Registration Dossier

Data platform availability banner - registered substances factsheets

Please be aware that this old REACH registration data factsheet is no longer maintained; it remains frozen as of 19th May 2023.

The new ECHA CHEM database has been released by ECHA, and it now contains all REACH registration data. There are more details on the transition of ECHA's published data to ECHA CHEM here.

Diss Factsheets

Administrative data

Key value for chemical safety assessment

Toxic effect type:
dose-dependent

Effects on fertility

Description of key information

BASF, 2010. OECD 422 Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening test in Wistar rats with N-Methylaminoethanol (CAS 109 -83 -1; BASF, 2010).

Weight-of-evidence approach (please refer to Section 'Additional information')

Link to relevant study records

Referenceopen allclose all

Endpoint:
screening for reproductive / developmental toxicity
Type of information:
experimental study
Adequacy of study:
key study
Study period:
2008-11-25 to 2010
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 422 (Combined Repeated Dose Toxicity Study with the Reproduction / Developmental Toxicity Screening Test)
Deviations:
no
Qualifier:
according to guideline
Guideline:
other: EPA OPPTS 870.3650 Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test
Deviations:
no
GLP compliance:
yes (incl. QA statement)
Limit test:
no
Species:
rat
Strain:
Wistar
Sex:
male/female
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH
- Age at study initiation: 10-11 weeks
- Weight at study initiation: The weight variation of the animals used did not exceed 20 percent of the mean weight of each sex.
- Fasting period before study: no data
- Housing: individually in type M III polycarbonate cages
- Diet (e.g. ad libitum): ad libitum
- Water (e.g. ad libitum): ad libitum
- Acclimation period: yes

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24 °C
- Humidity (%): 30-70%
- Air changes (per hr): 10
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS: The test substance was applied as a solution. To prepare the solution, the appropriate amount of test substance was weighed out depending on the desired concentration. Then the vehicle (highly deionized water) was filled up to the desired volume, subsequently mixed using a magnetic stirrer. The test-substance solutions were prepared in such intervals that the stability was guaranteed.

VEHICLE
- highly deionized water
- Concentration in vehicle:0.5, 1.5 and 4.5 g/100 mL
- Amount of vehicle (if gavage): 100 mL
Details on mating procedure:
- M/F ratio per cage: 1/1
- Length of cohabitation: overnight
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy

- Further matings after two unsuccessful attempts: no
- After successful mating each pregnant female was caged (Pregnant females were provided with nesting material (cellulose wadding) toward the end of pregnancy.)
- Any other deviations from standard protocol: no
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The analyses of the test-substance preparations were carried out at the Analytical Chemistry Laboratory of the Experimental Toxicology and Ecology of BASF SE. The stability of the test substance in highly deionized water at room temperature for a period of 10 days was proven before the start of the administration period (Project No.: 01Y0540/078008). The concentration control analyses revealed that the values were in the expected range of the target concentration, i.e. were in a range of about 90.1-102.2% of the nominal concentration.
Duration of treatment / exposure:
The duration of treatment covered a 2-week pre-mating and mating period in both sexes, approximately 1 week post-mating in males, and the entire gestation period as well as 4 days of lactation in females (35 days for males and 55 days for females).
Frequency of treatment:
daily at the same time in the morning
Details on study schedule:
- Age at mating of the mated animals in the study: 13-14 weeks
Dose / conc.:
0 mg/kg bw/day
Remarks:
Basis: nominal conc.
Dose / conc.:
50 mg/kg bw/day
Remarks:
Basis: nominal conc.
Dose / conc.:
150 mg/kg bw/day
Remarks:
Basis: nominal conc.
Dose / conc.:
450 mg/kg bw/day
Remarks:
Basis: nominal conc.
No. of animals per sex per dose:
10
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: no data
- Rationale for animal assignment (if not random): randomized
Positive control:
no
Parental animals: Observations and examinations:
CAGE SIDE OBSERVATIONS: Yes
A check for moribund and dead animals was made twice daily on working days and once daily on Saturdays, Sundays and public holidays. If animals were in a moribund state, they were sacrificed and necropsied.
- Time schedule: A cageside examination was conducted before and after treatment for any signs of morbidity, pertinent behavioral changes and signs of overt toxicity. Abnormalities and changes were documented for each animal.
- Cage side observations checked in table 1 were included.

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: Detailed clinical observations were performed in all animals prior to the administration period and thereafter at weekly intervals. The findings were ranked according to the degree of severity, if applicable. The animals were transferred to a standard arena (50 x 37.5 cm with side borders of 25 cm high).
- The parameters examined are listed in the table 1

BODY WEIGHT: Yes
- Time schedule for examinations: once a week at the same time of the day (in the morning).

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): no

Generally, food consumption was determined once a week (in a period of 7 days) for male and female parental animals, with the following exceptions:
• Food consumption was not determined during the mating period (male and female F0 animals).
• Food consumption of the F0 females with evidence of sperm was determined on GD 0, 7, 14 and 20.
• Food consumption of F0 females, which gave birth to a litter, was determined on PND 0 and 4.
• Food consumption was not determined in females without positive evidence of sperm (during the mating period of dams used in parallel) and females without litter (during the lactation period of dams used in parallel).

WATER CONSUMPTION AND COMPOUND INTAKE (if drinking water study): No
Oestrous cyclicity (parental animals):
not examined
Sperm parameters (parental animals):
Parameters examined in [P] male parental generations: testis weight, epididymis weight
Litter observations:
STANDARDISATION OF LITTERS
- Performed on day 4 postpartum: [no data]
- If yes, maximum of [all] pups/litter ; excess pups were killed and discarded.

PARAMETERS EXAMINED
The following parameters were examined in [F1] offspring:
number and sex of pups, stillbirths, live births, postnatal mortality, presence of gross anomalies, weight gain, physical or behavioural abnormalities
viability index was calculated as follows: (number of live pups on PND4/number of liveborn pups on the day of birth)x100.
The same for sex ratio: (number of live male or female pups on day 0/ 4/number of live male and female pups on day 0/ 4)x100
The live pups were examined daily for clinical symptoms (including gross-morphological findings) during the clinical inspection of the dams.
The pups were weighed one day after birth (PND 1) and on day 4 after birth.

GROSS EXAMINATION OF DEAD PUPS:
yes, for external and internal abnormalities; possible cause of death was determined for pups born or found dead.
Postmortem examinations (parental animals):
SACRIFICE
- Male animals: All surviving animals [after approximately 1 week post-mating period]
- Maternal animals: All surviving animals after PND 4

GROSS NECROPSY
- Gross necropsy consisted of external and internal examinations including the cervical, thoracic, and abdominal viscera. All parental animals were sacrificed by decapitation using isoflurane anaesthesia. The exsanguinated animals were necropsied and assessed by gross pathology; special attention was given to the reproductive organs. The animals, which died intercurrently or were sacrificed in a moribund state, were necropsied as soon as possible after their death and assessed by gross pathology. Organ weights were recorded (see table 2).

HISTOPATHOLOGY / ORGAN WEIGHTS
The tissues indicated in table 3 were prepared for microscopic examination and weighed, respectively.
Postmortem examinations (offspring):
SACRIFICE
- The F1 offspring were sacrificed at 4 days of age. All surviving pups (after sacrifice on PND 4 by means of CO2), all stillborn pups and those pups that died before schedule, were examined externally, eviscerated and their organs were assessed macroscopically. All pups without any notable findings or abnormalities were discarded after their macroscopic evaluation.
- These animals were subjected to postmortem examinations (macroscopic and/or microscopic examination) as follows: all gross laesions, lungs and spinal cord (cervical, thoracic and lumbar cord) were preserved in neutrally buffered 4 % formaldehyde solution and then analyzed.

GROSS NECROPSY
- Gross necropsy consisted of external and internal examinations including the cervical, thoracic, and abdominal viscera.

HISTOPATHOLOGY / ORGAN WEIGTHS
The tissues indicated in table 3 were prepared for microscopic examination and weighed, respectively.
Statistics:
Food consumption, body weight and body weight change (parental animals and pups (for the pup weights, the litter means were used)), number of mating days, duration of gestation, number of pups delivered per litter, implantation sites, post implantation loss: DUNNETT-test (two-sided)
Reproduction indices and urinalysis, except color, turbidity, volume and specific gravity, females with stillborn pups, females with all stillborn pups, live birth index, pups stillborn, pups died, pups cannibalized, pups sacrificed moribund, viability index, number of litters with affected pups at necropsy: FISHER'S EXACT test
Proportions of affected pups per litter with necropsy observations: WILCOXON-test (one-sided)
Faeces, rearing, grip strength of forelimbs and hindlimbs, landing foot-splay test, motor activity, clinical pathology parameters, urine volume, urine specific gravity and organ weights : KRUSKAL-WALLIS test (two-sided).
Reproductive indices:
Male mating index %: (number of males with confirmed mating* /number of males placed with females)x100; *- defined by a female with vaginal sperm or with implants in utero;
Male fertility index (%): (number of males proving their fertility */number of males placed with females)x100; * - defined by a female with implants in utero;
Female mating index (%): (number of females mated */ number of females placed with males)x100; * - defined as the number of females with vaginal sperm or with implants in utero;
Female fertility index (%): (number of females pregnant */number of females mated **)x100; * defined as the number of females with implants in utero; ** defined as the number of females with vaginal sperm or with implants in utero.
Gestation index (%): (number of females with live pups on the day of birth/number of females pregnant *); * - defined as the number of females with implants in utero;
Live birth index(%): (number of liveborn pups at birth/total number of pups born)x100;
Post implantation loss (%): (number of implantations number of pups delivered/number of implantations)x100
Offspring viability indices:
Viability index (%): (number of live pups on PND4/number of liveborn pups on the day of birth)x100. The same for sex ratio: (number of live male or female pups on day 0/ 4/number of live male and female pups on day 0/ 4)x100
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
In test group 3 (450 mg/kg bw/d) one male animal (animal no. 37) was found dead within the first week of the study. One male animal (animal no. 32) of test group 3 (450 mg/kg bw/d) was sacrificed in a moribund state in study week 2. In addition, one female animal (animal no. 126) of test group 2 (150 mg/kg bw/d) was sacrificed on GD 23 because of an inability to deliver.

In test group 3 (450 mg/kg bw/d), salivation after treatment was observed in study week 1 in one male animal (animal no. 36) and in study weeks 1, 6 and 7 in six female animals. Poor general state was observed in test group 3 (450 mg/kg bw/d) in study weeks 1 and 2 in two male animals (animal nos. 32 and 36) and in study weeks 1, 6 and 7 in two female animals (animal nos. 132 and 135). In test group 3 (450 mg/kg bw/d), apathy was observed in study week 2 in one male animal (animal no. 32). Clonic convulsion was observed in test group 3 (450 mg/kg bw/d) in study week 1 in one male animal (animal no. 39).

