Meeting

The potential risks from ergot alkaloids in the maternal diet: Discussion paper

TOX/2022/36

Last updated: 11 July 2022

 This is a paper for discussion.

This does not represent the views of the Committee and should not be cited.                                          

Introduction

1.             The Scientific Advisory Committee on Nutrition (SACN) last considered maternal diet and nutrition in relation to offspring health, in its reports on ‘The influence of maternal, fetal and child nutrition on the development of chronic disease in later life’ (SACN, 2011) and on ‘Feeding in the first year of life’ (SACN, 2018). In the latter report, the impact of breastfeeding on maternal health was also considered. In 2019, SACN agreed to conduct a risk assessment on nutrition and maternal health focusing on maternal outcomes during pregnancy, childbirth and up to 24 months after delivery; this would include the effects of chemical contaminants and excess nutrients in the diet.

2.             SACN agreed that, where appropriate, other expert Committees would be consulted and asked to complete relevant risk assessments e.g., in the area of food safety advice. This subject was initially discussed during the horizon scanning item at the January 2020 meeting with a scoping paper being presented to the Committee of Toxicity (COT) in July 2020. This included background information on a provisional list of chemicals proposed by SACN. It was noted that the provisional list of chemicals was subject to change following discussion by COT who would be guiding the toxicological risk assessment process: candidate chemicals or chemical classes can be added or removed as the COT considered appropriate. The list was brought back to the COT with additional information in September 2020. Following a discussion at the COT meeting in September 2020, it was agreed that papers on a number of components should be prioritised. For this paper, the advice of the COT is sought on whether exposure to ergot alkaloids (EAs) would pose a risk to maternal health.

Background

3.             Ergot alkaloids (EA) are secondary metabolites produced by the fungi families Clavicipitaceae and Trichocomaceae, with Claviceps purpurea being the most widespread Claviceps species in Europe. They are known parasites affecting more than 400 plant species, including some economically important cereal grains such as rye, wheat, triticale, barley, millet and oats.

4.             There are 80 different naturally occurring EAs (Schiff, 2006). Based on their occurrence and the available toxicological data the European Food Safety Authority (EFSA) considered six EAs in their risk assessment in 2005, namely: ergotamine, ergocornine, α-ergocryptine, ergosine, ergocristine (peptide ergot alkaloids) and ergometrine (a lysergic acid amide). EFSA further included both forms (-ine and inine) in their assessment, while the -inine forms are considered biologically inactive interconversion occurs under various conditions (EFSA, 2005, Tasker and Wipf, 2021). Bromocriptine is a synthetic ergoline derivate and it is used in the treatment of Parkinson’s disease and pituitary tumours (Herdman et all., 2001).

Toxicity

5.             The latest EFSA opinion (2012), was used as a starting point for the current assessment. Literature searches were conducted using PubMed and Scopus; the search terms are shown in Appendix A.

6.             Due to their structural similarities, EAs are suggested to be agonists or antagonists of noradrenaline, dopamine and serotonin neurotransmitters (Arroyo-Manzanares et al., 2017, Fitzgerald and Dinan, 2008). EAs have been reported to produce direct peripheral effects such as uterotonic action or vasoconstriction, indirect peripheral effects such as serotonin antagonism or adrenergic blockade, and central nervous system (CNS) effects such as induction of hypothermia and emesis (EFSA, 2012).

Toxicokinetics

7.             Data from the literature suggest that EAs are absorbed from the gastrointestinal (GI) tract and subjected to oxidative biotransformation, primarily by cytochrome P450 3A4 via hydroxylation in the liver. Although cytochrome P450 3A4 metabolizes EAs via hydroxylation, EAs have also been shown to bind to it as an inhibitor substrate. Furthermore, cytochrome P450 3A4 is involved in the biosynthetic pathway of EAs in Claviceps purpurea (Haarmann et al., 2006). Recent studies have shown that many EAs are derivatives of lysergic acid (LA) and that P450 monooxygenase and its cluster CloA plays a key role in its biosynthesis. (Gerhards et al., 2014, Haarmann et al., 2006; Mulac and Humpf, 2011).

8.             Following hepatic metabolism, biliary excretion represents the main elimination pathway in primates including humans (Althaus et al., 2008, EFSA, 2012).  However, there is less faecal excretion in cattle, possibly due to a ruminant specific absorption profile. For example, limited information available from experimental studies in cattle showed that approximately 96 % of EAs were excreted in urine (Stuedemann et al., 1998).

9.             No studies are available on the toxicokinetics of dietary EAs in humans. However, human data are available on ergotamine used as a pharmaceutical to treat Parkinson’s disease. Absorption of ergotamine from the GI tract is poor after oral/sublingual administration and bioavailability is further reduced by high pre-systemic hepatic metabolism. Ergotamine tartrate can also be given rectally, to improve absorption, yet bioavailability is still ≤ 5 %. Caffeine can be included in oral and rectal preparations to improve the absorption, the efficacy of this is however still unclear (Tfelt-Hansen et al., 2000; Silberstein and McCrory, 2003).

Acute Toxicity

10.          In vivo studies identified different sensitivities of EAs in various animals, with rabbits being the most sensitive species with LD50 values between 0.9 and 3.2 mg/kg bw (intravenous injection).

11.          The LD50s were determined in a series of experiments by Griffith et al. (1978) on naturally occurring and (semi-) synthetic EAs administered by intravenous (i.v.), subcutaneous (s.c.) and oral exposure (in 2 % gelatine) in mouse, rat and rabbit. Following administration, the animals were observed for 7 days and all the clinical signs, including mortality, were recorded. All naturally occurring EAs demonstrated a low oral acute toxicity compared to the i.v. and oral administrations (Table 1), supporting the observations of low absorption and high pre-systemic metabolism via the oral route. Rabbits were the most sensitive species. Based on the LD50s, EAs exhibit a moderate oral acute toxicity (EFSA, 2012).

Table 1: LD50s in mice, rats and rabbits by i.v. or oral exposure for EAs.

Substance

Species

Route

LD50 (mg/kg)

D-lysergic acid

Mouse

Rabbit

i.v.

i.v.

240

100

Ergometrine

Mouse

Mouse

Rat

Rat

Rabbit

Rabbit

i.v

Oral

i.v.

Oral

i.v.

Oral

160

460

120

671

3.2

27.8

Ergotamine

Mouse

Mouse

Rat

Rat

Rabbit

Rabbit

i.v.

Oral

i.v.

Oral

i.v.

Oral

265

3200

38

1300

3

550

Ergosine

Mouse

Rat

Rabbit

i.v.

i.v.

i.v.

33.5

30

1.23

Ergostine

Mouse

Mouse

Rat

Rat

Rabbit

Rabbit

i.v.

Oral

i.v.

Oral

i.v.

Oral

125

1700

47

>1000

1.2

͂1000

i.v.: intravenous

12.          An acute toxicity study by Griffith et al. (1978) identified ergometine as the least toxic and ergocryptine as the most toxic compound. However, repeat oral dose studies in rats demonstrated no significant differences in the toxicity of ergotamine, ergometrine and α-ergocryptine, with no-observed-adverse effect levels (NOAELs) ranging from 0.22 - 0.60 mg/kg bw per day (EFSA, 2012).  

