+ All Categories
Transcript
Page 1: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

http://informahealthcare.com/txcISSN: 1040-8444 (print), 1547-6898 (electronic)

Crit Rev Toxicol, Early Online: 1–26! 2014 Informa Healthcare USA, Inc. DOI: 10.3109/10408444.2013.861798

REVIEW ARTICLE

Food additive carrageenan: Part II: A critical review of carrageenanin vivo safety studies

Myra L. Weiner

Toxpertise, LLC, Princeton, NJ, USA

Abstract

Carrageenan (CGN) is a seaweed-derived high molecular weight (Mw) hydrocolloid, primarilyused as a stabilizer and thickener in food. The safety of CGN regarding its use in food isreviewed. Based on experimental studies in animals, ingested CGN is excreted quantitatively inthe feces. Studies have shown that CGN is not significantly degraded by low gastric pH ormicroflora in the gastrointestinal (GI) tract. Due to its Mw, structure and its stability whenbound to protein, CGN is not significantly absorbed or metabolized. CGN also does notsignificantly affect the absorption of nutrients. Subchronic and chronic feeding studies inrodents indicate that CGN at doses up to 5% in the diet does not induce any toxicologicaleffects other than soft stools or diarrhea, which are a common effect for non-digestible highmolecular weight compounds. Review of several studies from numerous species indicates thatfood grade CGN does not produce intestinal ulceration at doses up to 5% in the diet. Effects ofCGN on the immune system following parenteral administration are well known, but notrelevant to food additive uses. The majority of the studies evaluating the immunotoxicitypotential were conducted with CGN administered in drinking water or by oral gavage whereCGN exists in a random, open structured molecular conformation, particularly the lambda form;hence, it has more exposure to the intestinal mucosa than when bound to protein in food.Based on the many animal subchronic and chronic toxicity studies, CGN has not been found toaffect the immune system, as judged by lack of effects on organ histopathology, clinicalchemistry, hematology, normal health, and the lack of target organ toxicities. In these studies,animals consumed CGN at orders of magnitude above levels of CGN in the human diet:�1000 mg/kg/d in animals compared to 18–40 mg/kg/d estimated in the human diet. DietaryCGN has been shown to lack carcinogenic, tumor promoter, genotoxic, developmental, andreproductive effects in animal studies. CGN in infant formula has been shown to be safe ininfant baboons and in an epidemiology study on human infants at current use levels.

Keywords

Carrageenan, chronic toxicity, degradation,food additive, gastrointestinal tractevaluations, infant formula additive

History

Received 6 May 2013Revised 26 October 2013Accepted 30 October 2013Published online 27 January 2014

Table of Contents

Abstract ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 1Introduction ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 2

Importance of vehicle... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 3Acute toxicity studies ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 5

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 5Acute oral, dermal, and inhalation toxicity ... ... ... ... ... ... ... ... ... 5Eye irritation potential, skin irritation, and sensitization

potential ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 5Degradation, excretion, absorption, and metabolism ... ... ... ... ... ... ... 5

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 5Degradation and excretion ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 5Absorption ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 6

Rat ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 6Guinea pig ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 7Primate ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 7

Special studies on nutrient absorption ... ... ... ... ... ... ... ... ... ... ... 7Metabolism ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 7

Subchronic and long-term oral toxicity studies... ... ... ... ... ... ... ... ... ... 8

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 8Subchronic oral toxicity ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 8

Primates ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 8Rats ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 8Mice ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 9

Long-term oral toxicity ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 9Immune system effects ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 9

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 9Ulcerative effects ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 11

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 11Mice ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 14Rats ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 15Guinea pigs ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 15Hamsters ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 15Pigs ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 15Primates ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 15

Carcingenicity studies ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16

Genetic effects ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16

Tumor promotion ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 16

Reproductive and developmental toxicity studies ... ... ... ... ... ... ... 17Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 17

Address for correspondence: Dr. Myra L. Weiner, Owner and Principal,TOXpertise, LLC, 100 Jackson Avenue, Princeton, NJ 08540, USA.E-mail: [email protected]

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 2: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Studies on special age groups ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 18Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 18Young animals and humans ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 18

Primates ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 18Humans ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 18

Epidemiology studies ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19Regulation of dietary uses ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19

Summary ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19Adults ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19

Regulatory background ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19Dietary exposure ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19

Infants ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19Regulatory background ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 19Dietary exposure ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 20

Conclusions ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 21Acknowledgements ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 21Declaration of interest ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 21References ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 21Appendix 1... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 24Appendix 2... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... ... 25

Introduction

Carrageenan (CGN) has been widely used as a food additive

for decades and its safety is based on a large database of

studies. CGN is used in food processing primarily to bind

water and promote gel formation, to thicken and stabilize

structure for food products by binding protein, and to improve

palatability. CGN is used in a variety of foods, including dairy

products, water-dessert gels, meat and fish products, bever-

ages, condiments, infant formula, and pet food. In addition to

conventional foods, CGN is used in foods labeled as Kosher,

Halal, and processed foods labeled as ‘‘organic’’. CGN is

listed in the US FDA Inactive Ingredient Guide as an

excipient that is presently used in oral drug products

(capsules, syrups, and coatings), lotions, and dental pastes.

CGN is also used as an ingredient in personal care products.

Chemically, CGN is a high molecular weight sulfated

polygalactan derived from a number of species of red seaweeds

of the class Rhodophyceae. It is a polymer of the sugar,

galactose, composed of repeating galactose units that may have

sulfate groups attached. The prevalent copolymers in CGN are

designated as kappa- (k), iota- (E), and lambda (�). k-CGN is

mostly the alternating polymer of D-galactose-4-sulfate and

3,6-anhydro-D-galactose. E-CGN is similar, except that the 3,6-

anydrogalactose is sulfated at carbon 2. Between k-CGN and

E-CGN, there is a continuum of intermediate compositions

differing in degree of sulfation at carbon 2. In �-CGN, the

alternating monomeric units are mostly D-galactose-2-sulfate

(a-1,3-linked) and D-galactose-2,6-disulfate (b-1,4-linked).

The anionic ester sulfate groups on CGN bind to positively

charged groups on food proteins, imparting functionality as a

thickener and stabilizer in foods (Blakemore & Harpell, 2010).

JECFA (1999) considers the various forms of CGN (k, �, E)

derived from various species of seaweed to show no major

differences in terms of toxicology and safety in foods. Food

grade CGN is included in foods and infant formula in which

CGN is tightly bound to protein.

Detailed regulatory specifications outline the criteria for

identity, purity, functionality, composition, heavy metals, and

weight average molecular weight (Mw) of food grade CGN,

shown in Table 1 (Commission Regulation, 2012; Food and

Agriculture Organization of the United Nations, 2007; the

Food Chemicals Codex, 2013; Japan Food Additives

Association, 2009).

From a toxicological perspective, the Mw of CGN is

important for an understanding of toxicological effects. There

has been much confusion in the literature between CGN, the

high Mw polymer used as a food additive, and poligeenan,

also known as degraded CGN, a much lower Mw polymer

(10–20 kDa) generated by subjecting CGN to the extreme

conditions of acid hydrolysis at low pH (0.9–1.3) and high

temperatures (480 �C) for several hours (Bixler, 2013).

Poligeenan has been shown to produce ulceration of the

intestinal tract and gastrointestinal tumors in animals (IARC,

1983). Early literature studies used the term ‘‘degraded

CGN’’, now known as poligeenan, to describe the low Mw test

material used in toxicology studies and erroneously called this

low Mw material ‘‘CGN’’. These errors in chemical nomen-

clature have led to confusion over the toxicological properties

of CGN both in the literature and more widely on the Internet

via consumer and health advocacy groups (Bixler, 2013;

Carthew, 2002; Tobacman, 2001). Consumers are clearly

confused by the interchangeable terminology. To clarify the

two materials, ‘‘degraded CGN’’ and food additive CGN,

the US Adopted Names Council (USAN, 1988) assigned the

name ‘‘poligeenan’’ to the substance previously referred to as

‘‘degraded CGN’’. The USAN defines poligeenan as having

an average Mw 10–20 kDa. Unfortunately, the term ‘‘poligee-

nan’’ has not been substituted for the term ‘‘degraded CGN’’

on a consistent basis.

The conditions required to produce poligeenan do not exist

in vivo in the gastrointestinal (GI) tract; hence, poligeenan is

not generated from food additive CGN in humans or animals

by degradation at physiological pH and temperature or by gut

microflora. Poligeenan is not a food additive and has no

utility in food. The toxicological and purity profiles of CGN

and poligeenan differ significantly. See Appendix 1 for a

comparison of key aspects of CGN and poligeenan. In this

regard, it is important to properly identify and specify

the analytical profile and Mw of test material used in

toxicology studies.

CGN permitted for food use must meet a viscosity

specification, which has been shown to correlate well with

molecular weight. Viscosity is an easily determined property

of solutions; therefore, various regulatory agencies use

viscosity measurement as an indirect measure of molecular

weight. The viscosity test for CGN is carried out under

standard conditions using a 1.5% solution at 75 �C. Regulatory

specifications require a viscosity of not less than 5 cPs or

5 mPa s (cP or centipose is the same as mPa s or milliPascal

seconds, both units of viscosity) for all seaweed-derived food

grade CGN (Commission Regulation, 2012; Food and

Agriculture Organization of the United Nations, 2007; the

Food Chemicals Codex, 2013; Japan Food Additives

Association, 2009). This viscosity corresponds to a molecular

weight (Mw) of approximately 100–150 kiloDaltons (kDa). In

its review of CGN, the European Commission’s Scientific

Committee on Foods (SCF) acknowledged that there was no

evidence to indicate adverse effects from CGN exposure, but

2 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 3: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

the SCF concluded that ‘‘if feasible’’ a molecular weight limit

of not 45% below 50 kDa should be introduced into the

specification’’ to minimize ‘‘the presence of any degraded

carrageenan.’’ (European Commission, 2003). Even though a

validated analytical method did not exist, Commission

Directive (2004) amended the purity criteria for CGN to

include: ‘‘Low molecular weight carrageenan (molecular

weight fraction below 50 kDa) – not more than 5%’’.

Currently, a validated analytical method is still not available.

There is no upper Mw limit for CGN.

Commercial CGN has an average Mw 200–800 kDa and

often includes different proportions of E-, k-, and �-CGN,

depending on the desired functionality characteristics (Bixler,

2013; Blakemore & Harpell, 2010). Uno et al. (2001b)

surveyed the Mw distributions of 29 samples of food-grade

refined CGNs using high-performance liquid gel permeation

chromatography (GPC). The Mw of 29 food-grade CGN

samples ranged from 453 to 652 kDa with a mean of the

average molecular weights (Mw) for all 29 samples of

530 kDa. This analysis did not detect poligeenan (Mw

of 10–20 kDa) in the CGN samples at a detection limit

of 5% (weight/weight) (Uno et al., 2001b).

CGN was reviewed by the Joint FAO/WHO Expert

Committee on Food Additives in 1974, 1999, 2002, and

2008. It is currently assigned an Acceptable Daily Intake

(ADI) of ‘‘not specified’’ and permitted for use in foods

at the level of good manufacturing practices, determined

by functionality in a given product (JECFA, 2002, 2008).

See Section on ‘‘Regulation of dietary uses’’ for more

information.

The purpose of the present review is to clarify and

summarize the toxicological properties of food grade CGN

and to distinguish it from poligeenan. In this review, the term

‘‘poligeenan’’ will be used to identify the low Mw polymer,

previously denoted as degraded CGN, which was often not

distinguished in other reviews. This review discusses the

physical and functional properties of CGN and how they

relate to toxicity, and includes references not available in

previous reviews. It also discusses all aspects of toxicological

studies; thus, significantly adding to the Cohen & Ito (2002)

review, which covered only the rodent bioassay and tumor

promotion studies and the Michel & Macfarlane (1996)

review, which covered only the GI tract studies. This review

covers the published literature on CGN, searched by NERAC

on an on-going daily basis with search terms such as CGN,

carbohydrate polymer, hydrocolloid, molecular weight deter-

mination, molecular weight profiling, and low molecular

weight fraction for toxicology endpoints. A few unpublished

studies and reports are also included: Albany Medical College

(1975, 1983), Bailey & Morgareidge (1973); FDRL (1972a,

1972b), Friedman & Douglass (1960), Mankes (1977), and

Weiner (2007). The published studies often do not specify the

purity, molecular weight and/or compliance with food addi-

tive grade specifications of a test sample of CGN. For

example, see Watt & Marcus (1969), Onderdonk & Bartlett

(1979), or Bhattacharyya et al. (2012). It is critically

important to accurately describe the Mw and purity of the

test material used in toxicology studies. Appendix 2 shows the

identity of the food additive CGN samples used as test

materials in studies quoted in this review in order of section,

including the type of CGN. k- and E-CGNs exist as both

organized (presence of helical structures and associations)

and disorganized conformations (random coil) in the presence

of water, but �-CGN exists only in the disorganized

conformation (unable to form helices) in water. The inter-

pretation of toxicology studies on CGN (k, E, or �) should

consider the possible conformation under the given experi-

mental conditions. References in Appendix 2 are shown on

the first time only that they are cited in the text. In the

following sections, results of toxicology studies will include

the type of CGN and the vehicle used for clearer, more

accurate interpretation of results.

Importance of vehicle

The vehicle employed to administer CGN is critical to an

understanding of the toxicological effects and an interpret-

ation of their meaning because the conformation of the CGN

molecule changes depending on the vehicle. Most animal

studies administered CGN in diet or in drinking water with

some studies conducted by oral gavage in water. When CGN

is dissolved in water at dilute concentration (50.1%) with no

Table 1. Molecular weights of carrageenan and related materials.

Polymer material Molecular weight (Mw): Daltons and/or viscosity specifications Reference

Food grade carrageenanCommercial carrageenan 200 000–800 000 Blakemore & Harpell (2010)Regulatory specifications Viscosity not less than 5 mPa (1.5% solution at 75 �C) Commission Regulation (2012)

Not more than 5% below 50 000 Da molecular weight CGN incommercial CGN*

Commission Directive (2004)

Viscosity not less than 5 mPa s (1.5% solution at 75 �C) Food Chemicals Codex (2013)Viscosity not less than 5 cP at 75 �C (1.5% solution) Food and Agriculture Organization of the

United Nations (2007)Viscosity not less than 5 mPa s Japan Food Additives Association (2009)

Artificially formed polymer (not for food use)Poligeenany 10 000–20 000 USAN (1988)

*Currently, there is no validated analytical method to measure the low molecular weight tail (LMT). Using the best available technology, the LMTfraction of commercial CGN is typically 1.9–12% of the total commercial carrageenan product (Weiner et al., 2007). This LMT fraction ofcommercial carrageenan is primarily due to the incomplete molecular synthesis of the seaweed plants during natural growth prior to harvesting, andthe bulk of the LMT is within the 20 000–50 000 Da range, with generally less than 10% of the LMT being lower than 20 000 Da.yCompared to the LMT fraction of commercial carrageenan detailed above, generally over 90% of poligeenan is less than 20 000 Da. Poligeenan and the

LMT are completely different with respect to molecular weight profile.

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 3

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 4: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

added cations, dietary solids or protein, the polymer exists in

a disorganized random conformation (Blakemore & Harpell,

2010). Under these conditions, all the CGN molecules are

available for maximum interaction with the GI tract cells.

When CGN is dissolved in water at higher concentrations,

such as animal drinking water studies, the carragenan may be

in the ordered or disordered conformation, depending on the

type of CGN and cations present. For example, k-CGN in the

presence of potassium cations and E-CGN in the presence of

calcium ions will gel at concentrations above 0.1% in water

(Blakemore & Harpell, 2010). The cation levels necessary for

such gelation depend on the specific CGN salt. For example,

k- and E-CGN may exist as partially disorganized and partially

organized conformations, depending on the concentration and

cation balance; whereas �-CGN is always in the disorganized

or non-gelling conformation at all concentrations for all

cations (Blakemore & Harpell, 2010). The large number of

negatively charged sulfate moieties in the structure of �-CGN

precludes it from forming helices in water. This last point is

important because several studies use �-CGN in drinking

water, and it is this form that has an open molecular structure

in water when not bound to protein. In dietary studies, where

the CGN concentration is well below the protein content of

food, for example, in rodent chow and infant formula, then the

organized conformation will exist.

As shown in Figures 1 and 2, CGN has numerous

negative charges due to the sulfate groups; protein has both

negatively charged carboxyl groups and positively charged

amine groups. Below the isoelectric point of the protein, a

direct polyanion CGN and polycation protein interaction can

occur, resulting in gelation or precipitation. Also, this direct

CGN–protein bonding occurs above the isoelectric point, but

to a lesser degree as the pH is increased. However,

regardless of pH, all these CGN–protein bonds are strong

and difficult to break. Above the isoelectric point, positively

charged cations, such as calcium or other polyvalent cations,

form a bridge across the negatively charged sulfate groups

on CGN to the negatively charged carboxyl groups on the

protein, forming stable gels (Figure 1). In addition, CGN–

CGN molecules interact to form helical bridges, adding

strength to the gels. In the absence of protein, CGN forms a

loose random conformation in water solution rather than the

structured helical coil conformation formed in the presence

of protein (Figure 2) (Blakemore & Harpell, 2010). Based

on these properties, �-CGN in water will have more sulfate

groups free and exposed to the GI mucosa where irritation

or ulceration may occur. However, it has to be noted that,

even with the absence of helices, the molecular bonds

between �-CGN and protein (both direct and via cations) are

strong and difficult to break. The hypothesis that CGN

conformation has an impact on its toxicological activity is

supported by the numerous studies in this review.

Figure 2. Carrageenan reactivity with casein,a two-step process.

Carrageenan reactivity with casein,a 2-step process

Carrageenanchains

Casein micellewith kappa

casein on thesurface

Step 1.At high temperatures,carrageenan reactselectrostatically withkappa casein

Step 2.On cooling, carrageenanchains interact, creating anetwork

Coolingsol gel

Helical chain

NH2 CO-2 NH2 CO-

2 NH2 CO-2 NH2 CO-

2 NH2 CO-2

++ ++ Ca++ Ca++ Ca++Ca Ca

SO-4 SO-

4 SO-4 SO-

4 SO-4

NH+3 CO2H NH+

3 CO2H NH+3 CO2H NH+

3 CO2H NH+3 CO2H••••

••••••

••••••

•••

••••••

••••••

•••

••••••••

••••••••

••••••••

SO-4 SO-

4 SO-4 SO-

4 SO-4

SO-4 SO-

4 SO-4 SO-

4 SO-4

NH+3 CO2H NH+

3 CO2H NH+3

NH+3

NH+3CO-

2

CO-2

CO-2

Protein

Protein

CarrageenanA

CarrageenanB

CarrageenanC

ABOVE ISOELECTRIC POINT

BELOW ISOELECTRIC POINT

Protein

Figure 1. Carrageenan binding to protein. (A) Above isoelectric point:carrageenan–protein interaction – through divalent cation. (B) Aboveisoelectric point: carrageenan–partial protein interaction – direct.(C) Below isoelectric point: carrageenan–full protein interaction –direct. Figures 1 and 2 are used with kind permission of FMC Corporation.