The detailed clinical observations on study days 0, 7, 13, 21, 28 in males and females and additionally day 35, 42 and 49 in female animals did not reveal any additional abnormalities in animals of test groups 0-3 (0, 50, 150 and 450 mg/kg bw/d).
Mortality:
mortality observed, treatment-related
Description (incidence):
In test group 3 (450 mg/kg bw/d) one male animal (animal no. 37) was found dead within the first week of the study. One male animal (animal no. 32) of test group 3 (450 mg/kg bw/d) was sacrificed in a moribund state in study week 2. In addition, one female animal (animal no. 126) of test group 2 (150 mg/kg bw/d) was sacrificed on GD 23 because of an inability to deliver.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
In test group 3 (450 mg/kg bw/d) male animals’ body weight was significantly lower in week 4 and body weight change was already significantly lower between weeks 1-2 and in summary between weeks 0-4. In test group 2 (150 mg/kg bw/d) male animals’ body weight change was significantly lower between weeks 3-4. Body weights and body weight changes of all female animals treated with 50, 150 or 450 mg/kg bw/d were not significantly changed during premating.
During gestation body weights of female animals of test group 2 (150 mg/kg bw/d) were significantly lower on GD 14 and 20 and of test group 3 (450 mg/kg bw/d) body weight was even decreased on GD 20.
Body weight changes of female animals during gestation were significantly lower between GD 0-7 in test group 1 (50 mg/kg bw/d) as well as between GD 0-7 and GD 7-14 in test group 2 (150 mg/kg bw/d). A body weight loss could be detected between GD 14-20 in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Consequently, the overall body weight change between GD 0-20 was also significantly lower for these test groups.
Body weights and body weight changes of female animals treated with 50 mg/kg bw/d were not significantly changed during lactation. During lactation, a comparison of body weight data of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) to the control were not meaningful as only one litter consisting of one stillborn pup existed in test group 2 (150 mg/kg bw/d) and no pups were alive in test group 3 (450 mg/kg bw/d).
During the post-weaning period female body weights were significantly lower in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) in study week 6 and 7. The same was true for females of test group 1 (50 mg/kg bw/d) in study week 7. As the terminal mean body weight in this test group was unaffected (see section 4.4.1.1. Absolute organ weights) this change was assessed as incidental and not related to treatment.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
Significantly decreased food consumption of the male animals of test group 3 (450 mg/kg bw/d) was observed during the first two study weeks.
Food consumption of the female rats of test group 3 (450 mg/kg bw/d) was significantly decreased during the first study week.
During gestation the food consumption in test group 2 (150 mg/kg bw/d) was significantly decreased between GD 14 and 20.
During lactation food consumption in test group 2 (150 mg/kg bw/d) was significantly lower compared to the control.
Food efficiency:
not specified
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
effects observed, treatment-related
Description (incidence and severity):
At the end of the administration period red blood cell counts (RBC), haemoglobin concentrations and hematocrit values were decreased in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Additionally, the hematocrit values were significantly decreased in females and males of test group 1 (50 mg/kg bw/d). This decrease compared to the controls was below 10% (males: 5%; females 7%), and it was the only dose-dependently changed red blood cell parameter in this test group. Therefore, the hematocrit decrease in rats of test group 1 (50 mg/kg bw/d) was regarded as treatment-related but not adverse.
The mean corpuscular volume (MCV) was decreased in male rats of all treatment groups (not significantly changed in test group 3 [450 mg/kg bw/d]). The measured MCV and RBC values were used to calculate the hematocrit values. In male rats of test group 1 (50 mg/kg bw/d) the MCV reflected the decreased hematocrit value because the RBC was not changed. Therefore, the decreased MCV in these rats was regarded as treatment-related, but not adverse as mentioned above.
In female rats of test group 3 (450 mg/kg bw/d) the relative reticulocyte counts were increased. No significant change was observed in the total white blood cell counts (WBC) of treated rats. However, some changes in the relative and absolute differential blood cell counts were measured (males: increased relative neutrophil counts and decreased relative eosinophil counts in test group 3 [450 mg/kg bw/d], decreased relative monocyte counts in test group 2 [150 mg/kg bw/d]; females: decreased absolute eosinophil counts in test group 3 [450 mg/kg bw/d], decreased relative neutrophil counts and increased relative lymphocyte counts in test group 2 [150 mg/kg bw/d]). These changes were regarded as being incidental and not treatment-related because they were not dose-dependently changed and not consistent in both sexes.
The prothrombin time was shortened in rats of both sexes of test group 3 (450 mg/kg bw/d) and, additionally, in females of test group 2 (150 mg/kg bw/d).
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
Liver enzyme activity was not changed in male and female rats of any test substance-treated group.
The urea levels were increased in males of test group 2 (150 mg/kg bw/d) and in rats of both sexes in test group 3 (450 mg/kg bw/d).
The total bilirubin concentrations were significantly higher in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The total protein and the albumin levels were increased in females of test group 1 (50 mg/kg bw/d) and higher (total protein level was not significantly increased in test group 3 [450 mg/kg bw/d]), although the increases were not dose-dependent.
In males the total protein levels were significantly increased in test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) and the albumin concentrations in test group 2 (150 mg/kg bw/d), only. These parameters were not changed dose-dependently, and the deviated values were within the historical control ranges (total protein: 62.45-69.74 g/L; albumin 36.12-39.76 g/L). Therefore, these deviations were regarded as non-adverse effects.
The sodium concentrations were increased in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) and, additionally, in males of test group 1 (50 mg/kg bw/d). The sodium mean in males at least of the low dose group was within the historical control range (140.9-147.1 mmol/L). Apart from this, only this electrolyte level was deviated in test group 1 (50 mg/kg bw/d). Therefore, the sodium levels increase at least in males of the low dose group was regarded as a non-adverse effect.
In males of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) the cholesterol levels were decreased. The parameter was not changed dose-dependently, and such deviation was not observed in females. Therefore, the cholesterol levels decrease in males of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) was regarded as non-adverse.
In treated females the potassium concentrations were significantly higher in test group 1 (50 mg/kg bw/d), the creatinine levels were higher in test group 2 (150 mg/kg bw/d) and the magnesium concentrations were increased in test groups 1 and 2. These values were not changed dose-dependently, and the deviations of these parameters were not measured in male rats. Therefore, these changes were regarded as incidental rather than treatment-related.
Endocrine findings:
not specified
Urinalysis findings:
effects observed, treatment-related
Description (incidence and severity):
In rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) the incidence of blood (haemoglobin) was found higher compared to the controls (in females of test group 3 not significant). Additionally, the incidence of higher leucocyte counts in the urine sediment was significantly increased in males of test group 2 (150 mg/kg bw/d). However, no significantly higher leucocyte counts were found in the urine sediment of rats of both sexes of test group 3 (450 mg/kg bw/d). In males of test group 3 (450 mg/kg bw/d), the incidence of higher transitional cell counts was increased.
Behaviour (functional findings):
no effects observed
Description (incidence and severity):
Detailed clinical examinations in an open field, detailed observations in a functional observational battery (FOB) and measurements of motor activity did not reveal indications of test substance-induced effects in low, mid and high-dose rats. Therefore, the clonic convulsions were assessed as being incidental.
Immunological findings:
not specified
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Histopathological findings: non-neoplastic:
effects observed, treatment-related
Description (incidence and severity):
Kidneys: The graded severity of tubular degeneration was dose-related increased. The statistically significant increase of the relative kidney weights in animals of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) was considered to be caused by the tubular degeneration/ regeneration process.
Testes: The decrease of the absolute testes weight in males of test group 3 (450 mg/kg bw/d) was related to the diffuse tubular degeneration.
Ovaries: In ovaries, vacuoles of different size were observed in the sex cord stroma in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Incidence and severity was dose-related increased (see Table 10). In addition, one female of test group 1 (50 mg/kg bw/d), one female of test group 2 (150 mg/kg bw/d) and all females of test group 3 (450 mg/kg bw/d) showed ovarian cysts. The occurrence of cysts in females of test group 3 (450 mg/kg bw/d) was assessed as treatment-related.
The cysts in each one female of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) were considered to be rather incidental.
Although there was no clear histopathological correlate for the decreased absolute and relative ovarian weights in females of test group 3 (450 mg/kg bw/d), a test substance-related effect cannot be ruled out.
Spleen: Incidence and graded severity of extramedullary hematopoiesis were dose-related increased in males and females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). The increased relative spleen weights in males of test group 3 (450 mg/kg bw/d) as well as in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) were associated with these findings. (see Table 8)
Histopathological findings: neoplastic:
no effects observed
Other effects:
not examined
Reproductive function: oestrous cycle:
not examined
Reproductive function: sperm measures:
not examined
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
The male mating index was 100% in all test groups. Fertility was proven for most of the F0 parental males of test groups 0 (control) and 1 (50 mg/kg bw/d) within the scheduled mating interval for the F1 litter. One control male and one male of test group 1 did not generate F1 pups. Furthermore, six males of test group 2 and nine males of test group 3 did not generate F1 pups. Thus, the male fertility index ranged between 11% and 90% (see Tab.4 ). For test groups 0 (control) and 1 (50 mg/kg bw/d) these findings reflected the normal range of biological variation inherent in the strain of rats used for this study as all respective values were within the range of the historical control data (see PART III, Supplement). With regard to pathological findings in epididymidis and testis (see section 4.4. Pathology) the test substance did adversely affect reproduction of the F0 males in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The female mating index calculated after the mating period for F1 litter was 100% for all test groups. The mean duration until sperm was detected (GD 0) amounted to 2.4, 1.4, 2.5, and 2.9 days (0, 50, 150 and 450 mg/kg bw/d, respectively). Consequently, the differences between the test groups were assessed as being spontaneous in nature and without biological relevance. All sperm-positive rats of test groups 0 (control) and 1 (50 mg/kg bw/d) delivered pups or had implantations in utero with the following exceptions: one female (test group 0) and one female (50 mg/kg bw/d) did not become pregnant. 6 females of test group 2 (150 mg/kg bw/d), and 9 females of test group 3 (450 mg/kgbw/d) did not become pregnant. The fertility index varied between 10% and 90% (Tab. 5).
Implantation was not affected by the treatment in test group 1 (50 mg/kg bw/d) since the mean number of implantation sites was comparable test group 0 (0 mg/kg bw/d). In test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) a significant reduction with only 5 and 1 implantation sites was found.The mean duration of gestation, i.e. 22.1 and 22.2 days, was similar in test groups 0 (control) and 1 (50 mg/kg bw/d). No parturition was seen in test group 2 (150 mg/kg bw/d) except of female No. 126 which was sacrificed on GD 23 because of an inability to deliver. Gestation length was not calculable for test group 3 (450 mg/kg bw/d). The gestation index varied between 89% (control group) and 100% (50 mg/kg body weight/day). All values seen in test groups 0 (control) and 1 (50 mg/kg bw/d) reflect the normal range of biological variation inherent in the strain of rats used for this study. All respective values were within the range of the historical control data (PART III, Supplement) and did not show a relation to dosing. However, a clear relation to dosing was obtained for test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The mean number of F1 pups delivered per dam was not affected in test group 1 (50 mg/kg bw/d) whereas only one pup was delivered in test group 2 (150 mg/kg bw/d) and none in test group 3 (450 mg/kg bw/d).
The rate of liveborn pups was unaffected in test group 1 (50 mg/kg bw/d) and the live birth index was 96%. The rate of stillborn pups was not significantly different compared to the control group and within the range of the historical control data (PART III, Supplement). In test group 2 (150 mg/kg bw/d) the live birth index was 0 because only one stillborn pup was delivered.
CLINICAL SIGNS AND MORTALITY (PARENTAL ANIMALS)

In test group 3 (450 mg/kg bw/d) one male animal (animal no. 37) was found dead within the first week of the study. One male animal (animal no. 32) of test group 3 (450 mg/kg bw/d) was sacrificed in a moribund state in study week 2. In addition, one female animal (animal no. 126) of test group 2 (150 mg/kg bw/d) was sacrificed on GD 23 because of an inability to deliver.

In test group 3 (450 mg/kg bw/d), salivation after treatment was observed in study week 1 in one male animal (animal no. 36) and in study weeks 1, 6 and 7 in six female animals. Poor general state was observed in test group 3 (450 mg/kg bw/d) in study weeks 1 and 2 in two male animals (animal nos. 32 and 36) and in study weeks 1, 6 and 7 in two female animals (animal nos. 132 and 135). In test group 3 (450 mg/kg bw/d), apathy was observed in study week 2 in one male animal (animal no. 32). Clonic convulsion was observed in test group 3 (450 mg/kg bw/d) in study week 1 in one male animal (animal no. 39).

The detailed clinical observations on study days 0, 7, 13, 21, 28 in males and females and additionally day 35, 42 and 49 in female animals did not reveal any additional abnormalities in animals of test groups 0-3 (0, 50, 150 and 450 mg/kg bw/d).

BODY WEIGHT AND FOOD CONSUMPTION (PARENTAL ANIMALS)

In test group 3 (450 mg/kg bw/d) male animals’ body weight was significantly lower in week 4 and body weight change was already significantly lower between weeks 1-2 and in summary between weeks 0-4. In test group 2 (150 mg/kg bw/d) male animals’ body weight change was significantly lower between weeks 3-4. Body weights and body weight changes of all female animals treated with 50, 150 or 450 mg/kg bw/d were not significantly changed during premating.
During gestation body weights of female animals of test group 2 (150 mg/kg bw/d) were significantly lower on GD 14 and 20 and of test group 3 (450 mg/kg bw/d) body weight was even decreased on GD 20.
Body weight changes of female animals during gestation were significantly lower between GD 0-7 in test group 1 (50 mg/kg bw/d) as well as between GD 0-7 and GD 7-14 in test group 2 (150 mg/kg bw/d). A body weight loss could be detected between GD 14-20 in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Consequently, the overall body weight change between GD 0-20 was also significantly lower for these test groups.
Body weights and body weight changes of female animals treated with 50 mg/kg bw/d were not significantly changed during lactation. During lactation, a comparison of body weight data of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) to the control were not meaningful as only one litter consisting of one stillborn pup existed in test group 2 (150 mg/kg bw/d) and no pups were alive in test group 3 (450 mg/kg bw/d).
During the post-weaning period female body weights were significantly lower in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) in study week 6 and 7. The same was true for females of test group 1 (50 mg/kg bw/d) in study week 7. As the terminal mean body weight in this test group was unaffected (see section 4.4.1.1. Absolute organ weights) this change was assessed as incidental and not related to treatment.

