosapentaenoic acid on central systolic blood
pressure. Prostaglandins Leukot Essent Fatty Acids 2013;88(2):191-195.
Jain S, Gaiha M, Bhattacharjee J, Anuradha S. Effects of low-dose omega-3 fatty acid
substitution in type-2 diabetes mellitus with special reference to oxidative stress--a prospective
preliminary study. J Assoc Physicians India 2002;50:1028-1033.
Jarvinen R, Knekt P, Rissanen H, Reunanen A. Intake of fish and long-chain n-3 fatty acids and
the risk of coronary heart mortality in men and women. Br J Nutr 2006;95(4):824-829.
65
Jolly SE, Eilat-Adar S, Wang H, Mete M, Fabsitz RR, Devereux RB, Ebbesson SO, Umans JG,
Howard BV. Sex-specific associations of nutrition with hypertension and systolic blood pressure
in Alaska Natives findings from the GOCADAN study. Int J Circumpolar Health 2011;70:254-
65.
Kagawa Y, Nishizawa M, Suzuki M, Miyatake T, Hamamoto T, Goto K, Motonaga E,
Izumikawa H, Hirata H, Ebihara A. Eicosapolyenoic acids of serum lipids of Japanese islanders
with low incidence of cardiovascular diseases. J Nutr Sci Vitaminol 1982;28:441-453.
Kappus RM, Curry CD, McAnulty S, Welsh J, Morris D, Nieman DC, Soukup J, Collier SR. The
effects of a multiflavonoid supplement on vascular and hemodynamic parameters following
acute exercise. Oxid Med Cell Longev 2011:210798. doi: 10.1155/2011/210798.
Kasim SE, Stern B, Khilnani S, McLin P, Baciorowski S, Jen KL. Effects of omega-3 fish oils
on lipid metabolism, glycemic control, and blood pressure in type II diabetic patients. J Clin
Endocrinol Metab 1988;67:1-5.
Kelley DS, Siegel D, Vemuri M, Mackey BE. Docosahexaenoic acid supplementation improves
fasting and postprandial lipid profiles in hypertriglyceridemic men. Am J Clin Nutr 2007;86:
324-333.
Kenny D, Warltier DC, Pleuss JA, Hoffmann RG, Goodfriend TL, Egan BM. Effect of omega-3
fatty acids on the vascular response to angiotensin in normotensive men. Am J Cardiol
1992;70:1347-1352.
Kestin M, Clifton P, Belling GB, Nestel PJ. n-3 fatty acids of marine origin lower systolic blood
pressure and triglycerides but raise LDL cholesterol compared with n-3 and n-6 fatty acids from
plants. Am J Clin Nutr 1990;51:1028-1034.
Kho TL, Leunissen KM, Wirtz JJ, van den Dries PJ, Noordzij TZ, Rijnders A, Heidendahl GA,
Lijnen PJ, van Hooff JP. Cyclosporine and urinary prostaglandins.
Transplant Proc 1989;21(1 Pt 2):1504-1505.
Kim SH, Kim MK, Lee HY, Kang HJ, Kim YJ, Kim HS. Prospective randomized comparison
between omega-3 fatty acid supplements plus simvastatin versus simvastatin alone in Korean
patients with mixed dyslipidemia: Lipoprotein profiles and heart rate variability. Eur J Clin Nutr
2011;65(1):110-116.
Knapp HR, FitzGerald GA. The antihypertensive effects of fish oil. A controlled study of
polyunsaturated fatty acid supplements in essential hypertension. N Engl J Med 1989;320:1037-
1043.
Kristensen SD, Schmidt EB, Andersen HR, Dyerberg J. Fish oil in angina pectoris.
Atherosclerosis 1987;64(1):13-19.
66
Lahoz C, Alonso R, Porres A, Mata P. [Diets enriched with monounsaturated fatty acids and
omega-3 polyunsaturated fatty acids decrease blood pressure without changing the plasma
insulin concentration in healthy subjects]. Med Clin 1999;112(4):133-137.
Laidlaw M, Holub BJ. Effects of supplementation with fish oil-derived n-3 fatty acids and
gamma-linolenic acid on circulating plasma lipids and fatty acid profiles in women. Am J Clin
Nutr 2003;77(1):37-42.
Landmark K, Thaulow E, Hysing J, Mundal HH, Eritsland J, Hjermann I. Effects of fish oil,
nifedipine and their combination on blood pressure and lipids in primary hypertension. J Hum
Hypertens 1993;7:25-32.
Lara JJ, Economou M, Wallace AM, Rumley A, Lowe G, Slater C, Caslake M, Sattar N, Lean
ME. Benefits of salmon eating on traditional and novel vascular risk factors in young, non-obese
healthy subjects. Atherosclerosis 2007;193:213-221.
Larnkjaer A, Christensen JH, Michaelsen KF, Lauritzen L. Maternal fish oil supplementation
during lactation does not affect blood pressure, pulse wave velocity, or heart rate variability in
2.5-y-old children. J Nutr 2006;136(6):1539-1544.
Leary SD, Ness AR, Emmett PM, Davey Smith G, Headley JE, Team AS. Maternal diet in
pregnancy and offspring blood pressure. Arch Dis Child 2005;90(5):492-493.
Leng GC, Lee AJ, Fowkes FG, Jepson RG, Lowe GD, Skinner ER, Mowat BF. Randomized
controlled trial of gamma-linolenic acid and eicosapentaenoic acid in peripheral arterial disease.
Clin Nutr 1998;17(6):265-271.
Leng GC, Smith FB, Fowkes FG, Horrobin DF, Ells K, Morse-Fisher N, Lowe GD. Relationship
between plasma essential fatty acids and smoking, serum lipids, blood pressure and haemostatic
and rheological factors. Prostaglandins Leukot Essent Fatty Acids 1994;51(2):10110-8.