4 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 5: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Acute toxicity studies

Summary

CGN is not toxic by oral, dermal, and inhalation routes of

exposure; is non-irritating to eyes and skin; and is not a skin

sensitizer.

Acute oral, dermal, and inhalation toxicity

CGN is non-toxic by the oral, dermal, or inhalation routes of

exposure in acute toxicity studies. The oral LD50 in rats,

hamsters, and mice is 45000 mg/kg (FDA, 1972c) and

confirmed in rats using E-CGN by Weiner (1991). The oral

LD50 in rabbits ranges from 2280 to 5340 mg/kg (FDA,

1972c). Acute oral LD50 levels of45000 mg/kg are considered

non-toxic by regulatory authorities, including the FDA (2000).

The dermal LD50 in rabbits is 42000 mg/kg, indicating

that E-CGN is non-toxic via the dermal route (Weiner, 1991;

Weiner et al., 1989).

CGN also has been used in human topical preparations,

such as cosmetics, toothpaste, shampoos, and shaving creams

(Tye, 1991; Weiner, 1991). No adverse effects have been

attributed to the use of CGN in these products. Data

developed for potential occupational exposure found an

inhalation LC50 of 4931 mg/m3 (4 h, maximum attainable

concentration) in the rat, indicating that E-CGN is non-toxic

via inhalation exposure (Weiner, 1991; Weiner et al, 1989).

Gravimetric analysis indicated a mass median aerodynamic

diameter of 8.8� 2.1 mm with 62% of the particles less than

10 mm (Weiner et al., 1989).

Eye irritation potential, skin irritation, and sensitization

potential

Standard animal studies for eye irritation, skin irritation, and

skin sensitization potential were conducted to provide infor-

mation for workplace exposures. I> -CGN is non-irritating to

eyes which were not washed eyes and minimally irritating to

eyes that were washed in rabbits (Weiner, 1991).

k/�-CGN is non-irritating to intact skin and minimally

irritating to abraded skin in rabbits (Weiner, 1991; Weiner

et al., 1989). In a guinea pig skin sensitization study,

k/�-CGN was found to be non-sensitizing (Weiner, 1991;

Weiner et al., 1989). Thus, CGN is not predicted to produce

skin sensitization following dermal exposure in humans.

Based on the current state of scientific knowledge,

repeated dermal exposure to CGN in human preparations

intended for topical use (sunscreens, etc.) is not predicted to

cause skin irritation or skin sensitization reactions in humans.

Degradation, excretion, absorption, and metabolism

Summary

CGN is not significantly degraded in the GI tract and is

excreted unchanged in the feces. CGN is not significantly

absorbed and is not metabolized. CGN does not affect nutrient

absorption.

Degradation and excretion

In an early study, Hawkins & Yaphe (1965) found that

89–101% of ingested food grade k/�-CGN, fed at levels of

2–20% in the diet, was excreted in rat feces, using the

resorcinol method. The resorcinol method measures the 3,6-

anhydrogalactose content as an indication of the presence of

CGN in feces or other medium. Because it relies on a

colorimetric method, the accuracy of the method depends on

the level of clean-up of the solution used to measure the CGN

content and the use of appropriate controls with no CGN to

determine any background interference (such as untreated

feces or feces with CGN intentionally added). If clean-up of

the solution is not thorough, then some CGN will remain

bound to protein in feces and not be extracted. It is important

to insure that there are either no interfering compounds to the

colorimetric measurement, or to account for interfering

compounds by validation using appropriate blanks and CGN

spike and recovery controls.

Dewar & Maddy (1970) found that 80% of the E-CGN fed

to young rats at a level of 5% in the diet was excreted in feces

within 24 h. The CGN content of feces was also estimated by

measurement of the 3,6-anhydrogalactose content using the

resorcinol method. The authors suggest that the low value for

the percent of CGN excreted (�80%) may have been due to

inaccuracy or errors in measuring the daily feed intake and in

assessing the background level of the 3,6-anhydrogalactose

content of the feces from rats on control diet (Dewar &

Maddy, 1970). Tomarelli et al. (1974) administered k/�-CGN

at a level of 5% in diet or CGN prepared in heat sterilized

milk at a level of 4% in milk, to rats and quantitatively

measured CGN excreted in the feces. Excretion of k/�-CGN,

determined by the 3,6-anhydrogalactose content of the feces,

was 98.8� 2.7% from diet-fed rats and 98.3� 2.2% in

processed milk-fed rats (Tomarelli et al., 1974). Tomarelli

et al. (1974) modified the resorcinol method for greater

accuracy by introducing a correction ‘‘blank’’ for non-

specific color formation and subtraction of the optical density

of a sample treated with resorcinol-free reagent from the

complete color reagent. With these modifications, as well as

the inclusion of feces from rats fed milk-based diets

without CGN to measure ‘‘zero’’ background, and standard-

ization of the calibration curve with the same lot of CGN used

in the experimental samples, the authors were able to

accurately quantitate CGN in the feces of treated rats

(Tomarelli et al, 1974).

Pittman et al. (1976) found that the electrophoretic

mobility of CGN (k, �, E) present in the feces from rats,

guinea pigs, or monkeys was lower than that originally

administered, indicating that some degradation may have

occurred in the GI tract or that recovery was not adequate.

Due to variations in the electrophoresis from run-

to-run, the authors were not able to estimate the reduction

of the Mw of the dosed CGNs or to quantitate it.

Nevertheless, they conclude that excretion occurs primarily

in the feces.

In more recent studies, Arakawa et al. (1986, 1988) found

that dietary k-CGN with a Mw4100 000 was not metabolized

to lower Mw material in the feces of rats and that 97–98% of

the ingested CGN dose (4 g/kg) was quantitatively excreted in

the feces. Fecal CGN had a similar Sephacryl S-300 elution

pattern to that of the CGN sample added to the treated diet,

indicating identical molecular weight profiles (Arakawa et al.,

1988). No degradation of CGN excreted in the feces was

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 5

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 6: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

found. This work agrees with the earlier work of Hawkins &

Yaphe (1965) and Tomarelli et al. (1974).

Tache et al. (2000) fed 2.5% k-CGN in water as a gel to

conventional rats with normal rodent gut microflora and rats

with their gut microflora replaced by human fecal microflora.

When CGN is gelled, it has a gel-mode helical conformation,

and, thus is not absorbed from the GI tract. CGN gel is used in

food products that do not contain protein, such as water

dessert gels. The average Mw of the CGN found in feces was

the same as that administered to both conventional rats with

typical rodent microflora and rats with human fecal micro-

flora and also the same as that from rat feces with CGN added

ex vivo as an extraction test. The authors conclude that the

average Mw of CGN was not changed significantly during its

passage through the GI tract. The conclusion that the average

Mw of CGN was not significantly changed was shown when

human fecal microflora had the opportunity to act on ingested

CGN gel. Thus, CGN gel, like CGN in diet, is not degraded

in the GI tract.

Uno et al. (2001a) used GPC to separate high and low

Mw �-CGN components in feces and used ICP atomic

emission spectroscopy (ICP-AES) for direct detection of the

sulfur in feces, thus utilizing the sulfur to find the CGN

present in feces. Food grade �-CGN was included in the diet

of rats at a level of 5%. Animals were deprived of food for

one night, then given access to the treated diet for one day.

For two days after CGN intake, the rats were given access to

the untreated basal diet. Feces were collected in the same

rats at three time points: day 1 (after ingestion of CGN), day

2, and day 3 (following CGN ingestion, but on basal diet).

The Mws of the CGN sample itself and CGN found in the

feces were determined. The Mws of the CGN found in the

feces after day 1 and day 2 were nearly the same as that of

the CGN in the blended diet: 782 kDa in feces compared to

832 kDa in diet. No CGN was detected in feces excreted

on day 3. Uno et al. (2001a) concluded that dietary �-CGN

is excreted in the feces without any decomposition to lower

Mw fractions.

The work of Uno et al. (2001a) and Arakawa et al. (1988)

both used the GPC method to characterize the Mw of CGN

excreted in feces; whereas Pittman et al. (1976) used

electrophoresis for estimation of molecular weights. Uno

et al. (2001a) suggest that the conflicting results are due to

differences in these methods. In electrophoresis, the distance

of travel is determined by Mw and the charge that exists on the

molecules; hence, differences in electrophoretic mobility do

not necessarily indicate a change in Mw alone. According to

Uno et al. (2001a), the feasibility of simple electrophoresis of

high Mw polysaccharides of several hundred thousand Daltons

is questionable. In addition, Uno et al. (2001a) suggest that

the drying and storage of collected feces by Pittman et al.

(1976) may have resulted in decomposition of the CGN

excreted in the feces at the time of drying. Such drying would

strengthen CGN–protein bonds, resulting in making subse-

quent extraction of CGN more difficult. In addition, Pittman

et al. (1976) included boiling feces in water for 2 h, which

may not have completely removed the CGN because it is

strongly bound to protein in feces. Such heat treatment may

also cause some thermal degradation of the CGN, depending

on the pH of the feces.

Arakawa et al. (1988) also explain the difference in their

results from those of Pittman et al. (1976) by the different

composition of the diets used in the two studies and possible

changes in the physiochemical environment in the rat intestine

due to the diet (Arakawa et al., 1988).

In summary, studies on the excretion of food-grade CGN

in the feces indicate that CGN is excreted unchanged

without significant degradation to lower Mw fractions as

verified by GPC, ICP-AES and other methods (Arakawa

et al., 1986, 1988; Uno et al., 2001a). Excretion of CGN was

shown to be 498–100% of the ingested amount by most

researchers (Arakawa et al., 1986, 1988; Hawkins & Yaphe,

1965; Tomarelli et al., 1974; Uno et al., 2001a). JECFA

(1999, 2008) concluded that breakdown of CGN in the GI

tract is probably of limited toxicological significance since,

‘‘if native CGN were sufficiently degraded to cause

ulceration or tumor growth, this would be detected in

feeding studies’’.

Absorption

The absorption of CGN has been reviewed by Roch-Arveiller

& Giroud (1979) and Weiner (1988). Numerous studies

indicate that food grade CGN is not significantly absorbed

after oral exposure due to its high molecular weight.

Rat. Grasso et al. (1973) did not detect the presence of

CGN in the intestinal walls of rats fed 5% E-CGN, although

systemic absorption was not evaluated in this study. CGN

(k, E) was not stored in the livers of rats, following dietary

administration at levels of 1 or 5% for one year, as judged

by the absence of histochemical staining using toluidine blue

dye for the presence of metachromatic material, such as

CGN, followed by visualization by light and electron

microscopy (Albany Medical College, 1975). The toluidine

blue method for the detection of CGN was standardized for

heparin (MacIntosh, 1941) and refined for CGN for accuracy

and specificity (Roberts & Quemener, 1999). Pittman et al.

(1976) fed rats diets containing 5% k/�-CGN extracted from

C. crispus with a Mw488 000 for 13 weeks and

reported that no CGN was detected in the liver, hence not

absorbed.

Absorption of CGN was studied by Nicklin et al. (1988)

using radiolabeled 3H-E-CGN administered orally by gavage

in water to rats. Radiolabeling was achieved by incubating

CGN with galactose oxidase and tritiated borohydride. The

–CH2OH group receives tritium with one 3H tritium bonded

to the carbon atom which is stable and one 3H bonded tritium

to the oxygen atom which is labile and capable of exchanging

with body water. The majority of the radioactivity was

excreted in the feces (Nicklin et al., 1988). Some uptake of the

label into the intestinal wall, Peyer’s Patches, mesenteric

lymph node, cecal lymph node, and serum was observed.

However, the authors cautioned that due to the exchange of

the 3H tritium label with the hydrogen of body water, only

levels of radioactivity greater than those found in serum can

be considered significant. Thus, only the cecal lymph node,

Peyer’s patches, and possibly the intestinal wall exhibited the

presence of bound tritium. Autoradiographs of tissues

indicated that the bound radioactivity was apparently

6 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 7: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

associated with the macrophages. The authors suggest that

CGN probably enters the body via the macrophages of the

Peyer’s Patches and cecal lymph nodes.

Nicklin & Miller (1984) administered 0.5% CGN (k, E, �)

in drinking water to rats for 90 d and detected the presence

of small amounts of stained material within cells of the

intestinal villi, lamina propria, and basement membrane of

lymphatics, using Alcian blue stain for CGN. This method

relies on a color shift of the dye from green to purplish

green at 520 nm in the presence of CGN (Roberts &

Quemener, 1999). The amounts of CGN were not quanti-

tated and no difference in staining was seen between the

three forms of CGN. In addition, there were no histopatho-

logical findings or abnormal effects on the GI tract and no

alteration in the immune response to orally administered

antigen in treated animals.

The effect of CGN in diet on intestinal permeability to

macromolecular markers, polyethylene glycol (PEG) 900 and

4000, was evaluated in female rats (Elsenhans & Caspary,

1989). Following 4 weeks of feeding 20% CGN (an exces-

sively high dose which could result in nutritional deficiencies

due to replacement of a significant portion of the normal diet

by a non-nutrient additive), rats showed increased cecal and

intestine weights, increased intestinal length, and decreased

excretion of PEG-900, compared to rats on fiber-free diets.

The excretion of PEG-4000 was not affected by CGN

administration. These results are difficult to interpret.

It appears that CGN at an excessively high dose did not

increase the intestinal permeability to either marker and

actually decreased the permeability to the smaller marker.

Guinea pig. Pittman et al. (1976) detected E-CGN prepared

with a Mw range of 88–107 kDa, but not CGN prepared with a

Mw of 275 kDa, in the livers of guinea pigs after dietary

administration of specific isolated molecular weight fractions

(from 88 to 275 kDa). The majority of the administered CGN

was present in the feces. Detection of CGN in the urine of

guinea pigs administered artificially prepared fractions of

CGN in drinking water was found only in Mw fractions

�21 kDa (Pittman et al., 1976).

In contrast to CGN administered in the diet, Engster &

Abraham (1976) reported that CGN (k, �, E) with number

average molecular weight or Mn4145 000 administered as a

1% solution in drinking water was not retained in the guinea

pig cecum; whereas lower Mn forms, artificially prepared

from CGN (�107 kDa), were retained in the cecal lamina

propria and submucosal macrophages.

No CGN fragments with Mws439 000 were found in the

urine following administration of 1% CGN in drinking water

for 2–3 weeks to guinea pigs (Pittman et al., 1976). In both

these cases, it can be deduced, based on the work of others

using a broad spectrum of CGN Mw fractions (Engster &

Abraham, 1976; Pittman et al., 1976; Uno et al., 2001a) that

CGNs with Mw4100 kDa are not absorbed and are excreted

in the feces.

Primate. Rhesus monkeys given 1% k/�-CGN (average Mw

800 kDa) in drinking water for 7–11 weeks (with a subsequent

24-week recovery period) showed no evidence of CGN

located in the reticuloendothelial cells or Kupffer cells of the

liver after 11 weeks or after the recovery period, using

toluidine blue to stain for polysaccharides, in particular, CGN

(Abraham et al., 1972). In another study on rhesus monkeys,

Mankes & Abraham (1975) found no tissue storage of CGN

when the monkeys were given 1% k/�-CGN in the drinking

water for 10 weeks. Pittman et al. (1976) reported that

monkeys receiving daily doses of a k/�-CGN fraction (Mw

185 kDa) (500 mg/kg) by stomach tube for 15 months

excreted 12 mg of CGN per milliliter of urine. This value

(12 mg/ml) was reported to be at the limit of detection of the

method, and confirmation by isolation of the material from

urine was not made. Monkeys receiving daily doses of 500,

200, or 50 mg/kg of k/�-CGN in food for 7.5 years of oral

administration did not show evidence of the uptake or storage

of CGN in the liver, lymph nodes, small intestine, colon, or

cecum using toluidine blue staining by electron microscopy

(Albany Medical College, 1983).

Special studies on nutrient absorption

Five percent CGN in the diet had no effect on utilization of

casein, soybean protein, or other proteins by rats (Friedman &

Douglass, 1960). No difference in protein economy of the

growing rat was observed with a diet of 0.5–5% CGN and

either a high quality protein (casein) or a low quality

vegetable protein (JECFA, 1974). Three percent CGN in the

diet has been reported to reduce the plasma cholesterol level

in chicks by 50% (Riccardi & Fahrenbach, 1965). K/�-CGN

exhibited a cholesterol-lowering effect in rats; an increase in

feces weight and in the concentration and daily output of fecal

cholesterol and total bile acids after 33 weeks of feeding

(Reddy et al., 1980). When fed in a simulated milk powdered

diet to adult rats, k/�-CGN, at a dietary level of 4%, had no

influence (compared to glucose or cellulose) on growth rate,

diet energy efficiency, absorption of protein, fat, calcium,

blood coagulability, utilization of protein for growth, or

utilization of iron (Tomarelli et al., 1974). The fecal excretion

of calcium, iron, zinc, copper, chromium, and cobalt was

slightly higher in animals fed 10% k/�-CGN for 8–147 d

compared to control animals, indicating that CGN has no

chelating powers with respect to metallic ions (Harmuth-

Hoene & Schelenz, 1980). K/�-CGN in an infant formula fed

to rats did not inhibit calcium absorption (Koo et al., 1993).

Vitamin A (300 000 IU) was administered by Kasper et al.

(1979) to 11 young human female subjects, in a formula diet

containing 20 g of CGN. Under these experimental condi-

tions, CGN promotes the assimilation of vitamin A, since the

blood level of vitamin A was significantly higher some hours

after its administration. Similar effects were observed with

bran, microcrystalline cellulose, pectin, guar gum, and locust

bean gum. The authors did not interpret these experiments,

but hypothesized that possible modifications due to peristal-

sis, viscosity, transport, and enzymatic actions may occur in

the presence of these materials. Orally administered CGN

does not significantly affect the absorption of other nutrients.

