Risk-Benefit Analysis of Seafood Consumption: A Review

advertisement
Risk-Benefit Analysis of Seafood Consumption:
A Review
Rosalee S. Hellberg, Christina A. Mireles DeWitt, and Michael T. Morrissey
Abstract: Seafood, defined here as marine and freshwater fish and shellfish, is recognized as a healthy food choice
because it is a low-fat protein source that provides long-chain omega-3 fatty acids important for early development along
with eye and heart health. However, seafood is also known to contain certain contaminants, such as methylmercury
and persistent organic pollutants, which can have harmful effects on human health and development. In order to limit
exposure to contaminants while maximizing the benefits of seafood consumption, a number of quantitative and qualitative
risk-benefit analyses have been conducted for seafood consumption. This review paper provides a brief background on
risk-benefit analysis of foods, followed by a discussion of the risks and benefits associated with fish consumption. Next,
risk-benefit analyses are reviewed in an historical context. While risk-benefit analysis consists of three main elements
(that is, assessment, management, and communication), this review will primarily focus on risk-benefit assessments.
Overall, most studies have found that the benefits far outweigh the risks among the general population, especially when
a variety of fish is consumed at least twice per week. However, for certain populations (for example, pregnant women
and young children) a more targeted approach is warranted in order to ensure that these groups consume fish that are
low in contaminants but high in omega-3 fatty acids. The potentially harmful unintended consequences of risk-benefit
communication on the general population and certain groups are also discussed.
Introduction
Probably no food category has lent itself more to a risk-benefit
comparison than seafood. While much of this has been played
out in the popular press, the science of risk-benefit analysis has
steadily improved and has helped decision-makers and government
agencies develop consumption guidelines that influence individual consumption patterns as well as food policy. Seafood, defined
here as fish and shellfish from marine or fresh water, farmed or
wild, has been an important part of the human diet for a long
time and now represents 16.7% of the global population’s animal
protein intake. In 2009, worldwide production of seafood reached
145 million metric tons (MMT), divided between capture fisheries
at 90 MMT and aquaculture at 55.1 MMT (FAO 2009). Intake
of seafood and long-chain fatty acids of marine origin has been
associated with many benefits, such as reduced risk of coronary
heart disease (CHD) and improved neurodevelopment; however,
there are also several contaminants present in seafood, such as
MS 20120450 Submitted 3/22/2012, Accepted 5/23/2012. Author Hellberg is
with the U.S. Food and Drug Administration, Pacific Regional Laboratory Southwest,
19701 Fairchild, Irvine, CA 92612, U.S.A. Author DeWitt is with Oregon State
Univ. Seafood Research and Education Center, 2001 Marine Blvd., Astoria, OR
97103, U.S.A. Author Morrissey is with the Oregon State Univ. Food Innovation
Center, 1207 NW Naito Parkway, Portland, OR 97209, U.S.A. Direct inquiries to
author Morrissey (E-mail: michael.morrissey@oregonstate.edu).
Disclaimer: The views presented in this article do not necessarily reflect those
of the Food and Drug Administration.
methylmercury (MeHg) and persistent organic pollutants (POPs),
that have been associated with adverse health effects. In order
to address this dilemma, numerous studies have been published
examining various aspects of the risks and benefits of seafood consumption (see Appendix A). Health organizations worldwide have
also released guidance values and advisories to help consumers
manage risks and maximize benefits. The purpose of this review
is to provide a historical overview of risk-benefit analysis work
that has been carried out thus far regarding seafood consumption.
This will include a discussion of the identification and characterization of the health effects of seafood and risk-benefit assessments
related to seafood. Risk-benefit management and communication
advisories will also be discussed. In order to familiarize the reader
with the common terms and methods that will be discussed, an
initial overview of risk-benefit analysis of food is provided.
Methods of Risk-Benefit Analysis of Food
Risk analysis is a well-established field that is comprised of
3 major elements: risk assessment, risk management, and risk
communication (FAO/WHO 1997). The risk assessment aspect
involves hazard identification, hazard characterization (that is,
dose-response assessment), exposure assessment, and risk characterization. Risk management and communication are often performed by public health organizations and include risk evaluation,
option assessment, option implementation, monitoring and review, and dissemination of information (FAO/WHO 1997). Traditionally, analysis of risks in foods has been carried out by toxicologists, while nutritionists have evaluated the benefits making a
490 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
doi: 10.1111/j.1541-4337.2012.00200.x
Risk-benefit analysis of seafood consumption . . .
Positive Health/ Reduced
Adverse Health Effect
Identification
Hazard Identification
Hazard
Characterization
Exposure
Assessment
Risk
Characterization
Positive Health/ Reduced
Adverse Health Effect
Characterization
Benefit
Characterization
Risk Benefit
Comparison
Qualitative assessments
of risks and benefits
Assessments of risks
and benefits using
common metrics
Assessments of risks
and benefits using
composite metrics
Figure 1–Major steps in risk-benefit assessment, as recommended by the EFSA Scientific Committee and based on the discussions of the EFSA scientific
colloquium on risk-benefit analysis of foods. Risks and benefits are first assessed separately and then the results of the 2 assessments are compared in
the final stage of risk-benefit assessment, termed “risk-benefit comparison.” Figure modified from Barlow and others (2010).
combined risk-benefit analysis difficult (Fransen and others 2010).
When a food exerts both positive and negative effects on health,
it becomes important for risk managers to be able to assess both
the benefits and the risks (Barlow and others 2010). Despite the
importance of risk-benefit analysis of foods, there is currently no
scientific consensus regarding the underlying methodology and
general principles for this type of analysis (EFSA 2006; Barlow
and others 2010; Fransen and others 2010). However, there is
increased interest and recognition regarding the importance of
risk-benefit analysis of foods, and scientists at the European Food
Safety Authority (EFSA) have proposed a framework based on
risk analysis that incorporates risk-benefit assessment, risk-benefit
management, and risk-benefit communication (EFSA 2006).
An EFSA Scientific Committee recently presented a guidance
document outlining a methodology for risk-benefit assessments of
food (Barlow and others 2010). They recommended that riskbenefit assessments be comprised of 3 elements: risk characterization, benefit characterization, and risk-benefit comparison
(Figure 1). The Committee outlined 4 steps on the benefit assessment side that were designed to mirror those involved in risk
assessment: identification of positive health effects/reduced adverse
effects, characterization of those health effects (dose-response assessment), exposure assessment, and characterization of benefits.
The identification of risks or benefits involves the description
of adverse or positive health effects of the food or food components based on human observational or occupational studies,
animal model studies, or in vitro studies. During health effect characterization, a dose-response curve is developed that describes
the relationship between these effects and intake of the food or
food component. This step often results in the establishment of a
health-based guidance value, such as a tolerable daily intake (TDI)
for a hazardous compound or a reference daily intake (RDI) for
c 2012 Institute of Food Technologists®
a beneficial compound. The intake of the compounds from foods
is estimated during the exposure assessment step, based on food
consumption data obtained from food frequency questionnaires,
food diaries, food surveys, and so on. Next, risks or benefits can be
characterized by combining the dose-response relationship with
exposure assessment to evaluate the probability of a health risk or
benefit occurring in response to intake of a specific food or food
compound. The results of the separate risk and benefit assessments
can then be compared in the final stage of risk-benefit assessment,
termed "risk-benefit comparison." This assessment should be carried out for the general population and for any subpopulations that
show increased sensitivity to the food or food component(s) being
evaluated. All risk-benefit assessments should include recognition
of the assumptions, uncertainties, strengths, and weaknesses of the
method. Recently, another approach to risk-benefit assessment of
foods was presented by Fransen and others (2010). The authors
were in general agreement with the approach proposed by EFSA
scientists, with the exception that their tiered approach inserted a
number of "stop" points, where a decision is made as to whether
or not the assessment should be continued, and they recommend
conducting the exposure assessment prior to characterization of
health effects. The advantage of conducting an exposure assessment early on is to first determine whether or not current levels
of exposure (or lack of exposure) are of concern prior to carrying out an extensive literature search and dose-response modeling
during the health effects characterization step.
In addition to providing a framework for risk-benefit assessment of foods, EFSA scientists also outlined 3 different levels of
risk-benefit assessments that might be considered in a stepwise
approach: (1) separate assessments for risks and benefits to determine whether the risks far outweigh the benefits or vice versa,
(2) assessments of risks and benefits using common metrics that
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 491
Risk-benefit analysis of seafood consumption . . .
express risks and benefits in the same unit and can be directly
and quantitatively compared, and (3) assessments of risks and benefits using composite metrics (Barlow and others 2010). Some
examples of common metrics include estimates of the proportion
of the population that is not within the health-based guidance
value or estimates of the incidence of disease or mortality occurring at a certain exposure level and the impact of changing
that exposure level. On the other hand, composite metrics, such
as disability-adjusted life years (DALYs) and quality-adjusted life
years (QALYs), are designed to integrate increases or decreases in
2 or more of the following components: morbidity, mortality, disease burden, and quality of life (Barlow and others 2010). QALYs
were originally introduced in 1976 and are an important part of
health interventions and economic evaluations by regulatory agencies. They allow, as part of a cost-effectiveness analysis, the ability
to assess the improvement in quality-adjusted life expectancy that
may be obtained through specific health intervention (Ponce and
others 2000; Sassi 2006). Similarly, DALYs are used to quantify the
burden of disease across a population, essentially measuring the gap
between the current level of health and an ideal health situation
(WHO 2010). These tools have been applied in risk-benefit analysis of seafood consumption, as discussed in a later section (Ponce
and others 2000; Cohen and others 2005a; Guevel and others
2008). Consistent with the stepwise approach proposed by EFSA
scientists, the risk-benefit assessment portion of this paper will be
organized to first present qualitative research comparing risks and
benefits, followed by studies that used common metrics, and then
studies using composite metrics to examine the risks and benefits
of seafood and its components.
Identification and Characterization of the Health
Effects of Seafood
Positive or reduced adverse health effects of seafood
Interestingly, fish was initially recognized as a healthy food
choice, because it is a low-fat protein source, and not for its
health-promoting lipids (Anderson and Wiener 1995). High-fat
diets were associated with an increased risk of CHD and some cancers, and the National Research Council (NRC) recommended
substituting fish for fatty meats and whole-milk dairy products as
a way of reducing fat and cholesterol intake (NRC 1989). Early
studies with the Greenland Inuits (Bang and Dyerberg 1972, 1980)
and Danish men (Kromhout and others 1985) suggested an association between fish consumption and reductions in death from
CHD. For example, Kromhout and others (1985) reported a 44%
reduction in CHD death for men consuming 1 to 2 servings (100
to 200 g) of fish per week compared to men not eating fish.
Research over the next few decades supported the association between fish consumption and reductions in CHD incidence and
mortality (Whelton and others 2004; König and others 2005),
and identified two omega-3 polyunsaturated fatty acids (PUFAs)
that have a major role in these beneficial effects: eicosapentaenoic
acid (EPA) and docosahexaenoic acid (DHA) (Simopoulos 1991;
Wang and others 2006; Mozaffarian 2008). EPA and DHA cannot be synthesized in substantial amounts by the human body and
must be obtained through the diet, with the main source being
seafood, especially fatty fish (Williams and Burdge 2006). Another
omega-3 fatty acid found in plant-based oils, alpha-linolenic acid
(ALA), is a precursor to EPA and DHA but is converted at very low
rates in the human body (<10%) (Williams and Burdge 2006). In
addition to reduced CHD risk, a number of other health benefits
have been strongly associated with EPA/DHA intake and fish consumption and have been characterized in large-scale epidemiolog-
ical studies, reviews, and meta-analyses (Kris-Etherton and others
2002; IOM 2005; Mozaffarian and Rimm 2006). These include
reduced risk of other cardiovascular disease outcomes such as sudden death (Wang and others 2006; Mozaffarian 2008) and stroke
(He and others 2004a; Bouzan and others 2005), increased duration of gestation (IOM 2005), and improved visual and cognitive
development (Fleith and Clandinin 2005; IOM 2005; Brenna and
Lapillonne 2009). Some other health effects that have been associated with fish consumption and EPA/DHA include alleviation of
colitis (Hudert and others 2006) and rheumatoid arthritis (Kremer
2000), reduced cognitive decline, dementia, depression, and suicides (Morris and others 2005; Sontrop and Campbell 2006; Calon
and Cole 2007; van Gelder and others 2007; Hibbeln 2009), and
decreased likelihood of macular degeneration (SanGiovanni and
others 2007). Seafood also contains a number of vitamins (for
example, A, B-complex, and D) and minerals (such as selenium,
iodine, iron, and zinc) that have been linked to various health benefits (Delange and Lecomte 2000; Rayman 2000; WHO 2002).
Selenium in seafood has been evaluated for its potential to exert
protective effects in terms of reducing accumulation of mercury
in fish (Paulsson and Lundbergh 1989) and in humans (Seppänen
and others 2000). Dietary intake of selenium has also been reported to be inversely related to MeHg-induced adverse health
effects in rats, although a human study into this relationship was
inconclusive (Choi and others 2008; Ralston and others 2008).
In terms of dose-response relationships, meta-analyses have reported that reduced risks for CHD were found to occur with
intakes of at least 250 mg/d of EPA+DHA, and maximum benefits were found with about 500 mg/d of EPA+DHA (Mozaffarian
and Rimm 2006; Harris and others 2008; Harris and others 2009).
With regard to neurodevelopmental benefits of seafood intake during pregnancy, one observational cohort study reported beneficial
effects to children when maternal seafood intake exceeded 340
g (about 3 to 4 servings) per week (Hibbeln and others 2007)
and others have reported increased infant cognition scores when
maternal weekly seafood intake was 1 serving or more (Daniels
and others 2004), 1.5 to 3.5 servings (Oken and others 2008a), or
greater than 2 servings (Oken and others 2005; Oken and others
2008b), compared to mothers not consuming seafood.
Based on the identification and characterization of the health
benefits of seafood and EPA/DHA intake, a number of healthbased guidance values and nutritional recommendations have been
developed by various health organizations (Table 1). Scientific
committees from organizations such as the Institute of Medicine
(IOM), the Food and Agriculture Organization of the United Nations (FAO)/World Health Organization (WHO), and the Dietary
Guidelines Advisory Committee (DGAC) of the U.S. Department
of Agriculture (USDA) and the U.S. Department of Health and
Human Services (HHS) have recommended consumption of 2
servings per week of a variety of seafood (IOM 2005; DGAC
2010) or about 250 mg of EPA + DHA per day (FAO/WHO
2008; DGAC 2010). The American Heart Association (AHA)
recommends that all adults consume fish (particularly fatty fish)
twice a week and that patients with CHD obtain 1000 mg of
EPA+DHA per day (Kris-Etherton and others 2003). Other organizations, such as the International Society for the Study of
Fatty Acids and Lipids (ISSFAL 2004) and the French Food Safety
Agency (AFFSA 2010) have a recommended intake of 500 mg of
EPA + DHA per day for adults. The FAO/WHO expert consultation also recommended specific EPA + DHA intakes for pregnant
or nursing women of 300 mg/d, at least 200 mg of which should
be DHA (FAO/WHO 2008).
492 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
Table 1–Health guidance values for beneficial compounds worldwide.
Guidance
term
RI
RDI
RDI
RI
–
Compound
DHA + EPA
DHA + EPA/seafood
RI
Omega-3 fatty acids from fish
Long-chain omega-3 fatty acids
AI
RDA
Fish
–
PUFA
ALA: 10% EPA + DHA
–
n-3 PUFAs
DHA
–
–
–
Guidance value
500 mg/d
250 mg/d
300 mg/d∗
1000 mg/d
800 mg
EPA + DHA
Two, 113-g servings of seafood per week,
equivalent to about 250 mg/d
450 mg/d
500 mg/d
250 mg DHA and 500 mg EPA + DHA
Two servings of fish per week, especially
fatty fish
500 mg/d long-chain PUFA
1.6 g/d approximately 10%
EPA + DHA
1.1 g/d approximately 10%
EPA + DHA
1% to 2% of energy/d
200 mg DHA
Target
population
1a
1c
4a
5
1b
1b
All
1b4b
Organization/reference
ISSFAL 2004
FAO/WHO 2008
FAO/WHO 2008
AHA (Kris-Etherton and others 2003)
NATO (Simopoulos 1989)
DGAC 2010
HCN (Netherlands 2006)
AFFSA (AFFSA 2010)
1b
AHA (Kris-Etherton and others 2003)
1b
23
ADA (Kris-Etherton 2007)
IOM 2005a
1b
4c
WHO (WHO/FAO 2002)
WAPM (Koletzko and others 2007)
∗ of which at least 200 mg/d should be DHA.
1.
2.
3.
4.
5.
6.
(a) Healthy adults; (b) general adult population; (c) adult males and nonpregnant/nonlactating adult females.
Adult men.
Adult women.
(a) Adult pregnant and lactating females; (b) pregnant women; (c) pregnant and lactating women.
Patients with cardiovascular disease.
See Appendix B for abbreviations/acronyms.
Table 2–Health guidance values for contaminants worldwide.
Compound
MeHg
Hg in seafood
Cadmium
Lead
Hexachlorobenzene
Dioxins and dl PCBs
Dioxins (PCDD/F)i
Dioxins and dl PCBsi
PCBs
PCBsi
Guidance
term
RfD
PTDI
PTWI
MRL
GL
ML
EU ML
AL
PTWI
PTWI
TDI
PTMIb
PTWI
GL
EU ML
TDI
EU ML
TDI
RFDc
CO MRLc
TDI
TLg
Guidance value
0.1 µg/kg bw/d
0.2 µg/kg bw/d; 0.47 µg/kg bw/d
0.23 µg/kg bw/d∗
0.3 µg/kg bw/d
0.23 µg/kg bw/da ; 0.47 µg/kg bw/da
0.5 ppm, 1.0 ppm for six fishd
0.5 ppm; 1.0 ppme
1.0 ppm
0.83 µg/kg of body wt/da
3.6 µg/kg of body wt/daf
0.17 µg/kg bw/d; 0.16 µg/kg bw/d
2.33 pg TEQ/kg body wt/da
2 pg TEQ/kg body wt/da
2 pg TEQ/kg bw/d; 8 pg TEQ/kg bw/d
4 pg TEQg
1 pg TEQ/kg bw/d
8 pg TEQg
0.13 µg/kg bw/d
0.02 µg/kg bw/d
0.02 µg/kg bw/d
0.02 µg/kg bw/d (0.01 for iPCBs)
2.0 ppm
Target
population
1c
3; 1b
1c
1d
2; 4a
1
1
1
–
–
5; 6
–
4b
4b; 4a
1
1
1
1c
1
4c
1
Organization/reference
EPA 2001; Rice and others 2003
HC (Dabeka and others 2004)
FAO/WHO (JECFA 2003)
ASTDR 1999
SACN/COT 2004
HC (2007)
EC (2006)
FDA (2011)
JECFA 2010
JECFA 2010
IPCS 1997
JECFA 2001
SCF 2001a; SCF 2001b
SACN/COT 2004
EC 2006
FAO/WHO (JECFA 2003)
EC 2006
HC (Health Canada 2007)
EPA 1999
ATSDR 2009
AFSSA 2007
FDA/EPA (FDA 2011)
a Originally expressed on a weekly or monthly basis; b Adjusted to TDI here based on 30 d/mo; c Only in regards to Aroclor 1016, 1248, and 1254; d Escolar, orange roughy, marlin, fresh and frozen tuna, shark,
and swordfish; e For predatory fish species; f Withdrawn in 2010, no new PTWI established; g Per gram muscle meat of fish and fishery products, excluding eel; i In fish.
1. (a) All; (b) general population; (c) all (with special groups in mind); (d) all potentially exposed populations.
2. Pregnant women and women who might become pregnant within a year.
3. Children and women of childbearing age.
4. (a) For protection against nondevelopmental adverse effects (that is, increased cancer risk); (b) For protection of developing male reproductive system; (c) For protection of developmental effects.
5. For noncancer effects.
6. For neoplastic effects.
7. See Appendix B for abbreviations/acronyms.
Hazards and negative health effects of seafood
Despite the demonstrated positive health effects of seafood, there
are also several potential hazards (Table 2) that have been found in
seafood, including pathogens, marine toxins, environmental pollutants, and heavy metals (Yasumoto and Murata 1993; Plessi and
others 2001; Storelli and others 2003; Iwamoto and others 2010).
Although the greatest risk to human health is from pathogens
in seafood, this can be overcome through proper cooking, handling, and storage; therefore, negative health effects from seafood
pathogens are not discussed here. The hazards that are generally
considered in risk-benefit assessments are heavy metals, especially
mercury, and POPs. Mercury is released into the environment
c 2012 Institute of Food Technologists®
from both natural and anthropogenic sources and is converted
into MeHg by aquatic microorganisms (Rasmussen and others
2005). MeHg can bioaccumulate through the aquatic food chain,
resulting in higher levels in large, predatory species such as sharks
and swordfish, and the most common form of human exposure to
MeHg is from fish consumption (Gunderson 1995). A correction
factor of 0.85 can be applied to measurements to account for the
proportion of MeHg versus total Hg in seafood. MeHg was first
recognized as a hazard in seafood as a result of large-scale industrial
poisonings in the 1950s in Minamata Bay, Japan, where recorded
mercury levels in the local seafood reached 36 ppm (Harada 1995;
Eto 2000). A series of 3 additional large-scale poisonings occurred
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 493
Risk-benefit analysis of seafood consumption . . .
in Iraq between 1955 and 1972 due to the consumption of wheat
seeds coated with alkylmercury fungicide, where mercury levels
in maternal hair ranged from 18 to 598 ppm (Bakir and others
1980; Marsh and others 1987; WHO 1990; Watanabe and Satoh
1996). The events of Minamata Bay and Iraq demonstrated the
detrimental effects that organic mercury can have on the nervous
system and the heightened sensitivity of the fetus to high levels of
exposure. While the mothers were only slightly affected, they gave
birth to infants with severe neurological problems (Watanabe and
Satoh 1996). Many of the victims had hair mercury levels above
50 ppm and fish and shellfish showed a range from 5.61 to 35.7
ppm of Hg. Normal exposure levels to MeHg in the United States
are at a much lower level; for example, a mean mercury level in hair
of 0.38 ppm has been reported for U.S. women of childbearing
age consuming 3 or more servings of fish per month (McDowell
and others 2004), and mercury levels in most U.S. commercial fish
are below 0.5 ppm (FDA 2009c).