Levinson PD, Iosiphidis AH, Saritelli AL, Herbert PN, Steiner M. Effects of n-3 fatty acids in
essential hypertension. Am J Hypertens 1990;3:754-760.
Lindqvist HM, Langkilde AM, Undeland I, Sandberg AS. Herring (Clupea harengus) intake
influences lipoproteins but not inflammatory and oxidation markers in overweight men. Br J
Nutr 2009;101:383-390.
Liu JC, Conklin SM, Manuck SB, Yao JK, Muldoon MF. Long-chain omega-3 fatty acids and
blood pressure. Am J Hypertens 2011;24(10):1121-1126.
Lofgren RP, Wilt TJ, Nichol KL, Crespin L, Pluhar R, Eckfeldt J. The effect of fish oil
supplements on blood pressure. Am J Pub Health 1993;83:267-269.
67
Lok CE, Moist L, Hemmelgarn BR, Tonelli M, Vazquez MA, Dorval M, Oliver M, Donnelly S,
Allon M, Stanley K. Effect of fish oil supplementation on graft patency and cardiovascular
events among patients with new synthetic arteriovenous hemodialysis grafts: a randomized
controlled trial. J Am Med Assoc 2012;307(17):1809-1816.
Lungershausen YK, Abbey M, Nestel PJ, Howe PR. Reduction of blood pressure and plasma
triglycerides by omega-3 fatty acids in treated hypertensives. J Hypertens 1994;12(9):1041-1045.
Lungershausen YK, Howe PR, Clifton PM, Hughes CR, Phillips P, Graham JJ, Thomas DW.
Evaluation of an omega-3 fatty acid supplement in diabetics with microalbuminuria. Ann N Y
Acad Sci 1997;827:369-381.
Lungershausen YK, Howe PR. Improved detection of a blood pressure response to dietary
intervention with 24-hour ambulatory monitoring. Am J Hypertens 1994;7(12):1115-1117.
Mackness MI, Bhatnagar D, Durrington PN, Prais H, Haynes B, Morgan J, Borthwick L. Effects
of a new fish oil concentrate on plasma lipids and lipoproteins in patients with
hypertriglyceridaemia. Eur J Clin Nutr 1994;48:859-865.
Madden J, Brunner A, Dastur ND, Tan RM, Nash GB, Rainger GE, Shearman CP, Calder PC,
Grimble RF. Fish oil induced increase in walking distance, but not ankle brachial pressure index,
in peripheral arterial disease is dependent on both body mass index and inflammatory genotype.
Prostaglandins Leukot Essent Fatty Acids 2007;76(6):331-340.
Maki KC, Reeves MS, Farmer M, Griinari M, Berge K, Vik H, Hubacher R, Rains TM. Krill oil
supplementation increases plasma concentrations of eicosapentaenoic and docosahexaenoic acids
in overweight and obese men and women. Nutr Res 2009;29:609-615.
Manor I, Magen A, Keidar D, Rosen S, Tasker H, Cohen T, Richter Y, Zaaroor-Regev D, Manor
Y, Weizman A. Safety of phosphatidylserine containing omega3 fatty acids in ADHD children:
A double-blind placebo-controlled trial followed by an open-label extension. Eur Psychiatry
2013;28:386-391..
Margolin G, Huster G, Glueck CJ, Speirs J, Vandegrift J, Illig E, Wu J, Streicher P, Tracy T.
Blood pressure lowering in elderly subjects: a double-blind crossover study of omega-3 and
omega-6 fatty acids. Am J Clin Nutr 1991;53(2):562-572.
Mas E, Woodman RJ, Burke V, Puddey IB, Beilin LJ, Durand T, Mori TA. The omega-3 fatty
acids EPA and DHA decrease plasma F2- isoprostanes: Results from two placebo-controlled
interventions. Free Radic Res2010;44(9):983-990.
Matsumura K, Noguchi H, Nishi D, Matsuoka Y. The effect of omega-3 fatty acids on
psychophysiological assessment for the secondary prevention of posttraumatic stress disorder: an
open-label pilot study. Glob J Health Sci 2012;4(1):3-9.
68
Mayneris-Perxachs J, Bondia-Pons I, Serra-Majem L, Lopez-Sabater MC. Long-chain n-3 fatty
acids and classical cardiovascular disease risk factors among the Catalan population. Food Chem
2010;119:54-61.
McVeigh GE, Brennan GM, Cohn JN, Finkelstein SM, Hayes RJ, Johnston GD. Fish oil
improves arterial compliance in non-insulin-dependent diabetes mellitus. Arterioscler Thromb
1994;14:1425-1429.
McVeigh GE, Brennan GM, Johnston GD, McDermott BJ, McGrath LT, Henry WR, Andrews
JW, Hayes JR. Dietary fish oil augments nitric oxide production or release in patients with type 2
(non-insulin-dependent) diabetes mellitus. Diabetologia 1993;36(1):33-38.
Mehta JL, Lopez LM, Lawson D, Wargovich TJ, Williams LL. Dietary supplementation with
omega-3 polyunsaturated fatty acids in patients with stable coronary heart disease. Effects on
indices of platelet and neutrophil function and exercise performance. Am J Med 1988;84(1):45-
52.
Meland E, Fugelli P, Laerum E, Ronneberg R, Sandvik L. Effect of fish oil on blood pressure
and blood lipids in men with mild to moderate hypertension. Scand J Prim Health Care
1989;7:131-135.
Meyer BJ, Lane AE, Mann NJ. Comparison of seal oil to tuna oil on plasma lipid levels and
blood pressure in hypertriglyceridaemic subjects. Lipid 2009;44(9):827-835.
Micallef MA, Garg ML. Anti-inflammatory and cardioprotective effects of n-3 polyunsaturated
fatty acids and plant sterols in hyperlipidemic individuals. Atherosclerosis 2009;204:476-482.