Significantly decreased food consumption of the male animals of test group 3 (450 mg/kg bw/d) was observed during the first two study weeks.
Food consumption of the female rats of test group 3 (450 mg/kg bw/d) was significantly decreased during the first study week.
During gestation the food consumption in test group 2 (150 mg/kg bw/d) was significantly decreased between GD 14 and 20.
During lactation food consumption in test group 2 (150 mg/kg bw/d) was significantly lower compared to the control.

HEMATOLOGICAL FINDINGS
At the end of the administration period red blood cell counts (RBC), haemoglobin concentrations and hematocrit values were decreased in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Additionally, the hematocrit values were significantly decreased in females and males of test group 1 (50 mg/kg bw/d). This decrease compared to the controls was below 10% (males: 5%; females 7%), and it was the only dose-dependently changed red blood cell parameter in this test group. Therefore, the hematocrit decrease in rats of test group 1 (50 mg/kg bw/d) was regarded as treatment-related but not adverse.
The mean corpuscular volume (MCV) was decreased in male rats of all treatment groups (not significantly changed in test group 3 [450 mg/kg bw/d]). The measured MCV and RBC values were used to calculate the hematocrit values. In male rats of test group 1 (50 mg/kg bw/d) the MCV reflected the decreased hematocrit value because the RBC was not changed. Therefore, the decreased MCV in these rats was regarded as treatment-related, but not adverse as mentioned above.
In female rats of test group 3 (450 mg/kg bw/d) the relative reticulocyte counts were increased. No significant change was observed in the total white blood cell counts (WBC) of treated rats. However, some changes in the relative and absolute differential blood cell counts were measured (males: increased relative neutrophil counts and decreased relative eosinophil counts in test group 3 [450 mg/kg bw/d], decreased relative monocyte counts in test group 2 [150 mg/kg bw/d]; females: decreased absolute eosinophil counts in test group 3 [450 mg/kg bw/d], decreased relative neutrophil counts and increased relative lymphocyte counts in test group 2 [150 mg/kg bw/d]). These changes were regarded as being incidental and not treatment-related because they were not dose-dependently changed and not consistent in both sexes.
The prothrombin time was shortened in rats of both sexes of test group 3 (450 mg/kg bw/d) and, additionally, in females of test group 2 (150 mg/kg bw/d).

CLINICAL BIOCHEMISTRY FINDINGS
Liver enzyme activity was not changed in male and female rats of any test substance-treated group.
The urea levels were increased in males of test group 2 (150 mg/kg bw/d) and in rats of both sexes in test group 3 (450 mg/kg bw/d).
The total bilirubin concentrations were significantly higher in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The total protein and the albumin levels were increased in females of test group 1 (50 mg/kg bw/d) and higher (total protein level was not significantly increased in test group 3 [450 mg/kg bw/d]), although the increases were not dose-dependent.
In males the total protein levels were significantly increased in test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) and the albumin concentrations in test group 2 (150 mg/kg bw/d), only. These parameters were not changed dose-dependently, and the deviated values were within the historical control ranges (total protein: 62.45-69.74 g/L; albumin 36.12-39.76 g/L). Therefore, these deviations were regarded as non-adverse effects.
The sodium concentrations were increased in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) and, additionally, in males of test group 1 (50 mg/kg bw/d). The sodium mean in males at least of the low dose group was within the historical control range (140.9-147.1 mmol/L). Apart from this, only this electrolyte level was deviated in test group 1 (50 mg/kg bw/d). Therefore, the sodium levels increase at least in males of the low dose group was regarded as a non-adverse effect.
In males of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) the cholesterol levels were decreased. The parameter was not changed dose-dependently, and such deviation was not observed in females. Therefore, the cholesterol levels decrease in males of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) was regarded as non-adverse.
In treated females the potassium concentrations were significantly higher in test group 1 (50 mg/kg bw/d), the creatinine levels were higher in test group 2 (150 mg/kg bw/d) and the magnesium concentrations were increased in test groups 1 and 2. These values were not changed dose-dependently, and the deviations of these parameters were not measured in male rats. Therefore, these changes were regarded as incidental rather than treatment-related.

URINANALYSIS FINDINGS
In rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) the incidence of blood (haemoglobin) was found higher compared to the controls (in females of test group 3 not significant). Additionally, the incidence of higher leucocyte counts in the urine sediment was significantly increased in males of test group 2 (150 mg/kg bw/d). However, no significantly higher leucocyte counts were found in the urine sediment of rats of both sexes of test group 3 (450 mg/kg bw/d). In males of test group 3 (450 mg/kg bw/d), the incidence of higher transitional cell counts was increased.

TEST SUBSTANCE INTAKE (PARENTAL ANIMALS) not applicable

REPRODUCTIVE FUNCTION: ESTROUS CYCLE (PARENTAL ANIMALS) not examined

REPRODUCTIVE FUNCTION: SPERM MEASURES (PARENTAL ANIMALS) not examined

REPRODUCTIVE PERFORMANCE (PARENTAL ANIMALS)

The male mating index was 100% in all test groups. Fertility was proven for most of the F0 parental males of test groups 0 (control) and 1 (50 mg/kg bw/d) within the scheduled mating interval for the F1 litter. One control male and one male of test group 1 did not generate F1 pups. Furthermore, six males of test group 2 and nine males of test group 3 did not generate F1 pups. Thus, the male fertility index ranged between 11% and 90% (see Tab.4 ). For test groups 0 (control) and 1 (50 mg/kg bw/d) these findings reflected the normal range of biological variation inherent in the strain of rats used for this study as all respective values were within the range of the historical control data (see PART III, Supplement). With regard to pathological findings in epididymidis and testis (see section 4.4. Pathology) the test substance did adversely affect reproduction of the F0 males in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The female mating index calculated after the mating period for F1 litter was 100% for all test groups. The mean duration until sperm was detected (GD 0) amounted to 2.4, 1.4, 2.5, and 2.9 days (0, 50, 150 and 450 mg/kg bw/d, respectively). Consequently, the differences between the test groups were assessed as being spontaneous in nature and without biological relevance. All sperm-positive rats of test groups 0 (control) and 1 (50 mg/kg bw/d) delivered pups or had implantations in utero with the following exceptions: one female (test group 0) and one female (50 mg/kg bw/d) did not become pregnant. 6 females of test group 2 (150 mg/kg bw/d), and 9 females of test group 3 (450 mg/kgbw/d) did not become pregnant. The fertility index varied between 10% and 90% (Tab. 5).
Implantation was not affected by the treatment in test group 1 (50 mg/kg bw/d) since the mean number of implantation sites was comparable test group 0 (0 mg/kg bw/d). In test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) a significant reduction with only 5 and 1 implantation sites was found.The mean duration of gestation, i.e. 22.1 and 22.2 days, was similar in test groups 0 (control) and 1 (50 mg/kg bw/d). No parturition was seen in test group 2 (150 mg/kg bw/d) except of female No. 126 which was sacrificed on GD 23 because of an inability to deliver. Gestation length was not calculable for test group 3 (450 mg/kg bw/d). The gestation index varied between 89% (control group) and 100% (50 mg/kg body weight/day). All values seen in test groups 0 (control) and 1 (50 mg/kg bw/d) reflect the normal range of biological variation inherent in the strain of rats used for this study. All respective values were within the range of the historical control data (PART III, Supplement) and did not show a relation to dosing. However, a clear relation to dosing was obtained for test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d).
The mean number of F1 pups delivered per dam was not affected in test group 1 (50 mg/kg bw/d) whereas only one pup was delivered in test group 2 (150 mg/kg bw/d) and none in test group 3 (450 mg/kg bw/d).
The rate of liveborn pups was unaffected in test group 1 (50 mg/kg bw/d) and the live birth index was 96%. The rate of stillborn pups was not significantly different compared to the control group and within the range of the historical control data (PART III, Supplement). In test group 2 (150 mg/kg bw/d) the live birth index was 0 because only one stillborn pup was delivered.

ORGAN WEIGHTS (PARENTAL ANIMALS)

Absolute organ weights: When compared to control group 0 (set to 100%), the mean absolute weights of the organs listed in the Table 8 were significantly increased or decreased. All other mean absolute weight parameters did not show significant differences when compared to test group 0 (control).
Relative organ weights: The terminal body weight was significantly decreased in males of test group 3 (450 mg/kg bw/d) and in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) resulting in significant, secondary weight changes in various organs (Table 9)

GROSS PATHOLOGY (PARENTAL ANIMALS)
Three males of test group 3 (450 mg/kg bw/d) showed erosions or ulcers in the glandular stomach. The liver was enlarged in three males and one female of test group 2 (150 mg/kg bw/d) as well as in three males and five females of test group 3 (450 mg/kg bw/d). Four males of test group 1 (50 mg/kg bw/d) and four males of test group 2 (150 mg/kg bw/d) showed a prominent acinar pattern of the liver. The mesenteric lymph nodes were red discolored in one female of test group 2 (150 mg/kg bw/d) and in two females of test group 3 (450 mg/kg bw/d). All other gross lesions occurred either singly or were biologically equally distributed over the control group and the treatment groups. They were considered to be incidental.

HISTOPATHOLOGY (PARENTAL ANIMALS) (see Table 8)
Kidneys: The graded severity of tubular degeneration was dose-related increased. The statistically significant increase of the relative kidney weights in animals of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) was considered to be caused by the tubular degeneration/ regeneration process.
Testes: The decrease of the absolute testes weight in males of test group 3 (450 mg/kg bw/d) was related to the diffuse tubular degeneration.
Ovaries: In ovaries, vacuoles of different size were observed in the sex cord stroma in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). Incidence and severity was dose-related increased (see Table 10). In addition, one female of test group 1 (50 mg/kg bw/d), one female of test group 2 (150 mg/kg bw/d) and all females of test group 3 (450 mg/kg bw/d) showed ovarian cysts. The occurrence of cysts in females of test group 3 (450 mg/kg bw/d) was assessed as treatmentrelated.
The cysts in each one female of test groups 1 (50 mg/kg bw/d) and 2 (150 mg/kg bw/d) were considered to be rather incidental.
Although there was no clear histopathological correlate for the decreased absolute and relative ovarian weights in females of test group 3 (450 mg/kg bw/d), a test substance-related effect cannot be ruled out.
Spleen: Incidence and graded severity of extramedullary hematopoiesis were dose-related increased in males and females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). The increased relative spleen weights in males of test group 3 (450 mg/kg bw/d) as well as in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) were associated with these findings.
Dose descriptor:
NOAEL
Effect level:
50 mg/kg bw/day (nominal)
Based on:
act. ingr.
Sex:
female
Basis for effect level:
organ weights and organ / body weight ratios
histopathology: non-neoplastic
Dose descriptor:
LOAEL
Effect level:
50 mg/kg bw/day (nominal)
Based on:
act. ingr.
Sex:
male
Basis for effect level:
histopathology: non-neoplastic
Key result
Dose descriptor:
NOAEL
Effect level:
50 mg/kg bw/day (nominal)
Based on:
act. ingr.
Sex:
male/female
Basis for effect level:
reproductive performance
Critical effects observed:
not specified
Clinical signs:
no effects observed
Description (incidence and severity):
The F1 pups did not show adverse clinical signs up to scheduled sacrifice on PND 4. In one litter (dam No. 112 of test group 1) one pup showed a papilloma-like a skin flap. This single observation was considered to be spontaneous in nature and not to be adverse.
Mortality / viability:
mortality observed, treatment-related
Description (incidence and severity):
The mean number of delivered pups per dam and the rate of liveborn and stillborn pups were evenly distributed among test groups 0 (control) and 1 (50 mg/kg bw/d). The respective values reflect the normal range of biological variation inherent in the strain used in this study.
The viability index as indicator for pup mortality between PND 0-4 was 100% for test groups 0 (control) and 1 (50 mg/kg bw/d). No viable pups were observed in test group 2 (150 mg/kg bw/d) and test group 3 (450 mg/kg bw/d).
Body weight and weight changes:
no effects observed
Description (incidence and severity):
Mean pup body weights/pup body weight changes of all pups in test group (50 mg/kg bw/d) were comparable to the concurrent control values. The observable differences between the groups were assessed as being spontaneous in nature and without biological relevance.
One runt of each gender was seen in test group 0 (control) and 5 female runts were seen in test group 1 (50 mg/kg bw/d). Both values were within the range of the biological variation inherent in the strain of rats used for this study.
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Urinalysis findings:
not examined
Sexual maturation:
not examined
Organ weight findings including organ / body weight ratios:
not examined
Gross pathological findings:
no effects observed
Description (incidence and severity):
One stillborn pup of test group 1 (50 mg/kg bw/d) showed post mortem autolysis. In 3 pups of test group 1 (50 mg/kg bw/d) and in the single stillborn pup of test group 2 (150 mg/kg bw/d) the stomach was found empty.
Histopathological findings:
no effects observed
Description (incidence and severity):
no treatment -related effects in the low dose group
VIABILITY (OFFSPRING)
The mean number of delivered pups per dam and the rate of liveborn and stillborn pups were evenly distributed among test groups 0 (control) and 1 (50 mg/kg bw/d). The respective values reflect the normal range of biological variation inherent in the strain used in this study.
The viability index as indicator for pup mortality between PND 0-4 was 100% for test groups 0 (control) and 1 (50 mg/kg bw/d). No viable pups were observed in test group 2 (150 mg/kg bw/d) and test group 3 (450 mg/kg bw/d).