Metabolism

Dietary feeding studies have reported cecal enlargement and

decreased concentrations of cecal bacteria in the rat following

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 7

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 8: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

administration of high doses (5%) of hydrocolloid fibers, such

as E-CGN, pectin, guar gum or carboxymethyl cellulose

(Mallett et al., 1984, 1985). This enlargement may be

considered a bulking effect due to the osmotic pressure that

results from large amounts of non-digestible hydrocolloid in

the feces. There was a two- to three-fold enlargement of the

cecum as well as a significant decrease in the concentration of

gut microflora bacteria and alterations in several enzyme

activities of these bacteria in vitro following E-CGN admin-

istration to rats, mice and hamsters (Mallett et al., 1985).

Rowland & Mallet (1986) reviewed the effects of dietary

hydrocolloids on gut microflora in animals and concluded

that extrapolation to humans is difficult. They found that the

effects on gut microfloral enzyme activities (nitrate reductase

and b-glucuronidase) due to dietary pectin in rats were not

observed in humans following ingestion of 18 g pectin

(Rowland & Mallet, 1986). Rowland et al. (1986) compared

five enzyme activities (azoreductase, beta-glucosidase, beta-

glucuronidase, nitrate reductase, and nitroreductase) asso-

ciated with hindgut microflora in six species of animal (rat,

mouse, hamster, guinea-pig, marmoset, and man). They found

marked differences in the cecal activities of these enzymes in

the four rodents, with no one species exhibiting consistently

higher or lower enzyme activity. None of the animal species

provided an approximation of the enzyme profile associated

with human fecal microflora. They concluded that these

results indicate that it may not be valid to extrapolate the

results of bacterial metabolic studies between closely related

species, or from animals to man (Rowland et al, 1986).

Under the most drastic artificial stomach conditions, rapid

acidification for 3 h, breakdown of food-grade k/�-CGN was

very limited, with less than 10% of the CGN having a

Mw5100 kDa and less than 3.6% having a Mw550 kDa

(Capron et al., 1996). CGN was not fermented by any of

154 different anaerobic bacterial strains isolated from

human feces (Salyers et al., 1977). CGN and other sulfated

polysaccharides did produce hydrogen sulfide gas during a 48 h

in vitro incubation with human fecal slurries. However, CGN

fermentation is thought to provide only a very limited amount

of hydrogen sulfide in the large intestine (Michel &

MacFarlane, 1996).

In summary, following oral administration, CGN is not

metabolized and is not significantly broken down to lower Mw

material in the GI tract. Most of the CGN administered in the

diet (498%) is excreted unchanged in the feces.

Subchronic and long-term oral toxicity studies

Summary

Subchronic and chronic dietary studies demonstrate that CGN

does not result in target organ toxicities. The only effects

noted are soft stools and/or diarrhea, typically seen with high

levels of dietary fibers (5% or more in the diet). One study

finding an effect of CGN on glucose utilization in mice

employed �/k-CGN in drinking water (Bhattacharyya et al.,

2012), but does not agree with other publications.

Subchronic oral toxicity

Many of the earlier toxicology evaluations done with CGN

have been subchronic rodent feeding studies (reviewed by

FDA, 1973; Weiner, 1991). These have ranged from a few

weeks to 90 d of treatment using dietary concentrations of

1–25% CGN in rodents (Abraham et al., 1985; Grasso

et al., 1973; Mankes, 1977). The majority of these studies

have used dietary concentrations of 1% or 5%. In all

studies, a dietary level of 1% fed for 90 d has caused no

adverse effects. The only finding noted was soft stools

after dosing at the 5% dietary concentration. Very high

non-physiological dietary concentrations (25%) were given

to rats for 4 weeks (Mankes, 1977). The adverse effects

noted included growth retardation, inflammatory changes

in the gut, feces coated with fresh blood, and apparent

storage of CGN in the Kupffer cells of the liver (Mankes,

1977). It must be noted that these effects were not due to

direct toxicity, but rather to the replacement of a large

portion of the diet (25%) with an inert substance with a

strong likelihood to have nutritional effects, which will

confound the interpretation of any study. OECD guidelines

452 and 453 specify the highest dose of test material

tested in dietary feeding studies should be no more than

5% to avoid nutritional deficiencies for non-nutritive

additives: ‘‘For substances administered via the diet or

drinking water it is important to ensure that the quantities

of the test substance involved do not interfere with normal

nutrition or water balance. In long-term toxicity studies

using dietary administration, the concentration of the

chemical in the feed should not normally exceed an

upper limit of 5% of the total diet, in order to avoid

nutritional imbalances’’ (OECD, 2009a, 2009b).

Primates. Adult rhesus monkeys (10/sex) were adminis-

tered 1% high Mw k-CGN in drinking water for 11 weeks,

corresponding to an average daily intake of 1.3 g/kg/d

(Benitz et al., 1973). Animals were terminated after 7 or 11

weeks of treatment or were given plain tap water and

allowed to recover for 11 weeks. Because no adverse effects

were apparent after the recovery period, the animals then

received escalating oral doses of 50–1250 mg/kg/d in water

by stomach tube for up to 12 weeks. The only effects noted

were occasional soft or watery stools in treated and recovery

group animals (Benitz et al., 1973). Gross and microscopic

examination of the GI tract found no changes related to

treatment with high Mw CGN.

Rats. Groups of Fischer 344 rats (20/sex/group) were fed

control or treated diets at levels of 0, 2.5%, or 5.0%

k-CGN for 90 d in a standard FDA and OECD guideline

subchronic study conducted under GLP Guidelines (Weiner

et al., 2007). The sample of k-CGN used had an average

Mw range of 196–257 kDa, with a relatively high percent-

age of low molecular weight tail (LMT) (a mean of 7%

550 kDa with a range of 1.9–12.0% below 50 kDa). The

Mw of the LMT was determined by an experimental

method. Clinical observations were performed daily.

Individual feed consumption/body weight measurements

were made weekly. Ophthalmic exam was conducted prior

to and after treatment. Hematology, serum chemistry, and

urinalysis evaluations were done at necropsy, as were

organ weight determinations for adrenals, brain, heart,

8 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 9: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

kidneys, liver, ovaries, spleen, testes, and thyroid with

parathyroids. Full histopathological examination was con-

ducted on organs from animals in the control and high-

dose groups. In addition, the GI tract was evaluated in

detail using the Swiss roll technique, which permits more

extensive evaluation of the intestine than routine sections.

The results indicate no treatment-related effects on body

weight, urinalysis, hematology, or clinical chemistry par-

ameters, organ weights, or ophthalmic, macroscopic, or

microscopic findings. Even with the relatively higher

amount of low molecular weight material present in the

test material, there was no evidence of erosions, ulcer-

ations, inflammation, regeneration, hyperplasia, or any

other abnormalities of the entire GI tract (Weiner et al.,

2007). The NOAEL was at least 5.0% in the diet or

calculated k-CGN consumption of 3394� 706 mg/kg/d in

males and 3867� 647 mg/kg/d in females. The results of

the study provide evidence that it is not necessary to

characterize CGN by a food additive specification for a

LMT (less than 5% below 50 kDa Mw), as established by

the European Commission (Commission Directive 2004/45/

EC of 16 April 2004).

Mice. Bhattacharyya et al. (2012) gave C57BL/6 J male

mice (6/group) drinking water without or with �–k-CGN

(Sigma Chemical Company, St. Louis, MO, USA) at a level of

10 mg/L for 18 d to evaluate potential effects on glucose and

insulin tolerances. Glucose tolerance was performed at 12

weeks of age after the 18-d exposure to CGN. For the glucose

tolerance test, mice were fasted for 18 h prior to sampling

blood for measurement of glucose with and without an

intraperitoneal (ip) injection of dextrose (2 g/kg). For the

insulin tolerance test, mice were exposed to control or treated

drinking water for an additional 15 d (33 d total). Following a

2 h fast, insulin ip injection (0.75 U/kg) was given and blood

was sampled for glucose from time 0 min until 120 min post-

injection. There were no effects on body weight, water

consumption, activity, or clinical observations in mice with

drinking water with CGN, compared with controls. The

results indicate that ingestion of CGN in drinking water under

the experimental conditions described significantly impaired

glucose tolerance and significantly impaired the response to

an injection of insulin. There are a number of possible

explanations for these findings in this publication, which

makes conclusions difficult to interpret (see discussion in

McKim, 2014). A positive control group, such as insulin

injection to another group of animals, would have helped to

validate the findings. Since no GI tract inflammation or

histopathology was evaluated, it is not possible to determine if

the authors’ hypothesis that CGN acts via an inflammatory

mechanism is correct. The relevance of these results to human

diabetes is not clear. There are no other studies showing an

effect of CGN on glucose, either in short-term or long-term

studies, including chronic studies in rats and hamsters and a

7.5-year monkey study. CGN has not shown any effects on

hematological parameters or serum clinical chemistry par-

ameters, including blood glucose, when evaluated at the end

of the 7.5 year monkey study (Albany Medical College,

1983); a 40-week dietary study in rats (Abraham et al., 1985)

and a 90-d dietary study in rats (Weiner et al., 2007).

Thus, the results of this one study (Bhattacharyya et al., 2012)

are inconsistent with data from other toxicological studies

on CGN.

Long-term oral toxicity

Long-term administration of CGN to rodents (Abraham et al.,

1985; Rustia et al., 1980) and primates (Albany Medical

College, 1983) has resulted in essentially no adverse effects.

Rats and hamsters have been fed 0, 0.5%, 2.5%, or 5.0%

k-CGN for their lifetimes (Rustia et al., 1980). Survival was

not affected in either species. No statistically significant

pathologic effects were seen in either species at any of the

dietary levels used in the study. This study only evaluated

survival and tumor development and did not include other

parameters of chronic toxicity (Rustia et al., 1980).

Administration of 0, 1%, or 5% k/�/E-CGN in the diet for

39 weeks to two strains of rats had no effect on hematology

and clinical chemistry parameters, fecal examinations, liver

weights, and liver pathology (Abraham et al., 1985).

Rhesus monkeys have been fed 50, 200, or 500 mg/kg/d for

7.5 years; for the first 5 years k/�-CGN was administered by

oral gavage 6 d/week and for the final 2.5 years, CGN was

administered in dietary pellets. Parameters evaluated included

the following: body weight, clinical chemistry, hematology,

stools, liver and testes biopsies, organ weights (liver, kidneys,

heart, brain, adrenals, ovaries/testes, pituitary), gross nec-

ropsy, complete histopathology on all organs/tissues, histo-

chemistry with toluidine blue (small and large intestines,

cecum, liver, and lymph nodes). There were no effects on

hematological or clinical chemistry parameters, including

blood glucose. No pathologic effects attributable to the intake

of CGN were seen in the monkeys. No evidence of GI tract

ulceration or other abnormal pathology was observed. There

was no evidence of storage of CGN in the liver. The only

effects seen were soft stools or diarrhea and occasional fecal

occult blood, which were also seen in untreated controls

(Albany Medical College, 1983).

In summary, subchronic and chronic administration of

CGN in the diet to several species, in well-conducted studies,

did not result in adverse effects other than soft stools or

diarrhea. These effects are commonly seen when dietary

fibers are fed at high doses (Tungland & Meyer, 2002).

Immune system effects

Summary

Existing studies on immune system parameters were con-

ducted mainly with �-CGN in drinking water, which provide

data that are not directly applicable to dietary exposure where

CGN is tightly bound to protein. Based on the available

studies, conducted in diet, CGN has not been shown to

produce immunotoxicity.

CGN exhibits a number of biological effects following

systemic administration via intravenous (iv) or ip injection

that relate to the immune system (Nacife et al., 2004;

Thompson, 1978; Thompson & Fowler, 1981; Thompson

et al., 1979). When administered by a systemic route of

exposure (ip or iv), CGN has the capacity to induce acute and

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 9

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 10: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

chronic inflammation and granuloma formation; cause the

release of vasoactive amines and other humoral mediators

(kinins, prostaglandins, and cytokines) from immune system

cells; inhibit the hemolytic activity of complement; act as an

adjuvant, and suppress specific antibody- and cell-mediated

immunity. CGN has substantial effects on the immune system

when administered systemically, as shown in the mouse or rat

footpad model. Many of these models have been utilized for

the examination of basic immune processes. However, these

models all involve systemic administration, and are not

relevant to oral exposure of the high Mw CGN in the diet

(Cohen & Ito, 2002). It is important to emphasize that the

systemic routes of exposure have been used experimentally to

further pharmacological research in the management and

treatment of inflammatory diseases or conditions, and that

this work via systemic administration is not relevant to

consumption of food that contains CGN. CGN is not absorbed

from the GI tract in significant quantities (see the section

‘‘Degradation, excretion, absorption, and metabolism’’).

Bash & Vago (1980) gave rats k/�-CGN via oral gavage

in water at doses of approximately 5–500 mg/kg/d once or

daily in drinking water for 61–85 d followed by measurements

of immunity. No consistent dose–responses were observed.

A single oral dose of CGN (1 mg) in water by gavage

significantly suppressed splenic and lymph node T-cell

responses to mitogens; whereas a single dose of 10 mg was

not suppressive. Rats maintained on 0.1 or 1.0 mg/ml

solutions of CGN in drinking water for 61 d also exhibited

significant suppression of the spleen cell mitogenic response

at the lower dose only. These doses (0.1 and 1.0 mg/ml) are

stated to be equivalent to 2.3 and 23.5 mg/day/rat,

respectively. Responses returned to normal when adherent

cells (macrophages) were removed prior to mitogenic stimu-

lation. To further define the mechanism of action, macro-

phages were harvested from rats administered drinking water

for 85 d with 0.1 or 1.0 mg/ml/d of CGN, cultured for 72 h and

added to spleen cell cultures responding to phytohemagglu-

tinin (PHA). The degree of suppression of spleen cell

responses to mitogen was roughly equivalent to that seen in

control macrophage cultures pretreated in vitro with 10 mg/ml

of the same CGN preparation (Bash & Cochran, 1980). In

both the in vivo and in vitro studies, macrophage-induced

suppression of the mitogenic response was evident at the

lower doses only.

These responses (macrophage-induced suppression of

the T-cell mitogenic response) were not seen at the higher

CGN doses. In order to explain this inverse dose–response

phenomenon, the authors speculated that suppressor

macrophages were induced at the lower dose. This effect

is curious and has not been reported by other investiga-

tors, either in vitro, or in vivo via oral or systemic routes.

The �-CGN administered in drinking water or exposed to

cells in vitro exists in the random coil conformation and,

hence, not in the structured form found in food or liquids

(see Introduction).

Nicklin & Miller (1984) examined the systemic and local

(gut-associated) immune response of rats administered E-, k-,

or �-CGN in drinking water at a level of 0.5% or the

equivalent of approximately 150–250 mg/kg/d CGN for 90 d.

Gut-associated levels of IgA and of specific agglutinating

antibody to gut bacteria were not affected by CGN treatment.

Total bile IgA levels also were not affected. The response to

injected sheep red blood cells (RBCs) was reduced in animals

on drinking water containing CGN or guar gum.

Biliary anti-sheep RBCs or anti-CGN immune responses

also were not detected (Nicklin & Miller, 1984). In contrast to

orally administered antigen, the expected anti-sheep RBC

serum antibody responses following ip injection of anti-sheep

RBCs was somewhat delayed, relative to controls, after (k, �,

E) CGN administration. This was seen in the absence of any

specific T-cell effects on graft-versus-host reactivity. The

authors concluded that T-cell immunity was unaffected.

Nicklin et al. (1988) administered 0.25% E-CGN in drinking

water to rats for 184 d. Animals on tap water or CGN-

containing water were challenged with an ip injection of 1 ml

of 5% sheep RBCs. Serum was collected once a week for

4 weeks and analyzed for anti-sheep RBC antibody activity by

the hemagglutination method. Animals treated with CGN had

a delayed and reduced antibody response, compared to

controls.

Nicklin et al. (1988) demonstrated the presence of3H-radiolabeled E-CGN in the cecal lymph node (see section

‘‘Degradation, excretion, absorption, and metabolism’’). As

in their earlier report using Alcian blue staining (Nicklin &

Miller, 1984), autoradiographic analysis of the tissues

revealed that the bound radiolabel was associated with

macrophages. The pharmacokinetic data indicate that CGN

enters the body via the Peyer’s patches and cecal lymph nodes

and may be transported via resident macrophages to mesen-

teric lymph nodes. In oral feeding studies with the unlabeled

CGN, however, it took a relatively high oral dose (0.25% in

drinking water for 184 d) before suppression of immune

responses to T-cell-dependent antigens was noted. This CGN

dose level is consistent with that of earlier oral drinking water

studies. However, unlike their earlier study (Nicklin & Miller,

1984), accurate measurements of actual CGN consumption

were not made.

The relative potency of E-CGN to act as an adjuvant

following ip versus oral administration with the antigen,

ovalbumin, was studied by Coste et al. (1989). E-CGN elicited

reaginic and IgG–ovalbumin specific antibody responses

when admixed with 1 mg antigen (ovalbumin) and adminis-

tered ip to rats at doses equivalent to 5 mg/kg. When antigen

and E-CGN were administered orally via gavage in water, no

specific immune response was initiated. These results indir-

ectly support the studies of Nicklin & Miller (1984) who

demonstrated no oral adjuvant effects of CGN on local

immunity to intestinal antigens. Responses via the ip route are

not relevant to oral exposure to food containing CGN. Coste

et al. (1989) conclude that CGN is not active as an adjuvant

when administered orally in water.

More recently, there have been studies suggesting an effect

on the immune system following oral administration of

�-CGN (Frossard et al., 2001; Tsuji et al., 2003).

Unfortunately, in most of these studies, specific information

regarding the type of administered �-CGN is not provided.

Frossard et al. (2001) showed that oral gavage administration

of �-CGN in water (0.5 g/L) with low amounts of

b-lactoglobulin (BLG), a common milk antigen, could

prevent anaphylaxis in mice subsequently sensitized to BLG

10 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 11: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

together with the potent mucosal adjuvant cholera toxin (CT).

Tolerance was induced only when both CGN and BLG were

administered together prior to sensitization to BLG; but not

when CGN was administered alone. Since CGN and BLG

were administered simultaneously, there may have been an

effect of the CGN on the structure of BLG due to the known

binding of CGN to proteins. Similarly, Tsuji et al. (2003) also

showed a suppression of the allergic reaction when low doses

of �-CGN (0.001–0.005%) were administered to mice orally

in drinking water for six consecutive days, following

immunization by ovalbumin in alum intraperitoneally.

�-CGN suppressed both antibody production and mitogen-

induced T-cell proliferation optimally at 0.001%, compared to

higher doses of40.02%. Frossard et al. (2001) and Tsuji et al.

(2003) dosed �-CGN in water. As noted in the Introduction,

�-CGN exists only in the disorganized conformation in water,

so the relevance of these results to dietary exposure, where

CGN exists in the organized conformation, is questionable.

