Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-16T22:59:08.868Z Has data issue: false hasContentIssue false

Toxicity of acrylamide and evaluation of its exposure in baby foods

Published online by Cambridge University Press:  16 September 2010

Pınar Erkekoğlu
Affiliation:
Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
Terken Baydar*
Affiliation:
Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
*
*Corresponding author: Dr Terken Baydar, email tbaydar@hacettepe.edu.tr
Rights & Permissions [Opens in a new window]

Abstract

Contaminants are a vast subject area of food safety and quality and can be present in our food chain from raw materials to finished products. Acrylamide, an α,β-unsaturated (conjugated) reactive molecule, can be detected as a contaminant in several foodstuffs including baby foods and infant formulas. It is anticipated that children will generally have intakes that are two to three times those of adults when expressed on a body-weight basis. Though exposure to acrylamide is inevitable, it is necessary to protect infant and children from high exposure. The present review focuses on the several adverse health effects of acrylamide including mutagenicity, genotoxicity, carcinogenicity, neurotoxicity and reproductive toxicity, and the possible outcomes of childhood exposure from baby foods and infant formulas.

Type
Review Article
Copyright
Copyright © The Authors 2010

Introduction

Safe and adequate nutrition is crucial for the proper development of children. Other than breast milk, infant formulas and baby foods are the most important part of a baby's diet in the first year of life. Infant formulas, available in powder, liquid-concentrate and ready-to-feed forms, are artificial substitutes mimicking human breast milk. They are based on cows' milk or soya milk, designed for the consumption of infants. The medical community considers infant formulas as nutritionally acceptable for infants under the age of 1 year when breast-feeding is not possible. Infant formulas can be modified based on scientific evidence about the nutrient needs of infants(1, 2). Baby food is any food given to infants, with a soft, liquid texture, or that is chewed, from the age of 6 months to 2 years(2Reference Joeckel and Phillips4). Baby foods are also consumed for 3 years as a part of the child's diet(1, 2). Baby foods include several baby purées made from fruits, vegetables or vegetables plus meat (beef, chicken or fish) sold commercially.

Formulation, handling and storage of baby foods are important for the foods' nutritional quality and physico-chemical properties(Reference Nasirpour, Scher and Desobry3). There are several nutrients that an infant formula must contain according to the Food and Drug Administration through the advice of the American Academy of Pediatrics; it is also necessary to set upper limits for a nutrient in the formula(Reference O'Connor5, Reference Wharton6). On the other hand, infant formulas or baby foods can contain several contaminants that carry potential risk during the first years of life. A contaminant is an impurity; any material of an extraneous nature associated with a chemical, a pharmaceutical preparation, a physiological principle, or an infectious agent. Several contaminants, which can come from different stages of production and storage, may be present in infant formulas and baby foods. Food consumption is an important route of human exposure to contaminants such as acrylamide, pesticides and industrial pollutants(Reference Turconi, Guarcello and Livieri7, Reference Booker8). As acrylamide is an anticipated contaminant of baby foods and infant formulas, and as children generally have acrylamide intakes that are two to three times those of adults when expressed on a body-weight (BW) basis, we will focus on acrylamide and its presence in baby foods and infant formulas in the present review(9).

Properties of acrylamide

Acrylamide (acrylic amide; International Union of Pure and Applied Chemistry (IUPAC) name 2-propenamide), an α,β-unsaturated (conjugated) reactive molecule, has the chemical formula C3H5NO(10, Reference Fohgelberg, Rosén and Hellenäs11). It has a high mobility in soil and groundwater and is biodegradable(Reference Friedman12). It is also used in industry to manufacture polyacrylamide or its co-polymers(Reference Friedman12, Reference Wenzl, De La Calle and Anklam13).

Acrylamide is inadvertently found in starchy foods such as crisps, chips and bread(Reference Miller, Carter and Sipes14). Its discovery in heated foods has received a high level of attention from the public and the medical community. Acrylamide in fried and baked foods is produced by the reaction between asparagines and reducing sugars (fructose, glucose) or reactive carbonyls(Reference Viator and Muth15, Reference Choe and Min16). Coffee drinking and smoking are other major sources apart from the human diet(Reference Schettgen, Weiss and Drexler17, Reference Lantz, Ternité and Wilkens18). Boiling does not produce acrylamide, but baking, frying, deep-frying, over-cooking and microwaving produce large amounts of acrylamide in foodstuffs(Reference Miller, Carter and Sipes14). On the other hand, water is one of the major sources of exposure, as polyacrylamide is used mainly for the purification of drinking water as a flocculating agent(Reference Friedman12, Reference Wenzl, De La Calle and Anklam13).

Acrylamide is classified as ‘probably carcinogenic to humans (group 2A)’ by the International Agency for Research on Cancer. There is inadequate evidence in humans for the carcinogenicity of acrylamide(10).

Biotransformation of acrylamide

Absorption

According to data derived from animal studies, acrylamide is quickly absorbed by the skin and by the mucosa if inhaled. If taken by the oral route, it can diffuse evenly in the body because of its hydrosolubility(Reference Fohgelberg, Rosén and Hellenäs11).

Distribution

Tissue distribution is not significantly affected by dose or route of administration. The highest concentrations are found in erythrocytes. Despite the prominence of neurological effects, acrylamide is not concentrated in nervous system tissues(Reference Dearfield, Douglas and Ehling19). Acrylamide readily crosses the placenta(Reference Edwards20).

Metabolism

In blood, acrylamide has a half-life of approximately 2 h. In tissues, total acrylamide (parent compound and metabolites) exhibits biphasic elimination with an initial half-life of approximately 5 h and a terminal half-life of 8 d. Acrylamide does not accumulate in the body(Reference Miller, Carter and Sipes14, Reference Edwards20, Reference Bergmark, Calleman and He21).

At low doses 50 % of acrylamide is oxidised to a DNA-reactive epoxide, glycidamide, by cytochrome 2E1 (CYP2E1). CYP2E1 polymorphisms in mice can cause different amounts of glycidamide to be formed. Wild-type mice have been found to metabolise 50 % of the administered dose of acrylamide to glycidamide; however, CYP2E1-null mice could not metabolise acrylamide to glycidamide(Reference Jägerstad and Skog22). Glycidamide can be metabolised by epoxide hydrolase or can undergo conjugation with glutathione (GSH)(Reference Friedman12, Reference Bergmark, Calleman and He21, Reference Sumner, Fennell and Moore23Reference Carere25).