In order to examine the effects of low-level, prenatal exposure to MeHg through dietary consumption of seafood, several
large-scale epidemiological studies were conducted in the 1980s
and 1990s (Grandjean and others 1997; Crump and others 1998;
Davidson and others 1998; Myers and others 2003; Daniels and
others 2004; Wieslaw and others 2006); however, the outcomes of
these studies have been inconsistent (Spurgeon 2006). The 2 most
notable are the Faroe Islands study, n = 917 (Grandjean and others 1997), and the Seychelles Child Development Study, n = 711
(Myers and others 1995; Davidson and others 1998; Myers and
others 2003), which monitored families for over 10 y, but reported
conflicting results. For the purpose of developing health guidance
values for MeHg intake, the maternal hair-mercury concentration
equivalent to a no-observed-effect level (NOEL) or benchmark
dose lower confidence limit (BMDL) for neurobehavioural effects
has been calculated for each study. The calculated values were
12 ppm for the Faroe Islands studies and 15.3 ppm for the Seychelles Islands (JECFA 2003). Interestingly, while both populations
are known for their high-fish diets, the results of the Faroe Islands
study showed some adverse effects of mercury exposure on neuropsychological dysfunctions (such as with language, attention,
and memory); whereas no adverse effects were observed in the
Seychelles study, with the exception of one outlier. Inhabitants of
the Faroe Islands are also known to consume pilot whale, which
has been found to contain high levels of mercury and polychlorinated biphenyls (PCBs) in edible tissues ( Juhlshamn and others
1987; Longnecker and others 2003).
A third cohort study also evaluated scholastic and psychological
tests administered to 6- and 7-y-old children in New Zealand,
although their findings were highly influenced by a single child
whose mother’s mercury level was more than 4 times that of any
other women in the study. When this single outlier was removed,
the BMDL for mercury hair concentration was determined to
range from 7.4 to 10 ppm, and a mercury effect was found in
6 out of 26 tests administered. However, when the outlier was
included, there was no significant association between mercury
hair concentration and any of the tests. The authors had no reasons
to believe the outlier’s test results or mother’s hair mercury levels
were flawed and could not conclude which set of data was nearer
to the truth (Crump and others 1998). An epidemiological study
conducted more recently in the United States with 135 motherinfant pairs reported that infant cognition was favorably associated
with increased maternal fish intake; however, higher maternal hair
mercury levels were associated with lower infant cognition (Oken
and others 2005). Overall, the highest infant cognition scores were
obtained when women consumed 2 or more servings of fish per
week, but they had hair mercury levels ≤1.2 ppm.
In addition to neurodevelopmental effects, mercury has also
been examined in terms of its adverse cardiovascular effects
(Salonen and others 1995; Ahlqwist and others 1999; Hallgren
and others 2001; Guallar and others 2002; Yoshizawa and others
2002; Virtanen and others 2005), but the results of these studies have also been inconsistent. For example, Guallar and others
(2002) reported that toenail mercury levels were 15% higher in
684 male patients with myocardial infarction compared to controls, while Yoshizawa and others (2002) reported no significant
association between mercury levels in toenails and risk of CHD
for 470 male health professionals with documented cases of CHD,
compared to controls. According to Stern (2005), the strongest
basis for a formal risk assessment of the cardiovascular effects of
MeHg is provided by the study of Salonen and others (1995) who
reported a 2-fold increased risk of acute myocardial infarction and
a 2.9-fold increased risk of cardiovascular death for Finnish men
with hair levels of MeHg ≥ 2.0 ppm.
Focusing on management of risks associated with neurodevelopmental effects, health-based guidance values have been developed
for MeHg by a number of organizations worldwide. The U.S.
Environmental Protection Agency (EPA) developed a reference
dose (RfD) for MeHg of 0.1 µg/kg body weight (bw)/d, which
includes a 10-fold uncertainty factor (Rice and others 2003). RfD
is defined as the daily intake that is likely to be without appreciable
risk of deleterious effects during a lifetime. The RfD for MeHg
was originally calculated based on the Iraq wheat seed poisonings
(Marsh and others 1987) and was later re-assessed taking into consideration epidemiology studies from the Faroe Islands, Seychelles
Islands, and New Zealand (Rice and others 2003). Other guidance values put in place by various health organization are listed
in Table 2. In terms of risk management of MeHg concentrations
in seafood, the U.S. Food and Drug Administration (FDA) set an
action level of 0.5 ppm in 1969, which was adjusted to 1.0 ppm
in 1979 based on data from the Minamata Bay poisoning disaster.
This level corresponds to a daily intake of 0.5 µg/kg bw/d (NRC
2000; Burger and others 2005). Health Canada has set tolerance
limits of 0.5 ppm for most fish, with the exception of 1.0 ppm for
escolar, orange roughy, marlin, fresh and frozen tuna, shark and
swordfish (Health Canada 2007); and the European Commission
(EC) has set tolerance limits at 0.5 ppm for most fish and 1.0 ppm
for predatory fish species (EC 2006).
In 1994, the U.S. FDA advised pregnant women and women of
childbearing age who may become pregnant to limit consumption
of shark and swordfish to no more than one meal per month due
to the mercury levels in these fish (ASTDR 1999). Other consumers were advised to limit regular consumption of these species
to about 200 g per week and to limit consumption of fish with
mercury levels below 0.5 ppm to 400 g per week. With regard to
the top 10 species consumed in the U.S., consumption advice was
considered unnecessary because of the low levels of mercury in
these species and because few people eat more than the suggested
weekly limit of 2.2 pounds (1 kg) for the general contamination
level of these species. In 2001 the U.S. FDA updated a previous advisory and singled out infants, small children, pregnant or nursing
mothers, and women who may become pregnant to completely
avoid shark, swordfish, king mackerel, and tilefish; while stating
that, in general, consumers should limit their consumption of all
fish to no more than 340 g per week. This advisory was found
to reduce target consumers’ consumption by 15% to 30% or 1.4
servings per month, and it was associated with a 21.8% net drop in
494 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
canned fish consumption among households with young children
(Oken and others 2003; Schwarz and others 2007; Shimshack
and others 2007). In 2004, the U.S. FDA released a joint advisory with the U.S. EPA that targeted pregnant women, women
who might become pregnant, nursing mothers, and young children (FDA 2004a). These groups were advised to eat up to 340 g
(2 average meals) of seafood per week of a variety of fish and
shellfish that are lower in mercury, such as shrimp, canned light
tuna, salmon, pollock, and catfish. As part of the 340 g per week,
they were advised to eat only up to 170 g (1 average meal) of
albacore (“white”) tuna, due to slightly higher levels of mercury
in this species compared to canned light tuna. They were also
advised not to eat 4 types of fish due to higher levels of mercury:
shark, swordfish, king mackerel, and tilefish. Metals other than
mercury such as lead, manganese, chromium, cadmium, and arsenic may also be found in seafood, although studies have shown
that seafood does not seem to be a main route of exposure to these
metals. The geographic source of the fish is always a factor and
consuming a wide variety of seafood will reduce the risks involved
with the trace amount of metals found in different species and areas
(IOM 2005).
POPs, such as PCBs and dioxins, have also been identified as
hazards that are present in seafood. PCBs are a group of 209
congeners that were widely used in a variety of commercial and
industrial applications (Ross 2004). In the late 1960s and 1970s,
concerns over the health effects of PCBs were raised due to poisoning events resulting from the consumption of PCB-contaminated
food in Japan (Masuda and others 1996) and Taiwan (Ross 2004).
Due to these events and the persistence of PCBs in the environment, these chemicals were banned in the United States in
1979. Dioxin is a collective term for a group of toxic chemicals
with a similar mechanism of toxicity; it includes 7 congeners of
polychlorinated dibenzodioxins (PCDDs) and 10 congeners of
polychlorinated dibenzofurans (PCDFs). There are also 12 congeners of dioxin-like polychlorinated biphenyls (dl PCBs) that the
U.S. EPA includes within the term dioxin (EPA 2010), but they
can be listed separately (SCF 2001a). In Europe, indicator PCBs
(iPCBs) are a set of 7 PCB congeners used to estimate total PCBs
and have shown to be a good predictor of dl PCBs (Babut and
others 2010). For the purposes of this paper, the term dioxin will
refer to the 17 congeners of PCDDs and PCDFs, and the groups
of dl PCBs and iPCBs will be referred to separately. Although
the environmental levels of PCBs and dioxins have been declining, these compounds are widely distributed and current exposure
levels remain a concern. PCBs and dioxins can bioaccumulate
and have been characterized by the EPA as likely human carcinogens. In addition to the potential carcinogenic effects of PCBs and
dioxins, noncancer effects, including changes in hormone levels and fetal development, have been observed at levels of about
10 times above the normal background exposure (EPA 2010). The
most toxic dioxin congener, 2,3,7,8-tetrachlorodibenzo-p-dioxin
(TCDD), was classified as a known human carcinogen in 1997
and accounts for about 10% of the total background dioxin risk
(EPA 2010).
The toxicity of the various PCB and dioxin congeners is calculated relative to the toxicity of TCDD and is expressed as a
toxic equivalent factor (TEF), as established by WHO (Schecter
and others 2001). Most human exposure to dioxins and PCBs
is through the diet, specifically from animal fats found in meats,
seafood, and dairy products. To calculate the toxicity of a food
due to PCBs and dioxins, the level of each congener in the food is
multiplied by its TEF and the sum is reported as the total dioxin
c 2012 Institute of Food Technologists®
like toxic equivalency, or TEQ. As shown in Table 2, limits for
dioxin exposures range between 1 and 4 pg TEQ/kg bw/d for protection against developmental effects (JECFA 2001; SCF 2001b;
SACN/COT 2004) and for protection against increased cancer
risk, a limit has been set at 8 pg TEQ/kg bw/d (SACN/COT
2004). In general, adult men and women have daily TEQ intakes of 2.4 and 2.2 pg/kg bw, respectively. About 9% of dietary
exposure is from fish and shellfish (Schecter and others 2001), primarily from fish caught in fresh waters, estuaries, and near-shore
coastal waters rather than the open ocean. The EC has established
a maximum level for dl PCBs and dioxins in seafood of 8 pg TEQ
per g seafood (ppt TEQ) (EC 2006) and the U.S. FDA has set a
tolerance level for PCBs in seafood of 2.0 ppm (FDA 2011). A
review of numerous studies reporting levels of PCBs and dioxins
in a variety of fish and shellfish reported ranges of 0.5 to 100 ppb
for PCBs and 0.2 to 17 ppt TEQ for dioxins (Mozaffarian and
Rimm 2006), while international averages of dl PCBs and dioxins
in fish, cephalopods, and crustaceans have been reported to range
from 0.15 to 3.0 ppt TEQ (JECFA 2001; Storelli 2008).
The greatest risk to human health from POPs occurs when
fish is harvested recreationally from contaminated waters and consumed in large amounts by subsistence anglers, pregnant women,
and young children. While some studies have reported an association between prenatal exposure to PCBs and dioxins and childhood neurodevelopmental problems (Jacobson and others 1990;
Jacobson and Jacobson 1996; Patandin and others 1999;
Grandjean and others 2001; Ribas-Fito and others 2001; Schantz
and others 2003; Stewart and others 2003; Nakajima and others
2006; Stewart and others 2008), others have not found an effect
(Daniels and others 2003; Gray and others 2005). For example,
Grandjean and others (2001) reported an association between umbilical cord PCB concentration and deficits in 2 to 3 out of 17
neuropsychological tests among a cohort of 435 Faroese children.
Interestingly, PCB-associated deficits only occurred in children
that also had higher levels of mercury exposure, indicating a possible confounding effect. A cohort from the Great Lakes area in the
United States was examined for the relationship between prenatal
PCBs and Neonatal Behavioral Assessment Scale (NBAS) performance in babies born to women consuming Lake Ontario fish (n =
156). The authors concluded consumption of these contaminated
fish is associated with lower IQ in children (Lonky and others
1996; Stewart and others 2000; Stewart and others 2003; Stewart
and others 2008). On the other hand, in a series of studies published by Daniels and others (2003) and Gray and others (2005)
involving over 1200 mother-child pairs, there was no relationship between prenatal PCB exposure and mental or psychomotor
scores in 8-mo-old infants or IQ scores at 7 y of age.
EPA has developed a set of guidelines to assess the cancer risks of
certain compounds, such as POPs, in the population (EPA 2000,
2005). When making risk assessments, the EPA assumes there is
no “safe” lower threshold for carcinogenic compounds, signifying any exposure may pose some cancer risk. This cancer risk is
expressed as a cancer slope factor (CSF) or cancer potency factor
(CPF) with units of risk expressed in mg carcinogenic agent/kg
bw/d exposure. The initial data are dose-response data acquired
during epidemiological studies or chronic animal bioassays. These
data are then extrapolated to represent the lower doses encountered by the general public. The EPA has only identified CSFs for
compounds with enough data to justify development of a value,
such as arsenic, polycyclic aromatic hydrocarbons (PAHs), PCBs,
and dioxins/furans. CSFs can be calculated for both oral and inhalation exposure; as well as unit risks, for example, data specific
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 495
Risk-benefit analysis of seafood consumption . . .
for contaminants in a specific medium (air or water, for example) are expressed as risk per one unit of concentration of the
contaminant. Cancer potency is determined by fitting available
dose-response data to standard cancer risk extrapolation models.
The CSF is then estimated as the slope of the linear extrapolation
to the origin drawn from the 95% lower confidence limit in the
low-dose region. This provides a higher estimate of risk and the
actual risk can be much lower than or as low as zero. The CSFs can
then be useful in calculations of daily consumption limits. Most
CSFs for different compounds can be found on EPA’s Integrated
Risk Information System (http://www.epa.gov/IRIS/).
Qualitative Risk-Benefit Assessments
Incorporating the many risks and benefits that have been associated with seafood consumption into a risk-benefit assessment is
a challenging task. All studies involving characterization of health
effects have a certain degree of uncertainty and variation that must
be taken into account. Also, inconsistencies among the findings
of several studies that have investigated the same health endpoint
further complicate the issue. In order to provide a comprehensive assessment that includes consideration of the numerous health
endpoints associated with seafood, several large-scale, qualitative
risk-benefit assessments have been conducted. Many of these have
resulted in specific seafood consumption recommendations for
the general population and for target populations. For example, at
the request of the National Oceanic and Atmospheric Administration (NOAA), the Institute of Medicine (IOM) of the National
Academies put together an expert committee to examine relationships between the risks and benefits of seafood in order to help
consumers make informed choices (IOM 2005). The committee
carried out a 4-part qualitative protocol to assess and balance the
risks and benefits of seafood, involving (1) identification of the
magnitude of the risks and benefits, (2) identification of risks and
benefits that are important enough to be included in the balancing process, (3) evaluation of the changes in risks and benefits
associated with different consumption patterns, and (4) balancing of the risks and benefits to arrive at specific consumption
guidelines. In order to identify the health benefits of seafood and
EPA + DHA, the committee examined numerous studies and
other systematic reviews by the Agency for Health Research and
Quality (AHRQ) (Balk and others 2004; Schachter and others
2004; Wang and others 2004; Schachter and others 2005). Positive
health effects were examined for a number of areas, including benefits to women during and after pregnancy; duration of gestation
and birth weight; infant and child development; and cardiovascular disease, cardiovascular mortality, and all-cause morbidity and
mortality. Overall, the greatest evidence for health benefits was
related to reduced risk of cardiovascular disease, greater duration
of gestation, and improved visual and cognitive development associated with maternal seafood or EPA/DHA intake. However, the
committee also noted that the average amounts of seafood consumed by the U.S. population are below levels suggested by many
authoritative groups to achieve positive health effects. In terms
of hazards associated with seafood consumption, the committee
conducted an extensive evaluation of the health effects of MeHg
and other metals, such as cadmium; POPs; microbiological agents;
seafood allergens; and naturally occurring toxins. Based on their
review of the evidence, the committee concluded that among
the potential chemical contaminants in seafood, MeHg poses the
greatest concern for adverse health effects, whereas the risk associated with POPs remains uncertain, and risks associated with
microbial hazards, allergens, and toxins are persistent, yet more
controllable. Due to uncertainties in terms of the ability of the
committee to quantify risk-benefit tradeoffs based on the available
evidence, a qualitative risk-benefit assessment was conducted. After balancing the evidence with regard to cardiovascular benefits
and developmental risks and benefits of seafood, 4 target populations were identified and specific consumption guidelines were
developed for these populations. Briefly, the committee stated that
healthy adolescent and adult males and females (who will not become pregnant) may reduce their risk for cardiovascular disease
by consuming seafood regularly, making sure they chose from a
variety of seafood if consuming more than 2 servings per week.
Similar guidelines were given for adult males and females who
are at risk of cardiovascular disease, with the addition that this
group may benefit from including seafood high in EPA and DHA.
The committee stated that the other 2 target populations, females
who are or may become pregnant or who are breast-feeding and
children up to 12 y of age, may benefit from consuming seafood,
especially selections high in EPA and DHA, and they can safely
consume up to 340 g of seafood per week including up to 170 g
of albacore tuna, but that they should avoid large predatory species
such as shark, swordfish, tilefish, or king mackerel.
Another in-depth qualitative risk-benefit assessment of seafood
consumption was conducted by Mozaffarian and Rimm (2006)
from the Harvard School of Public Health and Harvard Medical School. The assessment involved examination of scientific
publications, government reports, systematic reviews and metaanalyses related to 4 categories: (1) association between intake
of fish or fish oils and reduced risk of cardiovascular events and
mortality, (2) effects of MeHg and fish oil on early neurodevelopment, (3) association between MeHg exposure and negative cardiovascular or neurologic effects in adults, and (4) health
risks of PCBs and dioxins in fish. The authors focused on human health studies from randomized trials and large prospective studies and, when possible, conducted meta-analyses to
better characterize risks and benefits. In terms of cardiovascular outcomes, the assessment showed that intake of about
250 mg/d EPA + DHA, or 1 to 2 servings/wk of fish high in these
fatty acids, was associated with risk reductions of 36% for CHD
death and 17% for total mortality. However, at higher intakes,
there was little additional risk reduction, suggesting a threshold
for maximum cardiovascular benefits of 250 mg/d EPA + DHA.
For developmental outcomes, the assessment revealed the importance of DHA for cognitive and visual development, but also
showed the potential negative effects of low-level MeHg exposure
on cognitive development. Based on these findings and doseresponse relationships, the authors recommended that women of
childbearing age and nursing mothers should consume 2 servings of seafood per week, with limited intake of selected species
that show elevated mercury levels. The assessment of MeHg and
cardiovascular outcomes in adults revealed uncertainty in terms
of the health effects of low-level exposure among adults. Therefore, Mozaffarian and Rimm (2006) suggested choosing from a
variety of seafood and noted that adults consuming ≥ 5 servings/wk should limit intake of high-mercury fish species. Levels
of PCBs and dioxins were reported to be relatively low in fish
and the health benefits of seafood intake were found to outweigh
any potential adverse effects from these contaminants. However,
women of childbearing age were advised to check regional risk
advisories prior to consuming locally caught freshwater fish. Overall, the authors concluded that the benefits of fish intake exceed the potential risks for major health outcomes among adults,
and for women of childbearing age, the benefits of modest fish
496 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
intake, excepting a few selected species, also outweigh the potential risks. They also cautioned that avoidance of fish due to
confusion surrounding the risks and benefits could lead to suboptimal neurodevelopment in children and thousands of additional
deaths annually from CHD. Although this risk-benefit assessment
has been criticized for discounting the negative health effects of
MeHg on cardiovascular disease in adults (Stern 2007), studies
examining this topic have reported conflicting results, as discussed
in an earlier section, and according to Mozaffarian and Rimm
(2006); even in studies reporting negative effects of MeHg, the
net effect of seafood consumption was still positive.
In Europe, FAO and WHO held an expert consultation in
January 2010 to compare the risks and benefits of seafood consumption, based on a request for scientific advice from the Codex
Alimentarius Commission (JECFA 2010). The consultation examined the levels of specific nutrients and contaminants (that
is, MeHg, dioxins, dl PCBs, and furans) in seafood, as well as
the scientific literature regarding the risks and benefits of seafood
consumption. This information was used to consider risk-benefit
assessments for certain health endpoints, including sensitive populations. The consultation compared the benefits of fish consumption related to neurodevelopment and prevention of cardiovascular
disease with the risks of fish consumption related to neurodevelopment, cardiovascular disease, and cancer. Overall, the consultation
recognized that fish was a source of energy, protein, and other
important nutrients, such as DHA and EPA, and that consumption of fish, especially oily fish, lowers the risk of CHD mortality
among the general adult population. They did not find convincing evidence of CHD risks from MeHg and determined that the
potential cancer risks of dioxins and dl PCBs were well below
established CHD benefits. With regard to neurodevelopmental
effects of omega-3 intake and MeHg exposure, the consultation
concluded that in most cases maternal fish consumption lowers
the risk of suboptimal neurodevelopment compared to no maternal fish consumption. Further, when maternal dioxin and dl PCB
intake is below the provisional tolerable monthly intake (PTMI) of
70 pg/kg bw/mo established by the JECFA, neurodevelopmental
risk was determined to be negligible; however, when intake exceeds the PTMI, the risk may no longer be negligible. Finally, the
consultation outlined a series of recommendations, including an
emphasis on the benefits of eating seafood in terms of reducing
CHD mortality and improving neurodevelopment, as well as the
development and evaluation of risk management and communication strategies related to seafood consumption that both minimize
risks and maximize benefits.