13.          In humans, acute effects are directly related to receptor antagonism and include diarrhoea, collapse, and vomiting. Exposure to EA contaminated cereal grains, can also lead to a condition called ergotism (Guggisberg, 2003). There are two main types of ergotism, gangrenous and convulsive. The two distinct types of ergotism may been considered as acute and chronic varieties. The gangrenous form is caused by the strong vasoconstrictive properties of some EAs, which result in restriction of blood flow to parts of the body (ischemia). As a result, parathesis (tingling) is felt in fingers and toes followed in many cases by dry gangrene of the limbs and consequently loss of limbs. In the convulsive form, tingling is followed by neurotoxic symptoms such as hallucinations, delirium, and epileptic-type seizures. It has been suggested that a deficiency in vitamin A together with high concentration of EAs could be a causative factor inducing convulsive ergotism (Arroyo-Manzanares et al., 2017; EFSA, 2012). Additional symptoms of ergotism are lethargy or depression.

14.          Limited data are available for individual EAs and their toxic effects on human cells. Most data consist of receptor interaction analysis for single substances in dopamine over-expressing cells or tumour cells. As an example, studies by Larson et al. (1995,1999) indicated that ergocryptine, ergocristine. ergotamine, ergonovine, ergovaline, and ergocornine increased baseline dopamine of about 30 µM. No toxic effects were shown in the studies.

15.          Because EAs act on several neurotransmitter receptors, particularly adrenergic, dopaminergic and serotonergic receptors, EFSA considered neurotoxicity the main acute effect with symptoms such as restlessness, miosis or mydriasis, muscular weakness, tremor and rigidity (see the Section 24 on neurotoxicity).

Chronic toxicity

Humans

16.          No data were available on the chronic toxicity of EAs from dietary exposure in humans. However, limited information was available from the use of ergot containing medications. Case studies on long-term use of EA medication for migraine headaches reported severe lower extremity claudication (pain in the limbs) due to chronic arterial insufficiency (Garcia et al., 2000; Bogun et al., 2011; Fröhlich et al., 2010; Silberstein and McCrory, 2003). In all instances treatment was discontinued and patients were also asked to discontinue the use of caffeine and cigarettes. Anti-platelet therapy was used to successfully reverse the symptoms.

17.          As noted above, ergotamine is used for the treatment of acute migraine. To minimize toxicity and avoid adverse effects such as nausea, vomiting, weakness, muscle pains, paraesthesiae and coldness of the extremities, the dosage is limited to no more than 10 mg per week (Orton and Richardson, 1982; Perrin et al., 1985). Bromocriptine is a synthetic compound with an affinity to dopamine receptors due to its structural similarities to a variety of EAs. It is therefore used as a treatment for Parkinson’s disease and type II diabetes. However, several studies with 790 patients in total, showed adverse side effects in 37 % of severe Parkinson’s patients at dose up to 100 mg/day (Lieberman at al., 1985; Bernard et al., 2015).

Animals

18.          A study by Valente et al. (2020) has shown that EAs are structurally similar to biogenic amines, such as 5-hydroxytryptamine (serotonin), which allows them to interact with serotonin receptors as a response to chronic exposure and act as agonists or antagonists when consumed by bovine.  In contrast, an earlier study by Kalkman et al. (1982) showed vasoconstriction based on adrenergic-like receptors in rats injected intravenously with ergometrine.

19.          A study by Korn et al. (2014) reported spontaneous alopecia, erosions, crusts and necrosis, specifically of the tail area in rabbits from a colony which was originally used in an approved breeding experiment. The lesions were found exclusively in young rabbits aged 113 ± 20 days (14 out of 103 rabbits) fed with hay and a commercial pelleted feed. Immunoassays on blood samples showed mean and maximum EA concentrations of 410 µg/kg and 1,700 µg/kg, respectively. In addition, EAs were detected in the faeces of the affected rabbits at levels up to 200 μg/kg. The mean and maximum dietary intakes of total EAs were 17 and 71 μg/kg bw, respectively. Other toxins, such as fusarium toxin, were also detected in the feed, but at levels which, according to the authors, did not explain the observed effects.

Genotoxicity and Carcinogenicity

20.          Data on the genotoxic and mutagenic effects of EAs are limited and the information available was contradictory. EFSA (2012) considered the available genotoxicity studies to be insufficient, except for ergotamine and concluded that the available data on ergotamine did not indicate any mutagenic potential; though conflicting reports of chromosome damaging effects in vitro. In a review by Uelger et al. (2020) it was noted that ergotamine was not mutagenic in mouse lymphoma cells but sister chromatid exchange had been observed in Chinese hamster ovary cells.

21.          A study by Roberts and Rand (1977) indicated that ergotamine induced chromosomal abnormalities in human lymphocytes and leukocytes. In a further study by Dighe and Vaidya (1988) ergotamine, ergonovine and methylergonovine effectively induced sister chromatid exchange (SCE) frequencies in vitro cultured Chinese hamster ovary (CHO) cells, while ergocristine and α-ergocryptine showed a weak and no effect, respectively.

22.          No genotoxicity or mutagenic effects in the Salmonella typhimurium (St) and mouse lymphoma TK+/- assay were demonstrated in a recent study on ergotamine tartrate (Et) (Seifried et al., 2006). Et was tested at the dose of 10-10000 µg/plate in the St assay; Et was incubated on plates containing St at 5 different concentrations for 48h at 37 C. In the mouse lymphoma TK+/- assay, Et was tested at the concentration of 7.7-108 µg/mL for 4h (Seifried et al., 2006).

23.          EAs are not considered carcinogenic and have not been classified by the International Agency for Research on Cancer (IARC). However, they are being studied as possible cytostatic agents (De Ruyck et al., 2015). Experiments in rodents showed that ergotamine, ergocryptine and ergocornine were able to suppress the growth of pituitary tumours in vivo (MacLeod and Lehmeyer, 1973). More recently a vast range of mRNA microarray studies investigated the cytotoxic activity on a range of human cancer cell types and reported strong inhibitory effects for 1-propylagroclavine and dihydroergocristine against genes associated with the progression of leukaemia. Further information is required to confirm the cytotoxic effect pathway, but preliminary results suggest that EAs have the potential to be used as possible treatment of otherwise drug-resistant and refractory tumours via the inhibition of prolactin release from the anterior pituitary gland (Cassady et al., 1974; Mrusek et at., 2015).

Neurotoxicity

24.          EFSA concluded that EAs induce neurotoxicity in mammals, with symptoms such as restlessness, miosis or mydriasis (contraction and dilation of the pupils), muscular weakness, tremor and rigidity. Repeated dosing with various EAs, resulted in ischaemia, particularly in the extremities (e.g., tails) of rats, decreased body weight gain and changes in the levels of some hormones. Tail gangrene was observed in rats 5 - 7 days after a single i.p. exposure to 25 mg/kg bw ergotoxine (a mixture including ergocornine, α- and β-ergocryptine, and ergocristine) (Griffith et al., 1978). The no-observed-adverse effect levels (NOAELs) was 0.22 - 0.60 mg/kg bw per day. No major quantitative difference in the toxicity of ergotamine, ergometrine and α-ergocryptine was observed (EFSA, 2012).