Excretion

The major pathway of metabolism for acrylamide is its conjugation with reduced GSH by glutathione S-transferase. Conditions such as malnutrition, oxidative stress and liver disease (alcoholic hepatitis, cirrhosis, and other malignant liver disorders) can decrease the GSH content of liver. Since a child's liver cannot carry as high a burden as an adult liver, especially under such conditions, a higher toxicity of acrylamide might be anticipated in children. Elimination occurs mainly in the urine as mercapturic acid conjugates. Greater than 90 % of absorbed acrylamide is excreted in the urine as metabolites. Less than 2 % is excreted as unchanged acrylamide. Smaller amounts are excreted in the bile and faeces. Approximately 60 % of an administered dose appears in the urine within 24 h(Reference Miller, Carter and Sipes14). Biotransformation of acrylamide is summarised in Fig. 1.

Fig. 1 Biotransformation mechanism of acrylamide. CYP2E1, cytochrome 2E1; GST, glutathione S-transferase.

Toxicity of acrylamide

There are several health risks caused by the intake of acrylamide from several routes. Smoking by the mother is a major way by which the neonate is exposed to acrylamide(Reference Hilbig, Freidank and Kersting26). In addition, contaminated foodstuffs and drinking water are two other main ways to expose the baby or child to acrylamide. As children, especially those under the age of 2 years, are the most sensitive population, acrylamide intake levels must be monitored particularly in infant formulas and baby foods. On the other hand, mothers should be attentive while choosing the water with which they prepare food for their child. The health risks of acrylamide can be classified as follows.

Mutagenic, genotoxic and carcinogenic properties of acrylamide

The genotoxic, mutagenic and carcinogenic potentials of acrylamide have been studied extensively. Acrylamide itself reacts rapidly with thiol (–SH) and amino groups; this explains why its primary targets are proteins(Reference Carere25). Acrylamide has been shown to bind DNA by a Michael-type process in vitro with low activity(Reference Bergmark, Calleman and He21, Reference Carere25). Nevertheless, there is sufficient evidence in the literature that both acrylamide and its metabolite glycidamide are mutagenic and clastogenic in mammalian cells(Reference Dearfield, Douglas and Ehling19, Reference Jägerstad and Skog22). Data suggest that mice are more vulnerable to acrylamide tumorigenicity. The metabolic activation of acrylamide is more efficient and the detoxification process is poorer in mice than rats, since mice have higher levels of glycidamide and lower levels of GSH–glycidamide conjugates(Reference Sumner, Selvaraj and Nauhaus27).

Acrylamide causes induction of the following genotoxic effects(Reference Rice28, Reference Hashimoto and Tanii29):

  1. (a) gene mutations and chromosomal aberrations in germ cells of mice in vivo;

  2. (b) chromosomal aberrations in germ cells of rats in vivo;

  3. (c) chromosomal aberrations in somatic cells of rodents in vivo;

  4. (d) gene mutations and chromosomal aberrations in cultured cells in vitro;

  5. (e) cell transformation in mouse cell lines;

  6. (f) somatic mutation in the spot test in vivo;

  7. (g) heritable translocation and specific locus mutations in mice and dominant lethal mutations in both mice and rats;

  8. (h) unscheduled DNA synthesis in rat spermatocytes in vivo; but not in rat hepatocytes. However, glycidamide induces unscheduled DNA synthesis in rat hepatocytes.

The studies on mutagenic, genotoxic and carcinogenic properties of acrylamide and glycidamide are summarised in Table 1(Reference Hashimoto and Tanii29Reference Mei, McDaniel and Dobrovolsky43).

Table 1 Summary of literature on mutagenicity, genotoxicity, carcinogenicity, neurotoxicity, reproductive and developmental toxicity of acrylamide

CHO, Chinese hamster ovary; CYP450, cytochrome 450; ppm, parts per million; GD, gestation day.

Adduct formation with Hb

Acrylamide forms different protein adducts, the most important of which are Hb adducts extensively formed at –SH groups and on the amino groups of the N-terminal valines in erythrocytes. The measurement of Hb adducts can give an integrated measure of the exposure in the previous 3–4 months, since the lifespan of erythrocytes is about 4 months(Reference Carere25, 44).

Adduct formation with acrylamide shows a linear dose–response relationship and glycidamide adducts generate a concave curve. This shows that the percentage of acrylamide metabolised to glycidamide is inversely proportional to the administered dose of acrylamide(Reference Calleman, Stern and Bergmark45, Reference Bergmark, Calleman and Costa46).

Neurotoxic effects of acrylamide

Early morphological studies suggested that neurological defects caused by acrylamide intoxication are mediated through distal axonal degeneration (also known as ‘dying-back’ neuropathy) in the peripheral nervous system and central peripheral nervous system(Reference Höke47, Reference Murray48). Later studies showed that nerve terminals and Purkinje neurons are the targets of acrylamide(Reference Cavanagh49). Nowadays, there are two important hypotheses regarding acrylamide neurotoxicity: (a) inhibition of kinesin-based fast axonal transport; (b) direct inhibition of neurotransmission(Reference LoPachin, Jones and Patterson50, Reference LoPachin51).

Acrylamide, the parent compound, is a soft electrophilic neurotoxicant, reacting on thiol groups of proteins (cysteine, homocysteine) and GSH as well as protein-bound –SH groups (kinesin, dynein), whereas the metabolite glycidamide is a harder electrophilic compound, reacting with nucleophilic centres of adenine and guanine in the DNA(Reference Ma, Wang and Yang37Reference Wolf, Niehaus-Rolf and Banduhn41). It has been shown that acrylamide inhibits the action of brain glutathione S-transferase and reduces the levels of brain GSH(Reference LoPachin, Ross and Reid52). It has also been suggested that acrylamide neurotoxicity is caused by its effects on heavy- and medium-weight neurofilaments, the change it causes on neurotransmitter receptor expression and through inhibition of neurotransmission(Reference Mei, McDaniel and Dobrovolsky43Reference Calleman, Stern and Bergmark45). Electrophilic neurotoxins, including acrylamide, may cause protein structure and function changes by oxidation and this may lead to pathway failure and finally nerve cell damage. Therefore such chemicals at low doses and long-term exposure might be a cause of neurodegenerative diseases such as Alzheimer's disease(Reference Friedman12, Reference Fu, Von Tungeln and Hammons35, Reference LoPachin and DeCaprio53, Reference LoPachin and Gavin54). The studies on neurotoxic effects of acrylamide and glycidamide are summarised in Table 1(Reference Hashimoto and Tanii29, Reference Fu, Von Tungeln and Hammons35, Reference Burek, Albee and Beyer55Reference Ghanayem, Bai and Kissling57).