CLINICAL SIGNS (OFFSPRING)
The F1 pups did not show adverse clinical signs up to scheduled sacrifice on PND 4. In one litter (dam No. 112 of test group 1) one pup showed a papilloma-like a skin flap. This single observation was considered to be spontaneous in nature and not to be adverse.

BODY WEIGHT (OFFSPRING)
Mean pup body weights/pup body weight changes of all pups in test group (50 mg/kg bw/d) were comparable to the concurrent control values. The observable differences between the groups were assessed as being spontaneous in nature and without biological relevance.
One runt of each gender was seen in test group 0 (control) and 5 female runts were seen in test group 1 (50 mg/kg bw/d). Both values were within the range of the biological variation inherent in the strain of rats used for this study.

SEXUAL MATURATION (OFFSPRING) not applicable
ORGAN WEIGHTS (OFFSPRING) not examined

GROSS PATHOLOGY (OFFSPRING)
One stillborn pup of test group 1 (50 mg/kg bw/d) showed post mortem autolysis. In 3 pups of test group 1 (50 mg/kg bw/d) and in the single stillborn pup of test group 2 (150 mg/kg bw/d) the stomach was found empty.

HISTOPATHOLOGY (OFFSPRING) no treatment -related effects in the low dose group
OTHER FINDINGS (OFFSPRING)
Dose descriptor:
NOAEL
Generation:
F1
Based on:
test mat.
Sex:
male/female
Remarks on result:
not measured/tested
Reproductive effects observed:
yes
Lowest effective dose / conc.:
50 mg/kg bw/day (nominal)
Treatment related:
yes
Relation to other toxic effects:
reproductive effects as a secondary non-specific consequence of other toxic effects
Dose response relationship:
yes
Relevant for humans:
yes

Table 4: Reproductive performance (male animals)

 

Test group 0

(0 mg/kg bw/d)

Test group 1

(50 mg/kg bw/d)

Test group 2

(150 mg/kg bw/d)

Test group 3

(450 mg/kg bw/d)

Male fertility

index [%]

90

90

40

11**

Table 5: Reproductive performance (female animals)

 

Test group 0

(0 mg/kg bw/d)

Test group 1

(50 mg/kg bw/d)

Test group 2

(150 mg/kg bw/d)

Test group 3

(450 mg/kg bw/d)

Female fertility

index [%]

90

90

40*

10**

* p ≤ 0.05; ** p ≤ 0.01

Table 6: Absolute organ weight (parental animals)

Male animals

Female animals

Test group (mg/kg bw/day)

1

(50)

2

(150

3

(450)

1

(50)

2

(150

3

(450)

Terminal body weight

101%

96%

86%**

95%

93%**

85%**

Adrenal glands

 

 

 

96%

90%

82%**

Brain

 

 

 

99%

100%

96%*

Epididymides

100%

90%

68%**

 

 

 

Liver

113%*

121%**

129%**

105%

123%**

124%**

Ovaries

 

 

 

97%

99%

74%**

Testes

103%

105%

78%**

 

 

 

Thymus

98%

92%

67%**

88%

83%*

69%

Table 7: Relative organ weight (parental animals)

 

Male animals

Female animals

Test group (mg/kg bw/day)

1

(50)

2

(150

3

(450)

1

(50)

2

(150

3

(450)

Adrenal glands

104%

102%

128%*

 

 

 

Brain

98%

104%

114%*

104%*

107%*

113%**

Epydidymides

94%

98%

80%**

 

 

 

Heart

96%

106%*

122%**

98%

105%*

116%**

Kidney

101%

110%*

126%**

108%

116%**

132%**

Liver

111%**

127%**

150%**

111%**

133%**

146%**

Ovaries

 

 

 

102%

106%

86%*

Seminal vesicle

104%

113%*

117%*

 

 

 

Spleen

102%

111%

144%**

102%

112%*

121%**

Testes

101%

110%*

91%

 

 

 

Thymus

97%

96%

79%*

 

 

 

* : p ≤ 0.05; **: p ≤ 0.01

Table 8: Histopathology (parental animals)

 

Male animals

Female animals

Test group (mg/kg bw/day)

1

(50)

2

(150

3

(450)

1

(50)

2

(150

3

(450)

Kidneys

Multifocal tubular degeneration

 

Multifocal tubular degeneration; increase of the kidney weight

 

increase of the kidney weight

 

 

 

Testes

 

diffuse tubular degeneration

 

 

 

Epididymides

 

Oligospermia

 

 

 

Ovaries

 

 

 

Ovarian cysts incidental

Ovarian cysts

Spleen

 

extramedullary hematopoiesis

 

extramedullary hematopoiesis;

hemosiderin storage

Liver

Fatty change of hepatocytes

 

enlarged livers

Fore- and glandular stomach

 

 

Erosions or ulcers

 

 

Erosions or ulcers

Mesenteric lymph node

 

 

Sinus erythrocytosis

 

Sinus erythrocytosis

Thymus

 

 

reduced cellularity of cortex

 

 

reduced cellularity of cortex

Conclusions:
Under the conditions of the present reproduction/developmental toxicity screening test the NOAEL (no observed adverse effect level) for reproductive performance and fertility was 50 mg/kg bw/d for the parental rats. The NOAEL for general, systemic toxicity of the test substance was 50 mg/kg bw/d for females and less than 50 mg/kg bw/d for male animals based on the tubular degeneration in the kidneys of six males.
Executive summary:

N-Methylaminoethanol was administered orally via gavage to groups of 10 male and 10 female Wistar rats (F0 animals) at dose levels of 50, 150 and 450 mg/kg bw/d.

The objective of the study was to detect possible effects of the test substance on the integrity and performance of the reproductive system of both sexes. Furthermore, it was intended to obtain information about the general toxicological profile including target organs and the no observed adverse effect level (NOAEL) after repeated oral administration. Control animals were dosed daily with the vehicle (highly deionized water). The duration of treatment covered a 2-week pre-mating and mating period in both sexes, approximately 1 week post-mating in males, and the entire gestation period as well as 4 days of lactation in females.

Regarding clinical examinations, signs of general systemic toxicity were only observed at a dose level of 450 mg/kg bw/d as there were significantly lower body weights in male and female parental animals accompanied with reduced food consumption and reduced general condition in single animals in several phases of the study. Reduced food consumption and body weights during gestation in females of test group 2 (150 mg/kg bw/d) were most likely related to implantation losses.

Detailed clinical examinations in an open field, detailed observations in a functional observational battery (FOB) and measurements of motor activity did not reveal indications of test substance-induced effects in low, mid and high-dose rats. Therefore, the clonic convulsions were assessed as being incidental.

Salivation was seen after dosing in all high-dose rats. From the temporary, short appearance immediately after dosing it is likely, that this finding was induced by a bad taste of the test substance or local affection of the upper digestive tract. Urine discoloration was also observed for all high-dose rats which was most likely related to the test compound. However, both types of findings were not considered to be adverse, toxicologically relevant effects.

Fertility was severely impaired by test-substance administration at dose levels of 150 and 450 mg/kg bw/d. Although mating (male and female mating indices) was not influenced no lifeborn pups were delivered for both test groups.

The deviated levels of clinical chemistry and haematology parameters pointed to anaemia and changed liver cell metabolism. The total protein and the albumin levels were significantly higher in female rats starting at test group 1 (50 mg/kg bw/d). As these were the only deviating parameters in females of this test group the changes were regarded as treatment-related, but non-adverse. The reason for the increase of the sodium concentrations in rats of both sexes in test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) remains unclear, but a test substance-related effect could not be excluded. The higher incidences of leucocytes in the urine of rats of both sexes in test group 3 (450 mg/kg bw/d) and, additionally, in males of the test group 2 (150 mg/kg bw/d) as well as the increased incidence of higher transitional cell counts in males of test group 3 (450 mg/kg bw/d) can be regarded as signs of an affection of the urinary tract in treated rats.

Regarding pathology, after administration of the test substance the terminal body weight was significantly lower in females of test group 2 (150 mg/kg bw/d) and in males and females of test group 3 (450 mg/kg bw/d). The body weight reduction resulted in weight changes of adrenal glands, brain, heart, seminal vesicle, and thymus. Target organs were the kidney, testes, epididymides, ovaries, liver, and spleen. In kidneys and testes, tubular degeneration was dose dependent and assessed as an adverse effect. In ovaries, the occurrence of cysts and vacuolization of sex cord stroma was related to treatment and was considered to be adverse. In test group 3 (450 mg/kg bw/d), the infertility was linked to the reduced number of sperms (oligospermia) caused by tubular degeneration in testes. In addition, the occurrence of ovarian cysts and vacuolization of the sex cord stroma in females may have influenced the fertility. In test group 2 (150 mg/kg bw/d), the severity of the findings in testes or ovaries was only minimal or slight and the findings did not occur in all infertile animals. Nevertheless, these lesions may have affected fertility. In the spleen, a dose-related increase in incidence and severity of extramedullary haematopoiesis occurred in males and females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d). In addition, in females of these test groups the severity of haemosiderin storage was increased. These findings are associated with the increased relative spleen weights in females of test group 2 (150 mg/kg bw/d) as well as in males and females of test group 3 (450 mg/kg bw/d). They were induced in response to anaemia and related to treatment. The liver weights were dose-related increased in males and females of all treatment groups. The liver was enlarged in three males and one female of test group 2 (150 mg/kg bw/d) as well as in three males and five females of test group 3 (450 mg/kg bw/d). In females, the liver enlargement correlated with a minimal central hepatocellular hypertrophy that was observed in five animals of test group 2 (150 mg/kg bw/d) and in 9 animals of test group 3 (450 mg/kg bw/d). In males, mainly a minimal fatty change of hepatocytes was observed in two animals of test group 1 (50 mg/kg bw/d), in 8 animals of test group 2 (150 mg/kg bw/d), and in 7 animals of test group 3 (450 mg/kg bw/d). The liver findings were related to treatment and considered to be adaptive. Although, there were no clear histopathological correlates for the increased liver weights in males of all treatment groups and in females of test group 1 (50 mg/kg bw/d), a test substance-related effect could not be ruled out. There was no correlation between erosion/ ulcer in the stomach and erythrocytosis of the mesenteric lymph node (findings occurred in different animals). However, a treatment-related effect could not be ruled out but was assessed as non-adverse. All further findings occurred either singly or were biologically equally distributed over the control group and the treatment groups. They were considered to be incidental or spontaneous in origin and without any relation to treatment.