Reproductive and Developmental toxicity

Animals

25.          Limited information was available on the effects of EAs exposure during pregnancy, in particular the effects on the vascular system supporting the growing fetus. A study by Duckett et al. (2014) examined fetal growth during maternal exposure to ergot alkaloids during gestation. Pregnant ewes (n = 16) were randomly assigned to one of two dietary treatments: (1) endophyte-infected (N. coenophialum) tall fescue seed (E+; 0.8 ug of ergovaline/g diet DM) and (2) endophyte-free tall fescue seed (E−; 0.0 ug of ergovaline/g diet DM). The results of the study demonstrated that exposure to EAs during mid and/or late gestation in ewes reduced fetal growth. A more recent study in ewes by Klotz et al. (2019) indicated that maternal blood supply to the placenta appeared to be shielded from adverse effects of EAs, but umbilical vasculature was not, which could adversely influence the normal fetal growth.

26.          Studies in livestock also reported reduced reproductive performance, particularly in female cattle, after EAs exposure (Poole and Poole, 2019). A regional vasoconstriction and corresponding decreased blood flow to reproductive tissues was observed, along with a decreased dry matter intake, and/or increased body temperature, leading the authors to conclude that the effect of EAs was both direct and indirect.

27.          In utero exposure to EAs in pregnant ewes, especially during phase two of gestation, alters fetal growth, muscle fibre formation, and miRNA expression (non-coding micro RNA that is associated with the control of gene expression, specifically glucose transport, insulin signalling, intracellular ATP, hypertension, or adipogenesis) (Greene et al., 2019). Ergovaline was shown to be a potent vasoconstrictor in the bovine umbilical and uterine arteries and reduces blood flow to developing placental tissues and fetuses (Klotz et al., 2015). Placental weight reduction is highly correlated with fetal birthweight and high exposure to EAs in ruminants can result in additional adverse effects such as hyperexcitability, hypermetria, and tremors (Klotz et al., 2015, Britt et al., 2019).

28.          Several studies have reported effects of EAs on the reproductive process in rodents and stallions, including prevention of pregnancy predominantly by poor sperm quality, interference with implantation, and embryotoxicity. Page et al., 2019 ; reported an in vitro study on bovine sperm in which sperm morphology and motility was adversely affected by incubation with three different EAS- ergotamine, dihydroergotamine and ergonovine. Fayrer-Hosken et al., 2013 reported that consumption of high levels of EAs decreased the gel free volume of sperm samples in stallion but did not result in statistically significant on the spermiogram (a test of fertility). These studies on adverse reproductive effects in males have been included for completeness.

Humans

29.          In a randomised clinical trial to evaluate the effects of EAs on milk secretion postpartum, 30 women received an injection of 0.2 mg methylergobasine immediately after delivery followed by 3 tablets of 1 mg of ergotamine tartrate given daily (orally) for 6 days post-partum. Results showed that the treatment had no significant effect on either the weight of the infant or the quantity of milk consumed (Jolivet et al., 1978). A study by Arroyo-Manzanares et al (2017) addressed the similarities of the actions of EAs to those of monoamine neurotransmitters and provided evidence that EAs have the ability to act on the secretion of adrenocorticotropic hormone (ACTH), prolactin (PRL), luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Prolactin has an important biological function in lactation, reproduction, immune responses, and metabolism and inhibition of milk production due to the inhibition of prolactin has been seen in humans, laboratory animals, and livestock animals (Arroyo-Manzanares et al., 2017; Prendiville et al., 2000).

30.          In utero exposure to the EA derivative methylergonovine has been associated with a case report of sirenomelia. Sirenomelia is a rare and deadly condition characterized by fusion of the lower limbs, lower spinal column defects, severe malformations of the urogenital and lower GI tract, and an aberrant abdominal umbilical artery. An alteration in the development of the caudal portion of the mesoderm (Duesterhoeft et al., 2007; Garrido-Allepuz et al. 2011) or an alteration of the growth of the umbilical vessels, with corresponding inadequate blood supply to the caudal portion (Cozzolino et al., 2016) has been suggested as reason for this congenital malformation at the critical stage of organogenesis.

31.           Data from trials on the use of EAs (ergometrine and methylergometrine) as uterotonic medication suggested that EAs may decrease mean blood loss from both mother and child by at least 500 mL and increase maternal haemoglobin levels in the blood. However, the results also suggested the treatment increased the incidence of adverse effects such as increased blood pressure and pain after birth (Liabsuetrakul et al., 2018).

32.           An epidemiological study linked the use of purified ergotamine to congenital abnormalities during pregnancy in humans. Ergotamine was used to treat migraine and a mean daily dose of 1.5 mg ergotamine during the 2nd month of pregnancy led to a higher risk for neural-tube defects, spina bifida, posterior cleft palate, congenital cataract and clubfoot (Czeizel, 1989). Two further case studies have reported an association between the use of ergotamine during early pregnancy and the development of Möbius sequence in children (Smets et al., 2004; Graf and Shepard, 1997). Möbius sequence is a rare congenital disorder defined by the paralysis of the 6th and 7th cranial nerves in combination with various odontological, craniofacial, ophthalmological and orthopaedic conditions (Kjeldgaard Pedersen et al., 2017). Vascular disruption has been suggested as one possible explanation for the pathogenesis of Möbius sequence. Ergotamine has also been reported to cause vasospasm and a prolonged and marked increase in uterine tone (Smets et al., 2004; Graf and Shepard, 1997).

33.          Interrogation of the UK Teratology Information Service database did not identify any information related to EAs.

Health-based guidance values

34.          EFSA (2012) considered the vasoconstrictive effect the critical effect for EAs, based on the finding of tail muscular atrophy in rats fed for 13 weeks with ergotamine. A BMDL10 of 0.33 mg/kg bw per day was derived and an uncertainty factor (UF) of 3 was applied to account for deficiencies in the database. Together with the default uncertainty factor of 100 for intra- and interspecies differences, EFSA applied an overall UF of 300 and established an acute reference dose (ARfD) of 1 μg/kg bw (rounded to one significant figure). In line with EFSA’s recommendations, an additional UF of 2 was applied for the extrapolation from a sub-chronic to a chronic study in their derivation of the tolerable daily intake (TDI). Therefore, an overall UF of 600 was applied to the same BMDL10 of 0.33 mg/kg bw per day to establish a TDI of 0.6 μg/kg bw per day. EFSA concluded that the available data were not sufficient to determine the relative potencies of individual EAs, but the limited data available for some EAs showed no apparent differences in potencies.

35.          In 2021, the Joint FAO/WHO Expert Committee on Food Additives (JEFCA), identified ergotamine maleate as the cause of uterine contractions in humans during late pregnancy and postpartum and decided to investigate the role of EAs in the diet (JEFCA, 2021). As ergometrine has the highest uterotonic effect and potency for uterine contractions JECFA established an ARfD based on the lowest oral therapeutic dose of 0.2 mg ergometrine maleate (equivalent to 2.5 μg/kg bw, expressed as ergometrine). An UF of 2 was applied for extrapolating a pharmacological LOEL to a NOEL. JECFA also applied an UF of 3.16 to account for possible interindividual toxicodynamic differences. Applying a composite UF of 6.3 (2 × 3.16) an ARfD of 0.4 μg ergometrine/kg bw was derived. JECFA also considered two 4-week studies on ergotamine tartrate and α-ergocryptine in rats and derived a reference point (BMDL10) of 1.3 mg/kg bw, based on muscular degeneration in the tail. However, JECFA considered the human pharmacological effect level of 2.5 μg/kg bw and resulting NOEL to provide a much more sensitive reference point than a downstream toxic effect in animals. A TDI of 1 μg/kg bw per day was initially established by selecting the lowest BMDL10 value of 0.6 mg/kg bw per day. However, JECFA concluded that a TDI should not be higher than the ARfD and hence decided to establish a group TDI for the sum of total EAs in the diet at the same value as the group ARfD of 0.4 μg/kg bw per day.