Reproductive and developmental toxicity of acrylamide

Reproductive toxicity has also been observed in laboratory animals exposed to high levels of acrylamide(Reference Burek, Albee and Beyer55). Several studies have been conducted on reproductive toxicity; these are presented in Table 1(Reference Zenick, Hope and Smith58Reference Tyl, Marr and Myers62).

There seems to be a relationship between the neurotoxicity and reproductive toxicity of acrylamide(Reference Chapin, Fail and George56, Reference Tyl, Marr and Myers62Reference Miller, Mulholland and Vogl65). One theory is that reproductive toxicity is related to neurotoxicity, as neurotoxicity influences mating behaviour. Several studies have clarified that some of the neurotoxic effects of acrylamide in rats are a weakness of the hind-limbs, reduced hind-limb grip strength, and increased foot splay(Reference Tyl, Marr and Myers62, Reference Sakamoto and Hashimoto63, Reference Miller, Mulholland and Vogl65). This reduced hind-limb function could cause impairment in mounting responses, copulatory behaviour, and intromission (entry)(Reference Zenick, Hope and Smith58). Dysfunctional intromission could also have an impact on the proper deposition of sperm in the vagina and uterus and subsequent hormonal events that cause the stimulation of reproductive hormones and implantation. In addition, erectile function could be decreased due to nerve damage in the penis(Reference Tyl and Friedman66). Another theory is that both types of toxicity are mediated through effects on the kinesin motor proteins(Reference Tyl, Marr and Myers62). These kinesin proteins are found in the flagella of sperm as well as the nervous system and other tissues(Reference Miller, Mulholland and Vogl65). Interference with these proteins could reduce sperm motility and fertilisation events(Reference Tyl, Marr and Myers62, Reference Tyl and Friedman66, Reference Friedman, Tyl and Marr67Reference Sickles, Brady and Testino70).

Other mechanisms of acrylamide on reproduction in rodents could be from the alkylation of sulfhydryl groups on unique proteins, such as protamine, in the sperm head and tail(Reference Jägerstad and Skog22, Reference Sega, Alcota and Tancongco71, Reference Sega72). This could affect sperm penetration and induce the pre-implantation losses seen in some dominant lethal studies(Reference Tyl, Marr and Myers62, Reference Lähdetie, Suutari and Sjöblom73).

No human data are present regarding the reproductive and developmental toxicity of acrylamide. However, data are sufficient to conclude that acrylamide is a reproductive and developmental toxicant in rodents.

Dose–response and dose–effect relationships of acrylamide

Numerous investigators have looked at dose–response and dose–effect relationships in a variety of animal models. There do not appear to be significant differences between the mammalian species studied. The lethal dose, 50 % (LD50) levels of acrylamide through various routes in different rodents are given in Table 2(Reference Manson, Brabec and Buelke-Sam74).

Table 2 Lethal dose, 50 % (LD50) levels of acrylamide through different routes in different rodents

The WHO states that acrylamide belongs to the group of chemicals thought to have no reliably identifiable ‘threshold’ of effects, meaning that very low concentrations will also result in very low risks, but not in zero risk: some risk is always present when the chemical is ingested. However, for carcinogens such as acrylamide, risk is thought to increase with increasing exposure(75).

In June 2002, the FAO of the UN and the WHO issued a report about the health implications of acrylamide in food. The Consultation concluded that the no observed adverse effect level (NOAEL) for acrylamide neuropathy is 0·5 mg/kg BW per d and the NOAEL for fertility changes is four times higher than for peripheral neuropathy and 2000-fold greater than estimated dietary exposures(Reference Dybing and Sanner24, Reference Konings, Baars and van Klaveren76, 77). The NOAEL for reproductive toxicity was estimated to be 2–5 mg/kg BW per d depending on the endpoint of fertility or embryonic death(77). No reproductive toxicities have been reported in humans. Therefore, it is highly unlikely that any reproductive toxicity in humans would result from dietary exposure to acrylamide, although concerns about the cumulative effects of low-level chronic exposure are increasing.

Evidence of neurological effects has been observed following single oral doses of 126 mg/kg in rats and rabbits and 100 mg/kg in dogs(Reference McCollister, Oyen and Rowe78, Reference Kuperman79). Using chronic dosing schedules, it has been observed that cumulative oral doses of 500–600 mg/kg using daily doses of 25–50 mg/kg per d are required to produce ataxia in rats, dogs and baboons(Reference McCollister, Oyen and Rowe78, Reference Thomann, Koella and Krinke80, Reference Hopkins81). Smaller daily doses do not produce a clinical effect until a larger cumulative dose is attained. It has been found that the administration of acrylamide at daily doses of 6 to 9 mg/kg does not produce evidence of neurotoxicity in rats until a cumulative dose of 1200 to 1800 mg/kg is attained, and that doses of up to 3 mg/kg per d for 90 d administered to rats do not result in adverse effects(Reference McCollister, Oyen and Rowe78, Reference Fullerton and Barnes82). Spencer et al. (Reference Spencer, Sabri and Schaumburg83) reported that Rhesus monkeys fed up to 2 mg/kg per d did not show any adverse clinical effects at 325 d.