Endpoint:
extended one-generation reproductive toxicity - basic test design (Cohorts 1A, and 1B without extension)
Data waiving:
study scientifically not necessary / other information available
Justification for data waiving:
the study does not need to be conducted because an extended one-generation reproductive toxicity study is available
Justification for type of information:
JUSTIFICATION FOR DATA WAIVING

According to Regulation EC no 1907/2006 (REACH) Annex IX, an "Extended One-Generation Reproductive Toxicity Study (B.56 of the Commission Regulation on test methods as specified in Article 13(3) or OECD 443), basic test design (cohorts 1A and 1B without extension to include a F2 generation), one species, most appropriate route of administration, having regard to the likely route of human exposure” is an information requirement, if the available repeated dose toxicity studies (e.g. 28-day or 90-day studies, OECD 421 or 422 screening studies) indicate adverse effects on reproductive organs or tissues or reveal other concerns in relation to reproductive toxicity.” This is the case for the registered substance N-Methylethanolamine (MMEA, CAS 109-83-1), because of the effects observed in the Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test according to OECD TG 422 in rats available for the registered substance MMEA. In this study the test item was administered to rats orally via gavage at doses of 50, 150 and 450 mg/kg bw/d (report no. 95R0540/07110). Among other systemic effects, fertility was severely impaired for MMEA in the screening test at dose levels of 150 and 450 mg/kg bw/d. In these groups no lifeborn pups were delivered. The pregnancy rate and the number of implantation sites were dramatically reduced. Corresponding histopathological effects were tubular degeneration in testes, oligospermia and the occurrence of cysts and vacuolization of sex cord stroma in ovaries. As such, an EOGRTS is required according to Annex IX. However, this study is not available for MMEA. To fulfil this requirement data sets for the analogue aliphatic amine substances monoethanolamine (MEA; CAS 141-43-5), diethanolamine (DEA; CAS 111-42-2; also known as 2,2’iminodiethanol), methyldiethanolamine (MDEA; CAS 105-59-9) and 2-dimethylaminoethanol (DMAE; CAS 108-01-0) have been evaluated within a weight-of-evidence approach in detail. Extensive robust study summaries of the studies evaluated in this approach are also included in the current dossier. A full description of the weight-of-evidence approach is found in the endpoint summary. As a result of this evaluation, it became evident that DEA is the most similar substance to MMEA with regard to the toxicological effects and their dose-response relationship. In addition, the observed effects do point to a similar mode-of-action of MMEA and DEA – a choline depletion-mediated toxicity. Based on this, the recent Extended One-Generation Reproductive Toxicity Study (EOGRTS) according to OECD TG 443 available for DEA (Extended One-Generation Reproductive Toxicity Study with the developmental neurotoxicity and immunotoxicity (DNT/DIT) cohorts but without the extension of Cohort 1B to mate the F1 animals to produce an F2 generation) explicitly contributes to the weight-of–evidence approach elaborated to address this endpoint for the registered substance MMEA.
To justify the use of this EOGRTS of DEA to fulfil the information requirement for MMEA, the systemic effects observed for MMEA in the screening study (OECD TG 422) and the ones observed for DEA in the EOGRTS (OECD TG 443) are compared in the following. Based on this a common mode-of-action is derived.
In the screening test according to OECD TG 422 in rats available for the registered substance MMEA, the test item was administered to rats orally via gavage at doses of 50, 150 and 450 mg/kg bw/d (report no. 95R0540/07110). The main toxicological effect of MMEA was the induction of anaemia. This was present in the mid- and high-dose group (150 and 450 mg/kg bw/d) of both sexes indicated by decreased red blood cell (RBC) counts, hematocrit and hemoglobin values as well as decreased mean corpuscular volume (MCV). Correspondingly, in the spleen a dose-related increase in incidence and severity of extramedullary haematopoiesis occurred. The increased relative spleen weights and adverse effects on the kidneys (i.e. tubular degeneration) are considered to be induced in response to anaemia. In addition to the effects linked to anaemia, liver weights were dose-related increased in males and females of all treatment groups. Liver enlargement was observed in the mid-and high-dose group and minimal fatty change of hepatocytes occurred in males of all dose groups.
The same general toxicity effects were observed for DEA in the EOGRTS according to OECD TG 443 (report no. 08R0295/14R078). In this study the test item DEA was administered to rats in drinking water at doses of 100, 300 and 1000 ppm (corresponds to 13, 38 and 128 mg/kg bw/d). Identical to MMEA, also DEA induced anaemia in the highest dose group of 128 mg/kg bw/d in males and females of the F0 and F1 generation. This was indicated by decreased red blood cell (RBC) counts, hematocrit and hemoglobin values, as well as decreased mean corpuscular volume (MCV). Also, corresponding signs of degeneration and regeneration in the proximal tubules of the kidneys were observed in the mid- and high-dose animals (38 and 128 mg/kg bw/d). Fatty change in the liver occurred in the mid- and high-dose animals (38 and 128 mg/kg bw/d).
Thus, with regard to general toxicity the same kind of effects were induced by DEA in the EOGRTS as observed for MMEA in the screening test at comparable doses. This provides strong evidence that the substances show a similar toxicological pattern and act via a common mode-of-action.
Moreover, the observed direct effects on the fertility are evaluated and discussed. With regard to toxic effects on reproduction, as already mentioned above, fertility was severely impaired for MMEA in the screening test at dose levels of 150 and 450 mg/kg bw/d. Although mating (male and female mating indices) was not influenced, no lifeborn pups were delivered in both test groups. Six females of test group 2 (150 mg/kg bw/d), and nine females of test group 3 (450 mg/kg bw/d) did not become pregnant. In test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) a significant reduction with only five and one implantation sites was found. In testes, tubular degeneration was dose-dependent and assessed as an adverse effect. In ovaries, the occurrence of cysts and vacuolization of sex cord stroma was related to treatment and was considered to be adverse. These were probably the key factors for the observed infertility. In test group 3 (450 mg/kg bw/d), the infertility was linked to the reduced number of sperms (oligospermia) caused by tubular degeneration in testes.
On the contrary, in its EOGRTS, DEA did not impede the fertility of the rats up to the highest dose of 1000 ppm (128 mg/kg bw/d). However, in the highest dose group a lower number of implants, prolonged/irregular estrous cycles as well as pathological changes in sexual organs (diffuse atrophy of the ovaries, luteal cysts, juvenile and immature testes, slight to moderate multifocal degeneration of the testicular tubules), pituitary and mammary glands of both genders were observed. As the dose of 128 mg/kg bw at which DEA shows first reprotoxic effects is below the dose of MMEA exerting the fertility effects (150 mg/kg bw), the toxicity profile for fertility effects cannot be fully compared. However, based on the described effects, which are highly similar between MMEA and DEA in correspondence with their structural similarity, it is very likely that they elicit their toxicology by the identical underlying mode-of-action. As various alkanolamines are known to produce choline-deficiency, effects on the choline-homeostasis (i.e. choline depletion) are assumed to be also a key factor for the toxicology of DEA and MMEA. This is underlined by the toxicological effects, as described in the following.
Choline is a vitamin-like compound with various physiological functions (i.e. building block of phospholipids and acetyl-choline, one-carbon-metabolism and DNA-methylation etc.). It could be demonstrated that certain alkanolamines exert an inhibitory effect on either choline-uptake and/or choline-metabolism. Thereby, alkanolamines cause a choline-depletion. As choline deficiency is associated with effects on membrane integrity, it results in a spectrum of toxic effects as anaemia, testes tubule degeneration, and possibly kidney tubule degeneration. These effects are completely in line with the observations in the studies with DEA and MMEA. Also, a hallmark of choline-depletion is a fatty liver change (Zeisel, 1994). In line with this, liver enlargement concurrent with an increase in absolute and relative liver weight has been reported in all dose groups after MMEA- and DEA-treatment. Furthermore, minimal fatty changes and central hepatocellular hypertrophy have been observed in parallel. Similar effects have also been reported in repeated dose toxicity tests published with DEA, where choline-deficiency caused liver and kidney effects (Melnick, 1992). To examine the effect of DEA on choline levels in the EOGRTS serum and liver choline levels were also analysed in this study. The analytical results demonstrated that DEA led to a reduction in the content of choline in plasma and liver. This effect appears to be dose-dependent, in that higher dose levels were associated with greater choline reduction. This effect was most clearly visible at lower dose levels (100 ppm and 300 ppm).
Based on the highly similar systemic toxicity (i.e. anaemia, kidney and liver effects), there is great evidence that the registered substance MMEA exerts its toxicity by the same mode-of action: choline depletion.
In conclusion, based on the similar toxicity profile and the same mode-of-action the EOGRTS for DEA is suitable to address the information requirement of an EOGRTS for MMEA.
The classification of DEA regarding reproductive toxicity (cat. 2; H361fd) was adopted accordingly for MMEA.


References

Lehman-McKeeman LD, Gamsky EA (2000). Choline supplementation inhibits diethanolamine-induced morphological transformation in Syrian hamster embryo cells: evidence for a carcinogenic mechanism. Toxicol Sci, 55, 303-10

Lehman-McKeeman, L. D. et al. (2002). Diethanolamine Induces Hepatic Choline Deficiency in Mice.Toxicological sciences 67, 39-45

Kamendulis LM and Klaunig JE (2005). Species differences in the induction of hepatocellular DNA synthesis by Diethanolamine. Toxicological Sciences 87(2),328-336.

Melnick R (1992). NTP technical report on the toxicity studies of Diethanolamine (CAS No. 111-42-2) Administered Topically and in Drinking Water to F344/N Rats and B6C3F1 Mice. Toxic Rep Ser. 20:1-D10.

Menjíar M, Cárdenas M, Ortiz G, Pedraza-Chaverrí J(2000). Fertility Diminution in Female Rats with Experimental Chronic Nephrosis. Biol reproduction 63, 1549–1554

Nazian SJ, Dietz JR (1987). Reproductive Changes during the Early Stages of Chronic Renal Insufficiency in the Mal Rat. Biol of Reproduction 37: 105-111

National Toxicology Program (1992). Toxicity Studies of Diethanolamine (CAS No. 111-42-2) Administered Topically and in Drinking Water to F344/N Rats and B6C3F1 Mice. Tech. Rep. Ser. No. 20; NIH Publication No. 92-3343), Department of Health and Human Services, Research Triangle Park, NC. Report no.: TR20.

Ortiz G, Vilchis F, Cárdenas M, Cruz C, Pedraza-Cahverris J, Menjívar M (1999). Reproduciton : Function in Male Rats with Chronic Nephrosis. Journal Reprod and Fertility 117: 223-228

Stott WT, Bartels MJ, Brzak KA, Mar M, Markham DA, Thornton CM, Zeisel SH (2000). Potential mechanisms of tumorigenic action of diethanolamine in mice. Toxicol. Lett., 114, 67-75.

Zeisel SH and Blasztajn JK (1994). Cholin and human nutrition. Ann. Rev. Nutr. 14: 269-296

Reason / purpose for cross-reference:
data waiving: supporting information
Reason / purpose for cross-reference:
data waiving: supporting information
Endpoint:
extended one-generation reproductive toxicity - with both developmental neuro- and immunotoxicity (Cohorts 1A, 1B without extension, 2A, 2B, and 3)
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
09 Dec 2014 - 29 Jan 2018
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
other: OECD Guideline 443 (Extended One-Generation Reproductive Toxicity Study) with the developmental neurotoxicity and immunotoxicity (DNT/DIT) cohorts but without the extension of Cohort 1B to mate the F1 animals to produce an F2 generation
GLP compliance:
yes
Limit test:
no
Justification for study design:
SPECIFICATION OF STUDY DESIGN FOR EXTENDED ONE-GENERATION REPRODUCTION TOXICITY STUDY WITH JUSTIFICATIONS [please address all points below]:

- Premating exposure duration for parental (P0) animals: 2 weeks
- Basis for dose level selection: dose-range finder similar or according to OECD 421
- Inclusion/exclusion of extension of Cohort 1B: yes
- Termination time for F2: no F2 generation was required
- Inclusion/exclusion of developmental neurotoxicity Cohorts 2A and 2B: inclusion of Cohorts 2A and 2B
- Inclusion/exclusion of developmental immunotoxicity Cohort 3: inclusion of Cohort 3
- Route of administration: raol via drinking water
- Other considerations, e.g. on choice of species, strain, vehicle and number of animals: regular strain in reproductive toxicity studies. The rat is the preferred animal species for reproduction studies according to test guidelines. This strain was selected since extensive historical control data were available for Wistar rats.

The design of the present OECD 443 Extended-one-genereation Reproductive Toxicity Study has been agreed with ECHA and eMSCA Germany in the according CORPA process and at MSC.

The objective of this multi-site study was to obtain general information on the possible effects
of 2,2’-iminodiethanol on the integrity and performance of the male and female reproductive
systems, including gonadal function, estrous cyclicity, mating behavior, conception, gestation,
parturition, lactation and weaning, as well as on growth and development of the offspring. The
study should also provide information about the effects on neonatal morbidity, mortality, target
organs of the pups and preliminary data on prenatal and postnatal developmental toxicity. As
part of this assessment, offspring were investigated for effects on the embryonic, fetal and
preadult development of the nervous and immune systems as well as alterations in endocrine
function (including thyroid pertubations). In addition choline was determined in blood plasma
and livers of F1 adolescents.
Specific details on test material used for the study:
SOURCE OF TEST MATERIAL
- Source and lot/batch No.of test material: 000STD77L0 v. 14.05.14 / Test substane number (internal): 14/0295-1
- Expiration date of the lot/batch: 14 May 2016
- Purity test date: regularly (also with respect to anaylsis of potential nitrosamine content)
- Purity: 99.9 corr. area-%

STABILITY AND STORAGE CONDITIONS OF TEST MATERIAL
- Storage condition of test material: Room temperature. The stability of the test substance under storage
conditions over the test period was guaranteed by the Sponsor, and the Sponsor holds this responsibility.
- Stability under test conditions: given, The stability of test substance in drinking water was demonstrated for a period of 9 days at
room temperature
- Solubility and stability of the test substance in the solvent/vehicle: given (soluble in water)
Due to the fact that the test substance preparations were true solutions, it was not considered
necessary to prove homogeneity through analytical procedures.