Sources of EA exposure

36.          EFSA’s Comprehensive European Food Consumption Database refers mainly to processed food and data available on human dietary exposure to EAs derived from consumption estimation are very limited. EFSAs estimated chronic dietary exposure in the adult population varied between 0.007 and 0.08 μg/kg body weight (bw) per day for average consumers and 0.014 and 0.19 μg/kg bw per day for high consumers. The acute dietary exposure in the adult population ranged between 0.02 and 0.23 μg/kg bw per day for average consumers, and between 0.06 and 0.73 μg/kg bw per day for high consumers. The highest exposure (chronic and acute) was in countries with relatively high consumption of rye bread and rolls. Assessment of the dietary exposure to EAs in specific groups of the population indicated no significant differences between vegetarians and the general population. However, a slightly higher dietary exposure to EAs was noted in consumers of unprocessed grains compared to the general population (EFSA, 2012).

37.          Caraballo et al. (2019) reported concentrations of up to 47 µg/kg in grains and grain-based composites. In cereal and flour, particularly rye, EA concentrations of over 7 mg/kg (Krska and Crews, 2008) have been reported.

38.          The European Union (EU) established maximum levels (ML) of ergot sclerotia and EAs, effective as of January 2022. For milled products derived from barley, wheat, spelt or oats with an ash content of less than 900 mg/100 g, an EAs limit of 100 µg/kg applies, being further reduced to 50 µg/kg in July 2024. For the same types of grain products with a higher ash content or sold directly to the end consumer, the maximum level of EAs was set at 150 µg/kg. The maximum level of EAs in wheat gluten is 400 µg/kg. As an open pollination species, rye is generally more susceptible to infestation, which is accounted for by a higher maximum level. Milled rye products are subject to an EAs limit of 500 µg/kg, further reduced to 250 µg/kg in July 2024. A maximum level of 20 µg/kg for EAs in grain-based food for infants and toddlers has also been introduced.

39.          The German Federal Institute for Risk Assessment (BfR) based their risk assessment on the consumption of rye flour contaminated with ergotamine and ergometrine. The BfR estimated that on average, ergotamine accounted for a maximum of 46 % of the total alkaloid content. The consumption of 250 g of the most contaminated rye flour would result in an intake of 834 µg ergotamine per day per person. The consumption of highly contaminated rye flour therefore exceeds the maximum therapeutic daily dose tolerated for a month-long therapy of 670 µg ergotamine tartrate per day (BfR, 2004).

40.          A research group at the University of Ghent (Arroyo-Manzanares, 2017) carried out an extensive survey on European products and tested 1,065 samples of cereals and cereal products (rye, wheat, and multigrain-based food that contain rye and wheat) intended for human consumption. In total, 59 % of samples tested positive for EAs, with EAs present in 84 % of rye, 67 % of wheat and 48 % of multigrain-based food. The levels ranged from 1 to 12,340 μg/kg.

41.          Storm et al. (2011) detected EAs in rye flour samples from Danish mills with an average and maximum concentration of 46 μg/kg and 234 μg/kg, respectively. Crews et al. (2007) detected EAs in 25 of 28 samples, including all 11 types of rye crispbreads with concentration up to 340 μg/kg while Müller et al. (2009) found EAs in 92 % of the tested rye products with a maximum content of 740 μg/kg. Reinhold et al. (2011) tested 500 food samples from Germany, approximately 50 % were positive for EAs with a highest concentration of 1,063 μg/kg. A more recent survey by Bryła et al. (2015) detecting EAs in 83 % of the tested rye grain, 94 % of rye flour, and 100 % of rye bran and flake samples. Ergocryptine, ergocristine, and ergotamine, including their C8-isomers, were the most common EAs detected in the majority of surveys and foods sampled.

42.          A study by Dusemund et al. (2006) concluded that ergometrine contributed 5 % of the total alkaloid content and that consumption of 250 g of the most contaminated rye flour would result in an intake of 91 µg ergometrine per day per person. This would be below the lowest therapeutic dose equivalent to 400 µg egometrine hydrogen maleate per day.

Exposure Assessment

43.          Exposure to EAs were derived using data from the 2014 Total Diet Study-Mycotoxin analysis and consumption data from the National Diet and Nutrition Survey (NDNS).

44.          The TDS data (Stratton et al., 2017) was based on 28 food groups which were further divided to produce 49 food groups; all food groups in which EAs were detected and hence included in this assessment can be seen in Table 2 (for all food groups included in the survey, please see Annex B, Table 5). Total EAs and epimers (ergocristine, ergotamine, ergocornine, ergosine, ergocryptine, ergometrine, ergocristinine, ergotaminine, ergocorninine, ergosinine, ergocryptinine and ergometrinine) were determined by LC/MS/MS (Carbonell-Rozas et al., 2021). Those twelve EAs are the most frequent forms detected. More data on each specific subset are available in the Diet Study (TDS) – Mycotoxin Analysis Report, 2017 Diet Study (TDS) – Mycotoxin Analysis: Final report (food.gov.uk)

Table 2: Foods groups considered

Food groups

 

White sliced bread

White unsliced bread

Brown bread

Wholemeal and granary bread

Other bread

Misc cereals FLOUR

Misc cereals Buns cakes and pastries

Misc cereals Savoury biscuits

Misc cereals Sweet biscuits

Misc cereals Chocolate biscuits

Misc cereals Breakfast cereals

Misc cereals RICE

Misc cereals Other cereal products

Misc cereals PASTA

Misc cereals Pizza

45.          The food groups contributing most to EAs exposure were a) wholemeal and granary bread, b) white sliced bread and c) other bread. Mean and 97.5th percentile estimated exposure to EAs from the individual food groups for women of child-bearing age (16- 49 years) can be found in Table 3 (acute) and Table 4 (chronic).

Table 3: Acute exposure to ergot alkaloids in women of childbearing age; food groups not containing EAs have been removed.

 

Exposure (µg/kg bw) LB - UB

Exposure (µg/kg bw) LB - UB

Food groups

Mean

P97.5

     

White sliced bread

0.0092-0.0093

0.041-0.041

White unsliced bread

0.0060-0.0061

0.034-0.035

Brown bread

0.0023

0.033

Wholemeal and granary bread

0.024

0.091

Other bread

0.015

0.068

Misc cereals FLOUR

0.0015-0.0017

0.012-0.013

Misc cereals Buns cakes and pastries

0.0019-0.0027

0.0094-0.013

Misc cereals Savoury biscuits

0.00091-0.00093

0.0076-0.0077

Misc cereals Sweet biscuits

0.0015-0.0016

0.0084-0.0090

Misc cereals Chocolate biscuits

0.00056-0.00061

0.0054-0.0058

Misc cereals Breakfast cereals

0.003562-0.00359

0.0183-0.0184

Misc cereals RICE

0-0.0062

0-0.025

Misc cereals Other cereal products

0.00097-0.0017

0.0088-0.015

Misc cereals PASTA

0.0022-0.0065

0.0089-0.027

Misc cereals Pizza

0.0072-0.0075

0.054-0.056

Total

0.052-0.057

0.12-0.13

LB= lower bound; UB= upper bound.