Mills DE, Mah M, Ward RP, Morris BL, Floras JS. Alteration of baroreflex control of forearm
vascular resistance by dietary fatty acids. Amer J Physiol 1990;259:R1164-1171.
Mills DE, Prkachin KM, Harvey KA, Ward RP. Dietary fatty acid supplementation alters stress
reactivity and performance in man. J Hum Hypertens 1989;3:111-116.
Mirmiran P, Hosseinpour-Niazi S, Naderi Z, Bahadoran Z, Sadeghi M, Azizi F. Association
between interaction and ratio of (omega)-3 and (omega)-6 polyunsaturated fatty acid and the
metabolic syndrome in adults. Nutrition 2012;28(9):856-863.
Miyajima T, Tsuijino T, Saito K, Yokoyama M. Effects of eicosapentaenoic acid on blood
pressure, cell membrane fatty acids, and intracellular sodium concentration in essential
hypertension. Hypertens Res 2001;24(5):537-542.
Monahan KD, Wilson TE, Ray CA. Omega-3 fatty acid supplementation augments sympathetic
nerve activity responses to physiological stressors in humans. Hypertension 2004;44:732-738.
69
Monge-Rojas R, Campos H, Rojas XF. Saturated and cis- and trans-unsaturated fatty acids intake
in rural and urban Costa Rican adolescents. J Am Coll Nutr 2005;24(4):286-293.
Mori TA, Bao DQ, Burke V, Puddey IB, Beilin LJ. Docosahexaenoic acid but not
eicosapentaenoic acid lowers ambulatory blood pressure and heart rate in humans. Hypertension
1999;34(2):253-260.
Mori TA, Burke V, Puddey I, Irish A, Cowpland CA, Beilin L, Dogra G, Watts GF. The effects
of [omega]3 fatty acids and coenzyme Q10 on blood pressure and heart rate in chronic kidney
disease: a randomized controlled trial. J Hypertens 2009;27(9):1863-1872.
Mori TA, Woodman RJ, Burke V, Puddey IB, Croft KD, Beilin LJ. Effect of eicosapentaenoic
acid and docosahexaenoic acid on oxidative stress and inflammatory markers in treated-
hypertensive type 2 diabetic subjects. Free Radic Biol Med 2003;35(7):772-781.
Moriyama T, Iwasaki C, Tanaka K, Ochi A, Shimizu A, Shiohira S, Itabashi M, Takei T, Uchida
K, Tsuchiya K, Nitta K. Effects of combination therapy with renin-angiotensin
system inhibitors and eicosapentaenoic acid on IgA nephropathy. Intern Med 2013;52(2):193-
199.
Morris MC, Manson JE, Rosner B, Buring JE, Willett WC, Hennekens CH. Fish consumption
and cardiovascular disease in the physicians' health study: a prospective study. Am J Epidemiol
1995;142(2):166-175.
Morris MC, Taylor JO, Stampfer MJ, Rosner B, Sacks FM. The effect of fish oil on blood
pressure in mild hypertensive subjects: a randomized crossover trial. Am J Clin Nutr 1993;57:59-
64.
Mortensen JZ, Schmidt EB, Nielsen AH, Dyerberg J. The effect of N-6 and N-3 polyunsaturated
fatty acids on hemostasis, blood lipids and blood pressure. Thromb Haemost 1983;50:543-46.
Mundal HH, Gjesdal K, Landmark K. The effect of N-3 fatty acids and nifedipine on platelet
function in hypertensive males. Thromb Res 1993;72:257-262.
Murphy KJ, Meyer BJ, Mori TA, Burke V, Mansour J, Patch CS, Tapsell LC, Noakes M, Clifton
PA, Barden A, Puddey IB, Beilin LJ, Howe PR. Impact of foods enriched with n-3 long-chain
polyunsaturated fatty acids on erythrocyte n-3 levels and cardiovascular risk factors. Br J Nutr
2007;97:749-757.
Neff LM, Culiner J, Cunningham-Rundles S, Seidman C, Meehan D, Maturi J, Wittkowski KM,
Levine B, Breslow JL. Algal docosahexaenoic acid affects plasma lipoprotein particle size
distribution in overweight and obese adults. J Nutr 2011;141:207-213.
70
Ness AR, Whitley E, Burr ML, Elwood PC, Smith GD, Ebrahim S. The long-term effect of
advice to eat more fish on blood pressure in men with coronary disease: results from the diet and
reinfarction trial. J Hum Hypertens 1999;13(11):729-733.
Nestel P, Shige H, Pomeroy S, Cehun M, Abbey M, Raederstorff D. The n-3 fatty acids
eicosapentaenoic acid and docosahexaenoic acid increase systemic arterial compliance in
humans. Am J Clin Nutr 2002;76:326-330.
Nilsson A, Radeborg K, Salo I, Bjorck I. Effects of supplementation with n-3 polyunsaturated
fatty acids on cognitive performance and cardiometabolic risk markers in healthy 51 to 72 years
old subjects: A randomized controlled cross-over study. Nutr J 2012;11:99.
Nishizawa H, Hamazaki K, Hamazaki T, Fujioka S, Sawazaki S. The relationship between tissue
RBC n-3 fatty acids and pulse wave velocity. In Vivo 2006;20(2):307-310.
Nordoy A, Hansen JB, Brox J, Svensson B.Effects of atorvastatin and omega-3 fatty acids on
LDL subfractions and postprandial hyperlipemia in patients with combined hyperlipemia. Nutr
Metab Cardiovas 2001;11:7-16.
Noreen EE, Brandauer J. The effects of supplemental fish oil on blood pressure and morning
cortisol in normotensive adults: a pilot study. J Compl Integr Med 2012;9.