Acrylamide appears to be a multi-organ carcinogen in rodents. This is consistent with its distribution throughout the whole body. However, it is noteworthy that there are no corresponding target organs in mice and rats. Rat thyroid follicular cell tumours and mammary tumours from two studies are considered of possible relevance for human health. Modelling of these data has allowed the determination of benchmark doses and benchmark dose lower confidence limits(Reference Johnson, Gorzinski and Bodner84, Reference Friedman, Dulak and Stedham85). For a 10 % CI, the results were in the range of 300–1100 μg/kg per d for the mammary tumours and between 630 and 930 for the thyroid tumours using the Environmental Protection Agency's dose–response modelling for determination of a point of departure for a risk of 10 % lower exposure dose (LED10)(Reference Parzefall86, Reference Shipp, Lawrence and Gentry87). Relating the combined tumour data to Hb adduct data of acrylamide and glycidamide the doses 440 or 950 μg/kg per d were determined, respectively, which are close to the benchmark dose lower confidence limits(Reference Parzefall86Reference Exon88).

The estimated average chronic human dietary intake was calculated crudely as 0·3–0·7 μg/kg BW per d in 2002. However, the Food and Drug Administration calculated the exact intake as 0·4 μg/kg BW per d in 2003 and this value remained the same when the Food and Drug Administration updated this subject in 2004(89). This increase of acrylamide intake by age seems to reflect the increased consumption of fast food together with potato crisps or French fries.

As the NOAEL for neuropathy is given as 0·5 mg/kg BW per d, a woman weighing 132 lb (60 kg) could safely consume 30 mg acrylamide daily; a man weighing 180 lb (82 kg) could consume about 41 mg(Reference Rosén and Hellenäs90). On the other hand, if the same calculation is applied to the daily intake of a child, a child weighing 20 lb (9 kg) can consume food including 4·5 mg acrylamide. Whether it is appropriate to judge the daily intake of a child with the same calculation is a matter of debate considering that the susceptibility of a child towards several chemicals is much higher than that of an adult. It is anticipated that children will generally have intakes that are two to three times those of adults when expressed on a BW basis.

Studies on acrylamide contamination in baby foods and infant formulas and estimated daily exposures of acrylamide are given in Table 3(Reference Hilbig, Freidank and Kersting26, Reference Alexy, Sichert-Hellert and Kersting9194).

Table 3 The daily intake of acrylamide of children in several European countries

BW, body weight; SCF, Scientific Committee on Food.

Determination of acrylamide and limits in foods

Potentially toxic acrylamide is largely derived from heat-induced reactions between the amino group of the free amino acid asparagine and carbonyl groups of glucose and fructose in cereals, potatoes, and other plant-derived foods(Reference Friedman and Levin95). Several methods have been described that determine acrylamide in several foodstuffs and water. The first reported data on acrylamide in foods available on the market were developed by the Swedish National Food Administration using liquid chromatography/tandem mass spectroscopy (LC/MS/MS)(96). Since then, several analytical procedures have been developed, including LC/MS/MS and GC/MS with different sensitivity, cost, speed and applicability(Reference Rosén and Hellenäs90).

There are a limited number of studies concerning the daily intake of acrylamide from foodstuffs. In the year 2002, the Swedish National Food Agency showed that certain fried, baked and deep-fried foods and coffee had high concentrations of acrylamide(96). In a Norwegian study, acrylamide exposure from foods and coffee was estimated to be 0·49 and 0·46 μg/kg BW in males and females, respectively(97). The New Zealand National Nutrition Survey showed that with a typical Western diet, including hot chips (French fries) and potato crisps, the calculated daily intake of acrylamide is 0·3 μg/kg BW, which is below the NOAEL(98).

On the other hand, as babies and children are a more sensitive population than adults, their consumption of foods including acrylamide must be limited. The studies calculating the daily intake of acrylamide of children in several European countries are summarised in Table 4(Reference Fohgelberg, Rosén and Hellenäs11, Reference Rosén and Hellenäs90, Reference Jiao, Zhang and Ren99103).

Table 4 Studies on acrylamide contamination in baby foods and infant formulas and estimated daily exposures of acrylamide

LC/MS/MS, liquid chromatography/tandem mass spectroscopy.

The European Union gives high importance to acrylamide contamination in food. After the Commission Recommendation 2007/331/EC, twenty-one member states of the European Union presented the results of acrylamide in several foodstuffs (presented under the titles of ‘French fries’, ‘potato crisps’, ‘potato products for home cooking’, ‘bread’, ‘breakfast cereals’, ‘biscuits’, ‘roasted coffee’, ‘jarred baby foods’, ‘processed cereal-based baby foods’ and ‘other products’) to the European Food Safety Authority in 2007. There were 2715 results reported for foods sampled in 2007, with a minimum of seventy-six reported for ‘processed cereal-based baby foods’ and a maximum of 854 reported for ‘other products’. The arithmetic mean acrylamide content ranged from 44 μg/kg for ‘jarred baby foods’ to 628 μg/kg for ‘potato crisps’, with the equivalent geometric means of 31 and 366 μg/kg. The 2007 results were compared with results collected by the European Commission Joint Research Centre's Institute for Reference Materials and Measurements in the years 2003 to 2006. There were 9311 results reported for foods sampled in the 4 years; however, there were only eight results reported for the food category ‘jarred baby foods’ and they were not included in the comparison. The arithmetic mean of acrylamide content ranged from 55 μg/kg for ‘cereal-based baby foods’ to 678 μg/kg for ‘potato crisps’, with the equivalent geometric means of 35 and 514 μg/kg. The highest 95th percentile value was reported for ‘potato crisps’ at 1718 μg/kg and the highest maximum for ‘other products’ at 7834 μg/kg(102).

Conclusion

Infants and children are susceptible towards the effects of several xenobiotics. The knowledge of all factors that affect the formation of glycidamide is important with respect to estimating risk from this known carcinogen. It is known that the metabolism and clearance of drugs are higher in children because of the larger liver:BW ratio and the higher blood flow through the liver compared with those of older adults(Reference Greim and Snyder104). As the major metabolite of acrylamide, glycidamide, is formed through biotransformation, it is likely that glycidamide can be formed at an higher rate than adults in children. In addition, the detoxification pathway through conjugation with GSH may be less functional due to low levels of GSH in a child's liver. Therefore, it is possible that the toxicity of acrylamide might be higher in children.