In the United States, the 2010 DGAC, which was established
jointly by the Secretaries of the USDA and the U.S. Department
of Health and Human Services, compared the risks and benefits of seafood and recommended specific guidelines for both
seafood and EPA + DHA intake. Several health endpoints were
considered, including the association between EPA + DHA and
risk of CHD, the association between maternal intake of EPA
+ DHA and health outcomes in infants, as well as an overall
comparison of the risks and benefits of seafood consumption.
Twenty-five studies published since 2004 were examined in terms
of CHD benefits associated with seafood and EPA + DHA, including 6 systematic reviews/meta-analyses (He and others 2004b;
Whelton and others 2004; König and others 2005; Mozaffarian and Rimm 2006; Wang and others 2006; Mozaffarian 2008).
DGAC found consistency among these studies and made the conclusion that moderate evidence shows that about two 113-g servings per week of seafood (about 250 mg EPA + DHA per day) is as
c 2012 Institute of Food Technologists®
sociated with a reduced risk of cardiac mortality from CHD or sudden death in individuals with and without cardiovascular disease. In
their evaluation of developmental benefits, the DGAC considered
several expert opinions, including the Cochrane Database Systematic Review (Makrides and others 2006), American Dietetic
Association (ADA) Evidence Analysis (Kaiser and Allen 2008),
and the European Union Perinatal Lipid Intake Working Group
assessment (Koletzko and others 2007), as well as a background
paper that reviewed 23 trials (Brenna and Lapillonne 2009), and 9
additional papers discussing the effects of omega-3 fatty acids on
breast milk composition and infant health outcomes. Overall, the
DGAC concluded that moderate evidence indicated an association
between increased maternal intake (during pregnancy and lactation) of long-chain omega-3 fatty acids, especially DHA from at
least 2 servings of seafood per week, and increased levels of DHA
in breast milk as well as improved infant health outcomes, such as
visual acuity and cognitive development. In their overall assessment
of the risks and benefits of seafood, the DGAC examined 9 studies
that have explored a wide range of potential health effects along
numerous health endpoints: 3 quantitative risk-benefit assessment
studies (Guevel and others 2008; Sioen and others 2008; Ginsberg
and Toal 2009), 4 cross-sectional studies (Rawn and others 2002;
Huang and others 2006; Dewailly and others 2007; Verger and
others 2008), 1 meta-analysis (Gochfeld and Burger 2005), the
systematic review from Mozaffarian and Rimm (2006), and the
report from IOM (2005). Based on their review of these studies,
the DGAC concluded that moderate, consistent evidence suggests that the health benefits associated with 2 servings per week
of cooked seafood outweigh the potential risks from MeHg and
POPs exposure, even among women of childbearing age, pregnant
and nursing women, and children ages 12 and under. In agreement
with the IOM report, the DGAC stated that consumers can safely
eat at least 340 g of a variety of seafood every week, provided that
they also follow federal and local advisories and limit consumption
of large predatory fish. Overall, the DGAC felt that encouragement of seafood consumption in the United States is justified, as
intake levels continue to be below those recommended for health
by the Committee and by the IOM.
Quantitative Risk-Benefit Assessments
Assessments based on common metrics
Risk-benefit assessments with common metrics include the use
of single outcome measures, like incidences of mortality, morbidity, or exceeding/not meeting health-based guidance values. These
types of assessments are easy to comprehend and have limited data
needs, but they must be interpreted with caution because they
usually only provide some of the information of interest (Fransen
and others 2010). Further, many of these studies do not combine
the risks and benefits into a net health outcome, as do composite metrics, but can be a useful step in a full risk-benefit analysis
by presenting the information in the same units for comparison.
Risk-benefit assessments that have been carried out for seafood
using common metrics are reviewed in this section.
An early risk-benefit assessment of seafood by Anderson and
Wiener (1995) used a risk tradeoff analysis method to compare the
potential cancer risk of eating fish to the potential CHD risk of not
eating fish. The cardiovascular risk of not eating fish was derived
from the results of Kromhout and others (1985), described earlier,
where consumption of 1 to 14 g fish/person/d was estimated to
reduce the risk of CHD by 36% and consumption of 15 to 29 g
fish/person/d was estimated to reduce the risk of CHD by 44%.
The cancer-related risk of eating fish was estimated by calculating
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 497
Risk-benefit analysis of seafood consumption . . .
potential exposure to 6 carcinogenic compounds with an allowable concentration limit set by the FDA and a CSF determined by
the EPA: PCBs, dioxins, dichlorodiphenyltrichloroethane (DDT),
chlordane, dieldrin, and heptachlor. The analysis assumed that all
fish consumed were contaminated with all 6 compounds at levels
matching the FDA limits of 2.0 ppm (PCBs), 50 ppt (dioxins),
5.0 ppm (DDT), and 0.3 ppm (chlordane, dieldrin, and heptachlor). The CSFs associated with exposure to compounds at
these levels were calculated, assuming an average body weight of
70 kg and chronic exposure over a 70-y lifespan. The results of
this analysis revealed that the lifetime risk of getting cancer for
someone eating 1 g of contaminated fish per day over the course
of 70 y was estimated at 5.0 × 10−4 (5 in 10000) and consumption
of 20 g of contaminated fish per day (1.4, 100-g servings/wk) for
70 y was estimated to increase cancer risk to 1 in 100. The overall
risk for cancer for the average American is 25%, so based on these
predictions consumption of 20 g/d of contaminated fish would
increase the overall cancer risk to 26%. However, the cancer risk
of eating 20-g fish/d in the United States based on the actual levels
of contaminants in fish has been estimated to be much less (0.75 in
10000) than the risk of eating contaminated fish calculated using
the previous assumptions (IOM 1991). On the other hand, at the
time of this study the average total risk of CHD mortality was
35% and the average fish consumption was 15 g/d. The results of
Kromhout and others indicated that reducing fish consumption
from 15 to 29 g/d to 0 g/d would increase the risk of dying from
CHD by about 66%, meaning that the total risk of dying from
CHD would be predicted to increase from 35% to 58%. Based
on the results presented here, the cardiovascular benefits of fish
consumption were determined to far outweigh the carcinogenic
risks, even when fish contain all 6 carcinogenic contaminants at
the FDA limits and all cancers are assumed to be fatal.
Several studies have been carried out assessing the levels of organic contaminants in farmed and wild salmon and potential risks
to human health (Hites and others 2004a; Hites and others 2004b;
Foran and others 2005a; Huang and others 2006), with resulting
recommendations to limit consumption of salmon to only 0.2 to
8 meals per month, depending on the harvest location. However, salmon is a rich source of omega-3 fatty acids and reducing
consumption to these levels would be expected to concomitantly
increase the risk of cardiovascular disease and mortality, as discussed
above. To better understand the overall health effects of altering
consumption levels of farmed and wild salmon, risk-benefit assessments have been conducted by Foran and others (2005b) and
Dewailly and others (2007).
Foran and others (2005b) considered the cancer and noncancer
risks associated with exposure to organic contaminants when
enough salmon is consumed to provide 1 g of EPA + DHA per
day. Levels of organic contaminants and omega-3 fatty acids in
farmed Atlantic salmon and wild Pacific salmon were obtained
from previous work (Hites and others 2004a). Risks were calculated based on an average body weight of 70 kg and continuous
exposure to contaminants over a 70-y lifespan. Noncancer risk was
calculated for 14 contaminants (including PCBs, hexachlorobenzene (HCB), chlordane, dieldrin, heptachlor, MeHg, and others)
and was deemed acceptable when the ratio between the cumulative exposure level for all contaminants and the cumulative RfD
for all contaminants was <1. The acceptable cancer risk level resulting from the cumulative analysis of 11 contaminants with CSFs
(including PCBs, 2,3,7,8-TCDD, HCB, chlordane, dieldrin, heptachlor, and others) was set at 1 × 10−5 , which is the midpoint
of the acceptable range established by the EPA (1 × 10−4 to 1 ×
10−6 ). At consumption levels of two 100-g servings per week, the
cumulative noncancer risk from organic contaminants was predicted to remain within acceptable levels, with the greatest benefit
coming from wild salmon. On the other hand, salmon consumption at these levels was associated with an increased cancer risk,
with a cumulative risk of 8 × 10−5 (that is, 8 in 100000) for
wild salmon and 2.4 × 10−4 for farmed salmon. Based on these
results, Foran and others (2005b) made recommendations to limit
consumption of farmed and wild salmon to 4 or fewer meals per
month, depending on the source of the fish. However, when the
health outcomes of consuming 1 g EPA + DHA per day were
calculated, the number of lives that would be saved from CHD
mortality was reported to be about 7100 per 100000 people, which
outweighs the potential risk of cancer by a factor of 300 (farmed
salmon) to 900 (wild salmon). Additional age-adjusted analysis by
Mozaffarian and Rimm (2006) indicated that the CHD benefits
outweigh the cancer risks by 100- to 370-fold for farmed salmon
and 300- to more than 1000-fold for wild salmon. These factors
may be further increased if salmon is consumed at levels that provide about 250 mg EPA + DHA/d (that is, about 150 g of wild
salmon per week or 100 g of farmed salmon every 2 wk), which
would be predicted to provide similar protection against CHD
mortality as 1 g EPA + DHA/d while reducing lifetime cancer
risk from salmon consumption by about 75%. Further, the levels
of organic contaminants that form the basis of the risk calculations
included skin and disregarded the losses of PCBs that can occur
during cooking, so the actual risks of eating cooked salmon are
predicted to be less than that calculated by Foran (Santerre 2010).
The CSF method used by Foran and others (2005b) to calculate
cumulative cancer risk has been criticized as not being appropriate for the type of carcinogens found in salmon (that is, epigenetic
carcinogens) because it assumes a linear, nonthreshold relationship between risk and low-dose exposure (Dewailly and others
2007). Instead, Dewailly and others used health-based guidance
values for MeHg, PCBs, and dioxins to examine the reproductive
and developmental risks associated with consumption of enough
salmon to provide 500 mg EPA + DHA per day. Atlantic salmon
and rainbow trout samples were collected in Quebec, Canada, and
levels of contaminants and fatty acids were analyzed in skinless,
raw fillets with all subcutaneous or mesenteric fat removed. The
levels of EPA + DHA determined for farmed salmon in this study
were about 4-fold less than those reported by Foran and others
(2005b), and the results indicated that daily intake of 58 to 69 g (or
four to five, 100-g servings per week) of farmed salmon or trout
would provide 500 mg EPA + DHA. For risk-benefit assessment,
the authors considered the contaminant exposure associated with
consumption of two 180-g meals per week of farmed Atlantic
salmon, which provides about 440 mg EPA + DHA/d. Calculations were carried out based on 20- to 39-y-old women with an
average body weight of 60 kg. The predicted exposure to contaminants at this consumption rate was below the tolerable intakes
established in Canada and by the FAO/WHO in all cases, with
levels of 0.015 µg/kg bw/d for MeHg, 0.012 µg/kg bw/d for
PCBs, and 0.070 pg TEQ/kg bw/d. Although not all dl PCBs
were measured in this study, total TEQ intake was estimated to be
0.28 pg TEQ/kg bw/d, which was based on the assumption that
dioxins make up about 25% of the total TEQ exposure. This predicted exposure level is well below the FAO/WHO tolerable intake of 2.33 pg TEQ/kg body wt/d. Farmed trout showed similar
(for MeHg) or lower (for PCBs or dioxins) levels of contaminants
as compared to farmed salmon and consumption of 360 g/wk
would not be expected to result in excessive exposure. Overall, the
498 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
authors concluded that two 180-g meals per week of farmed
salmon or trout available in North American markets would be
expected to provide sufficient levels of EPA + DHA without concern over the putative health risks.
Combining exposure assessments with health-based
guidance values. A number of studies have assessed the risks
and benefits of seafood by comparing exposure assessments with
health-based guidance values for seafood (for example, TDI, RfD,
or recommended intake). Although this type of study does not
allow for a full quantitative risk-benefit assessment, it can serve
as a valuable first step in identifying whether a complete assessment is warranted (Fransen and others 2010). These studies are
generally carried out using either a deterministic (“worst case”
scenario) or probabilistic approach (Cardoso and others 2010).
The latter approach allows for an estimate of the probability of
the target population that is at risk of either exceeding a maximum safe limit or not reaching a recommended intake level. The
modeling of probability distributions takes into account the variability of the data and this approach has been increasingly used to
assess contaminants in foods. It is important to note that probability estimates are highly dependent on the tail behavior of the
distributions, since health-based guidance values tend to be at the
upper or lower range of most individual intakes, and the statistical
tools used for these assessments must be highly rigorous and reliable (Cardoso and others 2010). Some important variables to take
into account in both deterministic and probabilistic studies are the
food intake data and the levels of contaminants and nutrients in
these foods. Food intake data are generally obtained through food
consumption surveys such as food frequency questionnaires, 24-h
recall dietary surveys, and one- to seven-day food diaries (Barlow
and others 2010), while contaminants and nutrients in foods are
generally obtained through databases, such as the one described
by Sioen and others (2007a) which pools data from different publications. However, seafood is a complex food group with many
different categories and species, and food intake data are generally
not specific with regard to the seafood species consumed. Because
various seafood species are known to contain different levels of
nutrients and contaminants, exposure levels estimated for seafood
in these types of assessments may be associated with a high degree
of uncertainty. Studies that have compared exposure assessments
with health-based guidance values to assess risks and benefits have
been carried out specifically for consumers in countries such as
France (Crépet and others 2005; Leblanc and others 2006; Verger
and others 2008; Pouzaud and others 2009), Spain (Domingo and
others 2007a), The Netherlands (van der Voet and others 2007),
and Belgium (Sioen and others 2008), as well as on a broader
scale for consumers across Europe (Cardoso and others 2010) and
worldwide (Sioen and others 2009).
Several studies have been devoted to risk-benefit assessments
for French consumers living in coastal regions with high fish consumption. One such assessment, called the CALIPSO study, was
delegated by the General Food Directorate to the French Institute
for Agronomy Research (INRA) (Leblanc and others 2006). The
CALIPSO study examined dietary patterns among some 1000
French consumers living in 4 coastal regions with high seafood
consumption, evaluated blood and urinary biomarkers associated
with nutrient and contaminant intake for about half of the study
participants, and determined levels of nutrients (omega-3 fatty
acids) and contaminants (6 trace elements and 3 categories of
POPs) in a variety of seafood sampled in the study regions. The
study participants were limited to adults that consumed seafood at
least twice per week and consumption data were obtained with a
c 2012 Institute of Food Technologists®
food frequency questionnaire that listed 82 fishes, mollusks, crustaceans, and seafood-based dishes. Omega-3 fatty acid intakes were
compared to the French Recommended Daily Allowance (RDA)
of 400 to 500 mg/d for adults and exposure levels to trace elements and POPs were compared to provisional tolerable intakes
established by the JECFA, when available. The results indicated
that consumption of at least 2 servings of fish, including some
oily fish, per week would allow consumers to obtain the RDA
of omega-3 fatty acids. Male and female study participants in the
age range of 18 to 64 y consumed a weekly average of about 630
to 640 g of fresh and frozen fish, 260 to 270 g of mollusks and
crustaceans, and about 270 to 310 g of other types of seafood,
making total seafood consumption equivalent to about twelve,
100-g servings per week. EPA + DHA intake exceeded 500 mg/d
for 84% of the study participants, with an average level of 1240 ±
960 mg/d (Bemrah and others 2008). However, most of the types
of seafood that contributed strongly to omega-3 fatty acid intake
also accounted for greatest exposure to POPs, particularly salmon,
mackerel, and sardine. In terms of risks, the study found that only
individuals in the highest consumption group had a nonnegligible
risk of exceeding the maximum limits for MeHg, Cd, dioxins,
and PCBs. The average exposure to POPs among all study participants was 18.7 ± 19.6 pg TEQ/kg bw/wk for dioxins and
dl PCBs, 0.04 ± 0.06 µg/kg bw/wk for iPCBs, and 2.2 ± 1.8
ng/kg bw/d for PBDEs. Study participants had a 39% probability
of exceeding the provisional tolerable weekly intake (PTWI) for
dioxins and dl PCBs and 72% probability of exceeding the PTWI
for iPCBs. The average exposure level to MeHg from seafood
was 1.5 ± 1.2 MeHg/kg bw/wk, with some 34% of the study
participants exceeding the PTWI established by JECFA (1.6 µg
MeHg/kg bw/wk). None of the study participants exceeded the
PTWIs established for organic tin, arsenic, and lead, and only 8.5%
exceeded that established for Cd. In terms of blood levels of these
trace elements, most subjects (94% to 97%) had levels at or below
the standard level, and women of childbearing age had average
MeHg levels of 2.3 to 3.4 µg/L, which is well below the highest
exposure level at which adverse effects do not occur to the fetus
(56 µg/L). Even individuals consuming up to 4.5 kg seafood per
week with predicted exposure levels of 9.6 µg/kg bw/wk had
maximum blood MeHg levels of 18 µg/L. When blood levels
were converted to weekly exposure for women of childbearing
age, the data indicated an average exposure of 0.4 ± 0.3 µg/kg
bw/wk, as compared to a predicted average exposure of 1.3 ±
0.9 µg/kg bw/wk based on consumption and contamination data
for the same group of women. Overall, based on the study results, the authors concluded that the general population should
consume at least 2 servings of fish, especially oily fish, per week
and pregnant and nursing women should limit consumption of
predatory fish to once per week.
Other studies conducted among French consumers have reported that women of childbearing age had a 3% to 5% probability
of exceeding the PTWI for MeHg, based on data from exposure
assessments (Crépet and others 2005; Verger and others 2007). In
order to examine methods for reducing MeHg exposure, Crépet
and others (2005) assessed the probabilistic effects of 5 different risk management scenarios (assuming 100% compliance): (1)
no change in consumption patterns, (2) remove all predatory fish
above 1.0 ppm MeHg and all other fish above 0.5 ppm MeHg from
the market, (3) remove all fish exceeding 0.5 ppm MeHg from the
market, (4) remove 12 species of predatory fish from the market,
or (5) restrict consumption of predatory fish to an exact number of
portions per week. Current fish consumption and exposure levels
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 499
Risk-benefit analysis of seafood consumption . . .
were obtained from a previous survey (French INCA survey) that
used a 7-d food log obtained from a nationally representative sample of the French population. Crépet and others (2005) utilized
data from 1945 male and female adults and 848 children within
this data set, combined with mercury levels reported in previous
studies for 89 individual seafood items. Average consumption of
seafood varied for each age group, with children consuming 174
g/wk and adults consuming 285 g/wk. Mean body weights were
19 kg for children ages 3 to 6 y, 29 kg for children ages 7 to
10 y, and 58 kg for women of childbearing age. Based on these
data, under scenario 1 the probability of exceeding the PTWI for
MeHg was 4.4% for women of childbearing age and 6.7% for children (12.6% for ages 3 to 6 y; 5.0% for ages 7 to 10 y). Scenarios 2
and 3 did not significantly reduce exposure levels among children,
but scenario 3 (removing all fish above 0.5 ppm) did significantly
reduce probability of MeHg exposure among women of childbearing age to 0.6%. In scenario 4, where 12 predatory species
are removed from the market, the authors reported significant reductions in MeHg exposure for women of childbearing age (0%
probability of exceeding the PTWI for MeHg) and children ages
3 to 6 y (2.8% probability), but not among children aged 7 to 10
y (1.5% probability). Under scenario 5, the authors suggested an
exact number of portions of predatory fish per week that would
allow target groups to remain under the PTWI for MeHg. For example, a recommendation that women of childbearing age limit
consumption to two, 170-g portions (or 340 g) of predatory fish
per week or to 255 g per week if also consuming nonpredatory
fish. Overall, the authors suggested that risk management options
that provide advice on food consumption, such as scenario 5,
are more efficient compared to additional restrictions for MeHg
levels in fish. However, a later study examining the effects of a
risk-benefit advisory among French consumers reported that the
advisory did not lead to a decrease in predatory fish consumption,
but it did result in a significant decrease in overall fish consumption
(Verger and others 2007).
A subsequent study compared the exposure levels of PCBs and
dioxins to intake levels of omega-3 fatty acids among 401 French
fish consumers in western coastal areas (Verger and others 2008).
The consumption data used in this study were obtained previously
in the fish advisory study, which utilized a 1-mo food diary detailing types and frequency of seafood consumed for 195 men and
206 women (Verger and others 2007). The men had an average
body weight of 75 kg and ate seafood 3.3 times per week, while
the women had an average body weight of 62 kg and ate seafood
2.9 times per week. Exposure levels were calculated using data
from the French Ministry of Agriculture and Fisheries. Overall,
20% to 30% of the target population was estimated to exceed the
PTWI established by the Scientific Committee on Food (SCF)
for dioxins and dl PCBs of 14 pg TEQ/kg bw/wk. About 25%
of the total exposure was from PCDDs and PCDFs and the remaining 75% was from dl PCBs. On the benefit side, about 60%
of the target population obtained the recommended intake of 500
mg/d long-chain omega-3 fatty acids. When risks and benefits
were compared, only 41% of the study participants had an optimal
balance of meeting the RDA of omega-3 fatty acids while remaining below the PTWI for dioxins and dl PCBs: 19% of individuals
met the RDA but exceeded the PTWI, while another 38% of
individuals were below the PTWI but were also below the RDA.
The authors concluded that consuming the RDA for omega-3s
of 500 mg/d through seafood consumption was compatible with
the threshold for dioxins and dl PCBs, but that consumers with
omega-3 intakes above 1500 mg/d from seafood consumption
were likely to also be exceeding the PTWI for dioxins and dl
PCBs.