Norris PG, Jones CJ, Weston MJ. Effect of dietary supplementation with fish oil on systolic
blood pressure in mild essential hypertension. Br Med J 1986;293:104-105.
Novgorodtseva TP, Kantur TA, Karaman YK, Antonyuk MV, Zhukova NV. Modification of
fatty acids composition in erythrocytes lipids in arterial hypertension associated with
dyslipidemia. Lipids Health Dis 2011;10:18.
Oda E, Hatada K, Kimura J, Aizawa Y, Thanikachalam PV, Watanabe K. Relationships between
serum unsaturated fatty acids and coronary risk factors: negative relations between nervonic acid
and obesity-related risk factors. Int Heart J 2005;46(6):975-985.
Oh SY, Ryue J, Hsieh CH, Bell DE. Eggs enriched in omega-3 fatty acids and alterations in lipid
concentrations in plasma and lipoproteins and in blood pressure. Am J Clin Nutr 1991;54:689-
695.
O'Keefe JH, Jr., Abuissa H, Sastre A, Steinhaus DM, Harris WS. Effects of omega-3 fatty acids
on resting heart rate, heart rate recovery after exercise, and heart rate variability in men with
healed myocardial infarctions and depressed ejection fractions. Am J Cardiol 2006;97(8):1127-
1130.
Oken E, Ning Y, Rifas-Shiman SL, Rich-Edwards JW, Olsen SF, Gillman MW. Diet during
pregnancy and risk of preeclampsia or gestational hypertension. Ann Epidemiol 2007;17(9):663-
668.
71
Olendzki BC, Leung K, Van Buskirk S, Reed G, Zurier RB. Treatment of rheumatoid arthritis
with marine and botanical oils: Influence on serum lipids. Evid Based Complement Alternat Med
2011;2011:827286.
Olivieri O, Negri M, De Gironcoli M, Bassi A, Guarini P, Stanzial AM, Grigolini L, Ferrari S,
Corrocher R. Effects of dietary fish oil on malondialdehyde production and glutathione
peroxidase activity in hyperlipidaemic patients. Scand J Clin Lab Invest 1988;48:659-665.
Paniagua JA, Perez-Martinez P, Gjelstad IM, Tierney AC, Delgado-Lista J, Defoort C, Blaak EE,
Riserus U, Drevon CA, Kiec-Wilk B, Lovegrove JA, Roche HM, Lopez-Miranda J, Investigators
LS. A low-fat high-carbohydrate diet supplemented with long-chain n-3 PUFA reduces the risk
of the metabolic syndrome. Atherosclerosis 2011;218(2):443-450.
Parinyasiri U, Ong-Ajyooth L, Parichatikanond P, Ong-Ajyooth S, Liammongkolkul S, Kanyog
S. Effect of fish oil on oxidative stress, lipid profile and renal function in IgA nephropathy. J
Med Assoc Thai 2004;87(2):143-149.
Park Y, Oh SH, Rhee MY. Association between 24-hour ambulatory blood pressure and
erythrocyte n-3 polyunsaturated fatty acids in Korean subjects with hypertension. Nutr Res
2010;30(12):807-814.
Passfall J, Philipp T, Woermann F, Quass P, Thiede M, Haller H. Different effects of
eicosapentaenoic acid and olive oil on blood pressure, intracellular free platelet calcium, and
plasma lipids in patients with essential hypertension. Clin Investigator 1993;71:628-633.
Pettersson EE, Rekola S, Berglund L, Sundqvist KG, Angelin B, Diczfalusy U, Bjorkhem I,
Bergstrom J. Treatment of IgA nephropathy with omega-3-polyunsaturated fatty acids: a
prospective, double-blind, randomized study. Clin Nephtrol Invest 1994;41(4):183-190.
Prisco D, Paniccia R, Bandinelli B, Filippini M, Francalanci I, Giusti B, Giurlani L, Gensini GF,
Abbate R, Neri Serneri GG. Effect of medium-term supplementation with a moderate dose of n-3
polyunsaturated fatty acids on blood pressure in mild hypertensive patients. Thromb Res
1998;91:105-112.
Radack K, Deck C, Huster G. The effects of low doses of n-3 fatty acid supplementation on
blood pressure in hypertensive subjects. A randomized controlled trial. Arch Intern Med
1991;151:1173-1180.
Radaelli A, Cazzaniga M, Viola A, Balestri G, Janetti MB, Signorini MG, Castiglioni P,
Azzellino A, Mancia G, Ferrari AU. Enhanced Baroreceptor Control of the Cardiovascular
System by Polyunsaturated Fatty Acids in Heart Failure Patients. J Am CollCardiol
2006;48(8):1600-1606.
72
Ramel A, Martinez JA, Kiely M, Bandarra NM, Thorsdottir I. Moderate consumption of fatty
fish reduces diastolic blood pressure in overweight and obese European young adults during
energy restriction. Nutrition 2010;26:168-174.
Rasmussen BM, Vessby B, Uusitupa M, Berglund L, Pedersen E, Riccardi G, Rivellese AA,
Tapsell L, Hermansen K, Group KS. Effects of dietary saturated, monounsaturated, and n-3 fatty
acids on blood pressure in healthy subjects. Am J Clin Nutr 2006;83(2):221-226.
Rizza S, Tesauro M, Cardillo C, Galli A, Iantorno M, Gigli F, Sbraccia P, Federici M, Quon MJ,
Lauro D. Fish oil supplementation improves endothelial function in normoglycemic offspring of
patients with type 2 diabetes. Atherosclerosis 2009;206(2):569-574.
Rogers S, James KS, Butland BK. Effects of a fish oil supplement on serum lipids, blood
pressure, bleeding time, haemostatic and rheological variables. A double blind randomised
controlled trial in healthy volunteers. Atherosclerosis 1987;63(2-3):137-143.