OTHER SPECIFICS:
- All mean values for 2,2’-iminodiethanol were in the expected range of the target
concentrations (90 - 110%), demonstrating the correctness of the drinking water preparations
- Determination of total N-Nitrosamine content: No measurable content of total N-Nitrosamine (calculated as N-Nitrosodiethanolamine,
NDELA) was detected in the test item preparations
Species:
rat
Strain:
other: Crl:WI(Han)
Sex:
male/female
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH
- Age at study initiation: about 9 wks
- Housing: 1 animal per cage, polycarbonate cages type III with wooden gnawing blocks (Typ NGM E-022) and dust-free wooden bedding; during mating: 1 male/1 female per cage; during rearing up to weaning: 1 dam with her litter
- Diet: Ground Kliba maintenance diet mouse/rat “GLP”; ad libitum
- Water: Drinking water ad libitum
- Acclimation period: at least 5 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Air changes (per hr): 15
- Photoperiod (hrs dark / hrs light): 12 / 12

Food analyses
With regard to the analytical findings of chemical and microbiological contaminants and the
duration of application, the diet was found to be suitable. Fed. Reg. Vol. 44, No. 91 of 09 May
1979, p. 27354 (EPA), served as a guideline for maximum tolerable chemical contaminants.
The concentration of microorganisms did not exceed 1*105/g feed.

Drinking water analyses
On the basis of the analytical findings, the drinking water was found to be suitable. German
Drinking Water Regulation (Trinkwasserverordnung, Bundesgesetzblatt, 05 Dec 1990) served
as a guideline for maximum tolerable contaminants.

Bedding and Enrichment analyses
On the basis of the analytical findings, bedding and cage enrichment were found to be suitable.
Levels given in Lab Animal (Nov-Dec 1979, pp. 24-34) served as a guideline for maximum
tolerable contaminants.

Route of administration:
oral: drinking water
Vehicle:
unchanged (no vehicle)
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
For the test substance preparation, the specific amount of test substance and deionized water will be weighed in a vessel, depending on the dose group, and mixed until it is completely dissolved (details will be retained with the raw data).

Adjustment of pH: The pH of the administration solution of test groups 1-3 is adjusted to 7.3-7.5 by the addition of hydrochloric acid (1N, analytical grade) under pH meter control.

The test substance preparations will be prepared at intervals which guarantee that the test substance concentrations in the diet will remain stable. Stock solution will be stored at room temperature, protected from light and air.
Details on mating procedure:
- M/F ratio per cage: 1:1
- Length of cohabitation: until there is evidence of copulation or when a maximum period of 14 days has elapsed
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy
- After successful mating each pregnant female was caged (how): single
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The analytical investigations of the test substance preparations will be carried out at the Analytical Chemistry Laboratory of Experimental Toxicology and Ecology of BASF SE, Ludwigshafen, Germany, as a part of this study.

The analytical investigations will be performed according to the most recently authorized version of the control procedure 14/0295_1. The control procedure will be described in the raw data and the report.

The Analytical Report is included with the study report.
Duration of treatment / exposure:
The F0 animals will be offered drinking water preparations with test substance for approximately 2 weeks prior to breeding and continuing through breeding (up to two weeks), approximately 4 additional weeks (males) or gestation (three weeks) and lactation (three weeks) for females. Selected F1 offspring (cohorts 1A, 1B, 2A, 2B and 3) will be maintained on drinking water preparations with test substance until sacrifice.
Frequency of treatment:
daily
Details on study schedule:
F0 GENERATION PARENTAL ANIMALS AND F1 PUPS
Male and female rats, aged about 10 or 9 weeks when supplied, will be used as F0 generation parental animals. After an acclimatization period of at least 5 days, these rats will be kept for at least 2 weeks.
Then the F0 animals will be paired. The female F0 animals will be allowed to deliver and rear their pups (F1 generation pups) until postnatal days (PND) 4 or 21. The F0 generation parental animals will be sacrificed after weaning of the F1 generation pups. All F0 females will be sacrificed at about the same day after weaning (+/- 2 days).

F1 PUPS AND SELECTION OF COHORTS
Before weaning of the F1 generation pups on PND 21, 75 male and 75 females per group will be randomly selected, to be placed into cohorts. Obvious runts (those pups whose body weight is equal to or greater than 25% below the mean body weight of the control group, separate for sexes) will not be included.

Cohorts:
Cohort 1A: One male and one female/litter (20/sex/group)
Cohort 1B: One male and one female/litter (25/sex/group)
Cohort 2A: One male or one female/litter (10/sex/group)
Cohort 2B: One male or one female/litter (10/sex/group)
Cohort 3: One male or one female/litter (10/sex/group)

Selected F1 offspring will be maintained on drinking water preparations with test substance until sacrifice.

BREEDING OF THE F0 GENERATION PARENTAL ANIMALS
Male and female animals will be paired overnight in a 1:1 ratio until there is evidence of copulation or a maximum period of 14 days has elapsed. Throughout the mating period, each female will be paired with a predetermined male.

Normally, the female will be placed in the cage of her male partner about 16:00 h and separated from the male between 7:00 h and 9:00 h on the following morning. Deviations from the specified times are possible on Saturdays, Sundays and public holidays and will be documented in the raw data.

A vaginal smear will be prepared after each pairing and examined for sperm. If sperm are detected, pairing of the animals will be discontinued. The day on which sperm are detected will be referred to as gestation day (GD) 0 and the following day as GD 1.

STANDARDIZATION OF LITTERS (CULLING) OF F1 PUPS
On PND 4, all litters will be standardized to 10 pups per litter. Whenever possible, each litter contains 5 male and 5 female pups; as a rule, the first 5 surviving pups/sex in each litter will be used. If it is not possible to have 5 pups/sex in each litter, unequal numbers of males and females can be retained (e.g. 6 male and 4 female pups). Surplus animals will be sacrificed. Standardization of litters will not be carried out in litters with 10 pups or less.
Dose / conc.:
0 ppm (nominal)
Dose / conc.:
100 ppm (nominal)
Remarks:
approx. 12.75 mg/kg body weight/day (mg/kg bw/d)
Dose / conc.:
300 ppm (nominal)
Remarks:
approx. 37.68 mg/kg bw/d
Dose / conc.:
1 000 ppm (nominal)
Remarks:
pprox. 128.35 mg/kg bw/d
No. of animals per sex per dose:
F0 parental animals: 30
F1 rearing animals, cohort 1A: 20
F1 rearing animals, cohort 1B: 25
F1 rearing animals, cohort 2A: 10
F1 rearing animals, cohort 2B: 10
F1 rearing animals, cohort 3: 10
Control animals:
yes, concurrent no treatment
Details on study design:
- Dose selection rationale:
- Rationale for animal assignment (if not random):
- Other:
Positive control:
The positive control substance (for immune system response) will be used with the given specifications of the producer (Sigma-Aldrich, Taufkirchen, Germany). No further analyses will be conducted.

Name of positive control substance: Cyclophosphamide monohydrate
CAS No.: 6055-19-2
Test-substance No.: 05/0012-6
Batch identification: SLBC0666V
Purity: 102.3% (according to supplier)
Homogeneity: Given (visually)
Stability: Expire date: March 2015
The stability of the positive control substance under storage conditions over the test period was guaranteed by the manufacturer and the manufacturer hold this responsibility.
Physical state/color: Solid/white
Storage conditions: Refrigerator (2-8°C)
The stability of Cyclophosphamide monohydrate (positive control substance) in the vehicle for 7 days at room temperature and 32 days frozen was verified in a comparable batch (Project No. 01Y0012/058064).

TEST SUBSTANCE PREPARATIONS AND ADMINISTRATION
Route of administration: Oral administration by gavage using 3 or 5 mL syringes.
Frequency of administration: Once daily
Volume to be administered: 10 mL/kg body weight; the body weight determined most recently will be used to calculate the administration volume.
Dose to be administered: 4.5 mg/kg body weight; the body weight determined most recently will be used to calculate the administration dose.
Preparation: Details on the technical procedure will be laid down in a description of the method and reported correspondingly.
Preparation frequency: At least once before the administration period.
Storage conditions of the test-substance preparations: The positive control substance preparations will be split in daily aliquots and stored closed in a freezer at -18°C.
Duration of the study: 4 weeks

Immunization and examinations (Anti SRBC-IgM ELISA) will be conducted at the same time and in exactly the same manner as with cohort 3 animals.
Parental animals: Observations and examinations:
MORTALITY
A check for moribund and dead animals will be made twice daily from Mondays to Fridays and once daily on Saturdays, Sundays and public holidays.

CLINICAL SIGNS
A cageside examination will be conducted at least once daily for any signs of morbidity, pertinent behavioral changes and/or signs of overt toxicity. If such signs occur, the animals will be examined several times daily. Abnormalities and changes will be documented for each animal.
The parturition and lactation behavior of the dams will generally be evaluated in the morning in combination with the daily clinical inspection of the dams. Only particular findings (e.g. inability to deliver or umbilical cord not cut) will be documented on an individual dam basis.
On weekdays (except Saturdays, Sundays and public holidays) the parturition behavior of the dams will be inspected in the afternoons in addition to the evaluations in the mornings.
The day of parturition is considered to be the 24-hour period from about 15:00 h of one day until about 15:00 h of the following day. Departures from this procedure may occur on Saturdays, Sundays and public holidays.

DETAILED CLINICAL OBSERVATIONS (DCO)
All F0 parental animals will be subjected to detailed clinical observations (DCO) outside their cages at the administration period (day 0) and subsequently once per week (in the morning) by the same trained technicians, whenever possible. For observation, the animals will therefore be removed from their cages and placed in a standard arena (50 × 37.5 × 25 cm). The scope of examinations and the scoring of the findings observed will be based on the current index of findings in GROSSE-Reprotox software and includes but is not limited to the following parameters listed:

1. Abnormal behavior in handling
2. Fur
3. Skin
4. Posture
5. Salivation
6. Respiration
7. Activity/arousal level
8. Tremors
9. Convulsions
10. Abnormal movements
11. Gait abnormalities
12. Lacrimation
13. Palpebral closure
14. Exophthalmos (Protruding eyeball)
15. Assessment of the feces excreted during the examination (appearance/consistency)
16. Assessment of the urine excreted during the examination
17. Pupil size

FOOD CONSUMPTION
Generally, food consumption will be determined once a week for the male and female F0 parental animals, with the following exceptions:
• Food consumption will not be determined after the 2nd premating week (male F0 animals) and during the mating period (male and female F0 animals).
• Food consumption of the F0 females with evidence of sperm will be determined for GD 0-7, 7-14 and 14-20.
• Food consumption of the F0 females, which gave birth to a litter, will be determined for PND 1-4, 4-7, 7-14 and 14-21.
Food consumption will not be determined in the females without positive evidence of sperm during mating and gestation periods and in the females without litter during lactation period.

WATER CONSUMPTION
Generally, water consumption will be determined twice a week for male and female F0 parental animals.
• Additionally, after the 2nd premating week water consumption of the females during gestation (animals with evidence of sperm plugs) will be determined for GDs 0-1, 4-5, 7-8, 10-11, 14-15, 17-18 and 20-21.
• Water consumption of the F0 females, which gave birth to a litter, will be determined for PNDs 1-2, 4-5, 7-8, 10-11, 14-15, 17-18 and 20-21.
Water consumption will not be determined in the females without positive evidence of sperm during mating and gestation periods and in the females without litter during lactation period.

BODY WEIGHTS
In general, the body weight of the male and female F0 parental animals will be determined once a week at the same time of the day (in the morning). The following exceptions are notable for the female parental animals:
• During the mating period of the F0 parental animals, the females will be weighed on the day of positive evidence of sperm (GD 0) and on GD 7, 14 and 20.
• Females with litter will be weighed on the day after parturition (PND 1) and on PND 4, 7, 14 and 21.
Body weight will not be determined in the females without positive evidence of sperm during mating and gestation periods and in the females without litter during lactation period.

CLINICAL PATHOLOGY IN F0 PARENTAL ANIMALS
Samples will be withdrawn from 10 F0 parental males and females per group at termination.

Blood samples will be taken from animals by puncturing the retrobulbar venous plexus following isoflurane anesthesia. Blood sampling and blood examinations will be carried out in a randomized sequence. The list of randomization instructions will be compiled with a computer.

In the afternoon preceding the day of urinalysis, the animals will be individually transferred into metabolism cages (no food or drinking water provided); on the following morning, the individual urine specimens will be examined in a randomized sequence (the list of randomization instructions will be compiled with a computer).