Table 4: Chronic exposure to ergot alkaloids in women of childbearing age; food groups not containing EAs have been removed.

 

Exposure (µg/kg bw) LB - UB

Exposure (µg/kg bw) LB - UB

Food groups

Mean

P97.5th percentile

     

White sliced bread

0.0040-0.0041

0.02140-0.021479

White unsliced bread

0.0021-0.0021

0.012761-0.01291

Brown bread

0.00080

0.010

Wholemeal and granary bread

0.011

0.052

Other bread

0.0055

0.029

Misc cereals FLOUR

0.00055-0.00062

0.0047-0.0052

Misc cereals Buns cakes and pastries

0.00067-0.00092

0.0038-0.0052

Misc cereals Savoury biscuits

0.00033-0.00033

0.003059-0.003125

Misc cereals Sweet biscuits

0.00057-0.00061

0.0036-0.0039

Misc cereals Chocolate biscuits

0.00018-0.00020

0.0018-0.0019

Misc cereals Breakfast cereals

0.00189-0.00190

0.0089-0.0090

Misc cereals RICE

0-0.0024

0-0.012

Misc cereals Other cereal products

0.00029-0.00052

0.0028-0.0048

Misc cereals PASTA

0.00071-0.0021

0.0035-0.010

Misc cereals Pizza

0.0019-0.0020

0.015-0.015

Total

0.031-0.035

0.072-0.080

LB= lower bound; UB= upper bound.

Exposures in subpopulation groups

Vegans and Vegetarians

46.          The number of vegans and vegetarian among the total number of consumers (n= 2556) were relatively small with 112 and 10, respectively.

47.          The LB and UB mean and 97.5th percentile acute exposures were 0.064 – 0.070 µg/kg bw and 0.127 – 0.13 µg/kg bw for vegans, respectively. For vegetarians the LB and UB mean and 97.5th percentile exposures were 0.061 – 0.067 µg/kg bw and 0.135 – 0.14 µg/kg bw, respectively.

48.          The LB and UB mean and 97.5th percentile chronic exposures were 0.044 – 0.049 µg/kg bw and 0.084 – 0.087 µg/kg bw for vegans, respectively. For vegetarians the LB and UB mean and 97.5th percentile exposures were 0.038 – 0.043 µg/kg bw and 0.078 – 0.092 µg/kg bw, respectively.

Ethnicity

49.          Total acute and chronic exposures to EAs in women from different ethnic groups are presented in Table 5 and Table 6.

Table 5: Acute exposure to EAs in women of childbearing age by ethnicity

 

Exposure LB-UB (µg/kg bw)

Exposure LB-UB (µg/kg bw)

Ethnic group

Mean

P97.5

Asian or Asian British (n=135)

0.057 – 0.068

0.11 – 0.13

Black or Black British (n=82)

0.047 – 0.055

0.10 – 0.11

White (n = 2234)

0.052 – 0.056

0.12 – 0.13

LB= lower bound; UB= upper bound.

Table 6: Chronic exposure to EAs in women of childbearing age by ethnicity

 

Exposure LB-UB (µg/kg bw)

Exposure LB-UB (µg/kg bw)

Ethnic group

Mean

P97.5

Asian or Asian British (n=135)

0.034 – 0.046

0.071 – 0.097

Black or Black British (n=82)

0.027 – 0.033

0.073 - 0.085

White (n = 2234)

0.030 – 0.034

0.073 – 0.079

LB= lower bound; UB= upper bound.

Risk Characterisation

50.          The available data suggested that EAs produce direct peripheral effects (uterotonic action or vasoconstriction), indirect peripheral effects (serotonin antagonism or adrenergic blockade), and central nervous system effects (induction of hypothermia and emesis or control of the secretion of pituitary hormones). Due to their structural similarities EAs have been suggested to act as agonists or antagonists to noradrenaline, dopamine and serotonin neurotransmitters.

51.          Exposure to EAs has also been associate with pregnancy hindrance by interfering with eggs implantation and embryotoxicity in rodents, negative effects on maternal blood supply to the placenta in ewes and sirenomelia associate with in utero exposure in humans. EAs can also negatively affect lactation due to their hormone mimicking activity, in particular LH/FSH balance and prolactin (Della-Giustina eta al., 2005).

52.          EFSA (2012) established an ARfD of of 1 μg/kg bw and a TDI of 0.6 μg/kg bw per day for EAs. JEFCA establish a group TDI for the sum of total EAs in the diet at the same value as the group ARfD of 0.4 μg/kg bw per day.

53.          Mean and 97.5th percentile total acute estimated exposures were 0.052 to 0.057 and 0.12 to 0.13 µg/kg bw respectively, mean and 97.5th percentile total chronic estimated exposures were 0.031 to 0.035 and 0.072 to 0.080 µg/kg bw respectively. All estimated exposures are below the respective ARfD and TDI established by EFSA and are therefore not of toxicological concern. The estimated exposures are also below the HBGV established by JECFA.

54.          The food groups contributing most to the overall exposures were wholemeal and granary bread, white sliced bread and other bread. However, it should be noted that the dietary exposure estimates are based on a limited number of food groups and that data from ready-to-eat foods analysis are scarce. A contribution to overall EAs exposure from other foods can therefore not be excluded.

55.          The current assessment was based on consumption data from the NDNS for women of maternal/childbearing age and therefore may not be representative of maternal diet. The relatively small data set and limited number of EAs evaluated further as a level of uncertainty to the results.

56.          The lowest prescribed dose of ergotamine for acute migraine treatment is 13-26 μg ergotamine/kg bw, the maximum recommended oral therapeutic dose in adults for ergotamine over a period of 30 days (to avoid possible severe adverse effects such as peripheral vasoconstriction) is 8 μg/kg bw per day. EFSA concluded that 2 μg/kg b.w. ergometrine, used to induce uterine contraction, is likely to be close to a NOAEL and that the margin between this dose in a sensitive subpopulation and the group ARfD of 1 μg/kg bw is adequate. The estimate exposures here are below any therapeutical doses reported to have adverse effects.

Conclusions

57.          Applying occurrence data from the 2011 TDS for EAs and consumption data for woman of childbearing age, all estimated mean and 97.5th percentile exposures are below the respective ARfD of 1 µg/kg bw and TDI of 0.6 μg/kg bw and are therefore not of toxicological concern. These exposures are also below the reported therapeutical doses of natural or synthetic EAs.

58.          However, it should be noted that the assessment was based on a relatively small sample size (food groups, EAs tested) and that applying consumption data for woman of childbearing age may not be representative of the maternal diet.

Questions on which the views of Committee are sought

      I.         Do Members have any comments on the potential risk of consuming ergot alkaloids in the maternal diet?

      II.            Does the Committee have any further comments?

Secretariat

July 2022

 

Abbreviations

ACTH

Adrenocorticotropic hormone

ARfD

Acute reference dose

BfR

Bundesamt fuer Risikobewertung/German Federal Institute for Risk Assessment

BMDL

Benchmark Dose Lower Confidence Limit

CNS

Central nervous system

CONTAM

Panel on Contaminants in the Food Chain

COT

Committee on the Toxicity of Chemicals in Food, Consumer Products and the Environment

DM

Dry matter

DNA

Deoxyribonucleic acid

EAs

Ergot alkaloids

EC

European Commission

EFSA

European Food Safety Authority

EU

European Union

FAO

Food and Agriculture Organization of the United Nations

FSH

Follicle-stimulating hormone

GI tract

Gastrointestinal tract

HBGV

Health based guidance value

IARC

International Agency for Research on Cancer

i.v.