Pouzaud and others (2009) assessed seafood consumption patterns and intake of MeHg and omega-3 fatty acids among 161
pregnant French women living in a coastal region with high-fish
consumption. The authors used the food frequency questionnaire
from the CALIPSO study to obtain seafood consumption data for
women at both 12 and 32 wk of pregnancy. Portion sizes were
estimated based on a catalog of photos presented to the study
participant. At both time points, hair samples were obtained for
MeHg testing and body weights were recorded. At week 12, participants had a mean seafood consumption of 322 g/wk and an
average body weight of 60 kg, compared to a mean seafood consumption of 309 g/wk and average body weight of 73 kg at week
32. The mean dietary exposure to MeHg from seafood was not
significantly different at the 2 time points, with an overall range of
0.6 to 0.7 µg/kg bw/wk. There was also no significant difference
in the hair MeHg concentrations across the 2 time points, which
ranged from 0.1 to 3.7 ppm, with a mean of 0.8 ppm. Overall,
about 5% of the women were exceeding the PTWI for MeHg,
similar to results of previous studies on women of childbearing
age (Crépet and others 2005; Verger and others 2007), and about
50% of the women were not obtaining the RDA of 500 mg/d for
long-chain omega-3 PUFAs. The authors used a cluster analysis
tool to group the study participants into 5 different categories related to fish consumption and exposure levels, and found that only
women consuming a high proportion of fatty fish meet the RDA
for omega-3 fatty acids without exceeding the PTWI for MeHg.
Domingo and others (2007a) estimated dietary exposure to
DHA + EPA and chemical contaminants among Spanish consumers. The authors measured fatty acids, metals (Hg, Cd, Pb),
and organic pollutants (dioxins, dl PCBs, polybrominated diphenyl
ethers (PBDEs), polychlorinated diphenyl ethers (PCDEs), HCB,
polychlorinated naphthalene (PCNs), and (PAHs) in the edible
portions of the top 14 species consumed in Spain. Daily consumption rates of these 14 species were calculated for a 70-kg male based
on consumption data obtained previously and a standard meal size
of 227 g. The average EPA + DHA intake was determined to
be 244 mg/d, which is very close to the level recommended
by FAO/WHO (250 mg/d). The estimated intakes of total Hg
(0.14 µg/kg bw/d), Cd (0.02 µg/kg bw/d), and Pb (0.03 µg/kg
bw/d) were all below the provisional tolerable intakes established
by the FAO/WHO for these compounds (Table 2). When the correction factor of 0.85 is applied to the total Hg intake, the MeHg
intake can be estimated at 0.12 µg/kg bw/d, which is below the
PTWI but slightly above the RfD established by the U.S. EPA. The
estimated intakes of dioxins and dl PCBs (0.54 pg TEQ/kg bw/d)
as well as HCB (0.16 ng/kg bw/d) were all below the PTWIs
established by FAO/WHO for noncarcinogenic effects. The total
intake of 7 carcinogenic PAHs was associated with an increased
cancer risk of 0.27 × 10−6 (that is, 2.7 incidences of cancer per
10000000 people) resulting from chronic exposure over a 70-y life
span, based on EPA CSFs. Tolerable intake limits have not been
established for PBDEs, PCDEs, and PCNs, which had estimated
exposure levels of 0.30, 0.56, and 0.02 ng/kg bw/d, respectively.
The authors did not report the percentage of the target population that may be at risk from excess exposure to contaminants or
deficient intake of EPA + DHA. In order to help consumers remain below exposure limits for carcinogenic and noncarcinogenic
effects, the authors developed recommended monthly fish consumption guidelines for the top 14 fish in Spain. They determined
the greatest noncarcinogenic risk to be from MeHg exposure, and
500 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
recommended limiting consumption of tuna to 2 meals per month
and swordfish to 0.5 meals per month. The greatest carcinogenic
risk was determined to be from PAHs and dioxins. To remain
below PAH exposure limits, recommended consumption levels
were calculated to be between 0.5 (clam/mussel/shrimp) and 4
(hake/red mullet/sole/cuttlefish/squid) meals per month, depending on the species. To remain below dioxin exposure limits recommended consumption levels were calculated to be between 1 (red
mullet) and 16 (hake or cuttlefish) meals per month, depending
on fish species. However, the authors did not compare the risks of
reducing fish consumption to these levels in terms of the concomitant decreased intake of EPA + DHA that would occur and the
subsequent increases in risk of cardiovascular disease and mortality. In a companion paper, Domingo and others (2007b) presented
an interactive risk-benefit online tool that allows the consumer to
input their weight, meal size, and consumption frequency in order
to calculate their intake of EPA + DHA and exposure to metals
and organic pollutants from the 14 seafood types examined above.
A study from The Netherlands reported the development of a
probabilistic model to calculate simultaneous exposure to multiple
compounds from food and to predict different dietary scenarios
(van der Voet and others 2007). The model was used to assess longterm intake of EPA + DHA, dioxins, and dl PCBs from a total diet
perspective, as well as predict the effects of replacing other types of
food in the diet with seafood. Dietary patterns were derived from
the Dutch National Food Consumption Survey of 1997/1998,
in which body weight and food intake were recorded for 6250
Dutch individuals using a 2-d food diary that included amount
and frequency of consumption. The authors considered 18 food
types in the model, 11 of which were fish/shellfish, and combined
levels of dioxins, dl PCBs, and EPA + DHA in these foods with the
dietary information to estimate total intake of these compounds.
In order to compare the FAO/WHO TDI for dioxins and dl PCBs
with the Health Council of The Netherlands adequate intake (AI)
for EPA + DHA (450 mg/d), the AI was expressed in terms of
body weight for a 65-kg individual (7 mg/kg bw/d). Based on 500
random samples from 10000 Monte Carlo simulations, the results
of the dietary analysis showed that in most cases (98% to 99%) the
EPA + DHA intake was below the body weight-adjusted AI and
the dioxin and dl PCB exposure was below the TDI established by
FAO/WHO. Only about 2% of the population was above the body
weight-adjusted AI and below the TDI for dioxins and dl PCBs.
In addition to calculating the percentage of the sample population
that is meeting health-based guidelines, the authors also examined
the probable effects of replacing beef and pork consumption with
salmon, eel, or a mixture of fatty fish (salmon, eel, herring, and
mackerel) at levels of 10%, 25%, 50%, and 100%. The base rate for
frequency of fatty fish consumption reported in the food diaries
was 5.3%, compared to 68% for beef, 74% for pork, and 27% for
chicken. Overall, the best scenario in terms of meeting healthbased guidance values was found to be substitution with 10% to
25% salmon or a mix of fatty fish. At higher percentages (50%
to 100%), there was a lower frequency of maintaining dioxin and
dl PCB levels below the TDI. At 10% replacement with these
fish categories, about 50% of the population reached the AI for
EPA +DHA and only 1.3% of the population exceeded the dioxin
and dl PCB limit, whereas at 25% replacement, more than 90%
of the sample population was able to reach the AI for EPA +
DHA, while less than 5% were predicted to exceed the limits
for dioxins and dl PCBs. On the other hand, substitution of beef
and pork with 25% eel was predicted to result in about 99% of
the population reaching the AI, but also about 11% would be
c 2012 Institute of Food Technologists®
exceeding the TDI for dioxins and dl PCBs. The authors pointed
out that this analysis was meant to illustrate the use of the statistical
model, and that a more complete analysis should be carried out
that considers uncertainties, alternative data sets, and additional
dietary scenarios. The use of a total diet model, as presented
here, allows for a better overall picture of the dietary intake of
certain compounds and food replacement scenarios may be useful
in developing risk management and communication strategies.
Risk-benefit assessment in Belgium was carried out by Sioen
and others (2008) using a probabilistic model to assess simultaneous exposure to PBDEs and omega-3 fatty acids exclusively due
to fish consumption. Consumption data for some 800 Belgian fish
consumers representative of the Belgian adult population with respect to age and region was obtained from a SEAFOODplus food
frequency questionnaire in 2004 and body weights were incorporated based on previously determined age and sex distributions for
the Belgian population. These individuals consumed an average
of 216 ± 204 g seafood per week, as compared to the general
Belgian population, which consumes an average of 168 g/wk.
Based on a 100 to 150 g serving size, these levels are similar to the
recommendations of the Belgian Health Council to consume fish
1 to 2 times per week. Levels of EPA + DHA and PBDEs were
obtained for 10 fish commonly available on the Belgian market
(for example, cod, salmon, tuna, saithe, and sole) using previously
published data and exposure levels for 4 different dietary scenarios
were calculated: (1) base consumption (216 g/wk of a variety of
10 fish), (2) consumption of 150 g/wk of cod (lean fish) and 150
g/wk of salmon (fatty fish), (3) consumption of 300 g/wk salmon,
and (4) consumption of 150 g/wk salmon and 150 g/wk herring
(also a fatty fish). Monte Carlo simulations were used to estimate the variability of the intakes in terms of consumption, body
weight, and concentration of the contaminants and nutrients in
fish. EPA + DHA intake was adjusted for body weight and was
compared to a health-based guidance value of 9.7 mg/kg bw/d
(derived from a dietary reference intake of 681 mg/d for a 70-kg
individual consuming 2046 kcal/d). The results of the analysis revealed that scenario 1 was associated with the lowest mean intakes
of both EPA + DHA (3.54 mg/kg bw/d) and PBDEs (0.85 ng/kg
bw/d), while consumption of 2 servings per week of salmon in
scenario 3 allowed for the highest ratio of EPA + DHA (11.9
mg/kg bw/d) to PBDEs (1.28 ng/kg bw/d). The replacement
of 1 serving of salmon with herring in scenario 4 also provided
elevated levels of EPA + DHA (9.6 mg/kg bw/d) compared to
scenarios 1 and 2, but led to slightly higher intake of PBDE
(2.4 ng/kg bw/d). While there is no established tolerable intake
for PBDEs, the lowest observed adverse effect level associated with
this group of compounds has been reported to be 0.6 mg/kg bw
for the penta-BDEs (Darnerud 2003; Siddiqi and others 2003).
Overall, increased fish consumption was associated with increased
intake of EPA + DHA and PBDEs, with the greatest benefit: risk
ratio being from consumption of 2 servings of salmon per week.
In a subsequent study, Sioen and others (2009) used the probabilistic approach described above to assess exposure to several
nutrients (EPA + DHA, vitamin D, iodine) and contaminants
(MeHg, iPCBs, dioxins + dl PCBs) on a global scale. The authors
used consumption data for 7 different seafood categories gathered
by the Global Environment Monitoring System (WHO 2007),
which reports average food consumption for 13 regional "cluster"
diets representing 183 countries worldwide. General levels of nutrients and contaminants were obtained from databases developed
in previous studies (Sioen and others 2007a, b) and an intake assessment was performed for all 13 cluster diets with probability
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 501
Risk-benefit analysis of seafood consumption . . .
distributions fitted for each seafood category, nutrient, and contaminant. Exposure levels were based on the general adult population, with a mean body weight of 60 kg for most regions and
55 kg for individuals from Asian countries. The highest
seafood intake was reported for Cluster L, which included
Japan, Korea, Philippines, Madagascar, and others, with 69.0
g/person/d, followed by Cluster F (the Nordic-Baltic countries;
49.2 g/person/d), and Cluster G (Afghanistan, China, India, Thailand, and others; 45.0 g/person/d). The 2 clusters with the highest
seafood intake had the highest intakes of EPA + DHA (400 to 600
mg/person/d), iodine (30 to 40 µg/person/d), and vitamin D (3
to 4 µg/person/d). Differences in the dietary patterns between
regions were reflected in the types of contaminants present: Clusters L and F consume relatively high levels of pelagic fish and had
the highest exposure to MeHg (about 200 to 300 ng/kg bw/d),
whereas Cluster G, which consumed a greater proportion of freshwater fish, cephalopods, crustaceans, and mollusks, had lower exposure to MeHg (about 100 ng/kg bw/d) but the highest exposure
to iPCBs (about 60 ng/kg bw/d) and dioxins + dl PCBs (about
3.3 pg TEQ/kg bw/d). In order to combine data for nutrients
and contaminants, the intake of EPA + DHA was divided by the
dietary reference intake (DRI) of 500 mg/d and graphed against
the exposure to either MeHg or dioxins + dl PCBs divided by the
TDIs established by the JECFA. These plots revealed that most
clusters were not exposed to contaminants above the tolerable exposure levels; however, they also were not obtaining sufficient EPA
+ DHA to meet the DRI, with intake levels of about 100 to 300
mg/d. The only cluster diet (Cluster L) that obtained 500 mg/d of
EPA + DHA also exceeded the TDIs for MeHg and dioxins + dl
PCBs. Cluster G exceeded the TDI for dioxins + dl PCBs and had
a daily EPA + DHA intake of about 200 to 300 mg, while cluster F
was just below the DRI for EPA + DHA and had exposure at levels
around the TDIs for both MeHg and dioxins + dl PCBs. When
the authors compared the mean exposure levels for each cluster to
the tolerable intakes for MeHg and dioxins + dl PCBs established
by the Scientific Advisory Committee on Nutrition/Committee
on Toxicity out of the United Kingdom (SACN/COT) to protect
against nondevelopmental health problems (Table 2), none of the
clusters exceeded the guidance values. There are several uncertainties of this study that could influence the estimated nutrient and
contaminant exposure levels. For example, food consumption data
were gathered by dividing food availability for a given country by
the total population and it tends to overestimate consumption by
about 15%. Also, the nutrient and contaminant data were based on
seafood tested in Europe or North America and do not represent
regional seafood consumed by some cluster diets. In conclusion,
the authors noted that the benefits outweigh the risks of seafood
consumption when the focus is on nondevelopmental effects and
they called for a more in-depth international study that would
include local nutrient and contaminant concentration data.
Cardoso and others (2010) examined seafood consumption patterns across 8 European countries (Germany, France, United Kingdom, Italy, Spain, The Netherlands, Portugal, and Iceland) and
calculated the probability of exceeding the tolerable intake for
MeHg or being deficient in EPA + DHA for consumers in each
country. The authors took into account the 5 most-consumed
types of seafood for each country and calculated per capita weekly
consumption, assuming that two-thirds of the seafood weight was
edible and average body weight was 60 kg. Because detailed consumption surveys were not used, log-normal distributions were
constructed to reflect seafood consumption patterns among different consumers, including individuals that do not regularly eat
seafood, and the values of each distribution curve were randomly
sampled using a sample size of 10000 with the Monte Carlo
method. As was the case with Sioen and others (2009), the nutrient and contaminant data were not representative of the entire
study population, but rather were based on seafood collected in
Portugal. The probabilities of exceeding the weekly reference intakes for MeHg and EPA + DHA from consumption of each
seafood species were calculated using a tail-estimation estimator
for most cases and a plug-in estimator for the few cases with
high probabilities. Total per capita seafood consumption for the
8 countries ranged from 140 g/wk in the United Kingdom to
630 g/wk in Iceland, while consumption of the top 5 species
examined in this study for each country ranged from 80 g/wk
in the United Kingdom to 390 g/wk in Iceland. Based on total
seafood consumption, the probability of exceeding 500 mg/d of
EPA + DHA was estimated at 0.3% for the United Kingdom,
2.0% for Italy, 12.4% for Germany and The Netherlands, 20.3%
to 24.2% for France and Iceland, 61.1% for Spain, and 66.0% for
Portugal. Although Spain and Portugal consume less per capita
seafood (210 and 290 g/wk, respectively) than Iceland, these 2
countries include sardines among the top 5 species, which are a
rich source of EPA + DHA. The probability of exceeding the
PTWI established by JECFA for MeHg based on total seafood
consumption was below 5% for most countries and reached 6.7%
for Portugal and 9.6% for Iceland. Among the top 5 fish consumed
in Iceland, the highest probabilities of exceeding the PTWI for
MeHg based on a single fish species were with tuna (0.50%) and
haddock (0.34%), which had consumption levels of 69 and 158
g/person/wk, respectively. However, exceeding the PTWI based
on exclusive consumption of either of these fish would require
about five 100-g servings of tuna per week (7 times the current
consumption levels) or fourteen 100-g servings of haddock (9.2
times the current consumption levels). The results of this study
highlight the fact that selecting fish high in EPA + DHA and low
in MeHg will improve the benefit:risk ratio related to seafood
consumption.
Assessments using combined risk-benefit dose-response
models. In 2009, the U.S. FDA issued 2 draft reports examining
the health outcomes of seafood consumption with the purpose
of providing additional scientific information to help address concerns over risks and benefits of commercial seafood in the United
States (FDA 2009a, b). In one report, the beneficial effects of
seafood consumption and omega-3 fatty acids for certain neurodevelopmental and cardiovascular endpoints were summarized
(FDA 2009b) and in the other report, a quantitative risk-benefit
assessment was conducted for seafood consumption (FDA 2009a).
The risk-benefit assessment was focused on 3 health endpoints:
(1) fetal neurodevelopment, (2) risk of fatal CHD, and (3) risk of
fatal stroke. The consideration of both the benefits and the risks
of seafood in the same quantitative analysis was a novel approach
for the FDA, which has historically focused on quantifying the
risk but not the countervailing benefits of a particular food. Current levels of U.S. fish consumption (that is, amounts and species)
were estimated based on 3 sources of data: a 3-d food survey
conducted by the U.S. Department of Agriculture between 1989
and 1991 (USDA 1993), the 30-d National Health and Nutrition Examination Survey (NHANES) conducted in 2001 to 2002
(CDC 2004), and market share data on consumable commercial
fish in 2005 from the National Marine Fisheries Service (NMFS).
This information was combined with MeHg concentrations in
different fish species, as reported by FDA, EPA, and NMFS, to
calculate current levels of MeHg exposure from fish consumption.
502 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
The negative effects of MeHg on fetal neurodevelopment were
modeled based on verbal development measurements primarily in
children from the Iraq MeHg wheat poisoning event (Marsh and
others 1987), with some data from the Seychelles Islands study
(Myers and others 1995), while dose-response relationships regarding the positive effects of fish consumption were modeled
using data from the UK cognitive development study (Daniels
and others 2004). The effects of prenatal MeHg exposure and fish
consumption were combined to estimate a net effect of IQ size
equivalents in offspring using several hypothetical dietary scenarios involving women of childbearing age (15 to 45 y). IQ size
equivalents are IQ points based on Z-Score conversions, which
are statistical tools that measure the size of an effect and facilitate
the comparison of results from different models. At current consumption levels (about 5% of women eating ≥340 g of fish/week,
95% of women eating <340 g/wk), there is an estimated net neurodevelopmental benefit equivalent to 0.225 IQ point per child,
with 99% of the population likely to have a net benefit on IQ size
equivalents (in excess of 1 IQ point per child for about 5% of population), 0.9% of the population likely to have no net effect, and
0.1% of the population likely to experience a net negative effect,
equivalent to about 0.04 IQ point. In a scenario where women
do not change their current consumption amounts, but instead
eat only fish low in MeHg (≤0.12 ppm), an increase equivalent
to 0.02 IQ point was predicted. Another scenario in which 100%
of women were eating exactly 340 g of fish per week resulted in
a net benefit equivalent to 0.57 IQ point. On the other hand,
if women that are currently eating more than 340 g/wk were to
decrease their consumption to this level, a net decrease equivalent
to IQ point of 0.01 was predicted. Overall, these results indicate
the greatest net neurodevelopmental benefit for pregnant women
with increased fish consumption, especially fish that are low in
MeHg.
Cardiovascular effects in the FDA risk-benefit assessment (FDA
2009a) were assessed using 2 types of models (meta-analysis and
pooled analysis) that were developed based on studies that reported the effects of fish consumption, but not omega-3 fatty
acids or MeHg, on CHD or stroke fatalities. The CHD metaanalysis model was based on the meta-analysis conducted by He
and others (2004b), while the CHD-pooled analysis model included the studies in this meta-analysis as well as 3 additional
studies published later (Folsom and Demissie 2004; Nakamura
and others 2005; Iso and others 2006). The stroke meta-analysis
model was based on another meta-analysis conducted by Bouzan
and others (2005). The stroke-pooled analysis model utilized this
meta-analysis as well as studies by Mozaffarian and others (2005),
Nakamura and others (2005), and 3 additional studies that were
analyzed by He and others (2004a). Based on the central estimates
of these models, current levels of fish consumption were estimated to be averting approximately 31000 (meta-analysis model)
to 40000 (pooled analysis model) deaths per year from CHD and
approximately 22000 (meta-analysis) to 25000 (pooled analysis)
deaths per year from stroke. Results of dietary scenarios suggested
that if all women of childbearing age ate 340 g of fish per week,
there would be a predicted decrease of approximately 250 (metaanalyses) to 340 (pooled analyses) deaths per year from CHD and
stroke. If there were a 10% decrease in the amount of fish consumed by adult men and older women (> 46 y), there would be
a predicted increase of approximately 3500 (pooled analyses) to
4000 (meta-analyses) deaths per year from CHD and stroke; and if
all adult men and women were to increase their fish consumption
by 50%, there would be a predicted decrease of approximately
c 2012 Institute of Food Technologists®
11000 (pooled analyses) to 18000 (meta-analyses) deaths per year
from CHD and stroke. However, it should be noted that the confidence intervals for the pooled analyses models are, by design,
wider than those of the meta-analyses models and therefore they
did include a small possibility that current fish consumption is
associated with deaths in each age/gender category. Nevertheless,
the bulk of the probability distribution did indicate a beneficial
effect of fish consumption, making it more likely than not that
increased fish consumption leads to a decrease in cardiovascular
mortality.
The approach of combining the risks and benefits into a net
health effect for a given endpoint was also used in a study examining potential development of species-specific fish consumption
advice (Ginsberg and Toal 2009). The concentrations of MeHg
and omega-3 fatty acids in 16 species of fish commonly available
in the state of Connecticut, U.S.A, were used to predict the developmental and cardiovascular health effects associated with each
fish. The cardiovascular dose-response relationship was developed
based on the results of Guallar and others (2002), who found that
the relative risk for a 1st myocardial infarction increased by 23%
per 1 ppm hair mercury and on the findings of Mozaffarian and
Rimm (2006), which showed that increasing EPA + DHA intake from 100 to 250 mg/d was associated with a 14.6% decrease
in the risk of CHD death. Although these endpoints are a measure of cardiovascular health and they were compared equally in
this risk-benefit assessment, CHD mortality includes sudden death
and death from myocardial infarction, whereas a first myocardial
infarction is not necessarily fatal. The dose-response relationship
for neurodevelopmental health was based on the study of Oken
and others (2005), who reported a 2.0-point increase in infant
visual recognition memory (VRM) score per 100 mg fish oil/d
and a 7.5-point decrease in VRM score per 1 ppm hair mercury.