Children and adolescents should eat a balanced and assorted diet, which includes plenty of fruit and vegetables but also cereals. Also, parents should moderate their children's consumption of fried and fatty foods. Such a diet would also reduce the risk of high exposures to acrylamide, since acrylamide has not been determined in unheated or boiled foods(Reference Tareke, Rydberg and Karlsson105). Acrylamide presumably will also be present in low levels in a nutritionally balanced diet; however, it is in the power of parents to feed their children with more healthy foods. On the other hand, for small children who are merely fed with infant formula and baby food, it is obligatory for governments to take serious measures to monitor contaminants(89).

Taking into account all the knowledge given above, baby food and infant formulas should be routinely tested before use for the detection of acrylamide using improved techniques. Strict law enforcement should be undertaken regarding the detection of all the contaminants in infant formulas and baby foods. Furthermore, producers should be warned by regulatory authorities regarding good manufacturing practices to decrease the levels of contaminants, particularly acrylamide.

Acknowledgements

The present review was not supported by any funding.

P. E. and T. B. are working as faculty staff in the Department of Toxicology, Hacettepe University Faculty of Pharmacy and prepared this review together with equal contribution to the text.

There are no conflicts of interest.

References

1United Nations Treaty Collection (2009) Convention of the Rights of a Child. United Nations Treaty Collection.http://treaties.un.org/Pages/ViewDetails.aspx?src = TREATY&mtdsg_no = IV-11&chapter = 4&lang = en (accessed 25 January 2010).Google Scholar
2Anonymous (2009) Infant formula. http://www.answers.com/topic/infant-formula (accessed 25 January 2010).Google Scholar
3Nasirpour, A, Scher, J & Desobry, S (2006) Baby foods: formulations and interactions (a review). Crit Rev Food Sci Nutr 46, 665681.Google Scholar
4Joeckel, RJ & Phillips, SK (2009) Overview of infant and pediatric formulas. Nutr Clin Pract 24, 356362.CrossRefGoogle ScholarPubMed
5O'Connor, NR (2009) Infant formula. Am Fam Physician 79, 565570.Google ScholarPubMed
6Wharton, BA (1989) An approach to setting maxima in infant formulas. J Nutr 119, 17681772.CrossRefGoogle ScholarPubMed
7Turconi, G, Guarcello, M, Livieri, C, et al. . (2004) Evaluation of xenobiotics in human milk and ingestion by the newborn – an epidemiological survey in Lombardy (Northern Italy). Eur J Nutr 43, 191197.Google Scholar
8Booker, SM (2001) NTP center reports on phthalate concerns. Environ Health Perspect 109, A260A261.CrossRefGoogle ScholarPubMed
9Center for Science in the Public Interest (2003) Petition to establish interim acceptable levels for acrylamide in major food sources, submitted by the Center for Science in the Public Interest, 4 June 2003. http://cspinet.org/new/pdf/acrylamide_petition.pdf (accessed on 30 April 2010).Google Scholar
10International Agency for Research on Cancer (1994) Monographs on the Evaluation of Carcinogenic Risks to Humans: Some Industrial Chemicals no. 60.Lyon, France: IARC.Google Scholar
11Fohgelberg, P, Rosén, J, Hellenäs, KE, et al. . (2005) The acrylamide intake via some common baby food for children in Sweden during their first year of life – an improved method for analysis of acrylamide. Food Chem Toxicol 43, 951959.Google Scholar
12Friedman, M (2003) Chemistry, biochemistry, and safety of acrylamide. A review. Agric Food Chem 51, 45044526.CrossRefGoogle ScholarPubMed
13Wenzl, T, De La Calle, MB & Anklam, E (2003) Analytical methods for the determination of acrylamide in food products: a review. Food Addit Contam 20, 885902.Google Scholar
14Miller, MJ, Carter, DE & Sipes, IG (1982) Pharmacokinetics of acrylamide in Fisher-344 rats. Toxicol Appl Pharmacol 63, 3644.Google Scholar
15Viator, C & Muth, MK (2004) Acrylamide: the Next Food Safety Issue. Choices; 1st Quarter 2004. http://www.choicesmagazine.org/2004-1/2004-1-03.pdf (accessed 25 January 2010).Google Scholar
16Choe, E & Min, DB (2006) Chemistry and reactions of reactive oxygen species in foods. Crit Rev Food Sci Nutr 46, 122.Google Scholar
17Schettgen, T, Weiss, T, Drexler, H, et al. . (2003) A first approach to estimate the internal exposure to acrylamide in smoking and non-smoking adults from Germany. Int J Hyg Environ Health 206, 914.CrossRefGoogle ScholarPubMed
18Lantz, I, Ternité, R, Wilkens, J, et al. . (2006) Studies on acrylamide levels in roasting, storage and brewing of coffee. Mol Nutr Food Res 50, 10391046.CrossRefGoogle ScholarPubMed
19Dearfield, KL, Douglas, GR, Ehling, UH, et al. . (1995) Acrylamide: a review of its genotoxicity and an assessment of heritable genetic risk. Mutat Res 330, 7199.Google Scholar
20Edwards, PM (1976) The insensitivity of the developing rat fetus to the toxic effects of acrylamide. Chem Biol Interact 12, 1318.Google Scholar
21Bergmark, E, Calleman, CJ, He, F, et al. . (1993) Determination of hemoglobin adducts in humans occupationally exposed to acrylamide. Toxicol Appl Pharmacol 120, 4554.Google Scholar
22Jägerstad, M & Skog, K (2005) Genotoxicity of heat-processed foods. Mutat Res 574, 156172.CrossRefGoogle ScholarPubMed