Intravenous

JECFA

Joint FAO/WHO Expert Committee on Food Additives

LA

Lysergic acid

LB

Lower bound

LC

Liquid chromatography

LH

Luteinizing hormone

LOAEL

Lowest observed adverse effect level

Misc

Miscellaneous

MS

Mass spectrometry

NDNS

National Diet & Nutrition Survey

ng/g

nanograms per gram

NOAEL

No observed adverse effect level

PRL

Prolactin

SACN

Scientific Advisory Committee on Nutrition

sc

Subcutaneous

SCE

Sister chromatid exchange

TDI

Tolerable daily intake

TDS

Total Diet Study

UB

Upper bound

UF

Uncertainty factor

WHO

World Health Organisation

μg

μg = microgram

μg/g

microgram per gram

μg/kg

microgram per kilogram

μg/L

microgram per litre

 

References

Arroyo-Manzanares N., Gámiz-Gracia L., García-Campaña A.M.,  Di Mavungu J.D, De Saeger S. (2017). Ergot Alkaloids: Chemistry, Biosynthesis, Bioactivity, and Methods of Analysis. Fungal Metabolites 887-929

DOI 10.1007/978-3-319-25001-4_1

Althaus, M, Retzow A, Castell JV, Gomez-Lechon MJ, Amalou Z, Rose T, Appel K (2008). In vitro identification of the cytochrome P450 isoform responsible for the metabolism of a-dihydroergocryptine. Xenobiotica, 30(11): 1033-45.

DOI https://doi.org/10.1080/00498250010002261

Bernard N, Jantzem H, Becker M, Pecriaux C, Bénard-Laribière A, Montastruc JL, Descotes J, Vial T (2015). Severe adverse effects of bromocriptine in lactation inhibition: a pharmacovigilance survey. Royal College of Obstetricians and Gynaecologists, 122(9): 1244-51.

DOI https://doi.org/10.1111/1471-0528.13352

Bogun N, Mathies R, Baesecke J (2011). Angiospastic occlusion of the superficial femoral artery by chronic ergotamine intake. Deutsche Medizinische Wochenschrift, 136(1-2): 23-6.

DOI https://doi.org/10.1055/s-0030-1269435

Bryła M, Szymczyk K, Jędrzejczak R, Roszko M (2015). Application of liquid chromatography/ ion trap mass spectrometry technique to determine ergot alkaloids in grain products. Food Technol Biotechnol 53:18–28.

DOI https://doi.org/10.17113/ftb.53.01.15.3770

Britt JL, Greene MA, Bridges Jr WC, Klotz JL, Aiken GE, Andrae JG, Pratt SL, Long NM, Schrick FN, Strickland JR, Wilbanks SA, Miller Jr MF, Koch BM, Duckett SK (2019). Ergot alkaloid exposure during gestation alters. I. Maternal characteristics and placental development of pregnant ewes. Journal of Animal Science, 97(4):1874–90.

DOI https://doi.org/10.1093%2Fjas%2Fskz068

Caraballo D, Toloso J, Ferrer E, Berrada H (2019). Dietary exposure assessment to mycotoxins through total diet studies. A review. Food and Chemical Toxicology, 128: 8-20.

DOI https://doi.org/10.1016/j.fct.2019.03.033

Cassady JM, Li GS, Spitzner EB, Floss H, Clemens JA (1974). Ergot alkaloids. Ergolines and related compounds as inhibitors of prolactin release. J Med Chem. (3):300-7.

DOI https://doi.org/10.1021/jm00249a009

Chestnut D.H. Chronic Pain during and after Pregnancy (2020). Chestnut's Obstetric Anesthesia.

DOI Book

Cozzolino M, Riviello C, Fichtel G, Di Tommaso M (2016). Brief Report Exposure to Methylergonovine Maleate as a Cause of Sirenomelia. Birth Defects Research Part A: Clinical and Molecular Teratology, 106(7): 643-7.

DOI https://doi.org/10.1002/bdra.23503

COT (2017). Review of potential risks from mycotoxins in the diet of infants aged 0 to 12 months and children aged 1 to 5 years. Available: tox2017-30_0.pdf (food.gov.uk)

Crews C, Anderson WAC, Rees G, Krska R (2009). Ergot alkaloids in some rye-based UK cereal products. Food Addit Contam Part B Surveill 2:79–85.

DOI https://doi.org/10.1080/02652030903042509

Czeizel A (1989). Teratogenicity of ergotamine J Med Genet 26(1):69-70

DOI http://dx.doi.org/10.1136/jmg.26.1.69-a

Della-Giustina K and Chow G (2003). Medications in pregnancy and lactation. Emergency Medicine Clinics of North America, 21(3): 585-613.

DOI https://doi.org/10.1016/s0733-8627(03)00037-3

De Ruyck K, De Boevre M, Huybrechts I, De Saeger S (2015). Dietary mycotoxins, co-exposure, and carcinogenesis in humans: Short review. Mutation Research/Reviews in Mutation Research, 766: 32-41.

DOI https://doi.org/10.1016/j.mrrev.2015.07.003

Duckett SK, Andrae JG, Prat SL (2014). Exposure to Ergot alkaloids during gestation reduces fetal growth in sheep. Frontiers in Chemistry, 21: 2:68.

DOI https://doi.org/10.3389/fchem.2014.00068

Dighe, R., Vaidya, V.G., (1988). Induction of sister chromatid exchanges by ergot compounds in Chinese hamster ovary cells in vitro. Teratog. Carcinog. Mutagen. 8, 169–174.

DOI  https://doi.org/10.1002/tcm.1770080306

Duesterhoeft S.M., Ernst L.M., Siebert J.R., Kapur RK (2007). Five cases of caudal regression with an aberrant abdominal umbilical artery: Further support for a caudal regression-sirenomelia spectrum. Am J Med Genet A. 15;143A(24):3175-84.

DOI 10.1002/ajmg.a.32028

Dusemund B, Altmann H-J and Lampen A, (2006). II. Toxikologische Bewertung Mutterkornalkaloidkontaminierter Roggenmehle. Journal of Consumer Protection and Food Safety, 1, 150-152.

DOI https://link.springer.com/article/10.1007/s00003-006-0025-2

EFSA (2012). Scientific Opinion on Ergot alkaloids in food and feed. EFSA Journal, 10(7): 2798. Available: Scientific Opinion on Ergot alkaloids in food and feed | EFSA (europa.eu)

Commission Regulation (EU) 2021/1399 of 24 August 2021 amending Regulation (EC) No 1881/2006 as regards maximum levels of ergot sclerotia and ergot alkaloids in certain foodstuffs (Text with EEA relevance). Available: EUR-Lex - 32021R1399 - EN - EUR-Lex (europa.eu)

Fitzgerald P and Dinan TG (2008). Prolactin and dopamine: what is the connection? A review article. Journal of Psychopharmacology, 22(2 Suppl), 12-9.