The following parameters will be examined in all animals:
Hematology
1. Leukocytes
2. Erythrocytes
3. Hemoglobin
4. Hematocrit
5. Mean corpuscular volume (MCV)
6. Mean corpuscular hemoglobin (MCH)
7. Mean corpuscular hemoglobin concentration (MCHC)
8. Platelets
9. Differential blood count
10. Reticulocytes
11. Blood smear (only evaluated preparations will be archived)
12. Prothrombin time

Clinical chemistry
1. Alanine aminotransferase
2. Aspartate aminotransferase
3. Alkaline phosphatase
4. Serum  glutamyl transferase
5. Sodium
6. Potassium
7. Chloride
8. Inorg. phosphate
9. Calcium
10. Urea
11. Creatinine
12. Glucose
13. Total bilirubin
14. Total protein
15. Albumin
16. Globulins
17. Triglycerides
18. Cholesterol

Hormone evaluations
The following hormones will be determined in the serum samples:
1. T4 (thyroxine)
2. TSH
3.9.1.4. Urinalysis
1. Volume
2. Color
3. Turbidity
4. pH value
5. Protein
6. Glucose
7. Ketones
8. Urobilinogen
9. Bilirubin
10. Blood
11. Specific gravity
12. Microscopy of sediment
Oestrous cyclicity (parental animals):
For all F0 females, estrous cycle length and normality will be evaluated by preparing vaginal smears during a minimum of 2 weeks prior to mating and throughout cohabitation until there is evidence of sperm in the vaginal smear.

In all cohort 1A and 1B females, vaginal smears will be collected after vaginal opening until the first cornified smear (estrous) is recorded. The estrous cycle also will be evaluated in cohort 1A and 1B females for 2 weeks around PND 75.

Additionally, on the day of scheduled sacrifice, the estrous status will be determined in all female F0 animals and all females of cohorts 1A and 1B.
Sperm parameters (parental animals):
After the organ weight determination, the following parameters will be determined in the right testis or right epididymis of all male F0 parental animals and cohort 1A males sacrificed on schedule:

• Cauda epididymis sperm motility
• Sperm morphology
• Spermatid head count in the testis
• Sperm head count in the cauda epididymis

Initially, sperm morphology and sperm head count (cauda epididymis and testis) will be evaluated for the control and highest dose group, only. The intermediate doses will only be evaluated if deemed necessary based on the obtained results or other findings.

On completion of the final report, the testis and epididymis samples that have not been processed will be discarded. Sperm morphology slides will be archived.
Litter observations:
MORTALITY
A check for moribund and dead animals will be made twice daily from Mondays to Fridays and once daily on Saturdays, Sundays and public holidays.

DETAILED CLINICAL OBSERVATIONS (DCO)
All F1 animals in cohorts 1A, 1B, 2A and 3 will be subjected to detailed clinical observations (DCO) outside their cages at the administration period (day 0) and subsequently once per week (in the morning) by the same trained technicians, whenever possible. For observation, the animals will therefore be removed from their cages and placed in a standard arena (50 × 37.5 × 25 cm). The scope of examinations and the scoring of the findings observed will be based on the current index of findings in GROSSE-Reprotox software and includes but is not limited to the following parameters listed:
1. Abnormal behavior in handling
2. Fur
3. Skin
4. Posture
5. Salivation
6. Respiration
7. Activity/arousal level
8. Tremors
9. Convulsions
10. Abnormal movements
11. Gait abnormalities
12. Lacrimation
13. Palpebral closure
14. Exophthalmos (Protruding eyeball)
15. Assessment of the feces excreted during the examination (appearance/consistency)
16. Assessment of the urine excreted during the examination
17. Pupil size

FOOD CONSUMPTION
Generally, food consumption will be determined once a week for the F1 rearing animals.

WATER CONSUMPTION
Generally, water consumption will be determined twice a week for F1 rearing animals.

BODY WEIGHTS
In general, the body weight of the F1 rearing animals will be determined once a week at the same time of the day (in the morning).

AUDITORY STARTLE RESPONSE HABITUATION IN COHORT 2A ANIMALS
On PND 24±1, the auditory startle response test will be carried out in all animals of cohort 2A using the SR-LAB; STARTLE RESPONSE SYSTEM (San Diego Instruments, San Diego, CA, U.S.A.). The examinations are started in the morning. Age-appropriate sized enclosures are used. The animals are given a 5 minute acclimation period in the response chamber with a 70 dBA background noise. Then the startle response is recorded in 50 trials at a startle stimulus sound level of 120 dBA with a 5 - 10 second variable interval between the trials. Response is recorded for 50 milliseconds. Measurement is carried out with the light and ventilator switched on in the measurement chambers; no feed or water is provided during the test. Data (maximum amplitude, latency to the peak of the response) are analyzed in 5 blocks of 10 trials each.

On completion of measurement, the data will be printed. The printouts will be signed with the project No., date and name, and will be part of the raw data.

FUNCTIONAL OBSERVATIONAL BATTERY (FOB) IN COHORT 2A ANIMALS
The FOB will be carried out once, between PND 63-75, in all animals of cohort 2A. The examinations will generally start in the morning at about 10:00 h. The FOB will be carried out in a randomized sequence. The animals will not be transferred to new cages before the test, nor will food or drinking water be withdrawn. The FOB will start with passive observations without disturbing the rats, followed by removal from the home cage, open field observations in a standard arena and sensory motor tests as well as reflex tests. The findings will be ranked according to their degree or severity, if applicable.

Home cage observation
The animals will be observed for a short period (about 10-30 seconds) in their cages with the lids closed in the rack, while disturbing influences (touching of the cage and loud noises) are avoided. While other abnormalities will be recorded, particular attention will be paid to the following parameters:
1. Posture
2. Tremors
3. Convulsions
4. Abnormal movements
5. Gait
6. Other findings

Open field observation
For observation, the animals will be removed from their cages by the investigator and placed in a standard arena (50 × 50 × 25 cm). Besides noting other abnormalities, the following parameters will be assessed:
1. Behavior on removal from the cage
2. Fur
3. Skin
4. Salivation
5. Nasal discharge
6. Lacrimation
7. Eyes/ pupil size
8. Posture
9. Palpebral closure
10. Respiration
11. Tremors
12. Convulsions
13. Abnormal movements/ stereotypes
14. Gait
15. Activity/ arousal level
16. Feces (consistency/color) excreted during the examination (2 minutes)
17. Urine (amount/color) excreted during the examination (2 minutes)
18. Rearing within 2 minutes
19. Other findings

Sensory-motoric test/Reflexes
The animals will be removed from the open field and will be subjected to the sensory motor and reflex tests listed below:
1. Reaction to an object being moved towards the face (Approach response)
2. Touch sensitivity (Touch response)
3. Vision (Visual placing response)
4. Pupillary reflex
5. Pinna reflex
6. Audition (Startle response)
7. Coordination of movements (Righting response)
8. Behavior during handling
9. Vocalization
10. Pain perception (Tail pinch)
11. Other findings
12. Grip strength of forelimbs
13. Grip strength of hindlimbs
14. Landing foot-splay test

Motor activity measurement
The Measurement of motor activity (MA) will be carried out between PND 63-75, in all animals of cohort 2A. The MA will be measured from 14:00 h onwards on the same day as the FOB will be performed. The examinations will be performed using the TSE Labmaster System supplied by TSE Systems GmbH, Bad Homburg, Germany. For this purpose, the animals will be placed in clean polycarbonate cages with a small amount of bedding for the duration of the measurement. Eighteen beams are allocated per cage. The number of beam interrupts will be counted over 12 intervals for 5 minutes per interval. The sequence at which the animals are placed in the cages will be selected at random. Since the starting time of each animal will be vary by the time needed to place the animals in the cages, the measurement periods will be staggered accordingly, beginning at about 14:00 h for the first animal. The individual measurement period begins when the 1st beam is interrupted and finishes exactly 1 hour later. No food or water will be offered to the animals during these measurements. After the transfer of the last animal in each case, the measurement room will be darkened. The program requires a file name for the measured data to be stored. This name consists of the reference number and a serial number.

CLINICAL EXAMINATIONS OF F1 GENERATION PUPS
Pup status and litter size after birth
The status (sex, live-born or stillborn) and number of all pups delivered from the F0 parents will be determined as soon as possible after birth. At the same time, the pups will also be examined for gross-morphological changes.

Pup viability/mortality
In general, a check will be made for any dead or moribund pups twice daily on workdays (once in the morning and once in the afternoon) or as a rule, only in the morning on Saturdays, Sundays or public holidays.

Pups, which died before the first determination of their status on the day of birth, will be defined as stillborn pups.

Clinical signs
All live pups will be examined daily for clinical symptoms (including gross-morphological findings) during the clinical inspection of the dams. If pups show particular findings, these will be documented with the dam concerned.

Nipple/areola presence
All surviving F1 male pups will be examined for the presence of nipples/areolas on PND 12 and again towards the end (either PND 20 or 21) of the lactation phase.

Anogenital distance
Anogenital distance (defined as the distance from the anus [center of the anal opening] to the base of the genital tubercle) measurements will be conducted in a blind randomized fashion, using a measuring ocular, on all live F1 pups on postnatal day 1.

Anogenital index
The anogenital index will be calculated according to the following formula:

anogenital index = anogenital distance [mm] / cubic root of pup weight [g]

Pup body weights
The pups will be weighed on the day after birth (PND 1) and on PND 4, 7, 14 and 21. Whenever possible, the individual weights will always be determined at the same time of the day (in the morning) and on PND 4 before standardization of the litters.

Additionally, the body weight of all F1 rearing animals will be determined on the day of vaginal opening and preputial separation.

Puberty onset
Developmental landmarks of puberty onset will be recorded on the following days in all cohort 1A, 1B, 2A, and 3 animals:
- From PND 27 onward: vaginal patency
- From PND 38 onward: balanopreputial separation

Recording will be completed when all animals are sexually mature or latest on PND 66 in males and in females.

CLINICAL PATHOLOGY IN COHORT 1A ANIMALS
Samples will be withdrawn from 10 cohort 1A males and females per group at termination.

Blood samples will be taken from animals by puncturing the retrobulbar venous plexus following isoflurane anesthesia. Blood sampling and blood examinations will be carried out in a randomized sequence. The list of randomization instructions will be compiled with a computer.

In the afternoon preceding the day of urinalysis, the animals will be individually transferred into metabolism cages (no food or drinking water provided); on the following morning, the individual urine specimens will be examined in a randomized sequence (the list of randomization instructions will be compiled with a computer).

The following parameters will be examined in all animals:
Hematology
1. Leukocytes
2. Erythrocytes
3. Hemoglobin
4. Hematocrit
5. Mean corpuscular volume (MCV)
6. Mean corpuscular hemoglobin (MCH)
7. Mean corpuscular hemoglobin concentration (MCHC)
8. Platelets
9. Differential blood count
10. Reticulocytes
11. Blood smear (only evaluated preparations will be archived)
12. Prothrombin time

Clinical chemistry
1. Alanine aminotransferase
2. Aspartate aminotransferase
3. Alkaline phosphatase
4. Serum  glutamyl transferase
5. Sodium
6. Potassium
7. Chloride
8. Inorg. phosphate
9. Calcium
10. Urea
11. Creatinine
12. Glucose
13. Total bilirubin
14. Total protein
15. Albumin
16. Globulins
17. Triglycerides
18. Cholesterol

Hormone evaluations
The following hormones will be determined in the serum samples:
1. T4 (thyroxine)
2. TSH
3.9.1.4. Urinalysis
1. Volume
2. Color
3. Turbidity
4. pH value
5. Protein
6. Glucose
7. Ketones
8. Urobilinogen
9. Bilirubin
10. Blood
11. Specific gravity
12. Microscopy of sediment

HORMONES IN PND 4 AND 22 F1-OFFSPRING
Blood sampling
Blood samples will be withdrawn from 10 surplus (culled) PND 4 offspring (as far as possible of different litters) per sex and group. PND 4 samples may be pooled per sex and litter if the available amount is not sufficient for a hormone analysis.
Blood samples will be withdrawn from 10 surplus PND 22 offspring (as far as possible of different litters) per sex and group.
The blood samples will be collected after decapitation (following isoflurane anesthesia) from the Vena cava cranialis.

Hormone evaluations
The following hormones will be determined in the serum samples:
1. T4 (thyroxine)
2. TSH

BLOOD SAMPLING FOR CHOLINE DETERMINATION IN COHORT 1B ANIMALS
Blood will be sampled from all cohort 1B males and females shortly before sacrifice. Blood samples will be taken from non-fasted animals by puncturing the retrobulbar venous plexus under Isoflurane anesthesia. On the specific sampling days, samples will be withdrawn about 4 hours after lights turned on.

Blood samples (1 mL) will be collected in prefilled commercial EDTA tubes (e.g. Fa. Sarstedt). The samples will be centrifuged. The plasma will be separated. The preparation of the samples will be done under cooling. All samples will be stored at -80°C until analysis.