23Sumner, SC, Fennell, TR, Moore, TA, et al. . (1999) Role of cytochrome P450 2E1 in the metabolism of acrylamide and acrylonitrile in mice. Chem Res Toxicol 12, 11101116.Google Scholar
24Dybing, E & Sanner, T (2003) Risk assessment of acrylamide in foods. Toxicol Sci 75, 715.CrossRefGoogle ScholarPubMed
25Carere, A (2006) Genotoxicity and carcinogenicity of acrylamide: a critical review. Ann Ist Super Sanita 42, 144155.Google ScholarPubMed
26Hilbig, A, Freidank, N, Kersting, M, et al. . (2004) Estimation of the dietary intake of acrylamide by German infants, children and adolescents as calculated from dietary records and available data on acrylamide levels in food groups. Int J Hyg Environ Health 207, 463471.Google Scholar
27Sumner, SC, Selvaraj, L, Nauhaus, SK, et al. . (1997) Urinary metabolites from F344 rats and B6C3F1 mice coadministered acrylamide and acrylonitrile for 1 or 5 days. Chem Res Toxicol 10, 11521160.Google Scholar
28Rice, JM (2005) The carcinogenicity of acrylamide. Mutat Res 580, 320.CrossRefGoogle ScholarPubMed
29Hashimoto, K & Tanii, H (1985) Mutagenicity of acrylamide and its analogues in Salmonella typhimurium. Mutat Res 158, 129133.CrossRefGoogle ScholarPubMed
30National Toxicology Program (1989) NTP toxicology and carcinogenesis studies of N-methylolacrylamide (CAS no. 924-42-5) in F344/N rats and B6C3F1 mice (gavage studies). Natl Toxicol Program Tech Rep Ser 352, 1204.Google Scholar
31Russell, B, Hunsicker, PR, Cacheiro, NL, et al. . (1991) Induction of specific-locus mutations in male germ cells of the mouse by acrylamide monomer. Mutat Res 262, 101107.CrossRefGoogle ScholarPubMed
32Hoorn, AJ, Custer, LL, Myhr, BC, et al. . (1993) Detection of chemical mutagens using Muta Mouse: a transgenic mouse model. Mutagenesis 8, 710.Google Scholar
33Dearfield, KL, Abernathy, CO, Ottley, MS, et al. . (1988) Acrylamide: its metabolism, developmental and reproductive effects, genotoxicity, and carcinogenicity. Mutat Res 195, 4577.Google Scholar
34Segerbäck, D, Calleman, CJ, Schroeder, JL, et al. . (1995) Formation of N-7-(2-carbamoyl-2-hydroxyethyl)guanine in DNA of the mouse and the rat following intraperitoneal administration of [14C]acrylamide. Carcinogenesis 16, 11611165.Google Scholar
35Fu, PP, Von Tungeln, LS, Hammons, GJ, et al. . (2000) Metabolic activation capacity of neonatal mice in relation to the neonatal mouse tumorigenicity bioassay. Drug Metab Rev 32, 241266.Google Scholar
36Gamboa da Costa, G, Churchwell, MI, Hamilton, LP, et al. . (2003) DNA adduct formation from acrylamide via conversion to glycidamide in adult and neonatal mice. Chem Res Toxicol 16, 13281337.Google Scholar
37Ma, H, Wang, W, Yang, Y, et al. . (2008) Studies on DNA damage and repair induced by acrylamide in cells from mouse various organs (article in Chinese). Wei Sheng Yan Jiu 37, 612614.Google Scholar
38Cao, J, Liu, Y, Jia, L, et al. . (2008) Curcumin attenuates acrylamide-induced cytotoxicity and genotoxicity in HepG2 cells by ROS scavenging. J Agric Food Chem 56, 1205912063.CrossRefGoogle ScholarPubMed
39Zhang, X, Jiang, L, Geng, C, et al. . (2008) Inhibition of acrylamide genotoxicity in human liver-derived HepG2 cells by the antioxidant hydroxytyrosol. Chem Biol Interact 176, 173178.Google Scholar
40Baum, M, Loeppky, RN, Thielen, S, et al. . (2008) Genotoxicity of glycidamide in comparison to 3-N-nitroso-oxazolidin-2-one. J Agric Food Chem 56, 59895993.CrossRefGoogle ScholarPubMed
41Wolf, T, Niehaus-Rolf, C, Banduhn, N, et al. . (2008) The hen's egg test for micronucleus induction (HET-MN): novel analyses with a series of well-characterized substances support the further evaluation of the test system. Mutat Res 650, 150164.Google Scholar
42Recio, L, Hobbs, C, Caspary, W, et al. . (2010) Dose–response assessment of four genotoxic chemicals in a combined mouse and rat micronucleus (MN) and Comet assay protocol. J Toxicol Sci 35, 149162.Google Scholar
43Mei, N, McDaniel, LP, Dobrovolsky, VN, et al. . (2010) The genotoxicity of acrylamide and glycidamide in big blue rats. Toxicol Sci 15, 412421.CrossRefGoogle Scholar
44National Toxicology Program (2005) NTP-CERHR Monograph on the Potential Human Reproductive and Developmental Effects of Acrylamide. NTP CERHR MON 14, iIII76.Google Scholar
45Calleman, CJ, Stern, LG, Bergmark, E, et al. . (1992) Linear versus nonlinear models for hemoglobin adduct formation by acrylamide and its metabolite glycidamide: implications for risk estimation. Cancer Epidemiol Biomarkers Prev 1, 361368.Google ScholarPubMed
46Bergmark, E, Calleman, CJ & Costa, LG (1991) Formation of hemoglobin adducts of acrylamide and its epoxide metabolite glycidamide in the rat. Toxicol Appl Pharmacol 111, 352363.CrossRefGoogle ScholarPubMed
47Höke, A (2006) Neuroprotection in the peripheral nervous system: rationale for more effective therapies. Arch Neurol 63, 16811685.CrossRefGoogle ScholarPubMed
48Murray, L (1996) Acrylamide. http://www.inchem.org/documents/pims/chemical/pim652.htm (accessed 25 January 2010).Google Scholar
49Cavanagh, JB (1982) The pathokinetics of acrylamide intoxication: a reassessment of the problem. Neuropathol Appl Neurobiol 8, 315336.CrossRefGoogle ScholarPubMed
50LoPachin, RM, Jones, RC, Patterson, TA, et al. . (2003) Application of proteomics to the study of molecular mechanisms in neurotoxicology. Neurotoxicology 24, 761775.Google Scholar
51LoPachin, RM (2004) The changing view of acrylamide neurotoxicity. Neurotoxicology 25, 617630.Google Scholar