There is little evidence in humans suggesting an effect of

dietary exposure to food-grade CGN on the immune system.

However, there is one anecdotal report in the literature

describing a case of anaphylaxis secondary to exposure to a

barium medical imaging formulation (Tarlo et al., 1995). Based

on an allergy skin test, the Radioallergosorbent Test (RAST)

evaluation, the authors concluded that the allergic reaction

occurred due to CGN and no other substances present in the

barium medical imaging formulation. Furthermore, when the

patient was placed on what was purported to be a CGN-free

diet, the patient’s previous gastrointestinal symptoms abated.

The report indicates the use of sodium CGN in the barium

medical imaging formulation. It is widely known in the

industry that poligeenan, not CGN, was the material used in the

barium medical imaging formulation (Marinalg, 2006; JECFA,

2008). The term ‘‘poligeenan’’ was not in general use

internationally at the time of the reported incident of anaphyl-

axis. In the Tarlo et al. (1995) paper, the type of CGN used for

the skin test is not detailed. The skin prick test with the barium

medical imaging formulation was positive (10 mm mean wheal

diameter with flare). The individual ingredients of the barium

medical imaging formulation were tested separately in water at

concentrations used in the commercial imaging solution. The

only ingredient demonstrating a positive response was 0.4%

‘‘sodium CGN’’ (8 mm mean wheal diameter with flare);

however, a separate skin test with a commercial skin test

preparation of CGN gum (1:50 wt/vol) gave a borderline

response (2 mm mean wheal response) (Tarlo et al., 1995).

Moreover, there are no prior or subsequent case reports of an

allergic reaction to CGN in humans or in animals. Because

CGN is not functional in the barium medical imaging

formulation (Marinalg, 2006), the ‘‘sodium CGN’’ reported

by Tarlo et al. (1995) was certainly poligeenan.

By comparison, Mallett et al. (1985) reported that antibody

to cecal bacteria was increased in Sprague–Dawley rats fed

approximately 5% E/k-CGN in the diet for 30 d.

In summary, the majority of the studies in which

animals were exposed to CGN in drinking water or by oral

gavage in water (Bash & Vago, 1980; Coste et al, 1989;

Frossard et al., 2001; Nicklin & Miller, 1984; Nicklin et al.,

1988; Tsuji et al., 2003) show conflicting results. Several

studies showed immunosupression (Bash & Vago, 1980;

Frossard et al., 2001; Tsuji et al., 2003). The conformation of

CGN in water is different from that in food, where CGN is

bound to protein (Blakemore & Harpell, 2010) (see the

section ‘‘Introduction’’). As discussed, �-CGN exists in the

random conformation in water, and �-CGN was used in the

studies of Frossard et al. (2001) and Tsuji et al. (2003). The

studies of Bash & Vago (1980) and Nicklin et al. (1988) used

�- and E-CGNs, respectively. Therefore, the relevance of these

drinking water and gavage studies to human dietary con-

sumption is unclear. The single dietary study (Mallet et al.,

1985) conducted a very limited investigation with only one

endpoint evaluated at 5% in the diet.

The data from oral exposure studies show that small

amounts of CGN may be taken up by gut associated lymphoid

tissue and may be transported to regional lymph nodes by

macrophages following ingestion. Nevertheless, the absence of

toxicity and lack of significant amounts of the material outside

the gut in feeding studies, including the long-term primate

feeding studies (Albany Medical College, 1983), demonstrate

that food grade CGN is safe from a toxicological viewpoint. In

dietary studies in which complete organ histopathology,

clinical chemistry and hematology were evaluated, CGN did

not cause any significant treatment-related toxicity related to

the immune system (Abraham et al., 1985; Weiner et al., 2007).

Doses of CGN used in the subchronic dietary study (Weiner

et al., 2007) are orders of magnitude above levels of CGN in the

human diet: 3394–3867 mg/kg bw/d in females and males,

respectively, compared to 18–30 mg/kg/d in humans (Shah &

Huffman, 2003). The long and safe use of CGN as a food

additive also attests to a general lack of toxicity and a lack of

immunotoxicity via dietary exposure.

Ulcerative effects

Summary

Due to the known ulcerative effects of poligeenan, the

potential of CGN to cause GI tract ulceration has been

extensively studied. Food grade CGN administered in diet

does not cause ulceration on the GI tract.

Poligeenan has been reported to cause intestinal ulceration

in animals when administered in drinking water or diet (see

review by Michel & Macfarlane, 1996). The ulcerogenic

potential of CGN has been studied extensively. Studies of

CGN in diet found no evidence of intestinal ulceration in rats,

hamsters, primates, and pigs when fed at levels of 5% in the

diet or less. Exposure to very high, non-physiological levels

(415%) of CGN in the diet produced ulcerative effects

(Mankes, 1977; Watanabe et al., 1978). Benitz et al. (1973)

found that exposure to poligeenan, but not CGN, in drinking

water produced gastrointestinal ulceration in adult primates.

Newborn primates given infant formula with k/�-CGN also

showed no evidence of GI ulceration (McGill et al., 1977).

Negative studies for intestinal effects have also been reported

for the hamster (Rustia et al., 1980) and the pig (Poulsen,

1973). Conflicting results have been reported in guinea pigs

with CGN (Engster & Abraham, 1976; Grasso et al., 1973;

Watt & Marcus, 1969). Table 2 summarizes the results of

studies on poligeenan (termed degraded CGN, rather than

poligeenan) published prior to 1985 demonstrating intestinal

ulceration (Michel & Macfarlane, 1996). These findings are

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 11

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 12: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Tab

le2

.In

test

inal

effe

cts

of

po

ligee

nan

(Mic

hel

&M

acfa

rlan

e,1

99

6).

An

imal

Car

ragee

nan

Am

ou

nt

Ad

min

istr

atio

nD

ura

tio

nH

isto

pat

ho

log

ical

chan

ges

Ulc

erat

ion

Ch

ange

inin

test

inal

flo

raR

efer

ence

Deg

rad

edca

rrag

een

ans

Gu

inea

-pig

sD

egra

ded

i-ca

rrig

een

anfr

om

Eu

chem

asp

ino

sum

(Gla

xo

)S

O2

9%

5%

(ca.

2g

kg�

1)

Dri

nk

ing

wat

er2

0–

45

(Ce,

Co

,R

e)W

att

&M

arcu

s(1

96

9,

19

71

)

Gu

inea

-pig

sD

egra

ded

carr

igee

nan

(Gla

xo

)In

crea

sein

aero

bic

Gra

m-n

egat

ive

bac

teri

aV

and

erW

aaij

etal

.(1

97

4)

Co

nven

tio

nal

2%

(ca.

2.5

gk

g�

1)

þ5

%(c

a.2

.4g

kg�

1)

Dri

nk

ing

wat

er3

0–

44

dþþ

Incr

ease

inae

rob

icG

ram

-neg

ativ

ebac

teri

a

En

tero

ba

cter

iace

ae-

2%

�F

ree

5%

þG

uin

ea-p

igs

Deg

rad

edi-

carr

egee

nan

fro

mE

.S

pin

osu

mC

16

(Gla

xo

).S

O3

3%

;M

w:

ca.

30

00

0

0.0

2%

Dri

nk

ing

wat

er1

2m

on

ths�

�A

bra

ham

etal

.(1

97

4)

0.2

%D

rin

kin

gw

ater

10

mo

nth

s�

�2

%D

rin

kin

gw

ater

2w

eek

þþ

(Ce)

(Co

)

2%

Mil

kso

luti

on

s3

mo

nth

s�

�G

uin

ea-p

igs

Deg

rad

edca

rrag

een

anfr

om

E.

spin

osu

m(G

laxo

).S

O3

3%

;

0.2

5%

Dri

nk

ing

wat

er4

wee

ks

�–

Gra

sso

etal

.(1

97

5)

0.5

%4

wee

ks

þþ

(aft

er4

wee

ks)

0.2

5th

en0

%1

2th

en4

or

8w

eek

s�

0.5

then

0%

4th

en2

,5

or

8w

eek

then�

þth

en�

Gu

inea

-pig

sM

ixo

fk

and�

-DC

fro

mC

ho

nd

rus

cris

pu

s,i-

DC

fro

mG

igar

tin

aac

icu

leri

s

þþ

(12

/12

)*W

att

&M

arcu

s(1

97

8)

3%

(ca.

5.7

gk

g�

1)

Dri

nk

ing

wat

er4

–8

wee

ks

þþþ

(12

/12

)*

Deg

rad

edfu

rcel

lara

nfr

om

Fu

rcel

lari

(8/1

2)*

Gu

inea

-pig

sD

egra

ded

carr

agee

nan

(Gla

xo

)5

%þþþ

Incr

ease

into

tal

bac

teri

a,co

lifo

rms

and

Gra

m-n

ega-

tive

anae

rob

es

On

der

do

nk

&B

artl

ett

(19

79

)

5%þ

Gen

tam

icin

**

þþþ

5%þ

SM

TM

þþþ

5%þ

Van

com

yci

nþþþ

5%þ

Cli

nd

amyci

nD

rin

kin

gw

ater

7–

21

d�

5%þ

Met

ron

idaz

ole

�5

SA

SP

þG

uin

ea-p

igs

Deg

rad

edca

rrag

een

an(G

laxo

)þþþ

On

der

do

nk

&B

artl

ett

(19

79

)

Co

nven

tio

nal

�G

erm

-fre

e5

%D

rin

kin

gw

ater

7–

21

d�

Acc

ord

ing

tost

rain

s–

(bu

td

eath

of

anim

als)

Gn

oto

bio

tic

12 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 13: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Gu

inea

-pig

si-

DC

fro

mE

.sp

ino

sum

(Sig

ma)

3%

(ca.

4.3

gk

g�

1)

No

rris

etal

.(1

98

1)

k-D

Cfr

om

Eu

chem

aco

tto

nii

3%

(ca.

7.7

gk

g�

1)

Dri

nk

ing

wat

er3

wee

ks

�-D

Cfr

om

Gig

art

ina

aci

cula

ris

and

G.

pis

till

ata

3%

(ca.

7.1

gk

g�

1)

Gu

inea

-pig

sD

egra

ded

carr

agee

nan

3%

Dri

nk

ing

wat

er2

d�

�M

arcu

set

al.

(19

90

)

28

dM

uco

sal

atte

nu

atio

nan

dhy

per

pla

sia�

New

bo

rnra

ts*

**

Deg

rad

edi-

carr

agee

nan

fro

mE

.sp

ino

sum

SO

29

%5

%D

rin

kin

gw

ater

6m

on

ths

�(4

/12

)�

(4/1

2,

Ce)

*M

arcu

s&

Wat

t(1

97

1)

Rat

sD

egra

ded

carr

agee

nan

fro

mE

.sp

ino

sum

(Gla

xo

).S

O3

3%

0.2

5%

Dri

nk

ing

wat

er3

–1

2w

eek

s�

�A

bra

ham

etal

.(1

97

4)

and

Gra

sso

etal

.(1

97

5)

0.5

%�

�5

%O

edem

a(R

e)�

Rat

sD

egra

ded

i-ca

rrag

een

an(G

laxo

)S

O3

0%

;M

w:

20

00

0–

40

00

0

Hir

on

oet

al.

(19

81

)

Ger

m-f

ree

10

%D

iet

7–

63

(sq

.ep

ith

eliu

m)þþ

Co

nven

tio

nal

�þ

Rat

sD

egra

ded

i-ca

rrag

een

anfr

om

E.

spin

osu

m(G

laxo

).S

O4

28

%;

Mw

:2

00

00

–4

50

00

1%

6,

12

,1

8,

24

mo

nth

s�

�Is

hio

ka

etal

.(1

98

6)

5%

Die

(sq

.m

etap

lasi

a)þþ

(Re,

dC

)

10

(sq

.m

etap

lasi

a)þþ

(Re,

dC

)

Rat

sD

egra

ded

i-ca

rrag

een

anfr

om

E.

spin

osu

m(G

laxo

).S

O4

28

%;

Mw

:2

00

00

–4

50

00

5%

Dri

nk

ing

wat

er1

5m

on

thsþ

(sq

.m

etap

lasi

a)þþ

(Re,

dC

)Is

hio

ka

etal

.(1

98

6)

Rab

bit

sl-

deg

rad

edca

rrag

een

an0

.5%

Dri

nk

ing

wat

erL

on

gte

rmO

edem

aþþ

Incr

ease

inB

act

erio

des

fra

geli

sK

itan

o(1

97

9)

and

Kit

ano

etal

.(1

98

1)

Mw

:3

00

00

Tp

then

0.5

Mo

nkey

s(M

aca

cam

ula

tta

)D

egra

ded

i-ca

rrag

een

anfr

om

E.

spin

osu

m(C

16

,G

laxo

)S

O3

3%

;M

w:

ca.

25

00

0

0.5

%(c

a.0

.7g

kg�

1)

14

wee

ks

�A

bra

ham

etal

.(1

97

2)

and

Ben

itz

etal

.(1

97

3)

1%

(ca.

1.4

gk

g�

1)

Dri

nk

ing

wat

er1

4w

eek

2%

(ca.

2.9

gk

g�

1)

7–

24

wee

ks

þþ

then

0%

�M

on

key

s(M

aca

cam

ula

tta

)D

egra

ded

i-ca

rrag

een

anfr

om

E.

spin

osu

mC

16

(Gla

xo

)S

O3

3%

;M

w:

ca.

25

00

0

2%

Dri

nk

ing

wat

er1

0w

eek

s�

Ab

rah

amet

al.

(19

74

)

Man

Deg

rad

edca

rrag

een

an(E

bim

ar);

Mw

:1

80

00

–2

20

00

5g

d�

1D

rin

kin

gso

luti

on

6–

24

mo

nth

s�

Bo

nfi

ls(1

97

0)

10

gd�

1�

Tab

leuse

dw

ith

per

mis

sio

no

fM

ichel

&M

acfa

rlan

e(1

996).

*N

um

ber

of

anim

als

wit

hu

lcer

atio

ns/

tota

ln

um

ber

of

anim

als.

**

An

tim

icro

bia

lag

ent

trea

tmen

tw

asst

arte

d3

db

efo

reca

rrag

een

an.

**

*A

nim

als

bo

rnfr

om

pre

gn

ant

anim

als

alre

ady

adap

ted

tod

egra

ded

carr

agee

nan

,5

%in

dri

nk

ing

wat

er.

Ce,

Cae

cum

;C

o,

Co

lon

;D

C,

deg

rad

edca

rrag

een

ans;

dC

,d

ista

lco

lon

;R

e,re

ctu

m;

SA

SP,

sali

cyla

zosu

lph

apy

rid

ine;

SM

TM

,su

lph

amet

hoxaz

ole

trim

etho

pri

m;

Tp

,th

iam

ph

enic

ol.

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 13

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 14: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

important in light of the known ulcerative effects and

gastrointestinal carcinogenicity of poligeenan (IARC, 1983)

and to show the different responses of the GI tract to

poligeenan versus CGN.

Table 2 uses the term ‘‘degraded CGN’’ for poligeenan

and shows the large number of studies reporting GI tract

ulceration with this material. As can be seen, GI tract

ulceration was reported in guinea pigs, rats, rabbits, and

monkeys, but not humans, exposed to poligeenan. Grasso

et al. (1973) report that six patients suffering from malignant

disease of the colon were given 5 g of poligeenan (called

‘‘degraded CGN’’ in the paper) daily for 10 d. Sampling of

the colon found no evidence of ulceration due to poligeenan.

Similarly, a report on 200 patients receiving a preparation of

poligeenan (Ebimar) to treat peptic ulcer in France at a dose

of 5 g/d for 6 months–2 years found no signs of ulcerative

colitis by routine monitoring and radiological examinations

(Bonfils, 1970). These reports (Bonfils, 1970; Grasso et al.,

1973) demonstrate that humans receiving 5 g/d poligeenan

orally did not develop the significant intestinal ulceration

reported in laboratory animals, particularly the guinea pig,

given poligeenan.

The ulcerogenic potential of CGN has been studied in

several species, as summarized in Table 3. The weight of the

evidence indicates that CGN does not produce intestinal

ulceration in rats, hamsters, primates, and pigs unless fed to

animals at non-physiologic levels in diet (415%). Exposure to

very high, non-physiological levels (415%) of CGN in the

diet produced ulcerative effects (Mankes, 1977; Watanabe

et al., 1978). Conflicting results have been reported in guinea

pigs (Engster & Abraham, 1976; Grasso et al., 1973; Watt &

Marcus, 1969), but, the guinea pig seems to be the most

sensitive species responding to poligeenan. Negative studies

for intestinal effects have also been reported for the hamster

(Rustia et al., 1980) and pig (Poulsen, 1973). Long-term

ingestion of CGN in the drinking water or diet has not been

shown to produce GI ulceration in adult or infant primates

(Benitz et al., 1973; McGill et al., 1977). The results for each

species are summarized and discussed in more detail in

Table 3. These findings are important in light of the known

ulcerative effects and gastrointestinal carcinogenicity of

poligeenan (IARC, 1983).

Mice

Administration of �-CGN (Sigma, stated to be pure

‘‘undegraded’’ CGN that is not food grade) to mice

(5/group) for 10 weeks at a concentration of 1% or 4% in

the drinking water was reported to induce inflammatory

responses in the colon (Donnelly et al., 2004a). Inflammation

was evaluated histologically and scored as inflammatory

score, hyperplasia score and crypt loss score. All three scores

were greater in treated animals than controls, with more

severe effects noted at 4% compared to 1% �-CGN in

drinking water. The composition of the �-CGN utilized for

these studies had an extremely high viscosity, in the range

700–800 cps, compared with the compendial (including

regulatory) viscosity specification of a minimum of 5 cps

(equivalent to �100–150 kDa). Most commercial CGNs are

Table 3. Intestinal effects of carrageenan in different species.