DOI https://doi.org/10.1177/0269216307087148

Fayrer-Hosken R.A., Hill N.S., Heusner G.L., Traylor-Wiggins W., Turner K (2013). The effects of ergot alkaloids on the breeding stallion reproductive system. Equine Vet J Suppl 12 (45):44-7.

DOI https://doi.org/10.1111/evj.12164

Fröhlich G, Kaplan V, Amann-Vesti B (2010). Holy fire in an HIV-positive man: a case of 21st century ergotism. Canadian Medical Association Journal, 182 (4): 378-80.

DOI https://doi.org/10.1503%2Fcmaj.091362

FSA (2014). Monitoring the Presence of Ergot Alkaloids in Cereals and a Study of a Possible Relationship between Occurrence of Sclerotia Content and Levels of Ergot Alkaloids. Available: Monitoring the presence of ergot alkaloids in cereals | Food Standards Agency

Garcia GD, Goff Jr JM, Hadro NC, I-Donnell SD, Greatorex PS (2000). Chronic ergot toxicity: A rare cause of lower extremity ischemia. Journal of Vascular Surgery, 31(6): 1245-7.

DOI https://doi.org/10.1067/mva.2000.105668

Garrido-Allepuz C., Haro E.,  González-Lamuño D, Martínez-Frías M.L., (2011) Bertocchini F and. Ros M.A. A clinical and experimental overview of sirenomelia: insight into the mechanisms of congenital limb malformations. Disease Models & Mechanisms 4, 289-299.

DOI https://doi.org/10.1242/dmm.007732

Gerhards N, Neubauer L, Tudzynski P, Li SM (2014). Biosynthetic Pathways of Ergot Alkaloids. Toxins, 6(12):3281-95.

DOI https://doi.org/10.3390%2Ftoxins6123281

Greene MA, Britt JL, Powell RP, Feltus FA, Bridges WC, Bruce T, Klotz JL, Miller MF, Duckett SK (2019). Ergot alkaloid exposure during gestation alters: 3. Fetal growth, muscle fiber development, and miRNA transcriptome. Journal of Animal Science, 97(7):3153-68.

DOI https://doi.org/10.1093/jas/skz153

Graf WD and Shepard TH (1997). Uterine contraction in the development of Möbius syndrome. Journal of Child Neurology,12(3): 225-7.

DOI https://doi.org/10.1177/088307389701200315

Griffith, R.W., Grauwiler, J., Hodel, Ch., Leist, K.H., Matter, B., 1978. In: Berde, B., Schild, H.O. (Eds.), Toxicologic Considerations. Ergot Alkaloids and Related Compounds. Springer Verlag, Berlin, Heidelberg, New York, pp. 805–851.

DOI https://rd.springer.com/book/10.1007/978-3-642-66775-6

Guggisberg H (1954). Changes in the therapeutic use of ergot. Ther Umsch. 11(4):77-9.

DOI na

Haarmann T, Ortel I, Tudzynski P, Keller U (2006). Identification of the cytochrome P450 monooxygenase that bridges the clavine and ergoline alkaloid pathways. ChemBioChem – Chemistry Europe. 7(4):645-52.

DOI https://doi.org/10.1002/cbic.200500487

Hardman J, Limbird L, Gilman A (2001). The pharmacological basis of therapeutics. Goodman & Gilman’s 10th ed. New York: McGraw-Hill. Available: Goodman & Gilman’s The Pharmacological Basis of Therapeutics... : Anesthesia & Analgesia (lww.com)

JECFA (2021). Safety evaluation of certain food additives and contaminants. Summary report of the ninety-first meeting. Available: Ninety-first meeting - Joint FAO/WHO Expert Committee of Food Additives (JECFA)

Jolivet A., Robyn C., Huraux-Rendu C., Gautray J.P. (1978). Effect of ergot alkaloid derivatives on milk secretion in the immediate postpartum period. Journal de Gynecologie, Obstetrique et Biologie de la Reproduction 7(1):129-134.

DOI clinical trial

Kalkman, H.O.; Van Gelderen, E.M.; Timmermans, P.B.; Van Zwieten, P.A. (1982) Involvement of α1- and α2-adrenoceptors in the vasoconstriction caused by ergometrine. Eur. J. Pharmacol. 78, 107–111.

DOI https://doi.org/10.1016/0014-2999(82)90377-6

Klotz J. (2015).Activities and Effects of Ergot Alkaloids on Livestock Physiology and Production. Toxins (Basel) 7(8), 2801-2821.

DOI https://doi.org/10.3390/toxins7082801

Klotz JL, Britt JL, Miller MF, Snider MA, Aiken GE, Long NM, Pratt SL, Andrae GJ, Duckett SK (2019). Ergot alkaloid exposure during gestation alters: II. Uterine and umbilical artery vasoactivity. Journal of Animal Science, 97(4): 1891-1902.

DOI https://doi.org/10.1093/jas/skz069

Korn AK, Gross M, Usleber E Thom N, Köhler K, Erhardt G (2014). Dietary ergot alkaloids as a possible cause of tail necrosis in rabbits. Mycotoxin Residues, 30(4):241–50.

DOI https://doi.org/10.1007%2Fs12550-014-0208-0

Krska R and Crews C (2008). Significance, chemistry and determination of ergot alkaloids: a review. Food Additives and Contaminants: Part A, chemistry, analysis, control, exposure and risk assessment, 25(6):722-31.

DOI https://doi.org/10.1080/02652030701765756

Larson, B.T., Harmon, D.L., Piper, E.L., Griffis, L.M., Bush, L.P. (1999). Alkaloid binding and activation of D2 dopamine receptors in cell culture. J. Anim. Sci. 77, 942–947.

DOI https://doi.org/10.2527/1999.774942x

Larson, B.T., Samford, M.D., Camden, J.M., Piper, E.L., Kerley, M.S., Paterson, J.A., Turner, J.T., (1995). Ergovaline binding and activation of D2 dopamine receptors in GH4ZR7 cells. J. Anim. Sci. 73, 1396–1400.

DOI https://doi.org/10.2527/1995.7351396x

Liabsuetrakul T., Choobun T., Peeyananjarassri K.,  Monir Islam Q. (2018). Prophylactic use of ergot alkaloids in the third stage of labour. Cochrane Database Syst Rev 7;6(6):CD005456.

DOI https://doi.org/10.1002/14651858.CD005456.pub3

Lieberman AN and Goldstein M (1985). Bromocriptine in Parkinson disease. Pharmacological Review. 37(2) 217-27. Bromocriptine in Parkinson disease - PubMed (nih.gov)

MacLeod RM  and Lehmeyer J (1973). Suppression of Pituitary Tumor Growth and Function by Ergot Alkaloids. Cancer Research 33, 849-855,

Mrusek M et al (2015). Identification of cellular and molecular factors determining the response of cancer cells to six ergot alkaloids. Invest New Drugs. 33(1):32-44.

DOI https://doi.org/10.1007/s10637-014-0168-4

Mulac D and Humpf HU (2011). Cytotoxicity and accumulation of ergot alkaloids in human primary cells. Toxicology, 282(3):112–21.