52LoPachin, RM, Ross, JF, Reid, ML, et al. . (2002) Neurological evaluation of toxic axonopathies in rats: acrylamide and 2,5-hexanedione. Neurotoxicology 23, 95110.CrossRefGoogle ScholarPubMed
53LoPachin, RM & DeCaprio, AP (2005) Protein adduct formation as a molecular mechanism in neurotoxicity. Toxicol Sci 86, 214225.CrossRefGoogle ScholarPubMed
54LoPachin, RM & Gavin, T (2008) Acrylamide-induced nerve terminal damage: relevance to neurotoxic and neurodegenerative mechanisms. J Agric Food Chem 56, 59946003.Google Scholar
55Burek, JD, Albee, RR, Beyer, JE, et al. . (1980) Subchronic toxicity of acrylamide administered to rats in the drinking water followed by up to 144 days of recovery. J Environ Pathol Toxicol 4, 157182.Google ScholarPubMed
56Chapin, RE, Fail, PA, George, JD, et al. . (1995) The reproductive and neural toxicities of acrylamide and three analogues in Swiss mice, evaluated using the continuous breeding protocol. Fundam Appl Toxicol 27, 924.Google Scholar
57Ghanayem, BI, Bai, R, Kissling, GE, et al. . (2010) Diet-induced obesity in male mice is associated with reduced fertility and potentiation of acrylamide-induced reproductive toxicity. Biol Reprod 82, 96104.Google Scholar
58Zenick, H, Hope, E & Smith, MK (1986) Reproductive toxicity associated with acrylamide treatment in male and female rats. J Toxicol Environ Health 17, 457472.Google Scholar
59Sublet, VH, Zenick, H & Smith, MK (1989) Factors associated with reduced fertility and implantation rates in females mated to acrylamide-treated rats. Toxicology 55, 5367.Google Scholar
60Wise, LD, Gordon, LR, Soper, KA, et al. . (1995) Developmental neurotoxicity evaluation of acrylamide in Sprague–Dawley rats. Neurotoxicol Teratol 17, 189198.Google Scholar
61Field, EA, Price, CJ, Sleet, RB, et al. . (1990) Developmental toxicity evaluation of acrylamide in rats and mice. Fundam Appl Toxicol 14, 502512.CrossRefGoogle ScholarPubMed
62Tyl, RW, Marr, MC, Myers, CB, et al. . (2000) Relationship between acrylamide reproductive and neurotoxicity in male rats. Reprod Toxicol 14, 147157.Google Scholar
63Sakamoto, J & Hashimoto, K (1986) Reproductive toxicity of acrylamide and related compounds in mice – effects on fertility and sperm morphology. Arch Toxicol 59, 201205.CrossRefGoogle ScholarPubMed
64Costa, LG, Deng, H, Gregotti, C, et al. . (1992) Comparative studies on the neuro- and reproductive toxicity of acrylamide and its epoxide metabolite glycidamide in the rat. Neurotoxicology 13, 219224.Google ScholarPubMed
65Miller, MG, Mulholland, DJ & Vogl, AW (1999) Rat testis motor proteins associated with spermatid translocation (dynein) and spermatid flagella (kinesin-II). Biol Reprod 60, 10471056.Google Scholar
66Tyl, RW & Friedman, MA (2003) Effects of acrylamide on rodent reproductive performance. Reprod Toxicol 17, 113.Google Scholar
67Friedman, MA, Tyl, RW, Marr, MC, et al. . (1999) Effects of lactational administration of acrylamide on rat dams and offspring. Reprod Toxicol 13, 511520.Google Scholar
68Shiraishi, Y (1978) Chromosome aberrations induced by monomeric acrylamide in bone marrow and germ cells of mice. Mutat Res 57, 313324.CrossRefGoogle ScholarPubMed
69Adler, ID, Zouh, R & Schmid, E (1993) Perturbation of cell division by acrylamide in vitro and in vivo. Mutat Res 301, 249254.CrossRefGoogle ScholarPubMed
70Sickles, DW, Brady, ST, Testino, A, et al. . (1996) Direct effect of the neurotoxicant acrylamide on kinesin-based microtubule motility. J Neurosci Res 46, 717.Google Scholar
71Sega, GA, Alcota, RP, Tancongco, CP, et al. . (1989) Acrylamide binding to the DNA and protamine of spermiogenic stages in the mouse and its relationship to genetic damage. Mutat Res 216, 221230.CrossRefGoogle Scholar
72Sega, GA (1991) Adducts in sperm protamine and DNA vs. mutation frequency. Prog Clin Biol Res 372, 521530.Google ScholarPubMed
73Lähdetie, J, Suutari, A & Sjöblom, T (1994) The spermatid micronucleus test with the dissection technique detects the germ cell mutagenicity of acrylamide in rat meiotic cells. Mutat Res 309, 255262.CrossRefGoogle ScholarPubMed
74Manson, J, Brabec, MJ, Buelke-Sam, J, et al. . (2005) NTP-CERHR expert panel report on the reproductive and developmental toxicity of acrylamide. Birth Defects Res B Dev Reprod Toxicol 74, 17113.CrossRefGoogle Scholar
75World Health Organization (2010) Food Safety. Questions related to Cancer. http://www.who.int/foodsafety/publications/chem/acrylamide_faqs/en/index1.html (accessed 25 January 2010).Google Scholar
76Konings, EJ, Baars, AJ, van Klaveren, JD, et al. . (2003) Acrylamide exposure from foods of the Dutch population and an assessment of the consequent risks. Food Chem Toxicol 41, 15691579.Google Scholar
77World Health Organization (2002) Health implications of acrylamide in food. Joint FAO/WHO consultation, Geneva, Switzerland, 25–27 June 2002. http://www.who.int/foodsafety/publications/chem/acrylamide_june2002/en/index.html (accessed 25 January 2010).Google Scholar
78McCollister, DD, Oyen, F & Rowe, VK (1964) Toxicology of acrylamide. Toxicol Appl Pharmacol 103, 172181.Google Scholar
79Kuperman, AS (1958) Effects of acrylamide on the central nervous system of the cat. Pharmacol Exp Ther 123, 180192.Google ScholarPubMed
80Thomann, P, Koella, WP, Krinke, G, et al. . (1974) The assessment of peripheral neurotoxicity in dogs: comparative studies with acrylamide and clioquinol. Agent Actions 4, 4753.Google Scholar