Rolf N, Tenschert W, Lison AE. Results of a long-term administration of omega-3 fatty acids in
haemodialysis patients with dyslipoproteinaemia. Nephrol Dial Transplant 1990;5(9):797-801.
Rontoyanni VG, Hall WL, Pombo-Rodrigues S, Appleton A, Chung R, Sanders TAB. A
comparison of the changes in cardiac output and systemic vascular resistance during exercise
following high-fat meals containing DHA or EPA. Br J Nutr 2012;108(3):492-499.
Rose EL, Holub BJ. Effects of a liquid egg product containing fish oil on selected cardiovascular
disease risk factors: a randomized cross over trial. Food Res Int 2006;39:910-916.
Rossing P, Hansen BV, Nielsen FS, Myrup B, Holmer G, Parving HH. Fish oil in diabetic
nephropathy. Diabetes Care 1996;19(11):1214-1219.
Russo C, Olivieri O, Girelli D, Azzini M, Stanzial AM, Guarini P, Friso S, De Franceschi L,
Corrocher R. Omega-3 polyunsaturated fatty acid supplements and ambulatory blood pressure
monitoring parameters in patients with mild essential hypertension. J Hypertens 1995;13(12 Pt
2):1823-1826.
Rytter D, Christensen JH, Bech BH, Schmidt EB, Henriksen TB, Olsen SF. The effect of
maternal fish oil supplementation during the last trimester of pregnancy on blood pressure, heart
rate and heart rate variability in the 19-year-old offspring. Br J Nutr 2012;108(8):1475-1483.
Ryu J, Lerner J, Sullivan JM. Unresponsiveness of forearm hemodynamics to omega-3
polyunsaturated fatty acids and aspirin. Prostaglandins 1990;39:339-347.
Sacks FM, Hebert P, Appel LJ, Borhani NO, Applegate WB, Cohen JD, Cutler JA, Kirchner KA,
Kuller LH, Roth KJ, Taylor JO, Hennekens CH. Short report: the effect of fish oil on blood
pressure and high-density lipoprotein-cholesterol levels in phase I of the Trials of Hypertension
Prevention. J Hypertens 1994b (2):209-213.
73
Sacks FM, Hebert P, Appel LJ, Borhani NO, Applegate, WB, Cohen JD, Cutler JA, Kirchner
KA, Kuller LH, Roth KJ, Taylor JO, Hennekens CH. Short Report: The effect of fish oil on
blood pressure and high-density lipoprotein-cholesterol levels in phase I of the Trials of
Hypertension Prevention. Trials of Hypertension Prevention Collaborative Research Group. J
Hypertens Suppl 1994a;12(7):S23-31.
Sagara M, Njelekela M, Teramoto T, Taguchi T, Mori M, Armitage L, Birt N, Birt C, Yamori Y.
Effects of docosahexaenoic acid supplementation on blood pressure, heart rate, and serum lipids
in Scottish men with hypertension and hypercholesterolemia. Int J Hypertens 2011;2011:1-7.
Sakamoto N, Nishiike T, Iguchi H, Sakamoto K. Effects of eicosapentaenoic acid intake on
plasma fibrinolytic and coagulation activity by using physical load in the young. Nutrition
2000;16(1):11-14.
Sanders TA, Gleason K, Griffin B, Miller GJ. Influence of an algal triacylglycerol containing
docosahexaenoic acid (22 : 6n-3) and docosapentaenoic acid (22 : 5n-6) on cardiovascular risk
factors in healthy men and women. Br J Nutr 2006;95:525-31.
Sanders TA, Vickers M, Haines AP. Effect on blood lipids and haemostasis of a supplement of
cod-liver oil, rich in eicosapentaenoic and docosahexaenoic acids, in healthy young men. Clin
Sci 1981;61:317-324.
Sanders TA, Hall WL, Maniou Z, Lewis F, Seed PT, Chowienczyk PJ. Effect of low doses of
long-chain n-3 PUFAs on endothelial function and arterial stiffness: a randomized controlled
trial. Am J Clin Nutr 2011;94:973-980.
Sanders TAB, Hinds A. The influence of a fish oil high in docosahexaenoic acid on plasma
lipoprotein and vitamin E concentrations and haemostatic function in healthy male volunteers. Br
J Nutr 1992;68(1):163-173.
Sanders TAB, Vickers M, Haines AP. Effect on blood lipids and haemostasis of a supplement of
cod-liver oil, rich in eicosapentaenoic and docosahexaenoic acids, in healthy young men. Clin
Sci 1981;61(3):317-324.
Santos J, Queiros J, Silva F, Cabrita A, Rodrigues A, Henriques AC, Sarmento AM, Pereira MC,
Guimaraes S. Effects of fish oil in cyclosporine-treated renal transplant recipients.
Transplantation Proceedings 2000;32(8):2605-2608.
Satoh N, Shimatsu A, Kotani K, Himeno A, Majima T, Yamada K, Suganami T, Ogawa Y.
Highly purified eicosapentaenoic acid reduces cardio-ankle vascular index in association with
decreased serum amyloid A-LDL in metabolic syndrome. Hypertens Res 2009;32(11):1004-
1008.
74
Schaefer EJ, Lichtenstein AH, Lamon-Fava S, Contois JH, Li Z, Goldin BR, Rasmussen H,
McNamara JR, Ordovas JM. Effects of National Cholesterol Education Program Step 2 diets
relatively high or relatively low in fish-derived fatty acids on plasma lipoproteins in middle-aged
and elderly subjects. Am J Clin Nutr 1996;63(2):234-241.
Schmidt EB, Lervang HH, Varming K, Madsen P, Dyerberg J. Long-term supplementation with
n-3 fatty acids, I: Effect on blood lipids, haemostasis and blood pressure. Scand J Clin Lab Invest
1992;52(3):221-228.