DOI https://doi.org/10.1016/j.tox.2011.01.019

Müller C, Kemmlein S, Klaffke H, Krauthause W, Preiss-Weigert A, Wittkowski R (2009). A basic tool for risk assessment: a new method for the analysis of ergot alkaloids in rye and selected rye products. Mol Nutr Food Res 53:500–507

DOI https://doi.org/10.1002/mnfr.200800091

Kjeldgaard Pedersen L, Rikke Damkjær Maimburg R, Hertz JM, Gjørup H, Klit Pedersen T, Møller-Madsen B, Rosendahl Østergaard J (2017). Moebius sequence – a multidisciplinary clinical approach. Orphanet Journal of Rare Diseases, 12(1):4.

DOI https://doi.org/10.1186/s13023-016-0559-z

Orton DA and Richardson RJ (1982). Ergotamine absorption and toxicity. Postgraduate Medical Journal, 58(675): 6-11.

DOI https://doi.org/10.1136%2Fpgmj.58.675.6

Page R, Lester T, Rorie R, Rosenkrans Jr C (2019). Ergot Alkaloid Effects on Bovine Sperm Motility In Vitro. Advance in Reproductive Science, 7(1): 7-15.

DOI https://doi.org/10.4236/arsci.2019.71002

Poole R and Poole DH (2019). Impact of Ergot Alkaloids on Female Reproduction in Domestic Livestock Species. Toxins, 11(6): 364.

DOI https://doi.org/10.3390/toxins11060364

Prendiville WJ, Elbourne D, McDonals S (2000). Active versus expectant management in the third stage of labor. Cochrane Database Systematic Reviews, 3.

DOI https://doi.org/10.1002/14651858.cd000007

Perrin VL (1985). Clinical pharmacokinetics of ergotamine in migraine and cluster headache. Clinical Pharmacokinetics 10(4): 334–52.

DOI https://doi.org/10.2165/00003088-198510040-00004

Reinhold L, Reinhardt K (2011). Mycotoxins in foods in Lower Saxony (Germany): results of official control analyses performed in 2009. Mycotoxin Res 27:137–143.

DOI https://link.springer.com/article/10.1007/s12550-011-0086-7

Roberts G and Rand MJ (1977). Chromosomal damage induced by some ergot derivatives in vitro. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 48 (2):205-214.

DOI https://doi.org/10.1016/0027-5107(77)90162-2

Carbonell-Rozas L, Gamiz-Gracia L, Lara FJ, Garcia-Campana AM (2021). Determination of the main ergot alkaloids and their epimers in oat-based functional foods by ultra-high performance liquid chromatography tandem mass spectrometry. Molecules, 26(12): 3717.

DOI https://doi.org/10.3390/molecules26123717

Seifried, H.E., Seifried, R.M., Clarke, J.J., Junghans, T.B., San, R.H.C. (2006). A compilation of two decades of mutagenicity test results with the Ames Salmonella typhimurium and L5178Y mouse lymphoma cell mutation assays. Chem. Res. Toxicol. 19, 627–644.

DOI https://doi.org/10.1021/tx0503552

Silberstein SD and McCrory DC (2003). Ergotamine and dihydroergotamine: history, pharmacology, and efficacy. Headache, 43(2): 144-66.

DOI https://doi.org/10.1046/j.1526-4610.2003.03034.x

Smets K, Zecic A, Willems J (2004). Ergotamine as a possible cause of Möbius sequence: additional clinical observation. Journal of Child Neurology, 19(5): 398.

DOI https://doi.org/10.1177/088307380401900518

Storm ID, Have Rasmussen P, Strobel BW, Hansen HCB (2008). Ergot alkaloids in rye flour determined by solid-phase cation-exchange and high-pressure liquid chromatography with fluorescence detection. Food Addit Contam 25:338–346.

DOI https://doi.org/10.1080/02652030701551792

Stuedemann JA1, Hill NS, Thompson FN, Fayrer-Hosken RA, Hay WP, Dawe DL, Seman DH, Martin SA (1998). Urinary and biliary excretion of ergot alkaloids from steers that grazed endophyte-infected tall fescue. Journal of Animal Science, 76 (8) 2146–2154.

DOI https://doi.org/10.2527/1998.7682146x

Stratton J, Anderson S, Leon I, Hepworth P, Chapman S, Christy J, Jardine S, Philips D, Setter R, Clough J, MacDonals S (2017). Diet Study (TDS) – Mycotoxin Analysis. Final Report, FS102081. Fera Science Ltd, York (UK). Available: Diet Study (TDS) – Mycotoxin Analysis: Final report (food.gov.uk)

Tasker NR and Wipf P (2021). Biosynthesis, total synthesis, and biological profiles of Ergot alkaloids. Alkaloids Chemical Biology, 85:1-112.

DOI https://doi.org/10.1016/bs.alkal.2020.08.001

Tfelt-Hansen, P., Saxena, P.R., Dahlof, C., Pascual, J., Lainez, M., Henry, P.,Diener, H.,Schoenen, J., Ferrari, M.D., Goadsby, P.J., (2000). Ergotamine in the acute treatment of migraine: a review and European consensus. Brain 123 (Pt 1), 9–18.

DOI10.1093/brain/123.1.9

Uelger TG, Ucar A, Cakiroglu FP, Yilmaz S (2020). Genotoxic effects of mycotoxins. Toxicon 185: 104–13.

DOI https://doi.org/10.1016/j.toxicon.2020.07.004

Valente Eel, Klotz JL, Ahn G, McLeod KR, Herzing HM, King M, Harmon DL (2020) Ergot alkaloids reduce circulating serotonin in the bovine. Journal of Animal Science, 98(12).

DOI http://dx.doi.org/10.1093/jas/skaa362

 

Appendix A

Literature Search Terms for Ergot Alkaloids  (January 2022 - June 2022)

acute toxicity

chronic toxicity

reproductive toxicity

biomarkers (exposure/ toxicity)

maternal health

preconception

conception

pregnancy

post-natal

lactation

fetus/ foetus/ fetal /foetal

placenta

pre-term

preeclampsia

cancer/ carcinogen(icity)

teratogen(icity)

absorption

distribution

metabolis

Appendix B

Food groups analysed in the TDS for EAs. The information can be found in Table 22 of the report and summarised below.

Table 5: List of all food groups analysed for ergot alkaloids (EAs).

 

Food groups

Category

Occurrence data total EAs (µg/kg)

Bread

White sliced bread

14.08

Bread

White unsliced bread

11.88

Bread

Brown bread

27.29

Bread

Wholemeal and granary bread

33.69

Bread

Other bread

23.29

Miscellaneous cereals

Flour

19.46

Miscellaneous cereals

Buns cakes and pastries

8.25

Miscellaneous cereals

Savoury biscuits

2.23

Miscellaneous cereals

Sweet biscuits

9.34

Miscellaneous cereals

Chocolate biscuits

4.90

Miscellaneous cereals

Breakfast cereals

3.07

Miscellaneous cereals

Rice

7.08

Miscellaneous cereals

Other cereal products

0.00

Miscellaneous cereals

Pasta

0.64

Miscellaneous cereals

Pizza

0.00

Miscellaneous cereals

Group sample

6.94

Non-alcoholic beverages

Branded food drinks

4.30

(With bottles water)

Alternatives to milk

0.00

Alcoholic drinks

Beer

0.00

Alcoholic drinks

Cider

0.00

Snacks

Other snacks (not potato based)

0.00

Sandwiches

Sandwiches

3.65

Sandwiches

Group sample

13.46