Species VehicleCarrageenan

dose(s)Duration:

weeksHistopathological effects:

ulceration Reference

Mouse Drinking water* 1 and 4% 10 Mild inflammation at 1%,severe inflammation at 4%

Donnelly (2004a)

Rat Processed skimmedmilk

4% 24 No microscopic changes Tomarelli et al. (1974)

Diety 2.5 and 5% 13 No microscopic effects Weiner et al. (2007)Diet 5% 104 No microscopic effects Rustia et al. (1980)Dietz 15% 40 Chronic inflammatory changes

in the large intestinesWatanabe et al. (1978)

Dietz 25% 4 Pinpoint intestinal lesions[unpublished thesis]

Mankes (1977)

Diet 1 and 5% 13 and 40 No microscopic changes Abraham et al. (1985)Guinea pig Drinking waterx 1% 2 No microscopic effects Engster & Abraham (1976)

Dietx 2% 10 No microscopic effects Engster & Abraham (1976)Diet 5% 3–7 Cecal and colonic ulcerations –

no actual data providedGrasso et al. (1973)

Hamster Diet 5% 104 No microscopic effects Rustia et al. (1980)Pig Jelly in diet 500 mg/kg/d 12 No microscopic effects Poulsen (1973)Rhesus monkey Drinking water 1% 11 No microscopic effects Benitz et al. (1973)

Drinking water 1250 mg/kg/d 8 No microscopic effects Benitz et al. (1973)Baboon Infant formula 255 and 1220 mg/L

(analyzed)16 No microscopic effects McGill et al. (1977)

CGN had a Mw4145 kDa.*The study used �-CGN from Sigma Chemical Company (not food grade CGN).yCarrageenan sample used in this study was specifically altered to contain a low molecular weight tail (LMT) of 7% (mean) carrageenan to maximize

any potential effects of the lower molecular weight fraction on the gastrointestinal tract.zDoses greater than 5% test material in the diet are considered not to be nutritionally adequate because so high a percentage of the diet is replaced by an

inert substance without nutrient or caloric value. At levels of test material in the diet45%, animals will eat more food to compensate for the lowercaloric/nutritional intake. Inadequately controlled dietary variables may result in nutritional imbalances or caloric deprivation that couldconfound interpretation of the toxicity study results (FDA, 2000). The FDA suggests that if doses in dietary studies exceed 5% in the diet,additional controls may be needed (FDA, 2000) http://www.fda.gov/Food/GuidanceComplianceRegulatoryInformation/GuidanceDocuments/FoodIngredientsandPackaging/Redbook/ucm078345.htm.xCGN had a Mw445 kDa.

14 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 15: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

normally within the Mw range 200–800 kDa, with a typical

range of 400–600 kDa (Blakemore & Harpell, 2010; Uno

et al., 2001b). Under the conditions of the study, administra-

tion of �-CGN at 1% in water would have resulted in a very

thick solution and at 4% in water would have been a paste-like

material. These formulations would be expected to influence

the water consumption and water availability to the animals.

Such an effect was observed (depressed water consumption in

treated animals) (Donnelly et al., 2004a). The high concen-

tration in water, abnormal consistency of the dosing solutions,

and the fact that �-CGN exists in the disorganized conform-

ation in water, likely contributed to the induction of colitis

and raise serious concerns regarding the relevance of this

study to human dietary consumption.

Rats

Ulcerative effects in rats have been reported only after

exposure to extremely high, non-physiological concentrations

of CGN in the diet. Watanabe et al. (1978) reported chronic

inflammatory changes in the large intestines of rats that

received 15% k/�-CGN in the diet for 40 weeks. Multiple

pinpoint intestinal lesions were also reported in rats following

dietary exposure to 25% CGN for one month (Mankes, 1977).

This dose level greatly exceeds the recommended limit dose

for dietary studies (OECD, 2009a,b) and can result in

confounding effects due to nutritional deficiencies because

of the large percentage of the diet (25%) containing a non-

nutritive additive. Long-term or subchronic dietary adminis-

tration of CGN at concentrations of 5% or less has not

produced ulcerogenic lesions in the rat (Abraham et al., 1985;

Tomarelli et al., 1974; Rustia et al., 1980; Weiner et al.,

2007). Lifetime administration of k-CGN in the diet at

concentrations of 0, 0.5%, 2.5%, or 5% did not produce

intestinal lesions (Rustia et al., 1980). Similarly, 4% k/�-CGN

in a processed skim milk diet fed to rats for six months did not

produce gross or microscopic changes in the cecum or colon

(Tomarelli et al., 1974). The dose administered was estimated

at 1490 mg/kg/d: 14.9 g food/day with 4% CGN is 596 mg

CGN, divided by a body weight in males of 0.4 kg.

Histopathological evaluation of the GI tract in rats following

90-d of dietary exposure to 5% k-, �-, or E-CGN did not find

any evidence of intestinal ulceration or any other lesions

(Abraham et al., 1985; Weiner et al., 2007). Abraham et al.

(1985) also found no intestinal ulceration in rats fed CGN at

up to 5% in the diet for 40 weeks.

Guinea pigs

Early studies reported intestinal ulceration following admin-

istration of 1% E-CGN (molecular weight not specified) in the

drinking water for 20–30 d (Watt & Marcus, 1969). However,

later studies showed that exposure to high Mw fractions of

CGN (Mw4145 000 Da) did not produce intestinal lesions

when given in the drinking water at a concentration of 1%

E-CGN for two weeks (Engster & Abraham, 1976). In

addition, exposure to 2% E-CGN in the diet for 10 weeks

was not associated with ulcerative effects (Engster &

Abraham, 1976). Intentionally prepared fractions of CGN

(k, �, E) with Mw 21 kDa or 39 kDa did cause cecal ulceration

in the guinea pigs, and minimal erosion was noted at Mw

107 kDa fraction (Engster & Abraham, 1976). Fractions of

E-CGN with Mw of 88 kDa or 4145 kDa did not induce any

histopathological changes to the cecum. Engster & Abraham

(1976) suggest that the low Mw fractions, produced artificially

from CGN with Mws similar to poligeenan, caused cecal

ulceration by uptake of the fraction into the lysosomes of

lamina proprial macrophages, which stimulate release of

lysosomal enzymes (acid phosphatase and b-glucuronidase),

resulting in damage to the nearby tissues. Only the low Mw

fractions similar to poligeenan were able to cause cecal

ulceration and uptake into lamina propria macrophages, as

seen by toluidine-blue staining of the cecum. Interestingly,

neither �- nor k-CGN induced ulceration in guinea pigs given

1% drinking water solutions (Engster & Abraham, 1976).

In contrast, guinea pigs fed 5% E-CGN in the diet for

21–45 d developed multiple cecal and colonic ulcerations

(Grasso et al., 1973). More severe ulceration was reported in

the same study for poligeenan, including photomicrographs of

these lesions. No photomicrographs are provided to verify the

lesions reported after administration of CGN (Grasso et al.,

1973); therefore, this study is considered inconclusive due to

lack of documentation. The guinea pig and rabbit were more

sensitive to intestinal ulcerative effects due to poligeenan

exposure in drinking water than other species examined in

this study, including rat, ferret, squirrel monkey and hamster

(Grasso et al., 1973). The positive response of CGN at a high

dietary concentration in the guinea pig could be due to the

species sensitivity noted, but is not explained further by the

authors (Grasso et al., 1973).

Interestingly, Onderdonk & Bartlett (1979) have shown

that the intestinal ulceration observed with drinking water

administration of poligeenan (5% w/w) to guinea pigs can be

eliminated by using germ-free animals, rather than conven-

tional animals. Germ-free guinea pigs repopulated with

conventional guinea pig microflora developed cecal ulcer-

ations in response to poligeenan. These authors suggest that

bacteria may be a risk factor in the intestinal ulceration seen

with poligeenan (Onderdonk & Bartlett, 1979).

Hamsters

Hamsters fed 0, 0.5%, 2.5%, or 5% k-CGN in the diet for life

(100 weeks) did not exhibit intestinal alterations beyond that

shown by age-matched control animals (Rustia et al., 1980).

Pigs

In another study, Danish Landrace pigs received k-CGN

orally at levels of 0, 50, 200, and 500 mg/kg daily for 12

weeks. No intestinal ulceration was observed (Poulsen, 1973).

Primates

Administration of CGN in the drinking water of primates

has not produced any evidence of intestinal ulceration.

Rhesus monkeys administered 1% k-CGN in the drinking

water for 7–11 weeks showed no intestinal changes (Benitz

et al., 1973). An additional four animals were given CGN

for 7–11 weeks, followed by an 11-week recovery period;

the animals were then given escalating oral doses of

50–1250 mg/kg CGN daily for 12 weeks. The highest dose

of CGN, 1250 mg/kg, was administered daily for 8 of the

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 15

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 16: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

12 weeks. No intestinal ulceration was noted (Benitz et al.,

1973). Infant baboons fed 1220 mg/L k/�-CGN in infant

formula for 16 weeks showed no gastrointestinal lesions

(McGill et al., 1977).

In summary, administration of CGN in the diet to

numerous animal species (rat, mouse, pig, hamster and

monkey) did not induce intestinal ulcerations or any other

lesions when administered at a level up to 5%. Both positive

(Grasso et al, 1973) and negative effects (Engster & Abraham,

1976) on the GI tract were reported for the guinea pig, a more

sensitive species for ulcerative effects, following dietary

administration of CGN. The only report in which intestinal

ulcerations were reported was the Donnelly et al. (2004a)

study. In this study, mice given drinking water containing 4%

CGN with a high viscosity developed lesions of the intestine

which could be explained by confounding effects of dehy-

dration and the use of non-food grade test article. No such

effects were seen in numerous dietary studies or in drinking

water studies in guinea pigs and monkeys. Michel &

Macfarlane (1996) concluded that only poligeenan can

induce colorectal ulceration in animals, which is character-

istic of other sulfated polysaccharides, including amylopectin

sulfate and dextran sulfate. These polymers interact with the

gut mucosa in similar ways; and all possess sulfate,

low polymer size, and anti-peptic activity (Michel &

Macfarlane, 1996).

Carcingenicity studies

Summary

CGN is not carcinogenic based on chronic oncogenicity

studies in animals. CGN has been the subject of numerous

reviews (FDA, 1972c, 1973; Hopkins, 1981; IARC, 1983).

The International Agency for Research on Cancer (IARC)

published a review of CGN in 1983 and concluded that CGN

was not carcinogenic to experimental animals and that no

conclusion could be made regarding carcinogenicity to

humans due to the lack of epidemiological data (IARC, 1983).

k-CGN was found to lack carcinogenicity in chronic

feeding studies in rats and golden hamsters at levels up to 5%

(Rustia et al., 1980). Cohen & Ito (2002) reviewed in detail

the carcinogenicity testing of CGN and concluded that there

was no evidence of carcinogenicity of CGN in animal models.

Likewise, they concluded that there was no evidence of

tumor-promoting activity by CGN in animal models due to

flaws in the reported studies (Arakawa et al., 1986, 1988;

Corpet et al., 1997; Watanabe et al., 1978; Millet et al., 1997)

(see section ‘‘Tumor promotion’’).

Tobacman (2001, 2003) published a series of articles

questioning the interpretation of the carcinogenicity of CGNs

and other safety issues. She raises questions with respect to

carcinogenicity, tumor promotion and inflammatory and

immune effects. Tobacman (2001) often cites work on

poligeenan as if it applied to CGN. Carthew (2002) expressed

concerns with this paper in a letter to the editor. Tobacman

(2001) suggested that there is extensive breakdown of CGN

in vivo to small-molecular-weight products (poligeenan) in

the GI tract, which could be carcinogenic. As the conditions

in the human GI tract are not considered severe enough to

degrade CGN to poligeenan (see the section ‘‘Degradation,

excretion, absorption, and metabolism’’, and McKim, 2014),

these conclusions should be considered speculative. Orally

administered CGN is not digested into low molecular weight

forms; it is not absorbed or distributed systemically and shows

no evidence of toxicological effects at dose levels up to 5% in

the diet (Cohen & Ito, 2002; IARC, 1983; JECFA, 1999,

2008) (see the section ‘‘Degradation, excretion, absorption,

and metabolism’’). No evidence exists to suggest that food

grade CGN causes any carcinogenic effects in vivo.

Genetic effects

Summary

CGN is not genotoxic in in vitro and in vivo studies. Food

grade CGN has not been shown to be genotoxic following

extensive testing. The results of several in vitro mutagenesis

assays have shown that CGN was not mutagenic in the yeast

strain Saccharomyces cervisiae D4 (FDA, 1975), host

mediated assay (Sylianco et al., 1993) or the Ames test

(FDA, 1972a,b, 1975; Jackson, 1997; Mori et al., 1984;

Sylianco et al., 1993). CGN did not induce chromosomal

abnormalities in rat bone marrow cells in vitro (FDA,

1972a,b) or in an in vivo test, the mouse micronucleus

assay (Sylianco et al., 1993). Negative results were also

obtained in the dominant lethal assay in rats (FDA, 1972a,b).

Tumor promotion

Summary

CGN is not considered to be a tumor promoter. There were

some questions raised regarding possible tumor-promoting

activity of CGN in the past (JECFA, 1999). As discussed in

detail by Cohen & Ito (2002), the studies in which this

concern was raised had serious flaws and did not indicate any

evidence of tumor-promoting activity for the GI tract or any

other tissues. The concerns regarding potential tumor pro-

motion of CGN noted by JECFA (1999) were addressed by

Cohen & Ito (2002) and an ‘‘ADI not specified’’ was retained

(JECFA, 2008).

Cohen & Ito (2002) concluded that there clearly was no

evidence of tumor promoting activity when taking all these

studies into consideration (Corpet et al., 1997; Hagiwara et al,

2001; Tache et al., 2000). If one accepts the conclusions of the

investigators (Corpet et al., 1997), however, there is ques-

tionable significance to humans, since there was no tumor

promotion effect when human microflora replaced rat micro-

flora in the GI tract of rats.

More recently, Donnelly et al. (2004a–c, 2005) have

reported a series of experiments on colon crypt cell staining

when N-methyl-N-nitrosourea (MNU), a known mutagen, was

co-administered with �-CGN (stated as not food grade).

These investigators utilized persistent metallothionein over-

expression within single crypts as a biomarker of colonic

crypt stem cell mutations. It should be noted that a biomarker

for specific oncogene mutations in the stem cell fraction of

normal colonic mucosa is not available at present and the

model used does not directly address oncogene mutations

(Donnelly et al., 2004a). Although these investigators

reported an effect on DNA adduct formation and metallothio-

nein crypt-restricted immunopositive staining of colonic

16 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 17: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

crypts in the animals administered MNU alone (62.5 mg/kg

ip), no effects on these endpoints were seen with �-CGN

alone (1% or 4% �-CGN in drinking water for up to 20 weeks)

(Donnelly et al., 2004a, 2005). The addition of �-CGN

together with the mutagen, MNU, enhanced the response

observed with the mutagen alone on the number of aberrant

crypt foci, DNA adduct formation, and the metallothionein

immunopositive staining of colonic crypts (Donnelly et al.,

2004a, 2005).

The composition of the �-CGN utilized for these studies

had a considerably higher viscosity than accepted for food-

grade CGN specifications, the viscosity probably being close

to 700–800 cps per compendia viscosity test. Under the

conditions of the study, administration of this type of CGN

would be expected to form a very thick solution at 1% in water

and a paste-like material at 4% in water. These solutions

would be expected to influence the water consumption and

water availability for the animals. Such an effect was observed

(depressed water consumption in treated animals) (Donnelly

et al., 2004a). The AIN-76A diet used in these studies is

marginal in essential nutrients and the animals may become

stressed by the marginal nutrient content and exposure to

MNU and/or �-CGN. The AIN-76A diet would also cause

much of the water consumed by these animals to be absorbed,

as animals administered this diet tend to have extremely

small, compact, dry feces. The high concentration of

extremely high Mw �-CGN in water and the expected

abnormal consistency of the test solutions, along with a

nutritionally marginal diet, may have contributed to the noted

GI ulceration and raise doubts regarding the relevance of

these studies to humans. In addition, Donnelly et al. (2004a,

2005) do not describe the preparation of MNU or indicate

whether fresh solutions were prepared daily. Fresh MNU is

important because MNU can deteriorate, leading to artifacts

of the first group dosed, compared to the last group dosed on a

given day. The ip injection of MNU would be expected to

produce peritonitis, leading to confounding effects and

responses to �-CGN. The dose of MNU employed is far in

excess of human exposure to nitroso compounds (Donnelly

et al. 2004a, 2005), so the relevance to humans is questionable

based on the MNU dose alone.

Further complicating the interpretation of the results is the

finding of inflammation of the intestinal mucosa, including

ulceration (Donnelly et al., 2004a–c, 2005). As described

above, food-grade CGN in diet does not produce ulceration

(see section ‘‘Ulcerative effects’’; Cohen & Ito, 2002; Weiner

et al., 2007; see Table 3). The reports by Donnelly et al.

(2004a–c, 2005) raise concerns regarding the material that

was administered, as well as the diet employed and other

experimental factors and certainly complicate extrapolation

from the animal model to humans. In the paper in the British

Journal of Cancer (Donnelly et al., 2005), the authors indicate

that there were no statistically significant differences in large

crypts, only in smaller crypts. Other studies have shown that

large aberrant crypt foci are likely the ones that are most

meaningful with respect to the carcinogenic process

(Papanikolaou et al., 2000). Furthermore, there was no

consistent dose–response in the results for total immunopo-

sitive patches/104 crypts with �-CGN and MNU, whether the

CGN was administered for one week, one week repeated three

times, or continuous administration during the entire 20

weeks of the experiment. Donnelly et al. (2004a, 2005)

homogenized the whole intestinal tissue for DNA adduct

evaluation, whereas only a small percentage of the total cells

in the intestine, the epithelium and mucosa, are relevant to GI

tract dietary exposure. Any effect on the mucosal epithelium

would not be observed using the whole intestinal tissue

(Cohen & Ito, 2002). Most of the DNA extracted in their

procedure would have come from the inflammatory cells in

the lamina propria, normally present. Hence, the DNA adduct

formation results are not meaningful. Given all these

difficulties in interpretation, extrapolation to a conclusion of

a tumor-promoting effect of CGN in humans is highly

unlikely and not scientifically supported.

In conclusion, based on a variety of types of assays, there

continues to be no convincing evidence of tumor-promoting

activity of the GI tract for food-grade CGN in animal models.

The FDA (2012) in a response to the Citizen’s Petition by

Dr. Joanne K. Tobacman states that the use of CGN in foods

is safe and that CGN consumed in the diet is not a carcinogen,

tumor promoter or ulcerogenic substance in animal studies.

JECFA (2008) also critically reviewed the publications

on tumor promotion and also concluded that CGN is safe

in foods.

Reproductive and developmental toxicity studies

Summary

CGN is not a developmental or reproductive toxicant in

animal studies. The reproductive effects of CGN were

evaluated in a three-generation reproduction study in rats.

Groups of rats were fed k/�-CGN at concentrations of 0.5%,

1.0%, 2.5%, and 5% prior to mating, throughout pregnancy,

lactation, and weaning for three generations. The only effects

seen were soft stools at the 5.0% and 2.5% level and decreased

body weights for the dams. Decreased birth weight and body

weights at weaning were noted in the offspring of dams fed

1%, 2.5%, and 5%. No effects were seen at the 0.5% level

(Collins et al., 1977a). No effects were seen on reproductive

parameters at any dose level. A teratologic evaluation of the

F2C and F3C generations of the reproduction study showed

no teratological effects due to treatment with CGN (Collins

et al., 1977b).