Schmidt EB, Nielsen LK, Pederson JO, Kornerup HJ, Dyerberg J. The effect of n-3
polyunsaturated fatty acids on lipids, platelet function, coagulation, fibrinolysis and monocyte
chemotaxis in patients with hypertension. Clinica Chimica Acta 1990;189(1):25-32.
Schmitz PG, McCloud LK, Reikes ST, Leonard CL, Gellens ME. Prophylaxis of hemodialysis
graft thrombosis with fish oil: double-blind, randomized, prospective trial. J Am Soc Nephrol
2002;13(1):184-190.
Shah AP, Ichiuji AM, Han JK, Traina M, El-Bialy A, Meymandi SK, Wachsner RY.
Cardiovascular and endothelial effects of fish oil supplementation in healthy volunteers. J
Cardiovasc Pharmacol Ther 2007;12(3):213-219.
Shimizu H, Ohtani K, Tanaka Y, Sato N, Mori M, Shimomura Y. Long-term effect of
eicosapentaenoic acid ethyl (EPA-E) on albuminuria of non-insulin dependent diabetic patients.
Diabetes Res Clin Pract 1995;28(1):35-40.
Simao AN, Lozovoy MA, Bahls LD, Morimoto HK, Simao TN, Matsuo T, Dichi I. Blood
pressure decrease with ingestion of a soya product (kinako) or fish oil in women with the
metabolic syndrome: role of adiponectin and nitric oxide. Br J Nutr 2012;108:1435-42.
Simao ANC, Lozovoy MAB, Simao TNC, Dichi JB, Matsuo T, Dichi I. Nitric oxide
enhancement and blood pressure decrease in patients with metabolic syndrome using soy protein
or fish oil. Arq Bras Endocrinol Metabol 2010;54(6):540-545.
Simpson LO, McQueen EG, Shand BI, Herbison GP, Campbell AJ. Changes in red cell shape in
healthy elderly subjects taking low dose fish oil: pilot study. N Z Med J 1991;104:316-318.
Singer P, Berger I, Luck K, Taube C, Naumann E, Godicke W. Long-term effect of mackerel
diet on blood pressure, serum lipids and thromboxane formation in patients with mild essential
hypertension. Atherosclerosis 1986a;62:259-265.
Singer P, Berger I, Wirth M, Godicke W, Jaeger W, Voigt S. Slow desaturation and elongation
of linoleic and alpha-linolenic acids as a rationale of eicosapentaenoic acid-rich diet to lower
blood pressure and serum lipids in normal, hypertensive and hyperlipemic subjects.
Prostaglandins Leukot Med 1986b;24(2-3):173-193.
75
Singer P, Jaeger W, Voigt S, Thiel H. Defective desaturation and elongation of n-6 and n-3 fatty
acids in hypertensive patients. Prostaglandins Leukot Med 1984;15:159-165.
Singer P, Jaeger W, Wirth M. Lipid and blood pressure-lowering effect of mackerel diet in man.
Atherosclerosis 1983;49(1):99-108.
Singer P, Melzer S, Goschel M, Augustin S. Fish oil amplifies the effect of propranolol in mild
essential hypertension. Hypertension 1990b;16(6):682-691.
Singer P, Wirth M, Berger I. Influence on serum lipids, lipoproteins and blood pressure of
mackerel and herring diet in patients with type IV and V hyperlipoproteinemia. Atherosclerosis
1985b;56(1):111-118.
Singer P, Wirth M, Godicke W, Heine H. Blood pressure lowering effect of eicosapentaenoic
acid-rich diet in normotensive, hypertensive and hyperlipemic subjects. Experientia
1985c;41(4):462-464.
Singer P, Wirth M, Kretschmer H. Different changes of n-6 fatty acids in lipoproteins from
hyperlipemic subjects after diets supplemented with n-3 fatty acids. Prostaglandins Leukot
Essent Fatty Acids 1991;42(2):107-111.
Singer P, Wirth M, Voigt S. Blood pressure- and lipid-lowering effect of mackerel and herring
diet in patients with mild essential hypertension. Atherosclerosis 1985a;56(2):223-235.
Singer P, Wirth M, Voigt S. Clinical studies on the lipid and blood pressure lowering effect of an
eicosapentaenoic acid-rich diet. Biomedica Biochimica Acta 1984a;43(8-9):S421-S425.
Singer P. Different changes of N-6 fatty acids in lipoproteins from hyperlipaemic subjects after
diets supplemented with N-3 fatty acids. Aktuelle Ernahrungsmedizin Klinik und Praxis
1993;18(5):368-372.
Singer P. Low-sodium/high-potassium preparation of canned mackerel lowers blood pressure
more effectively than conventional canned mackerel. Nutr Res 1990a;10(9):949-964.
Sirtori CR, Paoletti R, Mancini M, Crepaldi G, Manzato E, Rivellese A, Pamparana F,
Stragliotto E. N-3 fatty acids do not lead to an increased diabetic risk in patients with
hyperlipidemia and abnormal glucose tolerance. Italian Fish Oil Multicenter Study. Am J Clin
Nutr 1997;65:1874-1881.
Sjoberg NJ, Milte CM, Buckley JD, Howe PR, Coates AM, Saint DA. Dose-dependent increases
in heart rate variability and arterial compliance in overweight and obese adults with DHA-rich
fish oil supplementation. Br J Nutr 2010;103:243-248.
76
Skulas-Ray AC, Kris-Etherton PM, Harris WS, West SG. Effects of marine-derived omega-3
fatty acids on systemic hemodynamics at rest and during stress: a dose-response study. Ann
Behav Med 2012;44(3):301-308.