81Hopkins, A (1970) The effect of acrylamide on the peripheral nervous system of the baboon. J Neurol Neurosurg Psychiatry 33, 805816.CrossRefGoogle ScholarPubMed
82Fullerton, PM & Barnes, JM (1966) Peripheral neuropathy in rats produced by acrylamide. Br J Ind Med 23, 210221.Google Scholar
83Spencer, PS, Sabri, MI, Schaumburg, HH, et al. . (1979) Does a defect in energy metabolism in the nerve fiber underlie axonal degeneration in polyneuropathies? Ann Neurol 5, 501507.Google Scholar
84Johnson, KA, Gorzinski, SJ, Bodner, KM, et al. . (1986) Chronic toxicity and oncogenicity study on acrylamide incorporated in the drinking water of Fischer 344 rats. Toxicol Appl Pharmacol 85, 154168.CrossRefGoogle Scholar
85Friedman, MA, Dulak, LH & Stedham, MA (1995) A lifetime oncogenicity study in rats with acrylamide. Fundam Appl Toxicol 27, 95105.CrossRefGoogle ScholarPubMed
86Parzefall, W (2008) Review on the toxicity of dietary acrylamide. Food Chem Toxicol 46, 13601364.Google Scholar
87Shipp, A, Lawrence, G, Gentry, R, et al. . (2006) Acrylamide: review of toxicity data and dose–response analyses for cancer and noncancer effects. Crit Rev Toxicol 36, 481608.Google Scholar
88Exon, JH (2006) A review of the toxicology of acrylamide. J Toxicol Environ Health B Crit Rev 9, 397412.Google Scholar
89World Health Organization (2002) FAO/WHO Consultation Report on the Health Implications of Acrylamide in Food, Summary Report. Geneva. 25–27 June 2002. Geneva: WHO.Google Scholar
90Rosén, J & Hellenäs, KE (2002) Analysis of acrylamide in cooked foods by liquid chromatography tandem mass spectrometry. Analyst 127, 880882.CrossRefGoogle ScholarPubMed
91Alexy, U, Sichert-Hellert, W, Kersting, M, et al. . (2001) The foods most consumed by German children and adolescents: results of the DONALD Study. Ann Nutr Metab 45, 128134.Google Scholar
92Wilhelm, M, Schrey, P, Wittsiepe, J, et al. . (2002) Dietary intake of persistent organic pollutants (POPs) by German children using duplicate portion sampling. Int J Hyg Environ Health 204, 359362.Google Scholar
93Scientific Committee of the Norwegian Food Control Authority (2002) Risk assessment of acrylamide intake from cereal-based baby foods. Report from the Scientific Committee of the Norwegian Food Control Authority. http://www.snt.no/nytt/tema/Akrylamid/RA_baby_food.pdf (accessed 30 April 2010).Google Scholar
94European Commission, Health and Consumer Protection Directorate (2003) Acrylamide. EU Summary of Activites. http://europa.eu.int/comm/food/fs/sfp/fcr/acrylamide/study_area2.pdf (accessed 30 April 2010).Google Scholar
95Friedman, M & Levin, CE (2008) Review of methods for the reduction of dietary content and toxicity of acrylamide. J Agric Food Chem 56, 61136140.CrossRefGoogle ScholarPubMed
96Swedish National Food Agency (2002) Press Conference. Uppsala, 21 April 2002. http://www.slv.se/upload/nfa/documents/about_us/org_nfa.pdf.Google Scholar
97Norwegian Food Agency (2002) Risk assessment of acrylamide intake from foods with emphasis on cancer risk. http://www.snt.no (accessed 25 January 2010).Google Scholar
98Food Standards Australia and New Zealand (2009) Acrylamide in food. http://www.foodstandards.gov.au/newsroom/factsheets/factsheets2009/acrylamideinfoodfebr4211.cfm (accessed 25 January 2010).Google Scholar
99Jiao, J, Zhang, Y, Ren, Y, et al. . (2005) Development of a quantitative method for determination of acrylamide in infant powdered milk and baby foods in jars using isotope dilution liquid chromatography/electrospray ionization tandem mass spectrometry. J Chromatogr A 1099, 198202.Google Scholar
100Şenyuva, HZ & Gökmen, V (2005) Survey of acrylamide in Turkish foods by an in-house validated LC-MS method. Food Addit Contam 22, 204209.Google Scholar
101Anonymous (2006) Estimation of the Scientific and Technological Research Council of Turkey for Acrylamide. http://www.kansernotlari.blogspot.com/2006_11_01_archive.html (accessed 25 January 2010).Google Scholar
102European Food Safety Authority (2009) Results on the monitoring of acrylamide levels in food. In The EFSA Scientific Report, no. 285, pp. 126. Parma, Italy: EFSA.Google Scholar
103Bundesamt für Verbraucherschutz und Lebensmittelsicherheit (2003) Acrylamidgehalte in Lebensmitteln. Daten vom 15.01. bis 25.03.2003 (Acrylamide levels in food. Data from 15 January to 25 March 2003). http://www.bvl.bund.de/dl/Acrylamid25.pdf (accessed 30 April 2010).Google Scholar
104Greim, H & Snyder, R (editors) (2008) Toxicology and Risk Assessment. A Comprehensive Introduction. Chichester, West Sussex: John Wiley & Sons.CrossRefGoogle Scholar
105Tareke, E, Rydberg, P, Karlsson, P, et al. . (2002) Analysis of acrylamide, a carcinogen formed in heated foodstuffs. J Agric Food Chem 50, 49985006.Google Scholar
Figure 0

Fig. 1 Biotransformation mechanism of acrylamide. CYP2E1, cytochrome 2E1; GST, glutathione S-transferase.

Figure 1

Table 1 Summary of literature on mutagenicity, genotoxicity, carcinogenicity, neurotoxicity, reproductive and developmental toxicity of acrylamide

Figure 2

Table 2 Lethal dose, 50 % (LD50) levels of acrylamide through different routes in different rodents

Figure 3

Table 3 The daily intake of acrylamide of children in several European countries

Figure 4

Table 4 Studies on acrylamide contamination in baby foods and infant formulas and estimated daily exposures of acrylamide