However, it should be noted that Oken and others (2005) only
observed adverse effects among participants that had hair mercury
levels above 1.2 ppm, and the maximum level of hair mercury
reported was 2.4 ppm. Interestingly, Ginsberg and Toal (2009) incorporated a threshold of 0.51 ppm hair mercury into the cardiovascular dose-response model as the level below which no adverse
effects were evident, but the hair mercury threshold for adverse
cognitive effects observed by Oken and others (2005) was not
incorporated into the neurodevelopmental dose-response model.
The level of mercury exposure resulting from consumption of
one 170-g meal was calculated for each fish species and then converted to hair mercury concentration using a one-compartment
model (Rice and others 2003). The results showed that when one
to two, 170-g meals per week are consumed, the estimated cardiovascular benefits from omega-3 fatty acids outweigh the risks
from MeHg for all species except shark, swordfish, and yellowfin
tuna. On the other hand, one 170-g meal per week from 9 out
of the 16 fish examined was predicted to result in adverse neurodevelopmental effects, even for some low-mercury fish like cod
(0.11 ppm) and canned light tuna (0.12 ppm). This is likely due
to the relatively low levels of omega-3 fatty acids in these fish and
the fact that the hair mercury threshold level for adverse cognitive effects was not incorporated into the model. Consumption
of these 9 fish at this level would not be expected to exceed the
RfD established by EPA. As expected, low-mercury fish with high
levels of omega-3 fatty acids, such as salmon, trout, and herring,
exhibited the greatest net benefits to neurodevelopment. The authors used the results of the risk-benefit assessments and acceptable
body burdens based on the RfD to suggest species-specific fish
consumption advisories. For example, for individuals concerned
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 503
Risk-benefit analysis of seafood consumption . . .
with neurodevelopmental effects, the authors recommended unlimited consumption (up to one, 170-g meal per day) for 7 of
the fish species; limited consumption of canned light tuna and
cod (two, 170-g meals per week), limited consumption of 4 other
species (that is, canned white tuna, tuna steak, halibut, sea bass, and
lobster) to just one, 170-g meal per week, and complete avoidance
of shark and swordfish. However, the authors note that the focus
of this study was to present a framework for risk-benefit assessment
and the uncertainties in the dose-response relationships presented
here make the conclusions tentative.
Assessments based on composite metrics
Composite metrics allow for the integration of risks and benefits for multiple health endpoints into a single net health impact.
These assessments can combine 2 or more types of common metrics, such as mortality, morbidity, or disease incidence, to quantify
the cumulative effect on health. Most studies assessing the risks
and benefits of seafood with composite metrics have utilized the
QALYs to express the net health outcome (Ponce and others 2000;
Cohen and others 2005a; Guevel and others 2008), and 1 study
used a monetary value based on standard EPA health benefit transfer figures (Shimshack and Ward 2010). These studies are reviewed
here.
Studies using QALYs. The earliest publication that applied
QALYs to risk-benefit assessment of seafood was presented by
Ponce and others (2000). Benefits of fish consumption were defined as a decrease in myocardial infarction mortality and the risks
were defined as neurodevelopmental delays associated with prenatal MeHg exposure. The benefit to dose-response relationship for
fish consumption was modeled by logistic regression from summary epidemiological data collected for an adult male population
in Chicago, IL (U.S.A.), over a 30-y period, which found an inverse relationship between fish consumption and the risk of death
from CHD, especially nonsudden death from myocardial infarction
(MI) (Daviglus and others 1997). A Weibul excess risk model was
used to develop a dose-response relationship for risks using data on
delayed talking incidence among children in Iraq with gestational
exposure to contaminated grain (Marsh and others 1987). Fish
consumption was examined over a range of intakes (0 to 300 g/d)
and at a range of mercury concentrations (0.5 to 2.0 ppm). The
net health impact of fish consumption was then calculated for
2 different population groups (n = 100000 individuals/
population): (1) all members of a population and (2) women of
childbearing age (defined by Ponce and others (2000) as 15 to 44
y) and their offspring. When myocardial infarction mortality was
assumed to be equal in severity to delayed talking (that is, starting
to talk at 24 mo of age), the net effect of consumption of two,
100-g servings of fish per week with mercury levels of 0.5 to 2.0
ppm was predicted to be positive for the total population (2000
to 5000 QALYs), but negative for the subpopulation of women of
childbearing age and their offspring (−250 to 2000 QALYs). The
maximum benefits occurred among the total population when
more than ten, 100-g servings of fish with 0.5 ppm mercury were
consumed per week, with a net gain of about 15000 QALYs. On
the other hand, the subpopulation of women and children exhibited a net loss of 250 to 2000 QALYs for 2 servings of fish
per week. When myocardial infarction mortality was weighted as
more severe than delayed talking, two, 100-g servings of fish per
week were linked to a net benefit of about 5000 QALYs for the
total population, regardless of the mercury concentration in the
fish (0.5 to 2.0 ppm). The results of unequal weighting were reported to continue to result in a net negative health impact for
the subpopulation of women and children (QALYs not reported).
However, the benefits of fish consumption to neurodevelopment,
which would be expected to greatly improve the net health impact, were not considered in this model. Further, the mercury
levels for fish used in this model were higher than those in most
commonly consumed fish, which generally have concentrations of
<0.05 to 0.35 ppm, with the exception of a few large predatory
fish, such as swordfish, shark, and king mackerel (FDA 2009c).
To improve future analyses, the authors suggested the inclusion
of a greater number of health endpoints and scenarios comparing
fish-based diets with other types of diets, as well as the substitution
of fish with low MeHg levels.
To this regard, a comprehensive risk-benefit assessment was carried out for a range of fish consumption scenarios using composite metrics that incorporated dose-response relationships from
4 different studies (Cohen and others 2005a). An expert panel
convened by the Harvard Center for Risk Analysis published a
series of 4 papers developing dose-response relationships for fish
consumption and CHD mortality (König and others 2005); fish
consumption and stroke (Bouzan and others 2005); prenatal DHA
intake and cognitive development (Cohen and others 2005b); and
prenatal MeHg exposure and cognitive development (Cohen and
others 2005c). The dose-response relationships were only developed for health endpoints that were expected to be substantially
affected by changes in fish consumption and for which there were
sufficient data for a quantitative analysis. In the study determining
a relationship between fish consumption and heart disease mortality, including CHD and nonfatal MI, the authors identified 7
observational studies (Kromhout and others 1985; Ascherio and
others 1995; Daviglus and others 1997; Albert and others 1998;
Oomen and others 2000; Hu and others 2002; Mozaffarian and
others 2002) of individuals with no pre-existing CHD for use in
the dose-response analysis (König and others 2005). To develop a
dose-response relationship between fish consumption and stroke
risk, the authors combined relative risk results from 6 studies (5
prospective cohort studies and 1 case-controlled study) (Gillum
and others 1996; Orencia and others 1996; Iso and others 2001;
Caicoya 2002; He and others 2002; Bouzan and others 2005).
The dose-response relationship between prenatal intake of n-3
PUFAs and cognitive development utilized 8 randomized control trials (RCTs) (Agostoni and others 1997; Willatts and others
1998; Lucas and others 1999; Birch and others 2000; Makrides
and others 2000; Auestad and others 2001, 2003; Helland and
others 2003) comparing cognitive development for children or
mothers receiving n-3 PUFA supplementation (Cohen and others
2005b). The dose-response relationship between prenatal MeHg
and cognitive effects was determined by aggregating results from
3 major epidemiology studies conducted in the Faroe Islands,
Seychelles Islands, and New Zealand (Cohen and others 2005c).
The dose-response relationships developed in these studies were
then used to calculate the net public health impact of fish consumption patterns related to risk-benefit advisories. The impacts
of changes in fish consumption on MeHg exposure, omega-3
fatty acid intake, and servings of fish per week were estimated
using a modified version of a previously developed exposure assessment model (Carrington and Bolger 2002; Carrington and
others 2004). This model assumes that 10% to 20% of the population does not eat fish, and therefore health impacts due to changes
in fish consumption are assumed to affect 85% of the population.
Consumption rates for 42 types of fish were estimated using data
from the USDA Continuing Survey of Food Intake by Individuals
(CSFII) (USDA 1998) and NHANES data from 1999 to 2000
504 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
(CDC 2003). Levels of MeHg in fish were obtained from the
FDA and NMFS and levels of omega-3 fatty acids were derived
from the USDA Agricultural Research Service Nutrient Data Laboratory (http://www.nal.usda.gov/fnic/foodcomp/search/). The
average maternal body weight was estimated at 60 kg and the
baseline fish consumption was 18.7 g/d (130 g/wk) for women of
childbearing age (15 to 44 y) and 23.1 g/d (160 g/wk) for other
population members ≥15 y of age. The public health impacts of 5
dietary scenarios were compared to the baseline fish consumption
for the U.S. population: (1) women of childbearing age maintain
current amounts of fish consumption, but only eat fish with mercury levels ≤ 0.13 ppm, (2) women of childbearing age decrease
total fish consumption by 17%, regardless of mercury content, (3)
in addition to women of childbearing age, other members of the
population also reduce fish consumption by 17%, (4) all females
not of childbearing age and all males increase fish consumption
by 50%, and (5) all adult females (including those of childbearing
age) and males increase fish consumption by 50%. When women
of childbearing age maintained current consumption levels but
only ate fish lower in mercury (scenario 1), a net benefit of 49000
QALYs per year was predicted for the total population, primarily
due to the cognitive benefits of DHA, which would contribute an
average of 0.1 IQ point per child born. Scenario 2 was based on
a study that reported a 17% decrease in overall fish consumption
by pregnant women following the 2001 FDA advisory (Oken and
others 2003). This dietary scenario was associated with a substantially smaller net benefit to health as compared with scenario 1,
with + 0.02 IQ point per child and a net impact of 9700 QALYs
per year. In scenario 3, where all members of the population decrease fish consumption by 17%, the net health impact was negative
(−41000 QALYs per year), with the greatest losses experienced
by elderly males (ages 75 to 84), whose annual CHD mortality
risk would be increased by about 2 in 10000. On the other hand,
the greatest net benefit to health (+120000 QALYs per year) occurred in scenario 4 when fish consumption was increased by 50%
among all females not of childbearing age and all males, with a
reduced risk for CHD mortality of 5 in 10000 among elderly
men. In the case where all adult males and females increased fish
consumption by 50%, the total benefits were offset slightly by the
negative impact from MeHg on cognitive development (−0.07
IQ point/child), with a net health impact of +90000 QALYs per
year. The effects of POPs were not considered in this risk-benefit
assessment because they were not expected to be major contributors to the net health impact; for example, a sample calculation for
scenario 4 based on the data reported by Hites and others (2004a)
showed that exposure to organic contaminants was associated with
a loss of about 600 QALYs per year, compared to the net benefits
of 120000 QALYs per year. The overall results of this risk-benefit
assessment suggest the importance of fish consumption in terms
of the net health impact for the total population. When comparing the predicted results of scenarios 1 and 2, the importance of
correctly following advisories for women of childbearing age to
consume low-mercury fish, but not reduce total fish consumption is also apparent. The results of scenario 3, where the total
population reduces fish consumption, indicate the potential for
substantial reductions in public health and increased risks of CHD
mortality when advisories targeted at a specific population inadvertently discourage fish consumption among other population
groups.
Guevel and others (2008) utilized a QALY approach to assess the
risks and benefits of high fish consumers in France. Consumption
data for individuals consuming 2 or more servings of seafood per
c 2012 Institute of Food Technologists®
week were obtained from the CALIPSO study (Leblanc and others
2006; Bemrah-Aouachria and others 2008) and the most common
types of fish and seafood were sampled locally and analyzed for
fatty acids and MeHg. This information was combined to determine the exposure levels for these compounds among the target
populations and then to compare risks and benefits for consumers
with medium and high EPA + DHA intake. Consumers in the
1st quintile of the CALIPSO study (medium EPA + DHA intake)
had an average fish consumption of 334 g/wk, an average EPA
+ DHA intake of 391 mg/d, and an estimated MeHg exposure
of 0.8 µg/kg bw/wk. On the other hand, consumers in the 5th
quintile (high EPA + DHA intake) consumed an average of 1104
g seafood/wk, with a daily EPA + DHA intake of 2700 mg and a
weekly MeHg exposure of 2.6 µg/kg bw. The net QALYs associated with increasing EPA + DHA intake from medium to high levels were calculated for the adult population in France using doseresponse curves developed previously linking fish consumption to
cognitive and cardiovascular endpoints (Bouzan and others 2005;
Cohen and others 2005a, b, c; König and others 2005). Additional
dose-response curves linking EPA + DHA intake to the same cardiovascular endpoints were also developed by the authors based on
previous studies (Dolecek and Grandits 1991; Iso and others 2001;
He and others 2002; Hu and others 2002; Mozaffarian and others
2002). The net result of increasing EPA + DHA intake from 391 to
2700 mg/d was beneficial for both cognitive (+5949 QALYs) and
cardiovascular endpoints (91229 to 114475 QALYs), regardless of
the dose-response model. When all endpoints were combined, the
total QALYs associated with increasing fish-derived EPA + DHA
intake were 97248 using the EPA + DHA loglinear model developed by Guevel and others (2008); 116800 using the fish linear
model from the Harvard Center for Risk Analysis (Cohen and
others 2005a); and 285774 using the EPA + DHA exponential
model developed by Guevel and others (2008). Despite the net
benefits associated with increasing fish consumption and EPA +
DHA intake, the potential effects of MeHg on neurodevelopment
resulted in a negative lower bound of the 95% confidence intervals,
ranging from −104380 QALYs for the EPA + DHA exponential
model to −278665 for the EPA + DHA loglinear model. Overall,
these results were in agreement with Cohen and others (2005a)
that the benefits of fish consumption outweigh the risks; however,
the magnitude of these effects is influenced by the type of doseresponse curve utilized. Integration of additional risks and benefits
into the QALY model was recommended for future studies on this
topic.
Health-based monetary impacts. The use of monetary values to quantify risks and benefits of seafood consumption was
explored in a study that considered the effects of mercury advisories on dietary patterns (Shimshack and Ward 2010). The dietary
changes following the 2001 U.S. FDA advisory on mercury in
seafood were examined using household-level seafood consumption data obtained from the Information Resources, Inc.’s InfoScan Consumer Network database. The data used by Shimshack
and Ward were collected from close to 15000 consumers that
were asked to scan the universal product codes for all products purchased from all stores upon returning home over a 3-y
period (2000 to 2002). Changes in seafood types and amounts
were investigated following the 2001 advisory for “at-risk” households with pregnant women, nursing women, or children under
6. Similar to the results reported by Oken and others (2003), a
17% decrease in fish consumption by pregnant women was observed following this advisory (Shimshack and Ward 2010). Of
those who decreased their fish consumption, “at-risk” households
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 505
Risk-benefit analysis of seafood consumption . . .
decreased fish consumption 21.4% and there was a 60% increase
in the number of consumers with no significant fish and shellfish consumption. Overall, there was no evidence of differential avoidance of high mercury fish, with at-risk groups reducing
consumption of low-mercury seafood like salmon (27.9% reduction) and shrimp (17.5% reduction). Consumption of white tuna
and light tuna fell by 14.0% and 19.5%, respectively. However,
when education level was considered, households with a college
degree did show selective avoidance of high-mercury fish, with
an overall decrease in MeHg exposure of 27.9% (compared to
0.003% for less educated households), but there was also a substantial decrease of about 20% in the n-3 fatty acid intake among
both education groups. The health impacts of these declines in
seafood consumption were calculated using the cognitive and cardiovascular dose-response models developed previously (Bouzan
and others 2005; Cohen and others 2005a, b, c; König and others 2005). However, rather than using a QALY approach, the net
health impacts were expressed in terms of monetary values using U.S. EPA benefit transfer figures of $13084 per IQ point and
$7.52 million for the value of statistical life, based on 2007 U.S.
dollar amounts. Overall, the 21.4% reduction in seafood consumption following the 2001 U.S. FDA advisory was estimated to have a
net health impact of U.S. −$30. Declines in MeHg exposure were
associated with a benefit of +0.012 IQ points/child, while declines in the EPA + DHA intake were associated with −0.008 IQ
points/child, resulting in a net benefit of 0.004 IQ points/child
following the advisory (equivalent to U.S. $61). On the other
hand, the decline in EPA + DHA intake was also associated with
a net increase in CHD and stroke mortality of +0.63 deaths per
100000 adults (equivalent to U.S. −$91). The effects of an idealized scenario presented by Cohen and others (2005a; scenario
1), in which at-risk households maintain overall fish consumption
amounts but only eat low-mercury fish, were also examined by
Shimshack and Ward (2010) in terms of risks and benefits to neurodevelopment. This scenario resulted in a net benefit of 0.039
IQ points/child (U.S. $587). These results indicate the importance
of targeted strategies for reducing MeHg exposure without concomitantly reducing n-3 fatty acid intake in order to receive the
greatest health benefits from seafood.
Conclusions
While the application of risk-benefit analysis to specific foods
is a relatively new field of study, it represents a powerful tool that
allows decision makers to set policy that can have a universal beneficial effect. Although science-based, risk-benefit analysis remains
complex but will continually improve with increased analyses, new
methodologies, and larger studies. It also provides new research
opportunities for investigating the multiple components of food as
well as their physiological and nutritional interactions in populations. There are continuing efforts to maximize benefits and minimize risks through optimization of food consumption patterns.
Consumers are informed about what foods to avoid, while being
encouraged to consume others that will, hopefully, promote better health. Seafood is somewhat unique as few other foods present
so many deleterious and beneficial components in one packet.
Many of the studies highlighted in this review show the importance of maximizing the benefit: risk ratio by consuming fish that
are high in EPA + DHA but low in contaminants. Although the
benefits of EPA + DHA consumption can be negated by excessive consumption of high-MeHg seafood by sensitive populations,
such as pregnant women and young children, under-consumption
of EPA + DHA across both the at-risk and general population
groups is of equal concern. Studies also demonstrate that attempts
to “protect” an at-risk population can have negative unintended
consequences on that group and on the much larger overall population. It is therefore prudent that agencies consider the impact
of unintended consequences that messaging can create. Regardless of the risk-benefit method utilized, this review indicates that
results from studies overwhelmingly support the idea that messaging regarding seafood consumption risks requires a targeted
strategy that does not discount the benefits. Sensitive populations
will likely benefit by following risk advisories and reducing consumption of high-mercury fish; however, there is a need to be
careful about how these advisories are worded and released to
the public. Studies have shown that messaging reduces fish consumption in general, in both the target and nontarget populations,
resulting in an overall reduction in the potential health benefits
derived from EPA + DHA. This is a cause for concern as most
populations are not currently meeting the recommended intake
levels for EPA + DHA. While a majority of research has shown
that seafood consumption greatly outweighs the risks, it is important to keep in mind that this field of science is just at the
incipient stages of determining how to accurately assess the everyday choices we make in our diet and how these ultimately affect
our lives.
Acknowledgments
This work was partially supported by a grant from the
National Food Safety Initiative (Grant Nr 2007–5110-03815)
of the National Inst. of Food and Agriculture, U.S. Dept. of
Agriculture.
References
AFFSA. 2010. Opinion of the French Food Safety Agency on the update of
French population reference intakes (ANCs) for fatty acids. Available from:
http://www.afssa.fr/Documents/NUT2006sa0359EN.pdf. Accessed July
12, 2012.
Agostoni C, Trojan S, Bellu R, Riva E, Bruzzese MG, Giovannini M. 1997.
Developmental quotient at 24 months and fatty acid composition of diet in
early infancy: a follow-up study. Arch Dis Child 76:421–4.
Ahlqwist M, Bengtsson C, Lapidus L, Gergdahl IA, Schutz A. 1999. Serum
mercury concentration in relation to survival, symptoms, and diseases:
results from the prospective population study of women in Gothenburg,
Sweden. Acta Odontol Scand 57:168–74.
Albert CM, Hennekens CH, O’Donnell CJ. 1998. Fish consumption and
risk of sudden cardiac death. JAMA 279:23–8.
Anderson PD, Wiener JB. 1995. Eating fish. In: Graham JD, Wiener, JB,
editors. Risk vs. risk: tradeoffs in protecting health and the environment.
Cambridge, MA: Harvard University Press. p 104–23.
Ascherio A, Rimm EB, Stampfer MJ, Giovannucci EL, Willett WC. 1995.
Dietary intake of marine n-3 fatty acids, fish intake, and the risk of coronary
disease among men. N Engl J Med 332:977–82.
ASTDR. 1999. Agency for Toxic Substances and Disease Registry.
Toxicological profile for mercury. Available from: http://www.
atsdr.cdc.gov/toxprofiles/tp.asp?id=115&tid=24. Accessed July 12, 2012.
Auestad N, Halter R, Hall RT. 2001. Growth and development in term
infants fed long-chain polyunsaturated fatty acids: a double-masked,
randomized, parallel, prospective, multivariate study. Pediatrics 108:
372–81.
Auestad N, Scott DT, Janowsky J. 2003. Visual, cognitive, and language
assessments at 39 months: a follow-up study of children fed formulas
containing long-chain polyunsaturated fatty acids to 1 year of age. Pediatrics
112:E177–183.
Babut M, Miege C, Villeneuve B, Abarnou A, Duchemin J, Marchand P,
Narbonne JF. 2010. Correlations between dioxin-like and indicators PCBs:
potential consequences for environmental studies involving fish or sediment.