Stark KD, Holub BJ. Differential eicosapentaenoic acid elevations and altered cardiovascular
disease risk factor responses after supplementation with docosahexaenoic acid in
postmenopausal women receiving and not receiving hormone replacement therapy. Am J Clin
Nutr 2005; 79:765-773.
Steiner A, Oertel R, Battig B, Pletscher W, Weiss B, Greminger P, Vetter W. Effect of fish oil on
blood pressure and serum lipids in hypertension and hyperlipidaemia. J Hypertens 1989;7:S73-
S76.
Surai PF, MacPherson A, Speake BK, Sparks NH. Designer egg evaluation in a controlled trial.
Eur J Clin Nutr 2000;54(4):298-305.
Svensson M, Christensen JH, Solling J, Schmidt EB. The effect of n-3 fatty acids on plasma
lipids and lipoproteins and blood pressure in patients with CRF. Am J Kidney Dis
2004;44(1):77-83.
Szabo de Edelenyi F, Vergnaud AC, Ahluwalia N, Julia C, Hercberg S, Blacher J, Galan P.
Effect of B-vitamins and n-3 PUFA supplementation for 5 years on blood pressure in patients
with CVD. Br J of Nutr 2012;107(6):921-927.
Takaki A, Umemoto S, Ono K, Seki K, Ryoke T, Fujii A, Itagaki T, Harada M, Tanaka M,
Yonezawa T, Ogawa H, Matsuzaki M. Add-on therapy of EPA reduces oxidative stress and
inhibits the progression of aortic stiffness in patients with coronary artery disease and statin
therapy: a randomized controlled study. J Atheroscler Thromb 2011;18(10):857-866.
Theobald HE, Goodall AH, Sattar N, Talbot DC, Chowienczyk PJ, Sanders TA. Low-dose
docosahexaenoic acid lowers diastolic blood pressure in middle-aged men and women. J Nutr
2007;137:973-978.
Titova OE, Sjogren P, Brooks SJ, Kullberg J, Ax E, Kilander L, Riserus U, Cederholm T,
Larsson EM, Johansson L, Ahlström H, Lind L, Schiöth HB, Benedict C. Dietary intake of
eicosapentaenoic and docosahexaenoic acids is linked to gray matter volume and cognitive
function in elderly. Age 2013;35(4):1495-14505.
Toft I, Bønaa KH, Ingebretsen OC, Nordøy A, Jenssen T. Effects of n-3 polyunsaturated fatty
acids on glucose homeostasis and blood pressure in essential hypertension. A randomized,
controlled trial. Ann Intern Med 1995;123(12):911-918.
Ueshima H, Stamler J, Elliott P, Chan Q, Brown IJ, Carnethon MR, Daviglus ML, He K, Moag-
Stahlberg A, Rodriguez BL, Steffen LM, Van Horn L, Yarnell J, Zhou B. Food omega-3 fatty
77
acid intake of individuals (total, linolenic acid, long-chain) and their blood pressure:
INTERMAP study. Hypertension 2007;50:313-319.
Ulbak J, Lauritzen L, Hansen HS, Michaelsen KF. Diet and blood pressure in 2.5-y-old Danish
children. Am J Clin Nutr 2004;79(6):1095-1102.
Urakaze M, Hamazaki T, Kashiwabara H, Omori K, Fischer S, Yano S, Kumagai A. Favorable
effects of fish oil concentrate on risk factors for thrombosis in renal allograft recipients. Nephron
1989b;53(2):102-109.
Urakaze M, Hamazaki T, Yano S, Kashiwabara H, Oomori K, Yokoyama T. Effect of fish oil
concentrate on risk factors of cardiovascular complications in renal transplantation.
TransplantProc1989a;21(1 Pt 2):2134-2136.
Vakhapova V, Richter Y, Cohen T, Herzog Y, Korczyn AD. Safety of phosphatidylserine
containing omega-3 fatty acids in non-demented elderly: a double-blind placebo-controlled trial
followed by an open-label extension. BMC Neurol 2011;11:79.
Valdivielso P, Rioja J, Garcia-Arias C, Sanchez-Chaparro MA, Gonzalez-Santos P. Omega 3
fatty acids induce a marked reduction of apolipoprotein B48 when added to fluvastatin in
patients with type 2 diabetes and mixed hyperlipidemia: a preliminary report. Cardiovasc
Diabetol 2009;8:1.
van der Heide JJ, Bilo HJ, Donker JM, Wilmink JM, Tegzess AM. Effect of dietary fish oil on
renal function and rejection in cyclosporine-treated recipients of renal transplants. N Engl J Med
1993;329(11):769-773.
van der Heide JJ, Bilo HJ, Tegzess AM, Donker AJ. The effects of dietary supplementation with
fish oil on renal function in cyclosporine-treated renal transplant recipients. Transplantation
1990;49(3):523-527.
van der Heide JJ, Bilo HJ, Donker AJ, Wilmink JM, Sluiter WJ, Tegzess AM. The effects of
dietary supplementation with fish oil on renal function and the course of early postoperative
rejection episodes in cyclosporine-treated renal transplant recipients. Transplantation
1992;54(2):257-263.
Vandongen R, Mori TA, Burke V, Beilin LJ, Morris J, Ritchie J. Effects on blood pressure of
omega 3 fats in subjects at increased risk of cardiovascular disease. Hypertension
1993;22(3):371-379.
van Rossem L, Wijga AH, de Jongste JC, Koppelman GH, Oldenwening M, Postma DS,
Abrahamse-Berkeveld M, van de Heijning B, Brunekreef B, Smit HA. Blood pressure in 12-
year-old children is associated with fatty acid composition of human milk: the prevention and
incidence of asthma and mite allergy birth cohort. Hypertension 2012;60(4):1055-1060.