In a one-generation reproduction study in rats, CGN

caused no effects on physical or behavioral development of

offspring of parental animals fed 0.45% or 0.9% CGN prior to

mating, during gestation, and lactation and after weaning

(Vorhees et al., 1979).

CGN was studied for potential teratogenicity in four

species: mice, rats, rabbits, and hamsters by oral gavage

administration. k/�-CGN (source not specified) was admin-

istered during gestation: mice at 10–900 mg/kg on days 6–15;

rats at 40–600 mg/kg on days 6–15; rabbits at 40–600 mg/kg

on days 6–18, and hamsters at 10–900 mg/kg on days 6–10.

No treatment-related malformations of offspring were

observed in any species at any dose level (FDRL, 1972a,b).

Fetotoxicity was observed in mice at the 900 mg/kg dose by

gavage. Teratology studies were also conducted in rats and

hamsters via dietary administration at 1% and 5% k/�-CGN

during gestation days 6–15 and 6–10, respectively (Bailey &

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 17

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 18: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Morgareidge, 1973). Carrageenan was shown to lack terato-

genic effects in both species at a limit dose of 5% in the diet.

CGN has been tested in a teratology study in a non-

mammalian system. In a protocol not used to determine

teratogenicity in mammals, injection of 0.1% �-CGN into the

yolk-sac of chicken eggs has resulted in increased lethality, a

decrease in hatching weight (Rovasio & Monis, 1980) and

microscopic nervous system abnormalities (Rovasio & Monis,

1987). It is difficult to assess these results due to the lack of

concordance with other mammalian studies discussed above.

The exposure of developing chick embryos to �-CGN injected

directly into eggs in an aqueous milieu is not considered

relevant to human exposure to food grade CGN in the diet.

In the chick embryo system, the developing embryo is

exposed directly to �-CGN; whereas in mammalian studies,

CGN is not absorbed into the systemic circulation and does

not reach the embryo.

In conclusion, CGN is not considered to be a develop-

mental or reproductive toxicant or a teratogen via dietary

administration.

Studies on special age groups

Summary

Existing data on infant baboons demonstrate that CGN is safe

when fed at the maximum feasible levels in formula which

exceed levels used in infant formula. New questions raised by

JECFA (2008) regarding the use of CGN in infant formula are

currently being addressed by FMC Corporation and the

International Formula Council.

Young animals and humans

In the United States, CGN has been used for decades as a

stabilizer in infant formula. The CGN builds structure with

the casein proteins by strong binding, provides the desired

mouth-feel, and stabilizes the fat emulsion. The use level of

CGN recommended and used in casein-based infant formu-

lations for stabilization of the finished food is normally within

the range 200–400 ppm (Glicksman, 1969; Moirano, 1977;

Thomas, 1999), see Table 4.

CGN is permitted in infant formula at a level of

0.03 g/100 ml in regular milk and soy-based liquid infant

formula and at a level of 0.1 g/100 ml in hydrolyzed protein or

amino acid-based liquid infant formula (Canada Gazette,

2004). The higher use level in the hydrolyzed protein and

amino acid-based formula is necessary to adequately stabilize

and suspend the constituents, as discussed above. The higher

use rate infant formula is recommended for infants with

special needs, milk allergies, and medical conditions, as

recommended by the American Academy of Pediatrics

(2000). As noted in Table 4, a concentration of 300 ppm

CGN has the consistency of thick milk and 1000–3000 ppm

CGN has the consistency of custard. Thus, the level of CGN

that can be tested in animal infant formula feeding studies is

limited by the viscosity of high levels of CGN, which can

result in undesirable palatability.

Primates

McGill et al. (1977) fed newborn baboons infant formula with

up to 1220 mg k/�-CGN/liter formula (analyzed concentra-

tion) as their only diet for 112 d. There were no effects on

health, urine or blood parameters or any histopathological

effects on the gut tissues. Ingestion of CGN in infant formulas

by primates did not have any adverse health effects to suggest

altered intestinal permeability or immune responses (McGill

et al., 1977). The McGill et al. (1977) study provides data on

primates which are similar to humans with regard to gut

closure, and are, thus, an appropriate animal model for safety

assessment.

Humans

The potential immunosuppressive effects of CGN in human

infants were studied in a retrospective study reported by

Sherry et al. (1993, 1999). In this study, the frequency of

symptomatic upper respiratory tract infection (URI) was

studied in two populations of full-term infants in the first six

months of life: those exclusively fed infant formula

containing 0.03% k/�-CGN (n¼ 1269), and those exclu-

sively fed powdered (0% CGN) infant formula (n¼ 149).

The results of statistical analysis showed no difference

between frequencies of URIs in the two populations. In

addition, a slightly higher proportion of infants fed CGN

were illness-free for the first six months of life when

compared with infants who received powdered formula

(53.4% versus 47.6%). Thus, these results support the

conclusion that CGN-containing infant formulas do not

produce immunosuppression in infants.

In another study, groups of healthy newborn infants (0–9 d

of age) were fed either powdered (PWD) casein hydrolysate-

based formula (CHF) (95 infants) or liquid (RTF) CHF

formula (100 infants) until 112 d of age (Borsches et al.,

2002). Although the type of formula used in this study is

typically used only by special needs infants, for ethical

purposes, only healthy term infants were included in the

study. The study was a masked, randomized, parallel study

with intake and stool patterns and anthropometric measure-

ments monitored at entry and on days 14, 28, 56, 84, and 112

of the study. The liquid RTF CHF formula contained 0.1 g

k/�-CGN per 100 ml product as a stabilizer; whereas the

PWD CHF formula did not contain CGN (Personal

Communication by Ross Laboratories to author, 2007).

Stabilization of the formula with CGN at this use level is

necessary to keep the ingredients in the liquid formula

homogeneous and stable. On all measures, the infants had

similar growth, tolerance to formula, and stools in treated and

untreated groups. The use of CGN in human infants at a level

of 0.1% in formula has been demonstrated to be safe when

administered for up to 112 d (Borsches et al., 2002).

Table 4. Effect of carrageenan levels on food consistency*.

Carrageenan level in food: ppm Consistency of food

300–500 Thick chocolate milk400–800 Milk shake or eggnog700–1000 Sour cream or creamcheese1000–3000 Flan or custard

*References: Glicksman (1969), Moirano (1977), and Thomas (1999).

18 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 19: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Epidemiology studies

Tobacman (2001, 2002, 2003) published a hypothesis that

CGN is a cause of breast cancer. These studies are based

entirely on a comparison of gross consumption of CGN in the

United States with national breast cancer incidence rates. Their

analysis assumes an average consumption by all individuals in

the United States, an assumption that is incorrect, and has been

discussed at length regarding a variety of alternative methods

for assessing consumption (Munro & Danielewska-Nikiel,

2006). Evidence over the past decade indicates that fibers of

several kinds do not correlate with breast cancer incidence in

women despite previous claims that increased fiber ingestion

protected against breast cancer (Willett et al., 1992).

Furthermore, Tobacman and her colleagues describe a positive

correlation for several other fibers, including agar, alginate,

gum arabic, and locust bean. Three of these (agar, gum arabic,

and locust bean) have been thoroughly evaluated in carcino-

genicity assays by the National Toxicology Program and found

to be negative (Melnick et al., 1983). In summary, there is no

epidemiologic evidence for a carcinogenic, tumor promoting,

or inflammatory effect of CGN in humans.

Regulation of dietary uses

Summary

Exposure to CGN in the diet has been estimated to be below

the current numeric ADI of 0–75 mg/kg/d (SCF, 1978). The

JECFA ADI is ‘‘not specified’’ (JECFA, 2008). Exposure to

CGN in dietary foods is safe.

Adults

Regulatory background

Various regulatory agencies have completed risk assessments

and safety evaluations of the food additive CGN. The Joint

FAO/WHO Expert Committee on Food Additives (JECFA)

reaffirmed an ADI of ‘‘not specified’’ for CGN (JECFA,

2008). However, as discussed below, the ADI does not apply

to infants below 12 weeks of age (JECFA, 2008).

Other agencies have also completed risk assessments on

CGN. The European Scientific Committee on Foods estab-

lished an ADI for CGN at 0–75 mg/kg body weight/d for food

use (SCF, 1978; European Commission, 2003). CGN is

permitted in follow-on formula for infants older than six

months of age.

In general, the United States FDA permits CGN use in

foods based on the amount necessary to achieve functionality

as an emulsifier, a stabilizer, or a thickener in foods. In 1961,

an amendment to title 21 Code of Federal Regulations Part

121 listing CGN as a permitted emulsifier, stabilizer, and

thickener was published in the Federal Register (FR, 1961,

FDA, 1973) (currently listed as Part 172.620). In 1973, FDA

included ‘‘Chondrus extract (carrageenin)’’ (also known as

CGN) for use as a stabilizer to its list of substances Generally

Recognized As Safe (GRAS ID Code 9000-07-1); 21 CFR

Section 182.7255.

Dietary exposure

Data on dietary per-capita intakes in 1995 derived from

poundage data and per capita intakes in 1995 for Europe and

the USA ranged from 28 to 51 mg/d and 30 to 50 mg/d

(JECFA, 2002). The Seaweed Industry Association of

the Philippines reported similar estimates, based on sales:

44 mg/person per day for the populations of Canada and the

USA and 33 mg/person per day for European populations

(JECFA, 2002).

JECFA (2002) considers estimates derived from poundage

data to be consistent with those derived for the population of

the USA in model diets, with reported mean intakes of CGN

of 20 mg/d for consumers and 40 mg/d for consumers at the

90th percentile (derived by multiplying the mean by a factor

of 2). The intakes were derived from data on the food

consumption of individuals aged 2 years and over, available in

1976 from nutrition surveys in the USA, combined with the

results of a 2-week study by the Marketing Research

Corporation of America.

Shah & Huffman (2003) conducted a survey of the use of

CGN in foods at three large supermarket chains and two

major health food markets in the United States. They surveyed

140 North American food manufacturers to obtain recent and

accurate levels of use of CGN in a variety of food products.

They surveyed subjects from ages 417 to 485 years of age

(133 females and 65 males) in South Florida across a wide age

range and socioeconomic backgrounds to determine the use of

foods containing CGN. The results of this survey found that

the mean total CGN intake with standard deviation was

18.09� 17.96 mg/kg bw/d for males and 30.19� 31.61 mg/kg

bw/d for females, with the highest average intake reported in

people under 30 years of age (33.73� 31.61 mg/kg bw/d). An

analysis of CGN intake by body mass found that the highest

consumption on a body weight basis was found in under-

weight individuals with a body mass index of 519 kg/m2.

These individuals consumed 42.19� 25.34 mg/kg body

weight/d (Shah & Huffman, 2003). The consumption or

intake values reported by Shah & Huffman (2003) are higher

than those reported by JECFA (2002) by a factor of the

average body weight. JECFA (2008) has recommended that a

new dietary exposure evaluation be conducted, employing

specific food type and use level information.

The consumption levels of CGN reported in the literature

for the general population are well below the current ADI

in Europe (0–75 mg/kg bw/d) (European Commission, 2003)

based on the current survey information (JECFA, 2002, 2008;

Shah & Huffman, 2003).

Infants

Regulatory background

In Canada, CGN is approved at a maximum level of 0.05% in

formula as a suspension agent for calcium salts in lactose-free

infant formula, based on milk protein. Infant formula based on

isolated amino acids or protein hyrolysates or both may

contain a CGN level of up to 0.1% (Canada Gazette, 2004).

CGN is also used as a stabilizer in infant formula in the

United States. JECFA specifies that the ADI does not apply to

infants 12 weeks of age and younger: ‘‘As a general principle,

the Committee considers that the ADI is not applicable

to infants under the age of 12 weeks, in the absence of

specific data to demonstrate safety to this age group’’

(JECFA, 2008).

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 19

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 20: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

The Codex Committee on Nutrition and Foods for Special

Dietary Uses (CCNFSDU) initiated a program to review and

revise the Codex Standard 72-1981, the standard for infant

formula. The purpose was to evaluate the nutrient levels in

infant formula. The Committee on Nutrition of the European

Society for Pediatric Gastroenterology, Hematology and

Nutrition (ESPGHAN) undertook the task of coordinating

the evaluation at the request of the CCNFSDU. In its

conclusions, the ESPGHAN recommended that CGN be

excluded from infant formula (Koletzko et al., 2005). The

conclusion was based on ‘‘the lack of adequate information on

possible absorption of CGN by the immature gut in the young

infants and its biological effects in infancy.’’ Koletzko et al.

(2005) mistakenly attribute the reported allergic response to a

barium imaging agent reported by Tarlo et al. (1995) to CGN;

whereas the product in this report contained poligeenan (see

section ‘‘Immune system effects’’). This error was never

corrected, but is indicated in the review by JECFA (2008).

In its review of CGN, JECFA (2008) stated that, ‘‘The

European Commission’s Scientific Committee on Food

concluded that it could not be excluded that CGN might be

absorbed by the immature gut and that absorbed material

might affect the immune system in the infant. No new data

have been published addressing this issue.’’ In addition,

JECFA concluded that based on the information available, it

is not advisable to use CGN or processed Eucheuma seaweed

in infant formulas (JECFA, 2008). JECFA considers infor-

mation on the absorption of CGN from neonates to be lacking

and data on the effects of CGN in neonates to be limited.

JECFA (2008) considered the McGill et al. (1977) study and

thought that it should have included special staining proced-

ures, such as toluidine blue stain, for GI tract mast cells,

which can be mobilized by inflammation. McGill et al. (1977)

fed infant baboons (three males and five females per dose

group) formula from birth to 112 d of age at nominal dose

levels of 0, 300, and 1500 mg/L CGN. The high dose was

equivalent to 1220 mg/L CGN analytically, the highest

feasible level without effects on formula palatability or

about 4–5 times use level (300 mg/L). Although the staining

technology and necessity to include mast cell staining were

not used in this study, nevertheless, the study did include

complete histopathology of the GI tract and found no lesions

related to CGN exposure using standard techniques and stains

(hematoxylin & eosin, periodic acid-Schiff, and Prussian blue

stains) and no other treatment-related effects or indications of

inflammation (McGill et al., 1977). This study, McGill et al.

(1977), provides data to assure the safety of CGN use in infant

formula in primates at a level of CGN equivalent to

432 mg/kg bw/d. In response to the JECFA (2008) review,

the International Formula Council, as well as, FMC

Corporation, initiated a research program on the safety of

CGN, using an appropriate physiologic model of neonatal

pigs (Personal Communication, FMC Corporation). Results of

the research program are anticipated in 2014.

Dietary exposure

The average daily exposure to CGN from liquid infant

formulas was estimated to be 47 mg/kg bw per day for milk-

and soy-based formulas (0.03% CGN) and 160 mg/kg bw per

day for hydrolysed protein- and/or amino acid-based liquid

infant formulas (0.1% CGN) (JECFA, 2008). These exposure

estimates apply to infants fed exclusively on formula. The

table below summarizes estimates of infant exposure to CGN

in formula at two different use levels in very young infants

and to 12-month old infants, as well as a comparison to the

baboons evaluated for toxicity by McGill et al. (1977). These

exposure levels agree with those calculated for infants of

different age groups, based on formula consumption and

body weight values reported in the literature (Weiner, 2007)

(Table 5).

The International Programme on Chemical Safety (IPCS,

1987) provides some guidance for the conduct of studies to

support exposure of infants. IPCS indicates that toxico-

logical studies should include animals in the same period of

life, i.e. after birth during the time of nursing; however, they

state that it is difficult to recommend precise toxicological

testing procedures until more basic research has been

undertaken (IPCS, 1987). Thus, the use of adult animal

studies via drinking water or dietary exposure are not

appropriate studies for margin of exposure and risk assess-

ment calculations for the evaluation of the infant formula

use. IPCS (1987) suggested that an expert panel develop

guidelines for testing food additives in infants and neonates.

A more recent publication by IPCS also acknowledges that

there are gaps in the testing protocols for assessment of

developmental toxicity, including the limited exposure to

neonatal animals (IPCS, 2009). Currently, there are no

guidelines for the conduct of studies for constituents of

infant formula.

The infant baboon infant formula feeding by McGill et al.

(1977) is the best available study for supporting the safety of

Table 5. Carrageenan exposure to infants from the infant formula application.

SpeciesCarrageenan informula: ppm

Carrageenan:mg/kg bw/d Method of calculation Reference

Human 300 47 Assumes 100% formula fed infants JECFA (2008)Human 300 6 Assumes 12-month old infants consuming 13.7% of

caloric intake from formulaJECFA (2008)

Human 300 30.4 Assumes 100% formula fed infants, one to sixmonths old, using reported body weights

Sherry et al. (1993, 1999) andWeiner (2007)

Human 1000 160 Assumes 100% formula fed infants JECFA (2008)Human 1000 22 Assumes 12-month old infants consuming 13.7% of

caloric intake from formulaJECFA (2008)

Baboon 1220 432 100% formula fed from birth until 117 d of age, asdescribed by McGill et al. (1977)

McGill et al. (1977) andJECFA (2008)

20 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 21: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

CGN in infant formula. It meets several criteria for relevance

to this age group: baboons were dosed by bottle from birth

through 112 d of age. Levels of CGN were 432 mg/kg bw/d

and were at the limit of feasibility. These levels are considered

to be about five-fold higher than those ingested by human

infants. Higher concentrations of CGN in formula would have

resulted in gelling and would have changed the consistency of

the formula in a way that the baby baboons would not drink it

due to palatability differences. The highest dose tested was

well above expected infant exposure. In addition, the infant

baboon study (McGill et al., 1977) provides additional

support for the safety of CGN use in infant formula because

of the similarity of gut closure timing between non-human

primates and humans.