Environ Pollut 157:3451–6.
506 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
Bakir F, Rustam H, Tikriti S, Al-Damluji SF, Shihristani H. 1980. Clinical
and epidemiological aspects of methylmercury poisoning. Postgrad Med J
56(651):1–10.
Balk EM, Lichtenstein A, Chew P, Kupelnick B, Lawrence A, DeVine D,
Lau J. 2004. Effects of omega’3 fatty acids on cardiovascular risk factors and
intermediate markers of cardiovascular disease. Evidence Report/
Technology Assessment No. 93. Rockville, MD: AHRQ (prepared by the
Tufts-New England Medical Center Evidence-based Practice Center,
Boston, MA).
Bang HO, Dyerberg J. 1972. Plasma lipids and lipoprotein in Greenlandic
West Coast Eskimos. Acta Medica Scandinavica 192:85–94.
Bang HO, Dyerberg J. 1980. Lipid metabolism and ischemic heart disease in
Greenland Eskimos. In: Draper H, editor. Advances in nutrition research.
New York, NY: Plenum Press. p 1–22.
Barlow S, Chesson A, Collins JD, Flynn A, Galli CL, Hardy A, Jany K-D,
Jeger M-J, Knaap A, Kuiper H, Larsen J-C, Lovell D, Schlatter J, Silano V,
Smulders F, Vannier P. 2010. Guidance on human health risk-benefit
assessment of foods. EFSA J 8(7):1673.
Bemrah-Aouachria N, Sirot V, Leblanc JC, Volatier JL. 2008. Fish and
seafood consumption and omega 3 intake in French coastal populations:
CALIPSO survey. Public Health Nutr 12:599–608.
Bemrah N, Sirot V, Leblanc JC, Volatier JL. 2008. Fish and seafood
consumption and omega 3 intake in French coastal populations: CALIPSO
survey. Public Health Nutr 12:599–608.
Birch EE, Garfield S, Hoffman DR, Uauy R, Birch DG. 2000. A
randomized controlled trial of early dietary supply of LCPUFA and mental
development in term infants. Dev Med Child Neurol 42:174–81.
Bouzan C, Cohen JT, Connor WE, Kris-Etherton PM, Gray GM, König A,
Lawrence RS, Savitz DA, Teutsch SM. 2005. A quantitative analysis of fish
consumption and stroke risk. Am J Prev Med 29(4):347–52.
Brenna JT, Lapillonne A. 2009. Background paper on fat and fatty acid
requirements during pregnancy and lactation. Ann Nutr Metab
55(1–3):97–122.
Burger J, Stern AH, Gochfeld M. 2005. Mercury in commercial fish:
optimizing individual choices to reduce risk. Environ Health Perspect
113(3):266–71.
Caicoya M. 2002. Fish consumption and stroke: a community case-controlled
study in Asturias, Spain. Neuroepidemiology 21:107–14.
Calon F, Cole G. 2007. Neuroprotective action of omega-3 polyunsaturated
fatty acids against neurodegenerative diseases: evidence from animal studies.
Prostaglandins Leukot Essent Fatty Acids 77(5):287–93.
Health Canada. 2007. Canadian standards (“maximum limits”) for various
chemical contaminants in foods. Available from: http://www.hc-sc.gc.ca/fnan/securit/chem-chim/contaminants-guidelines-directives-eng.php.
Accessed July 12, 2012.
Cardoso C, Bandarra N, Lourenço H, Afonso C, Nunes M. 2010.
Methylmercury risks and EPA + DHA benefits associated with seafood
consumption in Europe. Risk Anal 30(5):827–40.
Carrington C, Bolger PM. 2002. An exposure assessment for methylmercury
from seafood for consumers in the United States. Risk Anal 22:689–99.
Carrington CD, Montwill B, Bolger PM. 2004. An intervention analysis
for the reduction of exposure to methylmercury from the consumption
of seafood by women of child-bearing age. Reg Toxicol Pharmacol 40:
272–80.
CDC. 2003. Centers for Disease Control and Prevention. National Center
for Health Statistics, National Health and Nutrition Examination Survey,
1999–2000. Available from: http://www.cdc.gov/nchs/nhanes.htm.
Accessed July 12, 2012.
CDC. 2004. Centers for Disease Control and Prevention. National Center
for Health Statistics, National Health and Nutrition Examination Survey,
2001–2002. Available from: http://www.cdc.gov/nchs/nhanes.htm.
Accessed July 12, 2012.
Choi AL, Budtz-Jorgernsen E, Jorgensen PJ, Steuerwald U, Debes F, Weihe
P, Grandjean P. 2008. Selenium as a potential protective factor against
mercury development neurotoxicity. Environ Res 107(1):
45–52.
Cohen JT, Bellinger DC, Connor WE, Kris-Etherton PM, Lawrence RS,
Savitz DA, Shaywitz BA, Teutsch SM, Gray GM. 2005a. A quantitative
risk-benefit analysis of changes in population fish consumption. Am J Prev
Med 29(4):325–34.
Cohen JT, Bellinger DC, Connor WE, Shaywitz BA. 2005b. A quantitative
analysis of prenatal intake of n-3 polyunsaturated fatty acids and cognitive
development. Am J Prev Med 29(4):366–74.
c 2012 Institute of Food Technologists®
Cohen JT, Bellinger DC, Shaywitz BA. 2005c. A quantitative analysis of
prenatal methyl mercury exposure and cognitive development. Am J Prev
Med 29(4):353–3.
Crépet A, Tressou J, Verger P, Leblanc JC. 2005. Management options to
reduce exposure to methyl mercury through the consumption of fish and
fishery products by the French population. Reg Toxicol Pharmacol
42(2):179–89.
Crump KS, Kjellstrom T, Shipp AM, Silvers A, Stewart A. 1998. Influence
of prenatal mercury exposure upon scholastic and psychological test
performance: benchmark analysis of a New Zealand cohort. Risk Anal
18(6):701–13.
Dabeka R, McKenzie AD, Forsyth DS, Conacher HB. 2004. Survey of total
mercury in some edible fish and shellfish species collected in Canada in
2002. Food Addit Contam 21:434–40.
Daniels JL, Longnecker MP, Klebanoff MA, Gray KA, Brock JW, Zhou H,
Chen Z, Needham LL. 2003. Prenatal exposure to low-level
polychlorinated biphenyls in relation to mental and motor development at
8 months. Am J Epidemiol 157:485–92.
Daniels JL, Longnecker MP, Rowland AS, Golding J. 2004. Fish intake
during pregnancy and early cognitive development of offspring.
Epidemiology 15(4):394–402.
Darnerud PO. 2003. Toxic effects of brominated flame retardants in man and
in wildlife. Environ Intl 29:841–53.
Davidson PW, Myers GJ, Cox C, Axtell C, Shamlaye C, Sloane-Reeves J,
Cernichiari E, Needham L, Choi A, Wang Y, Berlin M, Clarkson TW.
1998. Effects of prenatal and postnatal methylmercury exposure from fish
consumption on neurodevelopment: outcomes at 66 months of age in the
Seychelles Child Development Study. JAMA 280(8):701–7.
Daviglus ML, Stamler J, Orencia AJ, Dyer AR, Liu K, Greenland P, Walsh
MK, Morris D, Shekelle RB. 1997. Fish consumption and the 30-year risk
of myocardial infarction. N Engl J Med 336(15):1046–53.
Delange F, Lecomte P. 2000. Iodine supplementation: benefits outweigh
risks. Drug Safety 22(2):89–95.
Dewailly E, Ayotte P, Lucas M, Blanchet C. 2007. Risk and benefits from
consuming salmon and trout: a Canadian perspective. Food Chem Toxicol
45(8):1343–8.
DGAC. 2010. Report of the dietary guidelines advisory committee on
the dietary guidelines for Americans, 2010. United States Department
of Agriculture Center for Nutrition Policy and Promotion. Available from:
http://www.cnpp.usda.gov/dietaryguidelines.htm. Accessed July 12,
2012.
Dolecek TA, Grandits G. 1991. Dietary polyunsaturated fatty acids and
mortality in the multiple risk factor intervention trial (MRFIT). World Rev
Nutr Diet 66:205–16.
Domingo JL, Bocio A, Falcó G, Llobet JM. 2007a. Benefits and risks of fish
consumption: Part I. A quantitative analysis of the intake of omega-3 fatty
acids and chemical contaminants. Toxicology 230(2–3):219–26.
Domingo JL, Bocio A, Martı́-Cid R, Llobet JM. 2007b. Benefits and risks of
fish consumption: Part II. RIBEPEIX, a computer program to optimize the
balance between the intake of omega-3 fatty acids and chemical
contaminants. Toxicology 230(2–3):227–33.
EC. 2006. European Commission Regulation No. 1881/2006 of 19
December 2006. Setting maximum levels for certain contaminants in
foodstuffs. Available from: http://eur-lex.europa.eu/LexUriServ/
LexUriServ.do?uri=CONSLEG:2006R1881:20100701:EN:PDF. Accessed
July 12, 2012.
EFSA. 2006. European Food Safety Authority. Colloquium 6: Risk-benefit
analysis of foods: methods and approaches; 2006 Jul 13–14; Parma, Italy.
Available from: http://www.efsa.europa.eu/en/colloquiareports/
colloquiariskbenefit.htm. Accessed July 12, 2012.
EPA. 2000. National guidance: guidance for assessing chemical contaminant
data for use in fish advisories. Vol. 2: Risk assessment and fish consumption
limits,3rd ed. Available from: http://water.epa.gov/scitech/swguidance/
fishshellfish/techguidance/risk/volume2_index.cfm. Accessed July 12, 2012.
EPA. 2005. Guidelines for carcinogen risk assessment. Risk assessment
forum, U. S. Environmental Protection Agency, Washington, D.C. Available
from: http://www.epa.gov/cancerguidelines/. Accessed July 12, 2012.
EPA. 2010. U.S. Environmental Protection Agency. Persistent
bioaccumulative and toxic chemical program. Dioxins and furans. Available
from: http://www.epa.gov/pbt/pubs/dioxins.htm. Accessed July 12, 2012.
Eto K. 2000. Minamata disease. Neuropathology 20:S14–19. FAO. 2009.
Fishery and aquaculture statistics 2007. FAO yearbook. Rome: FAO
Fisheries and Aquaculture Information and Statistics Service. p 72.
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 507
Risk-benefit analysis of seafood consumption . . .
FAO/WHO. 1997. Report from a joint consultation: risk management and
food safety. Available from: ftp://ftp.fao.org/docrep/fao/w4982e/
w4982e00.pdf. Accessed July 12, 2012.
FAO/WHO. 2008. Interim summary of conclusions and dietary
recommendations on total fat, fatty acids. From the Joint FAO/WHO
Expert Consultation on Fats and Fatty Acids in Human Nutrition; 2008
Nov 10–14; WHO, Geneva. Available from:
http://www.who.int/nutrition/topics/FFA_interim_recommendations/en/
index.html. Accessed July 12, 2012.
FDA. 2004a. Backgrounder for the 2004 FDA/EPA consumer advisory: what
you need to know about mercury in fish and shellfish. Available from:
http://water.epa.gov/scitech/swguidance/fishshellfish/outreach/factsheet.
cfm#diet. Accessed July 12, 2012.
FDA. 2009a. Draft report of quantitative risk and benefit assessment of
consumption of commercial fish, focusing on fetal neurodevelopment effects
(measured by verbal development in children) and on coronary heart disease
and stroke in the general population. Available from: http://www.fda.gov/
Food/FoodSafety/Product-SpecificInformation/Seafood/
FoodbornePathogensContaminants/Methylmercury/ucm088794.htm.
Accessed July 12, 2012.
FDA. 2009b. Draft summary of published research on the beneficial effects of
fish consumption and omega-3 fatty acids for certain neurodevelopmental
and cardiovascular endpoints. Available from: http://www.fda.gov/Food/
FoodSafety/Product-SpecificInformation/Seafood/
FoodbornePathogensContaminants/Methylmercury/ucm153051.htm.
Accessed July 12, 2012.
FDA. 2009c. Mercury levels in commercial fish and shellfish. Available from:
http://www.fda.gov/Food/FoodSafety/Product-SpecificInformation/
Seafood/FoodbornePathogensContaminants/Methylmercury/ucm115644.
htm. Accessed July 12, 2012.
FDA. 2011. Environmental chemical contaminants and pesticides. Fish and
fisheries products hazards and controls guidance, 3rd ed. Chapter 9. Silver
Spring, Md.: U.S. Food and Drug Administration. Available from:
http://www.fda.gov/Food/GuidanceComplianceRegulatoryInformation/
GuidanceDocuments/Seafood/FishandFisheriesProductsHazardsand
ControlsGuide/default.htm. Accessed July 12, 2012.
Fleith M, Clandinin MT. 2005. Dietary PUFA for preterm and term infants:
review of clinical studies. Crit Rev Food Sci Nutr 45(3):205–29.
Folsom AR, Demissie Z. 2004. Fish intake, marine omega-3 fatty acids, and
mortality in a cohort of postmenopausal women. Am J Epidemiol
160:1005–10.
Foran JA, Carpenter DO, Hamilton MC, Knuth BA, Schwager SJ. 2005a.
Risk-based consumption advice for farmed Atlantic and wild Pacific salmon
contaminated with dioxins and dioxin-like compounds. Environ Health
Perspect 113(5):552–6.
Foran JA, Good DH, Carpenter DO, Hamilton MC, Knuth BA, Schwager
SJ. 2005b. Quantitative analysis of the benefits and risks of consuming
farmed and wild salmon. J Nutr 135:2639–40.
Fransen H, De Jong N, Hendriksen M, Mengelers M, Castenmiller J,
Hoekstra J, Van Leeuwen R, Verhagen H. 2010. A tiered approach for
risk-benefit assessment of foods. Risk Anal 30(5):808–16.
Gillum RF, Mussolino ME, Madans JH. 1996. The relationship between fish
consumption and stroke incidence: the NHANES I epidemiologic
follow-up study (National Health and Nutrition Examination Survey). Arch
Intern Med 156(5):537–42.
Ginsberg GL, Toal BF. 2009. Quantitative approach for incorporating
methylmercury risks and omega-3 fatty acid benefits in developing
species-specific fish consumption advice. Environ Health Perspect
117(2):267–75.
Gochfeld M, Burger J. 2005. Good fish/bad fish: a composite benefit-risk by
dose curve. Neurotoxicology 26(4):511–20.
Grandjean P, Weihe P, Burse VW, Needham LL, Storr-Hansen E, Heinzow
B, Debes F, Murata K, Simonsen H, Ellefsen P, Budtz-Jørgensen E, Keiding
N, White RF. 2001. Neurobehavioral deficits associated with PCB in
7-year-old children prenatally exposed to seafood neurotoxicants.
Neurotoxicol Teratol 23:305–17.
Grandjean P, Weihe P, White RF, Debes F, Araki S, Yokoyama K, Murata
K, Sorensen N, Dahl R, Jorgensen PJ. 1997. Cognitive deficit in 7-year-old
children with prenatal exposure to methylmercury. Neurotoxicol Teratol
19(6):417–28.
Gray KA, Klebanoff MA, Brock JW, Zhou H, Darden R, Needham L,
Longnecker MP. 2005. In utero exposure to background levels of
polychlorinated biphenyls and cognitive functioning among school-age
children. Am J Epidemiol 162:17–26.
Guallar E, Sanz-Gallardo MI, van’t Veer P, Bode P, Aro A, Gomez-Aracena
J, Kark JD, Riemersma RA, Martin-Moreno JM, Kok FJ. 2002. Heavy
Metals and Myocardial Infarction Study Group. Mercury, fish oils, and the
risk of myocardial infarction. N Engl J Med 347:1747–54.
Guevel MR, Sirot V, Volatier JL, Leblanc JC. 2008. A risk-benefit analysis of
French high fish consumption: a QALY approach. Risk Anal 28(1):
37–48.
Gunderson EL. 1995. Dietary intakes of pesticides, selected elements, and
other chemicals: FDA total diet study, June 1984-April 1986. J AOAC Intl
78(4):910–21.
Hallgren CG, Hallmans G, Jansson JH, Marklund SL, Huhtasaari F, Schutz
A, Stromberg U, Vessby B, Skerfving S. 2001. Markers of high fish intake
are associated with decreased risk of a first myocardial infarction. Br J Nutr
86:397–404.
Harada M. 1995. Minamata disease: methylmercury poisoning in Japan
caused by environmental pollution. Crit Rev Toxicol 25(1):1–24.
Harris WS, Kris-Etherton PM, Harris KA. 2008. Intakes of long-chain
omega-3 fatty acid associated with reduced risk for death from coronary
heart disease. Curr Atheroscler Rep 10:503–9.
Harris WS, Mozaffarian D, Lefevre M, Toner CD, Colombo J, Cunnane SC,
Holden JM, Klurfeld DM, Morris MC, Whelan J. 2009. Towards
establishing dietary reference intakes for eicosapentaenoic and
docosahexaenoic acids. J Nutr 139(4):804S-819.
He K, Rimm EB, Merchant A, Rosner BA, Stampfer MJ, Willett WC,
Ascherio A. 2002. Fish consumption and risk of stroke in men. JAMA
288:3130–6.
He K, Song Y, Daviglus ML, Liu K, Van Horn L, Dyer AR, Goldbourt U,
Greenland P. 2004a. Fish consumption and incidence of stroke: a
meta-analysis of cohort studies. Stroke 35:1538–42.
He K, Song Y, Daviglus ML, Liu K, Van Horn L, Dyer AR, Greenland P.
2004b. Accumulated evidence on fish consumption and coronary heart
diesase mortality: a meta-analysis of cohort studies. Circulation
109:2705–11.
Helland IB, Smith L, Saarem K, Saugstad OD, Drevon CA. 2003. Maternial
supplimentation with very long chain n-3 fatty acids during pregnancy and
lactation augment children’s IQ at 4 years of age. Pediatrics 111:
E39–44.
Hibbeln JR, Davis JM, Steer C, Emmett P, Rogers I, Williams C, Golding J.
2007. Maternal seafood consumption in pregnancy and neurodevelopmental
outcomes in childhood (ALSPAC study): an observational cohort study.
Lancet 369(9561):578–85.
Hibbeln JR. 2009. Depression, suicide and deficiencies of omega-3 essential
fatty acids in modern diets. In: Simopoulos AP, Bazan NG, editors.
Omega-3 fatty acids, the brain and retina. New York, N.Y.: Karger. p 17–30.
Hites RA, Foran JA, Carpenter DO, Hamilton MC, Knuth BA, Schwager
SJ. 2004a. Global assessment of organic contaminants in farmed salmon.
Science 303(January 9):226–9.
Hites RA, Foran JA, Schwager SJ, Knuth BA, Hamilton MC, Carpenter
DO. 2004b. Global assessment of polybrominated diphenyl ethers in farmed
and wild salmon. Environ Health Perspect 38:4945–9.
Hu FB, Bronner L, Willet WC, Stampfer MJ, Rexrode KM, Albert CM,
Hunter D, Manson JAE. 2002. Fish and omega-3 fatty acid intake and risk
of coronary disease in women. JAMA 287:1815–21.
Huang X, Hites RA, Foran JA, Hamilton C, Knuth BA, Schwager SJ,
Carpenter DO. 2006. Consumption advisories for salmon based on risk of
cancer and noncancer health effects. Environ Res 101(2):263–274.
Hudert CA, Weylandt KH, Lu Y, Wang J, Hong S, Dignass A, Serhan CN,
Kang JX. 2006. Transgenic mice rich in endogenous omega-3 fatty acids are
protected from colitis. Proc Natl Acad Sci USA 103(30):11276–81.
IOM. 1991. Seafood safety: committee on evaluation of the safety of fishery
products. Washington, D.C.: National Academy of Sciences.
IOM. 2005. Seafood choices: balancing benefits and risks/Committee on
Nutrient Relationships in Seafood: selections to balance benefits and risks,
food and nutrition board. Washington, D.C.: National Academy of Sciences.
IPCS. 1997. Environmental Health Criteria 195: Hexachlorobenzene.
Geneva, World Health Organization, International Programme on
Chemical Safety. Available from: http://www.inchem.org/documents/
ehc/ehc/ehc195.htm. Accessed July 12, 2012.
Iso H, Kobayishi M, Ishihara J, Sasaki S, Okada K, Kita Y, Kokuba Y,
Tsugane S. 2006. Intake of fish and n3 fatty acids and risk of coronary heart
disease among Japanese. The Japan Public Health Center-Based (JPHC)
Study Cohort I. Circulation 113:195–202.
508 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
Iso H, Rexrode KM, Stampfer MJ, Manson JAE, Colditz GA, Speizer FE,
Hennekens CH, Willett WC. 2001. Intake of fish and omega-3 fatty acids
and risk of stroke in women. JAMA 285:304–12.
ISSFAL. 2004. International Society for the Study of Fatty Acids and Lipids.
Report of the sub-committee on the recommendations for intake of
polyunsaturated fatty acids in healthy adults. Available from:
http://www.issfal.org/news-links/resources/publications/
PUFAIntakeReccomdFinalReport.pdf. Accessed July 12, 2012.
Iwamoto M, Ayers T, Mahon BE, Swerdlow DL. 2010. Epidemiology of
seafood-associated infections in the United States. Clin Microbiol Rev
23(2):399–411.
Jacobson JL, Jacobson SW. 1996. Intellectual impairment in children exposed
to polychlorinated biphenyls in utero. N Engl J Med 335:783–789.
Jacobson JL, Jacobson SW, Humphrey HE. 1990. Effects of in utero exposure
to polychlorinated biphenyls and related contaminants on cognitive
functioning in young children. J Pediatr 116:38–45.
JECFA. 2001. Joint FAO/WHO Expert Committee on Food Additives:
fifty-seventh meeting, Rome, 5–14 June. Available from: http://whqlibdoc.
who.int/trs/WHO_TRS_909.pdf. Accessed July 12, 2012.