78
van Woudenbergh GJ, van Ballegooijen AJ, Kuijsten A, Sijbrands EJ, van Rooij FJ, Geleijnse
JM, Hofman A, Witteman JC, Feskens EJ. Eating fish and risk of type 2 diabetes: A population-
based, prospective follow-up study. Diabetes Care 2009;32(11):2021-2026.
Ventura HO, Milani RV, Lavie CJ, Smart FW, Stapleton DD, Toups TS, Price HL.
Cyclosporine-induced hypertension. Efficacy of omega-3 fatty acids in patients after cardiac
transplantation. Circulation 1993;88(5 Pt 2):II281-285.
Vericel E, Calzada C, Chapuy P, Lagarde M. The influence of low intake of n-3 fatty acids on
platelets in elderly people. Atherosclerosis 1999;147:187-192.
Vernaglione L, Cristofano C, Chimienti S. Omega-3 polyunsaturated fatty acids and proxies of
cardiovascular disease in hemodialysis: a prospective cohort study. J Nephrol 2008;21(1):99-
105.
von Houwelingen R, Nordoy A, van der Beek E, Houtsmuller U, de Metz M, Hornstra G. Effect
of a moderate fish intake on blood pressure, bleeding time. hematology, and clinical chemistry in
healthy males. Am J Clin Nutr 1987;46:424-436.
Walser B, Giordano RM, Stebbins CL. Supplementation with omega-3 polyunsaturated fatty
acids augments brachial artery dilation and blood flow during forearm contraction. Eur J Appl
Physiol 2006;97:347-354.
Walser B, Stebbins CL. Omega-3 fatty acid supplementation enhances stroke volume and cardiac
output during dynamic exercise. Eur J Appl Physiol 2008;104:455-461.
Wang S, Ma AQ, Song SW, Quan QH, Zhao XF, Zheng XH. Fish oil supplementation improves
large arterial elasticity in overweight hypertensive patients. Eur J Clin Nutr 2008;62:1426-1431.
Wennberg M, Bergdahl IA, Stegmayr B, Hallmans G, Lundh T, Skerfving S, Strömberg U,
Vessby B, Jansson JH. Fish intake, mercury, long-chain n-3 polyunsaturated fatty acids and risk
of stroke in northern Sweden. Br J Nutr 2007;98(5):1038-1045.
Weisser B, Struck A, Gobel BO, Vetter H, Dusing R. Fish oil and baroreceptor function in man.
Klin Wochenschr 1990;68 Suppl 20:49-52.
Wing LM, Nestel PJ, Chalmers JP, Rouse I, West MJ, Bune AJ, Tonkin AL, Russell AE. Lack of
effect of fish oil supplementation on blood pressure in treated hypertensives. J Hypertens
1990;8(4):339-343.
Wong AT, Chan DC, Barrett PH, Adams LA, Watts GF. Supplementation with n3 Fatty Acid
Ethyl Esters Increases Large and Small Arterial Elasticity in Obese Adults on a Weight Loss
Diet. J Nutr 2013;143;437-441.
79
Wong CY, Yiu KH, Li SW, Tam S, Lau CP, Tse HF. Fish-oil supplement has neutral effects on
vascular and metabolic function but improves renal function in patients with Type 2 diabetes
mellitus. Diabet Med 2010;27:54-60.
Woodman RJ, Mori TA, Burke V, Puddey IB, Watts GF, Beilin LJ. Effects of purified
eicosapentaenoic and docosahexaenoic acids on glycemic control, blood pressure, and serum
lipids in type 2 diabetic patients with treated hypertension. Am Clin Nutr 2002;76:1007-1015.
Xun P, Hou N, Daviglus M, Liu K, Morris JS, Shikany JM, Sidney S, Jacobs DR, He K. Fish oil,
selenium and mercury in relation to incidence of hypertension: a 20-year follow-up study. J
Intern Med 2011;270:175-186.
Yam D, Friedman J, Bott-Kanner G, Genin I, Shinitzky M, Klainman E. Omega-3 fatty acids
reduce hyperlipidaemia, hyperinsulinaemia and hypertension in cardiovascular patients. J Clin
Basic Cardiol 2002;5(3):229-231.
Yamakawa K, Shimabukuro M, Higa N, Asahi T, Ohba K, Arasaki O, Higa M, Oshiro Y,
Yoshida H, Higa T, Saito T, Ueda S, Masuzaki H, Sata M. Eicosapentaenoic acid
supplementation changes fatty acid composition and corrects endothelial dysfunction in
hyperlipidemic patients. Cardiol Res Pract 2012;2012:754181.
Yosefy C, Viskoper JR, Laszt A, Priluk R, Guita E, Varon D, Illan Z, Berry EM, Savion N, Adan
Y, Lugassy G, Schneider R, Raz A. The effect of fish oil on hypertension, plasma lipids and
hemostasis in hypertensive, obese, dyslipidemic patients with and without diabetes mellitus.
Prostaglandins Leukot Essent Fatty Acids 1999;61(2):83-87.
Yosefy C, Viskoper JR, Varon D, Ilan Z, Pilpel D, Lugassy G, Schneider R, Savyon N, Adan Y,
Raz A. Repeated fasting and refeeding with 20:5, n-3 Eicosapentaenoic Acid (EPA): A novel
approach for rapid fatty acid exchanges and its effect on blood pressure, plasma lipids and
hemostasis. J Hum Hypertens 1996;10(SUPPL. 3):S135-S139.
Yoshimura T, Matsui K, Ito M. Effects of highly purified eicosapentaenoic acid on plasma beta
thromboglubulin level and vascular reactivity to angiotensin II. Artery 1987;14(5):295-303.
Zhang G, Sun Q, Hu FB, Ye X, Yu Z, Zong G, Li H, Zhou Y, Lin X. Erythrocyte n-3 fatty acids
and metabolic syndrome in middle-aged and older Chinese. J Clin Endocrinol Metab
2012;97(6):E973-977.