Conclusions

CGN has been used widely in food as an emulsifier, a

stabilizer, and a thickener for more than five decades. Food

grade CGN meets the specifications established by regulatory

authorities. Food grade CGN is a high Mw polymer (200–

800 kDa for commercial product) that is excreted unchanged

in the feces and is not metabolized or significantly absorbed

from the GI tract. CGN is acutely non-toxic by oral, dermal,

and inhalation routes. The only effects reported in animal

subchronic and chronic feeding studies at high dietary

concentrations (5%) of CGN are loose stools and diarrhea,

effects commonly seen with high levels of non-digestible

dietary fibers. Results of animal studies have shown that CGN

is not genotoxic, carcinogenic, a tumor promoter, or a

developmental or reproductive toxicant. Available studies

evaluating immune system responses have generally produced

inconsistent results because animals were dosed to CGN in

drinking water or by oral gavage in which �-CGN exists in a

random conformation, allowing greater contact with the

intestinal mucosa. Nevertheless, based on a large number of

dietary toxicity studies, CGN has not been shown to cause

adverse effects on the immune system. Such studies support

the safety of CGN in food at current regulatory levels. The

JECFA ADI is ‘‘not specified,’’ indicating a high level of

safety. Human exposure from food has been estimated at

approximately 20–40 mg/kg body weight/day or less. JECFA

identified data gaps about CGN when consumed by infants

0–12 weeks of age. These datagaps are being addressed by a

research program by FMC Corporation and the International

Formula Council. Infant exposure to CGN has been shown to

be safe at levels used in infant formula based on the infant

formula feeding study in baboons and studies in human

infants. Together, these studies indicate that no effects are

expected in human infants consuming formula containing

CGN at levels of 0.03–0.10%.

Acknowledgements

The author wishes to thank Ms Eunice Cuirle, Dr James

M. McKim, and Mr William R. Blakemore, F.R.S.C. (retired)

for their generous time in reviewing this manuscript and for

their helpful comments and valuable insights. Special thanks

to Dr Samuel M. Cohen for sharing his insights on tumor

promotion and CGN. Thanks to Ms Muriel Reva for expert

editorial and administrative assistance.

Declaration of interest

The author of this paper is identified on the cover page.

Myra Weiner is the owner and principal in TOXpertise, LLC,

a consulting firm providing advice on toxicological and risk

assessment issues to private firms. The current review was

prepared for FMC Corporation under a cost reimbursable

contract. FMC Corporation is a manufacturer of CGN and

products containing CGN. The review strategy, the review of

the literature, analyses, and conclusions reported in this paper

are the professional work product of the author. The FMC

Corporation was given the opportunity to review the paper

and offer comments on the paper. Those comments did not

alter the professional opinions of the author. The author has

not appeared in any legal proceedings related to the findings

reported in this paper. The conclusions drawn are not

necessarily those of the FMC Corporation.

References

Abraham R, Benitz KF, Mankes R, Rosenblum I. (1985). Chronic andsubchronic effects of various forms of carrageenan in rats. EcotoxicolEnviron Saf, 10, 173–83.

Abraham R, Fabian RJ, Golberg L, Coulston F. (1974). Role oflysosomes in carrageenan-induced cecal ulceration. Gastroenterology67, 1169–81.

Abraham R, Golberg L, Coulston F. (1972). Uptake and storage ofdegraded carrageenan in lysosomes of reticuloendothelial cells of theRhesus monkey, Macaca Mulatta. Exp Mol Path, 17, 77–93.

Albany Medical College, Institute of Comparative and HumanToxicology. (1975). Unpublished reports.

Albany Medical College, Institute of Comparative and HumanToxicology. (1983). Unpublished reports.

American Academy of Pediatrics. (2000). Hypoallergenic infant for-mulas. Committee on Nutrition. Pediatrics, 106, 346–9.

Arakawa S, Ito M, Tejima S. (1988). Promoter function of carrageenanon development of colonic tumors induced by 1,2-dimethylhydrazinein rats. J Nutr Sci Vitamin, 34, 577–85.

Arakawa S, Okumura M, Yamada S, et al. (1986). Enhancing effectof carrageenan on the induction of rat colonic tumors by 1,2-dimethylhydrazine and its relation to B-glucuronidase activities infeces and other tissues. J Nutr Sci Vitamin, 32, 481–5.

Bailey DE, Morgareidge K. (1973). Teratologic evaluation of carra-geenan salts in rats and hamsters. Food and Drug Research LaboratoryProject No. N-73-02, Waverly, NY, USA (Unpublished).

Bash JA, Cochran FR. (1980). Carrageenan-induced suppression of Tlymphocyte proliferation in the rat: in vitro production of a suppressorfactor by peritoneal macrophages. J Reticuloendothel Soc, 28, 203–11.

Bash JA, Vago JR. (1980). Carrageenan-induced suppression of Tlymphocyte proliferation in the rat: in vivo suppression induced byoral administration. J Reticuloendothel Soc, 28, 213–21.

Benitz KF, Golberg L, Coulston F. (1973). Intestinal effects ofcarrageenans in the Rhesus monkey. Food Cosmet Toxicol, 11,565–75.

Bhattacharyya S, O-Sullivan I, Katyal S, et al. (2012). Exposure to thecommon food additive carrageenan leads to glucose intolerance,insulin resistance and inhibition of insulin signalling in HepG2 cellsand C57BL/6J mice. Diabetologia, 55, 194–203.

Bixler H. (2013). Food safety: refuting myths about carrageenan. FoodProcessing. Available from http://www.foodprocessing.com/articles/2013/refuting-myths-about-carrageenan.html.

Blakemore WR, Harpell AR. (2010). Carrageenan. In: Imeson A, ed.Food Stabilisers, Thickeners and Gelling Agent, Chapter 5. Wiley-Blackwell, Blackwell Publishing Ltd. Published Online, 73–94.

Bonfils S. (1970). Carrageenan and the Human Gut. Lancet, 2, 414.Borsches MW, Barrett-Reis B, Baggs GE, Williams TA. (2002). Growth

of healthy term infants fed a powdered casein hydrolysate-basedformula (CHF). FASEB J, 16, A66 (Abstract 497.7).

Canada Gazette (2004). Vol. 138, No. 20. May 15, Item A.5. Availablefrom: http://www.hc-sc.gc.ca/fn-an/securit/addit/list/4-emulsif-eng.php.

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 21

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 22: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Capron I, Yvon M, Muller G. (1996). In vitro gastric stability ofcarrageenan. Food Hydrocolloids, 10, 239–44.

Carthew P. (2002). Safety of carrageenan in foods. Correspond EnvironHealth Perspect, 110, A176.

Code of Federal Regulations (CFR). Title 21 – Food and Drugs, Part172.620 Carrageenan.

Cohen S, Ito N. (2002). A critical review of the toxicological effects ofcarrageenan and processed eucheuma seaweed on the gastrointestinaltract. Crit Rev Toxicol, 32, 413–44.

Collins TFX, Black TN, Prew JH. (1977a). Long-term effects of calciumcarrageenan in rats. I. Effects on reproduction. Food Cosmet Toxicol,15, 533–8.

Collins TFX, Black TN, Prew JH. (1977b). Long-term effects of calciumcarrageenan in rats. II. Effects on fetal development. Food CosmetToxicol, 15, 539–45.

Commission Directive. (2004). 2004/45/EC of 16 April 2004 amendingDirective 96/77/EC laying down specific purity criteria on foodadditives other than colours and sweetners. Official J Eur Union, 47,20 April 2004, L113/19–L113/21.

Commission Regulation. (2012). 2031/2012 of 9 March, 2012. Official JEur Union L83, 55, L 83/140–1.

Corpet DE, Tache S, Preclaire M. (1997). Carrageenan given as a jelly,does not initiate, but promotes the growth of aberrant crypt foci in therat colon. Cancer Lett, 114, 53–5.

Coste M, Dubuquoy C, Tome D. (1989). Effect of systemic and orallyadministered iota-carrageenan on ovalbumin-specific antibodyresponse in the rat. Int Arch Allergy Appl Immunol, 88, 474–6.

Dewar ET, Maddy ML. (1970). Faecal excretion of degraded and nativecarrageenan by the young rat. J Pharm Pharmacol, 22, 791–3.

Donnelly ET, Bardwell H, Thomas GA, et al. (2004a). Modulation ofN-methyl-N-nitrosourea-induced crypt restricted metallothioneinimmunopositivity in mouse colon by a non-genotoxic diet-relatedchemical. Carcinogenesis, 25, 847–55.

Donnelly ET, Bardwell H, Thomas GA, et al. (2004b). Colonic cryptstem cell mutation indices (CCSCMI) in a predictive risk assessmentmodel for diet related chemicals. Gastroenterology, 126, A505.

Donnelly ET, Bardwell H, Thomas GA, et al. (2004c). Dietaryinteractions and inception of colonic tumourigenesis. Gut, 53, A121.

Donnelly ET, Bardwell H, Thomas GA, et al. (2005). Metallothioeincrypt-restricted immunopositivity indices (MTCRII) correlate withaberrant crypt foci (ACF) in mouse colon. Br J Cancer, 92, 2160–5.

Elsenhans B, Caspary WF. (1989). Differential changes in the urinaryexcretion of two orally administered polyethylene glycol markers(PEG 900 and PEG 4000) in rats after feeding various carbohydrategelling agents. J Nutr, 119, 380–7.

Engster M, Abraham R. (1976). Cecal response to different molecularweights and types of carrageenan in the guinea pig. Toxicol ApplPharmacol, 38, 265–82.

European Commission (2003). Health & Consumer ProtectionDirectorate-General, Opinion of the Scientific Committee on Foodon Carrageenan, SCF/CS/ADD/EMU/199 Final, Available from:http://ec.europa.eu/food/fs/sc/scf/out164_en.pdf.

Federal Register (FR). (1961). 26FR 9711.Food and Agriculture Organization of the United Nations. (2007).

Combined Compendium of Food Additive Specifications; CGN – 68thsession of JECFA, 2007.

Food and Drug Administration (FDA). (1972a). Mutagenic evaluation ofcompound FDA 71-3, sodium carrageenan. PB 254 471.

Food and Drug Administration (FDA). (1972b). Study of mutageniceffects of calcium carrageenan (FDA No. 71-5). PB 221 820.

Food and Drug Administration. (1972c). Generally Recognized As Safe(GRAS) Food Ingredients – Carrageenan. PB 221 206.

Food and Drug Administration (FDA). (1973). Evaluation of the healthaspects of carrageenan as a food ingredient. PB 266 877.

Food and Drug Administration (FDA). (1975). Mutagenic evaluationof compounds FDA 71-3, 977007-70-7, Sodium Carrageenan(Viscarin) PB 254 515.

Food and Drug Administration (FDA). (2000). Toxicological principlesfor the safety assessment of food ingredients: Redbook. Availablefrom: http://www.fda.gov/food/guidanceregulation/guidancedocumentsregulatoryinformation/ingredientsadditivesgraspackaging/ucm2006826.htm.

Food and Drug Administration (FDA). (2012). Letter to Dr. Joanne K.Tobacman, Docket FDA-2008-8P-0347, dated June 11, 2012, fromTed Eklin, Acting Deputy Director for Operations, Center for FoodSafety and Applied Nutrition.

Food and Drug Research Laboratories, Inc. (FDRL). (1972a).Teratologic Evaluation of FDA 71-3 (Sodium Carrageenate).Prepared for the Food and Drug Administration, PB 221 812.

Food and Drug Research Laboratories, Inc. (FDRL). (1972b).Teratologic Evaluation of FDA 71-5 (Calcium Carrageenate).Prepared for the Food and Drug Administration, PB 221 787.

Food Chemicals Codex. (2013). FCC monographs, 8th ed., 219–28.Friedman I, Douglas CD. (1960). Unpublished summary report

submitted by Marine Colloids, Inc.Frossard CP, Hauser C, Eigenmann PA. (2001). Oral carrageenan

induces antigen-dependent oral tolerance: prevention of Anaphylaxisand induction of lymphocyte anergy in a murine model of foodallergy. Pediatr Res, 49, 417–22.

Glicksman M. (ed.) (1969). Seaweed extracts. In: Gum Technology inthe Food Industry, Chapter 8, Academic Press, 214–235.

Grasso P, Gangolli SD, Butterworth KR, Wright MG. (1975). Studies ondegraded carrageenan in rats and guinea-pigs. Food Cosmet Toxicol,11, 195–201.

Grasso P, Sharratt M, Carpanini FMB, Gangolli SD. (1973). Studies oncarrageenan and large-bowel ulceration in mammals. Food CosmetToxicol, 11, 555–64.

Hagiwara A, Miyashia K, Nakanishi T, et al. (2001). Lack of tumorpromoting effects of carrageenan on1,2-dimethylhydrazine-inducedcolorectal carcinogenesis in male F344 rats. J Toxicol Pathol, 14,37–43.

Harmuth-Hoene AE, Schelenz R. (1980). Effect of dietary fiber onmineral absorption in growing rats. J Nutr, 110, 1774–84.

Hawkins WW, Yaphe WW. (1965). Carrageenan as a dietary constituentof the rat: faecal excretion, nitrogen absorption and growth. Can JBiochem, 43, 479–84.

Hirono I, Sumi Y, Kuhara K, Miyakawa M. (1981). Effect ofdegraded carrageenan on the intestine in germfree rats. ToxicolLett, 8, 207–12.

Hopkins J. (1981). Carcinogenicity of carrageenan. Food CosmetToxicol, 19, 779–88.

International Agency for Research on Cancer (IARC) Monograph.(1983). Carrageenan, 31, 79–94.

International Programme on Chemical Safety in cooperation with theJoint FAO/WHO Expert Committee on Food Additives (JECFA).(1987). Principles for the safety assessment of food additives andcontaminants in food, environmental heath criteria 70. Geneva: WHO.

International Programme on Chemical Safety (IPCS) EnvironmentalHealth Criteria 240. (2009). Principals and methods for the riskassessment of chemicals in food. Available from http://www.who.int/foodsafety/chem/principles/en/index1.html.

Ishioka T, Kuwabara N, Oohashi Y, Wakabayashi K. (1986). Induction ofcolorectal tumors in rats by sulfated polysaccharides. Crit RevToxicol, 17, 215–44.

Jackson LI. (1997). Salmonella mutation test (Ames test) with a semi-refined carrageenan from two sources. Unpublished BIBAR ReportNo. 3160/2/1/97 from BIBRA International, Carshalton, Surrey,United Kingdom. Submitted to WHO by Dr. H.J. Bixler, SeaweedIndustry Association of the Phillipines, Searsport, Maine, UnitedStates.

Japan Food Additives Association (2009). Japan’s Specifications forFood Additives, 8th ed., the Ministry of Health and Welfare, 344.

JECFA: Joint FAO/WHO Expert Committee on Food Additives. (1974).Toxicological evaluation of some food additives including anticakingagents, antimicrobials, antioxidants, emulsifiers and thickeningagents. WHO Food Add Ser, 5, 386–410.

JECFA: Joint FAO/WHO Expert Committee on Food Additives. (1999).Toxicological evaluation of certain food additives, including antic-aking agents, antimicroials, antioxidants, emulsifiers and thickeningagents. WHO Food Addi Ser, 42, 147–69.

JECFA: Joint FAO/WHO Expert Committee on Food Additives (2002).Fifty-seventh report of the Joint FAO/WHO Expert Committee onFood Additives. WHO Technical Report Series 909.

JECFA: Joint FAO/WHO Expert Committee on Food Additives.(2008). Sixty-eighth Report of the Joint FAO/WHO ExpertCommittee on Food Additives; Evaluation of Certain FoodAdditives and Contaminants. WHO Technical Report Series, 947,32–7.

Kasper H, Rabast U, Fassl H, Fehle F. (1979). The effect of dietary fiberon the postprandial serum vitamin A concentration in man. Am J ClinNutr, 32, 1837–49.

22 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 23: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Kitano A. (1979). Study of experimental ulcerative colitis induced byadministration of carrageenan into rabbits. J Osaka Cty Med Cntr, 28,537–55.

Kitano, A, Kobayashi, K, Oshiumi, H, et al. (1981). Studies ofexperimental ulcerative colitis induced by carrageenan in rabbits.Jap J Gastro, 78, 2104–11.

Koletzko B, Baker S, Cleghorn G, et al. (2005). Global standard for thecomposition of infant formula: recommendations of an ESPGHANcoordinated international expert group. J Pediatr Gastroenterol Nutr,41, 584–99.

Koo J, Weaver CM, Neylan MJ. (1993). Solubility of calcium salts andcarrageenan used in infant formulas did not influence calciumabsorption in rats. J Pediatr Gastroenterol Nutr, 17, 298–302.

MacIntosh, FC. (1941). A colorimetric method for the standardization ofheparin preparations. Biochem J, 35, 778–82.

Mallett AK, Rowland IR, Bearne CA, Nicklin S. (1985). Influence ofdietary carrageenans on microbial biotransformation activities in thececum of rodents and on gastrointestinal immune status in the rat.Toxicol Appl Pharmacol, 78, 377–85.

Mallett AK, Wise A, Rowland IR. (1984). Hydrocolloid food additivesand rat caecal microbial enzyme activities. Food Chem Toxicol, 22,415–8.

Mankes R, Abraham R. (1975). Lysosomal dysfunction in colonic,submucosal macrophages of Rhesus monkeys caused by degraded iotacarrageenan (38996). Proc Soc of Exp Biol Med, 150, 166–70.

Mankes RF. (1977). Safety evaluation of Iridaea carrageenans in the ratincluding a study of the mechanisms of liver atrophy [thesis].Unpublished. Albany Medical College.

Marcus SN, Kline TJ, Pothoulakis H, et al. (1990). Role of inflammatorymediators in the pre-ulcerative phase of carrageenan-inducedulcerative disease of the colon. Gastroenterology, 98, A461.

Marcus R, Watt J. (1971). Ulceration in young rats fed degradedcarrageenan. Lancet, 2, 763–6.

Marinalg. (2006). Carrageenan (INS 407) and Processed EuchemaSeaweed (INS 407a): Monograph In Response to Request for Data forthe 68th Meeting of JECFA, Brussels, Belgium, Nov. 2006.Unpublished report.

McGill Jr HC, McMahan CA, Wigodsky HS, Sprinz H. (1977).Carrageenan in formula and infant baboon development.Gastroenterology, 73, 512–7.

McKim JM. (2014). Food additive carrageenan: Part I: a critical reviewof carrageenan in vitro studies, potential pitfalls, and implications forhuman health and safety. Crit Rev Toxicol, in press.

Melnick RL, Huff J, Haseman JK, et al. (1983). Chronic effects of agar,guar gum, gum arabic, locust-bean gum, or tara gum in F344 rats andB6C3F1 mice. Food Chem Toxicol, 21, 305–11.

Michel C, MacFarlane GT. (1996). Digestive fates of soluble polysac-charides from marine macroalgae: involvement of the colonicmicroflora and physiological consequences for the host. J ApplBacteriol, 80, 349–69.

Millet AS, Verhaeghe E, Preclaire M, et al. (1997). Carrageenan K, afood gelling agent, promotes the growth of microadenomae in thecolon in conventional rats but not in gnotoxenic rats with human flora.In: Fifth Toulouse symposium, Munich, 30 April–4 May, 67.