JECFA. 2003. Joint FAO/WHO Expert Committee on Food Additives:
sixty-first meeting, Rome, 10–19 June. Summary and conclusions.
Available from: ftp://ftp.fao.org/es/esn/jecfa/jecfa61sc.pdf. Accessed July
12, 2012.
JECFA. 2010. Report of the Joint FAO/WHO Expert Consultation on the
Risks and Benefits of Fish Consumption, Rome, 25–29 January 2010.
Available from: http://www.fao.org/docrep/014/ba0136e/ba0136e00.pdf.
Accessed July 12, 2012.
Juhlshamn K, Andersen A, Ringdal O, Morkore J. 1987. Trace elements
intake in the Faroe Islands: I. Element levels in edible parts of pilot whales
(globicephalus meleanus). Sci Total Environ 65:53–62.
Kaiser L, Allen LH. 2008. Position of the American Dietetic Association:
nutrition and lifestyle for a healthy pregnancy outcome. J Am Diet Assoc
108:553–61.
Koletzko B, Cetin I, Brenna JT. 2007. Dietary fat intakes for pregnant and
lactating women. Br J Nutr 98:873–7.
König A, Bouzan C, Cohen JT, Connor WE, Kris-Etherton PM, Gray GM,
Lawrence RS, Savitz DA, Teutsch SM. 2005. A quantitative analysis of fish
consumption and coronary heart disease mortality. Am J Prev Med
29(4):335–46.
Kremer JM. 2000. n-3 Fatty acid supplements in rheumatoid arthritis. Am J
Clin Nutr 71(1):349S-351.
Kris-Etherton PM, Harris WS, Appel LJ. 2002. Fish consumption, fish oil,
omega-3 fatty acids, and cardiovascular disease. Circulation
106(21):2747–57.
Kris-Etherton PM, Harris WS, Appel LJ. 2003. Omega-3 fatty acids and
cardiovascular disease. Arterioscler Thromb Vasc Biol 23:151–2.
Kromhout D, Bosschieter EB, de Lezenne Coulander C. 1985. The inverse
relationship between fish consumption and 20-year mortality from coronary
heart disease. N Engl J Med 312(19):1205–09.
Leblanc JC, Sirot V, Volatier JL, Bemrah-Aouachria N. 2006. Fish and
seafood consumption study and biomarker of exposure to trace elements,
pollutants and omega-3 (CALIPSO). Available from: http://www.afssa.
fr/Documents/PASER-Ra-CalipsoEN.pdf. Accessed August 2, 2010.
Longnecker MP, Wolff MS, Gladen BC, Brock JW, Grandjean P, Jacobson
JL, Korrick SA, Rogan WJ, Weisglas-Kuperus N, Hertz-Picciotto I, Ayotte
P, Stewart P, Winneke G, Charles MJ, Jacobson SW, Dewailly E, Boersma
ER, Altshul LM, Heinzow B, Pagano J, Jensen AA. 2003. Comparison of
ploychlorinated biphenyl levels across studies of human neurodevelopment.
Environ Health Perspect 111(1):65–70.
Lonky E, Reihman J, Darvill T, Sr. JM, Daly H. 1996. Neonatal behavioral
assessment scale performance in humans influenced by maternal
consumption of environmentally contaminated lake ontario fish. J Great
Lakes Res 22(2):198–212.
Lucas A, Stafford M, Morley R. 1999. Efficacy and safety of long-chain
polyunsaturated fatty acid supplimentation of infant-formula milk: a
randomized trial. Lancet 354:1948–1954.
Makrides M, Duley L, Olsen SF. 2006. Marine oil, and other prostaglandin
precursors, supplementation for pregnancy uncomplicated by pre-eclampsia
or intrauterine growth restriction. Cochrane Database of Systematic
Reviews 2006, Issue 3. Art. No.: CD003402.
doi:10.1002/14651858.CD003402.pub2.
Makrides M, Neumann MA, Simmer K, Gibson RA. 2000. A critical
appraisal of the role of dietary long-chain polyunsaturated fatty acids on
c 2012 Institute of Food Technologists®
neural indices of infants: a randomized, controlled trial. Pediatrics 105:
32–8.
Marsh DO, Clarkson TW, Cox C, Myers GJ, Amin-Zaki L, Al-Tikriti S.
1987. Fetal methylmercury poisoning, relationship between concentration
in single strands of maternal hair and child effects. Arch Neurol 44:
1017–22.
Masuda Y, Schecter AJ, Päpke O. 1996. Concentration of PCBs, PCDFs and
PCDDs in the blood of Yusho patients and their toxic equivalent
contributions. Organohalogen Compounds 30:146–9.
McDowell MA, Dillon CF, Osterloh J, Bolger PM, Pellizzari E, Fernando
R, Montes de Oca R, Schober SE, Sinks T, Jones RL, Mahaffey KR. 2004.
Hair mercury levels in U.S. children and women of childbearing age:
reference range data from NHANES 1999–2000. Environ Health Perspect
112(11):1165–71.
Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS. 2005. Fish
consumption and cognitive decline with age in a large community study.
Arch Neurol 62:1849–53.
Mozaffarian D. 2008. Fish and n-3 fatty acids for the prevention of fatal
coronary heart disease and sudden cardiac death. Am J Clin Nutr
87(6):1991S-1996.
Mozaffarian D, Lemaitre RN, Kuller LH, Burke GL, Tracy RP, Siscovick
DS. 2002. Cardiac benefits of fish consumption may depend on the type of
fish meal consumed: The cardiovascular, health study. Circulation
107:1372–77.
Mozaffarian D, Longstreth T, Lemaitre RN, Manolio TA, Kuller LH, Burke
GL, Siscovick DS. 2005. Fish consumption and stroke risk in elderly
individuals: the cardiovascular health study. Arch Intl Med 65:200–6.
Mozaffarian D, Rimm EB. 2006. Fish intake, contaminants, and human
health: evaluating the risks and the benefits. JAMA 296:1885–99.
Myers GJ, Davidson PW, Cox C, Shamlaye CF, Palumbo D, Cernichiari E,
Sloane-Reeves J, Wilding GE, Kost J, Huang LS, Clarkson TW. 2003.
Prenatal methylmercury exposure from ocean fish consumption in the
Seychelles child development study. Lancet 361(9370):1686–92.
Myers GJ, Marsh DO, Davidson PW, Cox C, Samlaye CF, Tanner M, Choi
A, Cernichiari E, Choisy O, Clarkson TW. 1995. Main
neurodevelopmental study of Seychellois children following in utero
exposure to methylmercury from a maternal fish diet: outcome at six
months. Neurotoxicology 16(4):653–64.
Nakajima S, Saijo Y, Kato S, Sasaki S, Uno A, Kanagami N, Hirakawa H,
Hori T, Tobiishi K, Todaka T, Nakamura Y, Yanagiya S, Sengoku Y, Iida
T, Sata F, Kishi R. 2006. Effects of prenatal exposure to polychlorinated
biphenyls and dioxins on mental and motor development in Japanese
children at 6 months of age. Environ. Health Perspect 114:773–8.
Nakamura Y, Hirotsugu U, Okamura T, Kadowaki T, Hayakawa T, Kita Y,
Tamaki S, Okayama A. 2005. Association between fish consumption and
all-cause and cause-specific mortality in Japan: NIPPON DATA80,
1980–99. Am J Med 118:239–245.
NRC. 1989. Diet and health: implications for reducing chronic disease risk.
Washington, DC: National Academy Press.
NRC. 2000. Toxicological effects of methylmercury. Washington, DC:
National Academy Press.
Oken E, Kleinman KP, Berland WE, Simon SR, Rich-Edwards JW,
Gillman MW. 2003. Decline in fish consumption among pregnant women
after a national mercury advisory. Obstet Gynecol 102(2):346–351.
Oken E, Osterdal ML, Gillman MW, Knudsen VK, Halldorsson TI, Strom
M, Bellinger DC, Hadders-Algra M, Michaelsen KF, Olsen SF. 2008a.
Associations of maternal fish intake during pregnancy and breastfeeding
duration with attainment of developmental milestones in early childhood: a
study from the Danish national birth cohort. Am J Clin Nutr 88(3):
789–96.
Oken E, Radesky JS, Wright RO, Bellinger DC, Amarasiriwardena CJ,
Kleinman KP, Hu H, Gillman MW. 2008b. Maternal fish intake during
pregnancy, blood mercury levels, and child cognition at age 3 years in a US
cohort. Am J Epidemiol 167(10):1171–81.
Oken E, Wright RO, Kleinman KP, Bellinger D, Amarasiriwardena CJ, Hu
H, Rich-Edwards JW, Gillman MW. 2005. Maternal fish consumption, hair
mercury, and infant cognition in a U.S. Cohort. Environ Health Perspect
113(10):1376–80.
Oomen cM, Feskens EJ, Rasanen L. 2000. fish consumption and coronary
heart disease mortality in Finland, Italy, and The Netherlands. Am J
Epidemiol 151:999–1006.
Orencia AJ, Daviglus ML, Dyer AR, Shekelle RB, Stamler J. 1996. Fish
comsumption and stroke in men: 30-year findings of the Chicago Western
Electric Study. Stroke 27:204–9.
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 509
Risk-benefit analysis of seafood consumption . . .
Patandin S, Lanting CI, Mulder PG, Boersma ER, Sauer PJ,
Weisglas-Kuperus N. 1999. Effects of environmental exposure to
polychlorinated biphenyls and dioxins on cognitive abilities in Dutch
children at 42 months of age. J Pediatr 134:33–41.
Paulsson K & Lundbergh K. 1989. The selenium method for treatment of
lakes for elevated levels of mercury in fish. Sci Total Environ 87–88:495–507.
Plessi M, Bertelli D, Monzani A. 2001. Mercury and selenium content in
selected seafood. J Food Comp Anal 14(5):461–7.
Ponce RA, Bartell SM, Wong EY, LaFlamme D, Carrington C, Lee RC,
Patrick DL, Faustman EM, Bolger M. 2000. Use of quality-adjusted life year
weights with dose-response models for public health decisions: a case study
of the risks and benefits of fish consumption. Risk Anal 20(4):
529–42.
Pouzaud F, Ibbou A, Blanchemanche S, Grandjean P, Krempf M, Philippe
H-J, Verger P. 2009. Use of advanced cluster analysis to characterize fish
consumption patterns and methylmercury dietary exposures from fish and
other sea foods among pregnant women. J Expos Sci Environ Epidemiol
20(1):54–68.
Ralston NVC, Ralston CR, Blackwell III JL, Raymond LJ. 2008. Dietary
and tissue selenium in relation to methylmercury toxicity. Neurotoxicology
29(5):802–11.
Rasmussen RS, Nettleton JA, Morrissey MT. 2005. A review of mercury in
seafood: special focus on tuna. J Aquat Food Prod Tech 14(4):71–100.
Rawn DF, Forsyth DS, Ryan JJ, Breakell K, Verigin V, Nicolidakis H,
Hayward S, Laffey P, Conacher HB. 2002. PCB, PCDD and PCDF
residues in fin and non-fin fish products from the Canadian retail market.
Sci Total Environ 359(1–3):101–10.
Rayman MP. 2000. The importance of selenium to human health. Lancet
356:233–41.
Ribas-Fito N, Sala M, Kogevinas M, Sunyer J. 2001. Polychlorinated
biphenyls (PCBs) and neurological development in children: a systematic
review. J Epidemiol Community Health 55:537–46.
Rice DC, Schoeny R, Mahaffey K. 2003. Methods and rationale for
derivation of a reference dose for methylmercury by the U.S. EPA. Risk
Anal 23(1):107–15.
Ross G. 2004. The public health implications of polychlorinated biphenyls
(PCBs) in the environment. Ecotoxicol Environ Safety 59(3):275–91.
SACN/COT. 2004. Scientific Advisory Committee on Nutrition (SACN)
and Committee on Toxicity (COT), advice on fish consumption: benefits
and risks. London: The Stationary Office.
Salonen JT, Seppanen K, Nyyssonen K, Korpela H, Kauhanen J, Kantola M,
Tuomilehto J, Esterbauer H, Tatzber F, Salonen R. 1995. Intake of mercury
from fish, lipid peroxidation, and the risk of myocardial infarction and
coronary, cardiovascular, and any death in eastern Finnish men. Circulation
91:645–55.
SanGiovanni JP, Chew EY, Clemons TE, Davis MD, Ferris FLr, Gensler
GR, Kurinij N, Lindblad AS, Milton RC, Seddon JM, Sperduto RD. 2007.
The relationship of dietary lipid intake and age-related macular degeneration
in a case-control study: AREDS Report No. 20. Arch Ophthalmol
125(5):671–9.
Santerre CR. 2010. The risks and benefits of farmed fish. J World
Aquaculture Soc 41(2):250–7.
Sassi F. 2006. Calculating QALYs, comparing QALY and DALY calculations.
Health Policy Plann 21(5):402–8.
SCF. 2001a. Opinion of the Scientific Committee on Food on the Risk
Assessment of Dioxins and Dioxin-like PCBs in Food. Update based on
new scientific information available since the adoption of the SCF opinion
of 22nd November 2000. Available from: http://ec.europa.eu/food/fs/sc/
scf/out90_en.pdf. Accessed July 12, 2012.
SCF. 2001b. Scientific committee on foods. press releases: fact
sheet on dioxin in feed and food. MEMO/01/270. Available from: http://
europa.eu/rapid/pressReleasesAction.do?reference=MEMO/01/270&
format=HTML&aged=0&language=EN&guiLanguage=en. Accessed July
12, 2012.
Schachter H, Kourad K, Merali Z, Lumb A, Tran K, Miguelez M. 2005.
Effects of omega-3 fatty acids on mental health. Evidence
Report/Technology Assessment No. 116. Rockville, MD: AHRQ
(Prepared by the University of Ottawa Evidence Based Practice Center,
Ottawa, Canada).
Schachter H, Reisman J, Tran K, Dales B, Kourad K, Barnes D, Sampson M,
Morrison A, Gaboury I, Blackman J. 2004. Health effects of omega-3 fatty
acids on asthma. Technology Assessment No. 91. Rockville, MD: AHRQ
(Prepared by the University of Ottawa Evidence Based Practice Center,
Ottawa, Canada).
Schantz SL, Widholm JJ, Rice DC. 2003. Effects of PCB exposure on
neuropsychological function in children. Environ. Health Perspect
111:357–576.
Schecter A, Cramer P, Boggess K, Stanley J, Päpke O, Olson J, Silver A,
Schmitz M. 2001. Intake of dioxins and related compounds from food in the
U.S. population. J Toxicol Environ Health A 63(1):1–18.
Schwarz N, Sanna LJ, Skurnik I, Yoon C. 2007. Metacognitive experiences
and the intricacies of setting people straight: Implications for debiasing and
public information campaigns. Advances in experimental social psychology,
Vol. 39. San Diego: Elsevier Academic Press Inc. p 127–61.
Seppänen K, Kantola M, Laatikainen R, Nyyssönen K, Valkonen V-P,
Kaarlöpp V, Salonen JT. 2000. Effect of supplementation with organic
selenium on mercury status as measured by mercury in pubic hair. J Trace
Elem Med Biol 14:84–7.
Shimshack JP, Ward MB. 2010. Mercury advisories and household health
trade-offs. J Health Econ 29(5):674–85.
Shimshack JP, Ward MB, Beatty TKM. 2007. Mercury advisories:
information, education, and fish consumption. J Environ Econ Manage
53(2):158–79.
Siddiqi MA, Laessig RH, Reed KD. 2003. Polybrominated diphenyl ethers
(PBDEs): new pollutants-old diseases. Clin Med Res 1:281–90.
Simopoulos AP. 1991. Omega-3 fatty acids in health and disease and in
growth and development. Am J Clin Nutr 54:438–63.
Sioen I, Bilau M, Verdonck F, Verbeke W, Willems JL, de Henauw S, van
Camp J. 2008. Probabilistic intake assessment of polybrominated diphenyl
ethers and omega-3 fatty acids through fish consumption. Mol Nutr Food
Res 52(2):250–7.
Sioen I, de Henauw S, van Camp J, Volatier J-L, Leblanc J-C. 2009.
Comparison of the nutritional–toxicological conflict related to seafood
consumption in different regions worldwide. Reg Toxicol Pharmacol
55:219–28.
Sioen I, de Henauw S, Verdonck F, van Thuyne N, van Camp J. 2007a.
Development of a nutrient database and distributions for use in a
probabilistic risk-benefit analysis of human seafood consumption. J Food
Comp Anal 20(8):662–70.
Sioen I, van Camp J, Verdonck FAM, van Thuyne N, Willems JL, de
Henauw SWJ. 2007b. How to use secondary data on seafood contamination
for probabilistic exposure assessment purposes? Main problems and potential
solutions. Human Ecol Risk Assess 13(3):632–57.
Sontrop J, Campbell MK. 2006. Omega-3 polyunsaturated fatty acids and
depression: a review of the evidence and a methodological critique. Prev
Med 42(1):4–13.
Spurgeon A. 2006. Prenatal methylmercury exposure and developmental
outcomes: review of the evidence and discussion of future directions.
Environ Health Perspect 114(2):307–12.
Stern AH. 2007. Public health guidance on cardiovascular benefits and risks
related to fish consumption. Environ Health Available from:
http://www.ehjournal.net/content/pdf/1476-069X-6-31.pdf. Accessed
July 24, 2012.
Stern AH. 2005. A review of the studies of the cardiovascular health effects of
methylmercury with consideration of their suitability for risk assessment.
Environ Res 98(1):133–42.
Stewart P, Reihman J, Lonky E, Darvill T, Pagano J. 2000. Prenatal PCB
exposure and neonatal behavioral assessment scale (NBAS) performance.
Neurotoxicolo Teratol 22(1):21–9.
Stewart PW, Lonky E, Reihman J, Pagano J, Gump BB, Darvill T. 2008.
The relationship between prenatal PCB exposure and intelligence (IQ) in
9-year-old children. Environ Health Perspect 116(10):1416–22.
Stewart PW, Reihman J, Lonky EI, Darvill TJ, Pagano J. 2003. Cognitive
development in preschool children prenatally exposed to PCBs and MeHg. J
Neurotoxicol Teratol 25:11–22.
Storelli MM. 2008. Potential human health risks from metals (Hg, Cd, and
Pb) and polychlorinated biphenyls (PCBs) via seafood consumption:
estimation of target hazard quotients (THQs) and toxic equivalents (TEQs).
Food Chem Toxicol 46(8):2782–88.
Storelli MM, Giacominelli-Stuffler R, Storelli A, Marcotrigiano GO. 2003.
Polychlorinated biphenyls in seafood: contamination levels and human
dietary exposure. Food Chem 82(3):491–6.
USDA. 1993. U.S. Department of Agriculture, Agricultural Research
Service. The continuing survey of food intakes by individuals (CCFII) and
the diet and health knowledge survey (DHKS) 1989–91. Available from:
http://www.ars.usda.gov/Services/docs.htm?docid=14392. Accessed July
12, 2012.
510 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
USDA. 1998. 1989–91 Continuing Survey of Food Intake by Individuals
(CSFII) and 1989–91 Diet and Health Knowledge Survey. Hyattsville, MD:
Agricultural Research Service (CD-ROM; PB96–501747).
van der Voet H, de Mul A, van Klaveren JD. 2007. A probabilistic model for
simultaneous exposure to multiple compounds from food and its use for
risk-benefit assessment. Food Chem Toxicol 45(8):1496–506.
van Gelder BM, Tijhuis M, Kalmijn S, Kromhout D. 2007. Fish
consumption, n-3 fatty acids, and subsequent 5-y cognitive decline
in elderly men: the Zutphen Elderly Study. Am J Clin Nutr 85(4):
1142–47.
Verger P, Houdart S, Marette S, Roosen J, Blanchemanche S. 2007. Impact
of a risk-benefit advisory on fish consumption and dietary exposure to
methylmercury in France. Reg Toxicol Pharmacol 48(3):259–69.
Verger P, Khalfi N, Roy C, Blanchemanche S, Marette S, Roosen J. 2008.
Balancing the risk of dioxins and polychlorinated biphenyls (PCBs) and the
benefit of long-chain polyunsaturated fatty acids of the n-3 variety for
French fish consumers in western coastal areas. Food Addit Contam: Part A.
Chem Anal Control Expo Risk Assess 25(6):765–71.
Virtanen JK, Voutilainen S, Rissanen TH, Mursu J, Tuomainen TP,
Korhonen MJ, Valkonen VP, Seppanen K, Laukkanen JA, Salonen JT.
2005. Mercury, fish oils, and risk of acute coronary events and cardiovascular
disease, coronary heart disease, and all-cause mortality in men in eastern
Finland. Arterioscler Thromb Vasc Biol 25:228–233.
Wang C, Chung M, Balk EM, Kupelnick B, DeVine D, Lawrence A,
Lichtenstein A, Lau J. 2004. Effects of omega-3 fatty acids on cardiovascular
disease. Evidence Report/Technology Assessment No. 94. Rockville, MD:
AHRQ (prepared by the Tufts-New England Medical Center
Evidence-based Practice Center, Boston, MA under contract no.
290–02-0022).
Wang C, Harris WS, Chung M, Lichtenstein AH, Balk EM, Kupelnick B,
Jordan HS, Lau J. 2006. n-3 Fatty acids from fish or fish-oil supplements,
but not alpha-linolenic acid, benefit cardiovascular disease outcomes in
primary- and secondary-prevention studies: a systematic review. Am J Clin
Nutr 84(1):5–17.
c 2012 Institute of Food Technologists®
Watanabe C, Satoh H. 1996. Evolution of our understanding of
methylmercury as a health threat. Environ Health Perspect 104(Suppl
2):367–79.
Whelton SP, He J, Whelton PK, Muntner P. 2004. Meta-analysis of
observational studies on fish intake and coronary heart disease. Am J Cardiol
93(9):1119–23.
WHO. 1990. Effects on man. Environmental health criteria 101:
methylmercury. Geneva: World Health Organization. p 68–99.
WHO. 2002. Reducing risks, promoting healthy life. The World Health
Report 2002. Available from: http://www.who.int/whr/2002/en/.
Accessed July 12, 2012.
WHO. 2007. World Health Organization. Global Environment Monitoring
System – Food Contamination Monitoring and Assessment Programme
(GEMS/Food). Available from: http://www.who.int/foodsafety/chem/
gems/en/index.html. Accessed July 12, 2012.
WHO. 2010. Metrics: disability-adjusted life year (DALY). Available from:
http://www.who.int/healthinfo/global_burden_disease/metrics_daly/en/
index.html. Accessed July 12, 2012.
Wieslaw J, Jeffery J, Elzbieta F, Anita S, Elzbieta M, Elzbieta ST, Iwona LM,
Agnieszka SW, Virginia R, Zbigniew S, Irena K, Frederica P. 2006. Effects
of prenatal exposure to mercury on cognitive and psychomotor function in
one-year-old infants: epidemiologic cohort study in Poland. Ann Epidemiol
16(6):439–47.
Willatts P, Forsyth JS, DiModugno MK, Varma S, Colvin M. 1998. Effect of
long-chain polyunsaturated fatty acids in infant formula on problem solving
at 10 months of age. Lancet 352:688–91.
Williams CM, Burdge G. 2006. Long-chain n-3 PUFA: plant v. marine
sources. Proc Nutr Soc 65:42–50.
Yasumoto T, Murata M. 1993. Marine toxins. Chem Rev 93(5):1897–909.
Yoshizawa K, Rimm EB, Morris JS, Spate VL, Hsieh CC, Spiegelman D,
Stampfer MJ, Willett WC. 2002. Mercury and the risk of coronary heart
disease in men. N Engl J Med 347:1755–60.
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 511
512 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
Women in North America
aged 20 to 39 y with
an average body
weight of 60 kg
Farmed Atlantic salmon
and rainbow trout
82 types of fishes,
mollusks, crustaceans,
and seafood-based
dishes
MeHg, PCBs, and
dioxins
Contaminants (6
trace elements
and 3 categories
of POPs)
Reproductive and
developmental
risks
Probability of
exceeding PTWI
for each
contaminant
Cancer and
noncancer risks
from lifetime
exposure
French consumers with
high fish consumption
(n = 1000)
Organic
contaminants
(PCBs, dieldrin,
and so on)
Farmed and wild salmon
Risk
endpoints
Not specified
Risk
factors
Unspecified types of fish Six organic pollutants Risk of cancer
containing six
cancer-causing
compounds at
maximum allowable
levels according to FDA
Seafood
type
American adults and
subpopulation of
subsistence fishers
Target
population
Table A1–Risk-benefit assessments for seafood consumption using common metrics.
Appendix A: Summary of Risk-Benefit Assessment Studies
EPA + DHA
Omega-3 fatty acids
EPA + DHA
Fish consumption
Beneficial
factors
Intake of 500 mg EPA +
DHA/d
Probability of obtaining
RDA of 500 mg/d
Probability of reducing
sudden death from
CHD
Reduced risk of death
from CHD
Benefit
endpoints
Method of analysis
Results/conclusions
Risk tradeoff
CHD-related benefits of eating
analysis—probabilities
fish far outweigh cancer risks for
of each health
American adults and for
endpoint were
subsistence fishers.
calculated and
Consumption of 20 g fish per
compared
day was estimated to reduce
CHD risk by 35%, while
increasing cancer risk by 1% (1
× 10−2 ) if fish are
contaminated at maximum
levels, and a more realistic
increased cancer risk of
0.0075% (7.5 × 10−5 ) based
on actual contaminant levels in
fish.
U.S. EPA risk-based
Consumption of 1 g/d of
assessments for
EPA+DHA from salmon would
organic contaminants
save about 300 times the
compared to results of
number of lives that would be
clinical trials with n-3s
lost to cancer from exposure to
organic compounds. Individuals
can maximize benefit while
reducing
contaminant-associated risk by
choosing salmon with lower
contaminant concentrations.
Combined data obtained 84% of study participants were
through food
meeting the RDA for omega-3
frequency
fatty acids and 34% of
questionnaires with
participants were exceeding the
levels of contaminants
PTWI for MeHg. Participants
in various fish types to
had a 39% probability of
determine probability
exceeding the PTWI for dioxins
of each population
and dl PCBs and a 74%
group to meet the RDA
probability of exceeding the
for omega-3 fatty acids
PTWI for iPCBs. Blood levels of
and to exceed the PTWI
MeHg were well below the no
for contaminants.
adverse effect level, even in
individuals consuming 4.5 kg
seafood/wk. The authors
recommended that the general
population should consume at
least 2 servings of fish,
especially oily fish, per week
and pregnant and nursing
women should only consume
predatory fish once per week.
The EPA + DHA intake
Based on the results, two 180-g
with two, 180-g
meals per week of farmed
servings of Atlantic
salmon or trout available in
salmon per week
North American markets would
calculated to be 440
be expected to provide
mg/d. MeHg, PCBs
sufficient levels of EPA + DHA
and dioxins was
for the target population
determined and
without concern over exceeding
compared to the PTWIs
the PTWIs for MeHg or PCBs
for these
and dioxins.
contaminants.
Reference
(Continued)
Dewailly and others
(2007)
Leblanc and others
(2006)
Foran and others
(2005b)
Anderson and
Wiener (1995)
Risk-benefit analysis of seafood consumption . . .
c 2012 Institute of Food Technologists®
c 2012 Institute of Food Technologists®
10 types of fish
7 categories of fish and
seafood
Belgian adult fish
consumers (n = 821)
13 regional clusters of
the global adult
population
38 categories of fish and
seafood
Adult consumers (n =
401) in a coastal
region of France with
high fish consumption
EPA + DHA
EPA + DHA
Beneficial
factors
Exposure levels for
different dietary
scenarios
EPA + DHA, iodine, and
vitamin D
EPA + DHA
Exposure as related LC n-3 PUFAs
to PTWI for dioxins
and dioxin-like
PCBs of 14 pg
TEQ/kg body
wt/wk
Exposure as related
to PTWI, TDI, and
cancer and
noncancer risks
from lifetime
exposure
Probability that
dioxin exposure is
below TDI of 2 pg
WHO-TEQ/kg
bw/day
Risk
endpoints
MeHg, iPCBs, dioxin-like Exposure related to
PCBs, PCDDs, and total
PTWI
TEQ
PBDEs
Dioxins and PCBs
MeHg, Pb, Cd, and 7
categories of organic
pollutants
14 most consumed fish
species in Spain
70-kg adult consumer in
Spain
Risk
factors
11 types of seafood and 7 Dioxins (PCDDs, PCDFs,
other foods
and DL-PCBs)
Seafood
type
Dutch consumers (n =
6250)
Target
population
Table A1–(Continued)
Method of analysis
Intake levels for different Estimated exposure and
regional clusters as
intake levels worldwide
compared to dietary
and compared to
guidance values (500
dietary guidance
mg/d EPA + DHA; 5
values
µg/d vitamin D; 150
mg/d iodine)
Intake levels for different Calculated probable
dietary scenarios as
dietary exposure and
compared to reference
intake levels based on
intake of 9.7 mg/kg
food frequency
bw/d (681 mg/d)
questionnaire and
compared fish
consumption scenarios
Calculated daily intakes
of risk and benefit
factors based on
dietary data for top 14
fish species and
compared to dietary
guidance values
Intake of LC n-3 PUFAs as Calculated exposure and
compared to
intake levels based on
recommended level of
1-mo dietary notebook
500 mg/d
records and compared
to dietary guidance
values
Daily intake of EPA +
DHA
Probability that
Calculated exposure
EPA+DHA intake is
levels based on food
above AI: 7 mg
diaries and levels of
EPA+DHA/day/kg bw
compounds in each
(based on AI of 450
food type and
mg/day and average
compared to dietary
body wt of 65 kg)
guidance values
Benefit
endpoints
Results/conclusions
For most consumers, exposure to
dioxins and EPA+DHA was
below the TDI and AI,
respectively. A scenario where
10–25% of beef and pork was
replaced with fatty fish allowed
for the AI of EPA+DHA while
remaining below the TDI.
Higher percentages of
substitution (50–100%) led to
exceeding the TDI.
Estimated daily intakes for 70-kg
adult consumer provided 244
mg EPA + DHA and were below
risk levels for all categories,
except that PAHs exposure
resulted in an increased cancer
risk of 2.4 × 10−5
41% of the subjects achieved the
recommended level of LC n-3
PUFAs and remained below the
tolerance threshold for dioxins;
19% of subjects met the
nutritional recommendation
but exceeded the tolerance
threshold; 38% of subjects were
below both the tolerance
threshold and the nutritional
recommendations
Current consumption levels
showed mean PBDE and EPA +
DHA intakes of 0.85 ng/kg
bw/d and 3.5 mg/kg bw/d,
respectively. Consumption of
two 150-g portions of salmon
per week allowed for 11.9
mg/kg bw/d EPA + DHA
without major increases in
PBDE exposure (1.3 ng/kg
bw/d).
Most regional clusters were below
the PTWIs for contaminants,
but did not achieve the
recommended dietary guidance
values. The countries with the
highest seafood intake
exceeded the PTWIs for MeHg
or dioxin-like compounds. The
mean intake values for these
countries did not exceed the
reference values. proposed by
SACN/COT.
Reference
(Continued)
Sioen and others
(2009)
Sioen and others
(2008)
Verger and others
(2008)
Domingo and others
(2007)
van der Voet and
others (2007)
Risk-benefit analysis of seafood consumption . . .
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 513
514 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
48 types of fish and
seafood items
Top 5 consumed species
for each country
French pregnant women
(n = 161)
European consumers
from 8 countries
MeHg
MeHg
16 types of fish
MeHg
commonly available in
Connecticut markets
Adult population
(cardiovascular risk
group) and
subpopulation of
pregnant women
(neurodevelopmental
risk group)
MeHg
Different fish species
consumed in the U.S.
Seafood
type
U.S. consumers
Target
population
Table A1–(Continued)
Risk
factors
Seafood consumption
Beneficial
factors
Probability of
exceeding the
PTWI for MeHg
EPA + DHA
Exposure as related LC n-3 PUFAs
to PTWI for MeHg
Increase in risk of
EPA + DHA
adult MI; decrease
in infant VRM
score
Fetal neurodevelopment
Risk
endpoints
Method of analysis
Results/conclusions
Fetal neurodevelopment, Maternal info based on
Current fish consumption is
risk of fatal CHD, risk of
Iraq and Seychelles
contributing a net benefit to
fatal stroke
Islands and UK
neurodevelopment, and if
cognitive development
women of child-bearing age
study; CHD based on
increased fish consumption to
meta-analysis and
340 g/wk, neurodevelopment
pooled analysis
benefits would also increase
(0.225 IQ point); commercial
fish baseline consumption is
averting a central estimate of
over 30000 deaths per year
from coronary heart disease
and over 20000 deaths per
year from stroke.
Decrease in risk for adult Calculation of net
Species-specific consumption
CHD mortality;
changes in
recommendations varied
increase in infant VRM
cardiovascular risk and
depending on risk group. Both
score
infant VRM scores for
groups were recommended to
each fish species based
avoid shark and swordfish; there
on dose-response
were 7 to 9 fish species that
relationships
could be eaten more than twice
per week and 5 to 7 fish that
should be limited to one or two
6-oz meals per week.
Intake of LC n-3 PUFAs as Calculated dietary
About 50% of the subjects
compared to
exposure and intake
achieved the recommended
recommended level of
levels based on
nutritional intake and about
500 mg/d
responses to food
5% of subjects exceeded the
frequency
PTWI for MeHg
questionnaires and
compared to dietary
guidance values
Probability of exceeding Log-normal distribution Probability of exceeding 500
a daily intake of 500
for seafood
mg/d of EPA + DHA in each
mg/d of EPA + DHA
consumption patterns
country was 0.32% to 66.05%;
and plug-in (PI) or
probability of exceeding the
tail-estimation
PTWI for MeHg was 0.04% to
(TE)-based estimates
9.61%.
to calculate
probabilities of
exceeding dietary
guidance values
Benefit
endpoints
Reference
Cardoso and others
(2010)
Pouzaud and others
(2009)
Ginsberg and Toal
(2009)
FDA (2009a)
Risk-benefit analysis of seafood consumption . . .
c 2012 Institute of Food Technologists®
c 2012 Institute of Food Technologists®
Most consumed types of
fish and seafood
among target
populations
All seafood purchased
from 2000 to 2002
French adults consuming
high amounts of fish
and seafood (average
11 servings/wk)
U.S. households with
pregnant women or
young children
Mercury
MeHg
42 types of fish consumed MeHg exposure through
in the U.S.
fish consumption
Total U.S. adult
population and
subpopulation of
women of childbearing
age
Hazards
Unspecified types of fish MeHg
with average MeHg
levels of 0.5 to 2.0 ppm
Seafood
type
Total population and
subpopulation of
women of childbearing
age and their children
Target
population
Beneficial
factors
EPA + DHA
Fish consumption;
prenatal DHA intake
through fish
consumption
Impact on IQ points/child EPA + DHA, total fish
and corresponding
consumption
monetary value
Impact on IQ points
Change in IQ points
Increase in
Fish consumption
neurodevelopmental
delay (that is, talking
after 24 mo of age)
from prenatal exposure
Risk
endpoints
Table A2–Risk-benefit assessments for seafood consumption using composite metrics.
Method of analysis
QALY-adjusted dose
response modeling
examining 5 different
scenarios of fish
consumption
QALY-adjusted
dose-response
modeling examining
consumption of 0 to
300 g fish/d with
MeHg levels of 0.5,
1.0, and 2.0 ppm
Impact on IQ
Calculation of health
points/child, reduction
impacts based on
of CHD-related deaths
seafood purchased in
and corresponding
14800 households
monetary values
CHD mortality and stroke QALY-adjusted
mortality and
dose-response
morbility; impact on IQ
modeling examining
points
effect of increasing
EPA + DHA intake
from 391 to 2700
mg/d through fish
consumption
Changes in the relative
risks of CHD mortality
and stroke; change in
IQ points
Decrease in myocardial
infarction mortality
Benefit
endpoints
Results/conclusions
Total population showed
net benefits for fish
consumption (gain of
approximately 5000
QALYs for 2 servings∗
fish per week).
Subpopulation of
women of childbearing
age showed negative
effects of consumption
of fish with 0.5 to 2.0
ppm MeHg (loss of
approximately 250 to
2000 QALYs for 2
servings per week;
benefits of DHA on
neurodevelopment
were not considered).
Total population showed
a net negative impact
of reducing fish
consumption by 17%
(−41000 QALYs) and
a net benefit of
increasing fish
consumption by 50%
(+90000 QALYs).
Subpopulation of
women of childbearing
age showed
substantial benefits of
replacing fish high in
MeHg with fish low in
MeHg (+49000
QALYs/ +0.1 IQ
points).
Beneficial effects of
increasing EPA + DHA
intake through fish
consumption, with
QALYs totaling 97248
to 285774, depending
on the dose-response
model. Confidence
interval indicates
possible negative
effects from MeHg for
some individuals.
FDA’s 2011 mercury in
fish advisory resulted in
reduced fish
consumption and an
overall negative
impact on health,
valued at negative $30
USD 2007
Reference
Shimshack and Ward
(2010)
Guevel and others
(2008)
Cohen and others
(2005a)
Ponce and others
(2000)
Risk-benefit analysis of seafood consumption . . .
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 515
516 Comprehensive Reviews in Food Science and Food Safety r Vol. 11, 2012
All seafood
Adult population
and special
groups
All seafood
Total fish consumption
Adult population
All Americans
General
Seafood
type
All
Target
population
Hazards
POPs, MeHg
MeHg, dioxins, dl PCBs,
furans
MeHg, PCBs, dioxins
MeHg, other metals,
POPs, microbs, seafood
allergens and naturally
occurring toxins
Table A3–Qualitative risk-benefit assessments.
Neurodevelopment,
cardiovascular disease
and cancer
Neurologic, carcinogenic,
and other health risks
MeHg was the greatest
concern, POPs remain
uncertain
Risk
endpoints
EPA + DHA
Fish consumption; EPA +
DHA
Fish consumption and
EPA + DHA intake
EPA + DHA
Beneficial
factors
EPA + DHA and risk of
CHD; EPA + DHA and
health of infants
Neurodevelopment and
prevention of
cardiovascular disease
Neurologic
developmental
benefits, reduction in
CHD, and sudden
death
Benefits to women during
pregnancy, duration of
gestation and birth
weight, infant and
child development,
cardiovascular disease,
cardiovascular
mortality, and all-cause
morbidity and
mortality.
Benefit
endpoints
Method of analysis
Evaluation of experts in
the fields and
systematic reviews in
the areas
Evaluation of specific
nutrients and
contaminants in
seafood as well as
scientific literature
regarding the risks and
benefits of seafood
consumption
Evaluation of studies and
meta-analyses
evaluating human
health effects related
to fish intake,
contaminants, and
beneficial compounds.
4-part qualitative
protocol involving:
identification of the
magni-tude of R/B;
identify risk; evaluate
different consumption
patterns; balancing to
arrive at specific
guidelines
Results/conclusions
Benefits of seafood
consumption
outweighed the risks in
healthy adults and may
reduce their risk for
cardiovascular disease
by consuming seafood
regularly Adults as risk
for cardiovascular
disease may benefit
from seafood high in
EPA + DHA
pregnant/nursing
women and children
may benefit from
consuming seafood (up
to 12 oz per week)
Among adults, the
benefits of fish intake
exceed the potential
risks. For women of
childbearing age, the
benefits also outweigh
risks, with the
exception of a few
selected species.
Moderate fish
consumption reduces
coronary death by
36%.
Overall: good source of
nutrients, lowers CHD.
No evidence of CHD
from MeHg or cancer
from dl PCBs were
below CHD benefits.
Omega-3 intake lowers
risk of suboptimal
neurodevelopment.
Maternal dioxin/dl
PCB intake is below
PTMI
neurodevelopmental
risk is negligible.
Recommendations:
benefit of seafood on
CHD mortality and
improving
neurodevelopment
Seafood intake reduces
risk of CHD mortality or
sudden death;
omega-3s improved
infant health visual
acuity and cognitive
development. Overall,
health benefits from 2
servings per week
outweigh the risks from
MeHg and POPs
Reference
DGAC (2010)
JECFA FAO/ WHO
(2010)
Mozaffarian and
Rimm (2006)
IOM 2005 IOM
(2005b)
Risk-benefit analysis of seafood consumption . . .
c 2012 Institute of Food Technologists®
Risk-benefit analysis of seafood consumption . . .
Appendix B: Abbreviations/Acronyms.
AHA: American Heart Association
AI: adequate intake (The Netherlands)
AHRQ: Agency for Health Research and Quality
AL: action level
ALA: alpha-linolenic acid
ASTDR: Agency for Toxic Substances of Disease Registry
BMD: benchmark dose
BMDL: benchmark dose lower bound
Cd: cadmium
CHD: coronary heart disease
CPF: cancer potency factor
CSF: cancer slope factor
CVD: cardiovascular disease
CFSII: USDA Continuing Survey of Food Intake by Individuals
CO: chronic oral
DALY: disability-adjusted life years
DDT: dichlorodiphenyltrichloroethane
DGAC: Dietary Guidelines Advisory Committee
DHA: docosahexaenoic acid
dl PCB: dioxin-like polychlorinated biphenyls
DRI: dietary reference intake
EC: European Commission
EFSA: European Food Safety Authority
EPA: eicosapentaenoic acid
EPA: U.S. Environmental Protection Agency
EU: European Union
EUML: European Union maximum level
EVT: extreme value theory
FAO: Food & Agriculture Organization of the United Nations
FDA: U.S. Food and Drug Administration
GL: guidance level
HC: Health Canada
HCB: hexachlorobenzene
HCN: Health Council of The Netherlands
HHS: U.S. Department of Health and Human Services
IOM: Institute of Medicine
INRA: Institute for Agronomy Research (France)
ISSFAL: International Society for the Study of Fatty Acids and Lipids
JECFA: Joint FAO/WHO Expert Committee on Food Additives
c 2012 Institute of Food Technologists®
MeHg: methylmercury
MI: myocardial infarction
ML: maximum level
MRL: minimal risk level
mt: metric ton
NHANES: National Health and Nutrition Survey
NMFS: National Marine Fisheries Service
NRC: National Research Council
NOAA: National Oceanic and Atmospheric Administration
NOEL: no observed effect level
PAH: polycyclic aromatic hydrocarbon
PBDE: polybrominated diphenyl ether
PCB: polychlorinated biphenyl
PCDD: polychlorinated dibenzo dioxin
PCDE: polychlorinated diphenyl ether
PCDF: polychlorinated dibenzo furan
PCN: polychlorinated naphthalene
PMTDI: provisional maximum tolerable daily intake
POP: persistent organic pollutant
PTDI: provisional tolerable daily intake
PTMI: provisional tolerable monthly intake
PTWI: provisional tolerable weekly intake
PUFA: polyunsaturated fatty acid
QALY: quality-adjusted life years
RCT: randomized controlled trail
RDA: recommended daily allowance
RDI: reference daily intake
RfD: reference dose
SACN/COT: Scientific Advisory Committee on Nutrition/Committee on Toxicity (UK)
SCF: Scientific Committee on Food (France)
TCDD: 2,3,7,8-tetrachlorodibenzo-p-dioxin
TDI: tolerable daily intake
TEF: toxic equivalent factor
TEQ: total dioxin-like equivalency
TL: tolerance level
USDA: U.S. Department of Agriculture
VRM: visual recognition memory
WAPM: World Association of Perinatal Medicine
WHO: World Health Organization
Vol. 11, 2012 r Comprehensive Reviews in Food Science and Food Safety 517
Download