Moirano A. (1977). Sulfated seaweed polysaccharides. In: Graham H, ed.Food colloids. Chapter 8. New York: AVI Publishing Company,347–81.

Mori H, Ohbayashi F, Hirono I, et al. (1984). Absence of genotoxicity ofthe carcinogenic sulfated polysaccharides carrageenan and dextransulfate in mammalian DNA repair and bacterial mutagenicity assays.Nutr Cancer, 6, 92–7.

Munro IC, Danielewska-Nikiel B. (2006). Comparison of estimated dailyintakes of flavouring substances with no-observed-effect levels. FoodChem Toxicol, 44, 758–809.

Nacife VP, Soeiro M, Gomes RN, et al. (2004). Morphologicaland biochemical characterization of macrophages activated bycarrageenan and lipopolysaccharide in vivo. Cell Struct Funct, 29,27–34.

Nicklin S, Baker K, Miller K. (1988). Intestinal uptake of carrageenan:distribution and effects on humoral immune competence. Adv ExpMed Biol, 237, 813–20.

Nicklin S, Miller K. (1984). Effect of orally administered food-gradecarrageenans on antibody-mediated and cell-mediated immunity inthe inbred rat. Food Chem Toxicol, 22, 615–21.

Norris AA, Lewis AJ, Zeitlin IJ. (1981). Inability of degradedcarrageenan fractions to induce inflammatory bowel ulceration inthe guinea-pig. J Pharm Pharmacol, 33, 612–13.

Onderdonk AB, Bartlett JB. (1979). Bacteriological studies of experi-mental ulcerative colitis. Am J Clin Nutr, 32, 258–65.

Organization for Economic Co-operation and Development (OECD).(2009a). Guideline 452: chronic toxicity studies, adopted 7 September2009.

Organization for Economic Co-operation and Development (OECD).(2009b). Guideline 453: chronic toxicity\carcinogenicity studies,adopted 7 September 2009.

Papanikolaou A, Wang QS, Papanikolaou D, et al. (2000). Sequentialand morphological analysis of aberrant crypt foci formation in mice ofdiffering susceptibility to azoxymethane-induced colon carcinogen-esis. Carcinogenesis, 21, 1567–72.

Pittman KA, Golberg L, Coulston F. (1976). Carrageenan: the effect ofmolecular weight and polymer type on its uptake, excretion anddegradation in animals. Food Cosmet Toxicol, 14, 85–93.

Poulsen E. (1973). Short-term peroral toxicity of undegraded carra-geenan in pigs. Food Cosmet Toxicol, 11, 219–77.

Reddy BS, Watanabe K, Sheinfil A. (1980). Effect of dietary wheat bran,alfalfa, pectin and carrageenan on plasma cholesterol and fecal bileacid and neutral sterol excretion in rats. J Nutr, 110, 1247–54.

Riccardi BA, Fahrenbach MJ. (1965). Hypocholesterolemic activity ofmucilaginous polysaccharides in white leghorn cockerels. Fed Prod,24, 263.

Roberts MA, Quemener B. (1999). Measurement of carrageenans infood: challenges, progress, and trends in analysis. Food Sci Technol,70, 169–81.

Roch-Arveiller M, Giroud JP. (1979). Effects biologiques et pharmao-logiques da la carragenine. Path Biol, 27, 615–25.

Rovasio RA, Monis B. (1980). Lethal and teratogenic effects of lambdacarrageenan, a food additive, on the development of the chick embryo.Toxicol Pathol, 8, 14–9.

Rovasio RA, Monis B. (1987). Carrageenan induces anomalies in thechick embryo. A light microscope study. Toxicol Pathol, 15, 444–50.

Rowland IR, Mallett AK. (1986). Dietary fiber and the gut microflora –their effects on toxicity. Toxicol Environ Sci, 12, 125–38.

Rowland IR, Mallett AK, Bearne CA, Farthing MJ. (1986). Enzymeactivities of the hindgut microflora of laboratory animals and man.Xenobiotica, 16, 519–23.

Rustia M, Shubik P, Patil K. (1980). Lifespan carcinogenicity test withnative carrageenan in rats and hamsters. Cancer Lett, 11, 1–10.

Salyers AA, West SEH, Vercellotti J, Wilkins TD. (1977). Fermentationof mucins and plant polysaccharides by anaerobic bacteria from thehuman colon. Appl Environ Microbiol, 34, 529–33.

Scientific Committee for Food on Emulsifiers, Stabilizers, Thickenersand Gelling Agents, 30 November 1978.

Shah ZC, Huffman FG. (2003). Current availability and consumption ofcarrageenan-containing foods. Ecol Food Nutr, 42, 357–71.

Sherry B, Flewelling A, Smith AL. (1993). Carrageenan: an asset ordetriment in infant formula? Am J Clin Nutr, 58, 715.

Sherry B, Flewelling A, Smith AL. (1999). Carrageenan: an asset ordetriment in infant formula? Am J Clin Nutr, 58, 715, 1993. Erratum:Am J Clin Nutr, 69, 1293.

Sylianco CYL, Balboa J, Serrame E, Guantes E. (1993). Non-genotoxicand antigenotoxic activity of PNG carrageenan. Phillip J Sci, 122,139–53.

Tache S, Peiffer G, Millet AS, Corpet DE. (2000). Carrageenan gel andaberrant crypt foci in the colon of conventional and human flora-associated rats. Nutr Cancer, 37, 193–7.

Tarlo SM, Dolovich J, Listgarent C. (1995). Anaphylaxis to carrageenan:a pseudo-latex allergy. J Allergy Clin Immunol, 95, 933–6.

Thomas WR. (1999). In: Imeson A, Thickening and gelling agents forfood. Chapter 3. 2nd ed. Aspen Publishers, 45–59.

Thompson AW. (1978). Carrageenan and the immune response.Biomedicine, 28, 148–52.

Thompson AW, Fowler EF. (1981). Carrageenan: a review of its effectson the immune system. Agents Act, 11, 265–73.

Thompson AW, Fowler EF, Pugh-Humphreys RGP. (1979).Immunopharmacology of the macrophage-toxic agent carrageenan.Int J Immunopharm, 1, 247–61.

Tobacman JK. (2001). Review of harmful gastrointestinal effects ofcarrageenan in animal experiments. Environ Health Perspect, 109,983–94.

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 23

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 24: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Tobacman JK. (2002). Carrageenan in foods: response. Environ HealthPerspect, 110, A176–7.

Tobacman JK. (2003). Toxic considerations related to ingestion ofcarrageenan. Rev Food Nutr Toxicity, 1, 204–29.

Tomarelli RM, Tucker Jr WD, Baumann LM, et al. (1974). Nutritionalquality of processed milk containing carrageenan. J Agri Fd Chem, 22,819–24.

Tsuji RF, Hosino K, Noro Y, et al. (2003). Suppression of allergicreaction by �-carrageenan: toll-like receptor 4/MyD88-dependent and-independent modulation of immunity. Clin Exp Allergy, 33, 249–58.

Tungland BC, Meyer D. (2002). Nondigestible oligo- and polysaccah-rides (dietary fiber): their physiology and role in health and food.Comprehen Rev Food Sci Food Saf, 3, 73–92.

Tye RJ. (1991). Carrageenan as a product ingredient. DCI 28–33.United Stated Adopted Names Council (USAN). (1988). List No. 297

New Names, Poligeenan. Clin Pharmacol Therapeut, 44, 246–8.Uno Y, Omoto T, Goto Y, et al. (2001a). Molecular weight and fecal

excreted quantity of carrageenan administered to rats in blended feed.Jpn J Food Chem (JJFC), 8, 83–93.

Uno Y, Omoto T, Goto Y, et al. (2001b). Molecular weight distribution ofCarrageenans studied by a combined gel permeation/inductivelycoupled plasma (GPC/ICP) method. Food Add Contamin, 18, 763–72.

Van der Waaij, D, Cohen, BJ, Anver, MR. (1974). Mitigation ofexperimental inflammatory bowel disease in guinea-pigs by selectiveelimination of the aerobic gram-negative intestinal microflora.Gastroenterology 67, 460–72.

Vorhees CV, Butcher RE, Brunner RL, Sobotka JJ. (1979). A develop-mental test battery for neurobehavioral toxicity in rats: a preliminary

analysis using monosodium glutamate, calcium carrageenan andhydroxyurea. Toxicol Appl Pharmacol, 50, 267–82.

Watanabe K, Reddy BS, Wong CQ, Weisburger JH. (1978). Effect ofdietary undegraded carrageenan on colon carcinogenesis in F344 ratstreated with azoxymethane or methylnitrosourea. Cancer Res, 38,4427–30.

Watt J, Marcus R. (1969). Ulcerative colitis in the guinea pig caused byseaweed extract. J Pharm Pharmacol, 21, 187s–8s.

Watt J, Marcus R. (1971). Carrageenan-induced ulceration of the largeintestine in the guinea pig. Gut, 12, 164–71.

Watt J, Marcus AJ. (1978). Comparison of the effects of carrageeninsand Danish agar on the colon of guinea-pigs. Proc Nutr Soc, 37, 39A.

Weiner ML. (1988). Intestinal transport of some macromolecules infood. Food Chem Toxicol, 26, 867–80.

Weiner ML. (1991). Toxicological properties of carrageenan. AgentsAct, 32, 46–51.

Weiner ML, Freeman C, McCarty JD, Fletcher MJ. (1989). Acutetoxicity of carrageenan. Fifth International Congress of Toxicology,52, 16–21 July.

Weiner ML. (2007). Response to JECFA on the Exposure of Adults andInfants to Carrageenan in food and infant formula. Unpublished paper,JECFA, 2007.

Weiner, ML, Nuber D, Blakemore WR, et al. (2007). A 90-day dietarystudy of kappa carrageenan with emphasis on the gastrointestinal tract.Food Chem Toxicol, 45, 98–106.

Willett WC, Hunter DJ, Stampfer MJ, et al. (1992). Dietary fat andfiber in relation to risk of breast cancer. J Am Med Assoc, 268,2037–44.

Appendix 1

Comparison of carrageenan and poligeenan.

Aspect Carrageenan Poligeenan

CAS Number 9000-07-1 53973-98-1Manufacturing To preserve functionality as a food ingredient, red

seaweed is processed under alkaline conditions(�pH 9)

To degrade the complex polysaccharide into low-molecular weight pieces, acid hydrolysis occursat pH 0.9–1.3 and high temperatures

Average molecular weight range (Mw) 200 000–800 000 Da 10 000–20 000 DaFunction Primarily stabilizer, thickener, gelling agent, with

functionality at concentrations as low as 0.01% insome systems

Dispersing agent; no gelling properties even atconcentrations as high as 10.0%

Uses Food additive/GRAS, pharmaceutical excipient,personal care products

Medical imaging; NO food uses

Toxicological profile: Not genotoxicNot acutely toxicIARC (1983): not carcinogenic in animals IARC (1983): sufficient evidence of carcinogenicity

in animalsNot teratogenic or embryotoxicNot a reproductive toxicantNot harmful to GI tract Ulceration and irritation to GI tract

US FDA food additive or GRAS Yes NoEuropean Food Safety Authority Yes NoCodex Alimentarius Commission Yes No

24 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 25: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Appendix 2

Identification of carrageenan test materials in cited references.*

Reference Carrageenan type Animal species Comments

Section I: acute studiesWeiner et al. (1989) Iota, food grade Rat Acute testsWeiner (1991) Kappa/lambda food grade Rat, rabbit, rat Oral gavage, dermal expos-

ure, inhalationFDRL (1972a) Not reported Rat, mouse, hamster, rabbit Acute oral

Section II: degradation, excretion, absorption, and metabolismHawkins & Yaphe (1965) Kappa/lambda Rat Diet, 27–148 dDewar & Maddy (1970) Iota Rat Diet, 10 dTomarelli et al. (1974) Kappa/lambda Rat Processed heat sterilized/

diet, 6 monthsPittman et al. (1976) Kappa/lambda, Mw 88–276 kDa

IotaIota, or kappa or lambdaKappa/lambda

RatRatGuinea pigGuinea pigMonkey

Diet, 13 weeksGavage, 9 months diet7–10 weeks drinking water2–3 weeks drinking waterGavage, 11 months

Arakawa et al. (1986) Kappa Rat Diet, 24 weeksArakawa et al. (1988) Not reported Rat Diet, 3 weeksTache et al. (2000) Kappa, Sigma Chemical Co. (Sweden,

Switzerland), Mw of 345 000Rat 0.25 and 2.5% gel in

water for 100 dUno et al. (2001a) Lambda, Mw¼�832 kDa Rat Diet, one dayGrasso et al. (1973) Iota Guinea pig; rat Drinking water, 21–45 d;

diet, 3–7 weeksAlbany Medical College (1975) Like kappa, between iota and kappa Rat Diet, one yearNicklin et al. (1988) Iota, food grade; removed polymer

530 kDaRat Gavage, one time; drinking

water, 184 dNicklin & Miller (1984) Kappa, lambda, iota, food grade Rat Drinking water for 90 dElsenhans & Caspary (1989) Not reported Rat Diet, 4 weeksEngster & Abraham (1976) Kappa, Mw 5–145 kDa

Lambda, Mw 20.8–275 kDaIota, Mw 5–145 kDa

Guinea pigGuinea pigGuinea pig

Drinking water, 2 weeksDrinking water, 2 weeksDrinking water, 2 weeks;

diet for 10 weeksAbraham et al. (1972) Kappa/lambda Mw 800 kDa Rhesus monkey Drinking water, 7–11 weeksMankes & Abraham (1975) Kappa, lambda Monkey Drinking water, 10 weeksAlbany Medical College (1983) Kappa/lambda Monkey Diet, 7.5 yearsFriedman & Douglass (1960) Not reported Rat DietRiccardi & Fahrenbach (1965) Not reported Chicken DietReddy et al. (1980) Kappa/lambda Rat Diet, 33 weeksHarmuth-Hoene & Schelenz (1980) Kappa/lambda Rat Diet, 8–147 dKoo et al. (1993) Kappa/lambda Rat Infant formula, single doseKasper et al. (1979) Not reported Human Oral liquid dietMallett et al. (1984) Iota, Sigma Chemical Co. rat Diet, 4 weeksMallett et al. (1985) Iota, kappa, Sigma Chemical Co. rat Diet, 30 dCapron et al. (1996) Kappa/lambda Not applicable In vitroSalyers et al. (1977) Not reported Anaerobic bacteria

Section III: subchronic and long-term toxicity studiesBenitz et al. (1973) Kappa Monkey Drinking water, 11 weeksMankes (1977) Kappa/lambda Rat, mouse Diet, 4 weeksWeiner et al. (2007) Kappa, Mw 196–257 kDa with a mean

of 7%550 kDaRat Diet, 90 d

Bhattacharyya et al. (2012) Lambda-kappa, Sigma Chemical Co. Mouse Drinking water, 18 d, somefor 15 d more

Abraham et al. (1985) Kappa, lambda, iota, Rat Diet, 13–39 weeksRustia et al. (1980) Kappa, food grade Rat, hamster Diet, life time (90–100

weeks)

Section IV: immune system effectsBash & Vago (1980) Kappa/lambda Rat Gavage, once and drinking

water, 8 weeksBash & Cochran (1980) Kappa/lambda In vitroCoste et al. (1989) Iota Rat Gavage, intraperitonealFrossard et al. (2001) Lambda, Sigma Chemical Co. Mouse GavageTsuji et al. (2003) Lambda, Sigma Chemical Co.,

impurities removedMouse Gavage

(continued )

DOI: 10.3109/10408444.2013.861798 Food additive carrageenan: Part II 25

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.

Page 26: Food additive carrageenan: Part II: A critical review of ...foodsciencematters.com/wp-content/uploads/2015/02/... · w polymer used as a food additive, and poligeenan, also known

Continued

Reference Carrageenan type Animal species Comments

Tarlo et al. (1995) Sodium CGN Human Skin prick

Section V: ulcerative effectsDonnelly et al. (2004a) Lambda, Sigma Chemical Co.

not food grade statedMouse Drinking water, 10 weeks

Watanabe et al. (1978) Kappa/lambda Rat Diet, 40 weeksWatt & Marcus (1969) Iota Guinea pig Drinking water, 10–20 dPoulsen (1973) Kappa Pig Oral jelly 83 dMcGill et al. (1977) Kappa/lambda Infant baboon Infant formula, 112 d

Section VI: carcinogenicity studiesMillet et al. (1997) Kappa Rat Oral-water/gel 8 or 100 dCorpet et al. (1997) Kappa, Sigma Rat Oral-water/gel 8 or 100 d

Section VII: genetic effectsFDA (1975) Kappa/lambda In vitroSylianco et al. (1993) Not reported Mouse In vivoFDA (1972a,b) Kappa/lambda Rat In vivo, in vitroMori et al. (1984) Kappa/lambda In vitroJackson (1997) Semi-refined In vitro

Section VIII: tumor promotionTache et al. (2000) Kappa, Sigma Chemical Co.,

heated in steam at 120 �Cfor 20 min in tap water

Rat 0.25% in water or 2.5% gelfor 100 d

Donnelly et al. (2004a, 2005) Lambda, undegraded, SigmaChemical Co. (UK)

Mouse Drinking water at 1% and4% for 10 weeks (2004a)or 20 weeks (2005)

Section IX: reproductive and developmental toxicity studiesCollins et al. (1977a,b) Kappa/lambda food grade Rat Diet at 0.5–5.0% for 12

weeks before mating,through gestation for 3generations

Vorhees et al. (1979) Not reported Rat Diet at 0.45–1.8%, 2 weeksprior to mating throughgestation, lactation,weaning at 21–90 d ofage

FDRL (1972a,b) Kappa/lambda Rat, mouse, rabbit, hamster Gavage during gestationBailey & Morgareidge (1973) Kappa/lambda Rat, hamster Diet, 1% and 5% during

gestationRovasio & Monis, (1980, 1987) Lambda, food grade Chick embryo Inject in yolk sac, 0.1% in

saline

Section X: studies on special age groupsSherry et al. (1993, 1999) Kappa/lambday Babies Infant formula, 6 monthsBorsches et al. (2002) Kappa/lambday Babies Infant formula, 112 d

*References are cited from the first instance of citation and not repeated.yAlthough the seaweed source and carrageenan types are not specified, the infant formula producers know that kappa/lambda type CGN is used (FMC

Corporation, personal communication).

26 M. L. Weiner Crit Rev Toxicol, Early Online: 1–26

Cri

tical

Rev

iew

s in

Tox

icol

ogy

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

72.7

3.21

6.17

7 on

01/

28/1

4Fo

r pe

rson

al u

se o

nly.


Top Related