DEVELOPMENTAL IMMUNOTOXICITY EXAMINATIONS IN COHORT 3 ANIMALS
T-cell dependent antibody response
All males and females of cohort 3 will be used to assess the functional responsiveness of major components of the immune system to a T-cell dependent antigen, sheep red blood cells (SRBC). For this purpose, the Anti SRBC-IgM ELISA of Life Diagnostics Inc, West Chester, USA (cat. no. 4200-2), will be performed. Each sample will be diluted 1:500. SRBC-IgM concentrations outside the standard curve range will be measured in a second test run with an appropriate dilution. Generally, two in-house controls will be measured with each test run. The ELISA will be measured with a Sunrise MTP-reader, Tecan AG, Maennedorf, Switzerland, and evaluated with the Magellan-Software of the instrument producer.

Immunization:
Route of administration: Intraperitoneal, using 1 mL tuberculin-syringes
Preparation: Details of preparation of the sheep red blood cells will be described in the raw data (4×108 SRBC/mL)
Frequency of administration: twice (within one action)
Administration volume: 0.5 mL per animal, split into two portions of 0.25 mL
Five days after immunization blood samples will be taken by puncturing the retrobulbar venous plexus following isoflurane anesthesia. Blood sampling and blood examinations will be carried out in a randomized sequence. The list of randomization instructions will be compiled with a computer.

Splenic lymphocyte subpopulation analysis
Ten males and females per group of cohort 1A will be used to perform a splenic lymphocyte subpopulation analysis (CD4+ and CD8+ T lymphocytes, B lymphocytes, and natural killer cells) using one half of the spleen, the other half of the spleen being preserved for histopathological evaluation.

Cyclophosphamide dependent immune system response
Ten male and ten female offspring derived from test group 10 will be selected at weaning to become a positive control group in this study. These animals will be treated with Cyclophosphamide monohydrate to prove the functional responsiveness of major components of the immune system of the rats against an immunosuppressant.

The following dose level of Cyclophosphamide monohydrate was selected to be sufficient to cause immunosuppressive activity as positive control substance:
4.5 mg/kg body weight/day: as dose level with expected immunosuppressive effects

CHOLINE DETERMINATION
Choline content will be determined in the liver of the blood-sampled PND 4 and 22 offspring.
Choline content will be determined in blood plasma and liver of cohort 1B animals. Details of the sampling are specified above, and in the section on pathology.

The choline analysis will be carried out at the Analytical Chemistry Laboratory of Experimental Toxicology and Ecology of BASF SE, Ludwigshafen, Germany, as a part of this study.

Methods of analysis
The analytical investigations will be performed according to the most recently authorized version of the control procedure. The control procedure will be described in the raw data and the report.
Postmortem examinations (parental animals):
NECROPSY
All F0 parental animals will be sacrificed by decapitation under isoflurane anesthesia. The exsanguinated animals will be necropsied and assessed by gross pathology, special attention being given to the reproductive organs. Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology.

Organ weights
The following weights will be determined in all animals sacrificed on schedule:
1. Anesthetized animals
2. Adrenal glands
3. Brain
4. Cauda epididymis
5. Epididymides
6. Heart
7. Kidneys
8. Liver
9. Lymph nodes, axillary (10 animals per sex per group, Cohort 1A animals only)
10. Lymph nodes, mesenteric (10 animals per sex per group, Cohort 1A animals only)
11. Ovaries
12. Pituitary gland
13. Prostate
14. Testes
15. Seminal vesicles including coagulating gland
16. Spleen
17. Thymus
18. Thyroid glands (with parathyroid glands)
19. Uterus (with cervix)

Organ/Tissue fixation
The following organs or tissues will be fixed in 4% formaldehyde solution or in modified Davidson’s solution:
1. All gross lesions
2. Adrenal glands
3. Bone marrow (femur)
4. Brain
5. Cecum
6. Cervix uteri
7. Coagulating glands
8. Colon
9. Duodenum
10. Epididymis, left (fixed in modified Davidson´s solution)
11. Esophagus
12. Eyes with optic nerve (fixed in modified Davidson’s solution)
13. Heart
14. Ileum
15. Jejunum (with Peyer’s patches)
16. Kidneys
17. Liver
18. Lungs
19. Lymph nodes, axillary
20. Lymph nodes, mesenteric
21. Mammary gland (male and female)
22. Ovaries (fixed in modified Davidson´s solution)
23. Oviducts
24. Pancreas
25. Pituitary gland
26. Prostate
27. Rectum
28. Sciatic nerve
29. Seminal vesicles
30. Skeletal muscle
31. Spinal cord (cervical, thoracic and lumbar cord)
32. Spleen
33. Stomach (forestomach and glandular stomach)
34. Target organs
35. Testis, left (fixed in modified Davidson ´s solution)
36. Thymus
37. Thyroid glands (with parathyroid glands)
38. Trachea
39. Urinary bladder
40. Uterus
41. Vagina
42. Vas deferens

The testes, epididymides, ovaries and eyes with optic nerve of animals that die or will have to be sacrificed intercurrently will be fixed in 4% buffered formaldehyde solution.

The left testis and left epididymis of all male F0 parental animals sacrificed at scheduled dates will be fixed in modified Davidson’s solution, whereas the right testis and epididymis will be used for sperm parameters.

In case of macroscopic findings in the right testis or right epididymis, this testis as well as the corresponding epididymis will be fixed for histopathological examination and the left testis and epididymis will be used for sperm analysis.

The uteri of all cohabited female F0 parental animals will be examined for the presence and number of implantation sites. The uteri of apparently nonpregnant animals or empty uterus horns will be placed in 1% ammonium sulfide solutions for about 5 minutes in order to be able to identify early resorptions or implantations (SALEWSKI's method). Then the uteri will be rinsed carefully in physiologic salt solution (0.9 % NaCl). When the examinations are completed, the uteri will be transferred to the Pathology Laboratory for further processing.

Histopathology
Fixation will be followed by histotechnical processing, examination by light microscopy and assessment of findings.

Special attention will be given to stages of spermatogenesis in the male gonads.

Special attention will be given to the synchrony of the morphology in ovaries, uterus, cervix, and vagina to the estrous cycle status. Any morphological patterns of asynchrony will be reported.

Animals that die or are sacrificed in a moribund state will be processed histotechnically and assessed like control animals. Special stains of individual organs of individual animals will be prepared if required.

Reproductive organs of all low- and mid-dose F0 parental animals suspected of reduced fertility, or for which estrous cyclicity or sperm quality were affected, will be subjected to histopathological investigation. Organs demonstrating potential treatment–related changes will also be examined in the lower dose groups.

Further examinations or procedures will depend on the results obtained in the study.
Postmortem examinations (offspring):
POSTMORTEM EXAMINATION OF F1 GENERATION PUPS
Pups sacrificed on schedule
On PND 4, all surplus F1 pups as a result of standardization will be sacrificed by decapitation under isoflurane anesthesia and blood will be sampled for determination of serum thyroid hormone concentrations. After sacrifice, these pups will be examined externally, eviscerated and their organs will be assessed macroscopically.

On PND 22, the surplus F1 generation pups that will not be used for the formation of the cohorts will be sacrificed by decapitation under isoflurane anesthesia with CO2 and blood will be sampled for thyroid hormone analyses.

Pups showing clinical symptoms or gross-morphological findings may be further examined using appropriate methods. Organs/tissues with gross-morphological findings may be preserved in a suitable manner for potential histopathological examination.

All F1 pups not used for other purposes without any notable findings will be discarded after their macroscopic evaluation.

Prematurely dead or sacrificed pups
Pups that die or are sacrificed in a moribund state will be eviscerated and examined for possible defects and/or the cause of death using appropriate methods. These animals will be preserved for this purpose, if necessary.

Liver sampling for choline determination in PND 4 pups
Livers of 10 male and 10 female culled PND 4 offspring per group (as far as possible of different litters) will be sampled.

As soon as possible after dissection of the animals, the livers will be weighed in toto. The weight of the livers will be recorded but will not be reported. Immediately after weighing, the liver samples will be frozen in liquid nitrogen. Until analysis, the samples will be stored at -80°C.

NECROPSY
COHORT 1A
All cohort 1A animals will be sacrificed by decapitation under isoflurane anesthesia. The exsanguinated animals will be necropsied and assessed by gross pathology, special attention being given to the reproductive organs. Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology.

Organ weights
The following weights will be determined in all animals sacrificed on schedule:
1. Anesthetized animals
2. Adrenal glands
3. Brain
4. Cauda epididymis
5. Epididymides
6. Heart
7. Kidneys
8. Liver
9. Lymph nodes, axillary
10. Lymph nodes, mesenteric
11. Ovaries
12. Pituitary gland
13. Prostate
14. Testes
15. Seminal vesicles including coagulating gland
16. Spleen
17. Thymus
18. Thyroid glands (with parathyroid glands)
19. Uterus (with cervix)

Organ/Tissue fixation
The following organs or tissues will be fixed in 4% formaldehyde solution or in modified Davidson’s solution:
1. All gross lesions
2. Adrenal glands
3. Bone marrow (femur)
4. Brain
5. Cecum
6. Cervix uteri
7. Coagulating glands
8. Colon
9. Duodenum
10. Epididymis, left (fixed in modified Davidson´s solution)
11. Esophagus
12. Eyes with optic nerve (fixed in modified Davidson’s solution)
13. Heart
14. Ileum
15. Jejunum (with Peyer’s patches)
16. Kidneys
17. Liver
18. Lungs
19. Lymph nodes, axillary
20. Lymph nodes, mesenteric
21. Mammary gland (male and female)
22. Ovaries (fixed in modified Davidson´s solution)
23. Oviducts
24. Pancreas
25. Pituitary gland
26. Prostate
27. Rectum
28. Sciatic nerve
29. Seminal vesicles
30. Skeletal muscle
31. Spinal cord (cervical, thoracic and lumbar cord)
32. Spleen
33. Stomach (forestomach and glandular stomach)
34. Target organs
35. Testis, left (fixed in modified Davidson ´s solution)
36. Thymus
37. Thyroid glands (with parathyroid glands)
38. Trachea
39. Urinary bladder
40. Uterus
41. Vagina
42. Vas deferens

The testes, epididymides, ovaries and eyes with optic nerve of animals that die or will have to be sacrificed intercurrently will be fixed in 4% buffered formaldehyde solution.
The left testis and left epididymis of all male animals sacrificed at scheduled dates will be fixed in modified Davidson’s solution, whereas the right testis and epididymis will be used for sperm parameters.
In case of macroscopic findings in the right testis or right epididymis, this testis as well as the corresponding epididymis will be fixed for histopathological examination and the left testis and epididymis will be used for sperm analysis.
Spleens of 10 animals per sex per group of cohort 1A will be split in two comparable parts (transversally). One part of the spleen will be fixed in 4% buffered formaldehyde and afterwards be embedded in paraplast. The other part of the spleen will be frozen at -80°C, being used to perform a splenic lymphocyte subpopulation analysis (CD4+ and CD8+ T lymphocytes, B lymphocytes, and natural killer cells).

Histopathology
Fixation will be followed by histotechnical processing, examination by light microscopy and assessment of findings.
Special attention will be given to stages of spermatogenesis in the male gonads.
Special attention will be given to the synchrony of the morphology in ovaries, uterus, cervix, and vagina to the estrous cycle status. Any morphological patterns of asynchrony will be reported.
Animals that die or are sacrificed in a moribund state will be processed histotechnically and assessed like control animals. Special stains of individual organs of individual animals will be prepared if required.
A differential ovarian follicle count (DOFC) will be conducted in test groups 10 and 13 (Cohort 1A females) according to Plowchalk et.al..
Further examinations or procedures will depend on the results obtained in the study.

COHORT 1B
All cohort 1B animals will be sacrificed by decapitation under isoflurane anesthesia. The exsanguinated animals will be necropsied and assessed by gross pathology, special attention being given to the reproductive organs. Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology.
Organ weights
The following weights will be determined in all animals sacrificed on schedule:
1. Anesthetized animals
2. Cauda epididymis
3. Epididymides
4. Liver
5. Ovaries
6. Pituitary gland
7. Prostate
8. Target organs
9. Testes
10. Seminal vesicles including coagulating gland
11. Uterus (with cervix)
Organ/Tissue fixation
The following organs or tissues will be fixed in 4% formaldehyde solution or in modified Davidson’s solution:
1. All gross lesions
2. Cervix uteri
3. Coagulating glands
4. Epididymis, left (fixed in modified Davidson ´s solution)
5. Liver
6. Ovaries (fixed in modified Davidson´s solution)
7. Pituitary gland
8. Prostate
9. Seminal vesicles
10. Target organs
11. Testis, left (fixed in modified Davidson ´s solution)
12. Uterus
13. Vagina

The testes, epididymides and ovaries of animals that die or will have to be sacrificed intercurrently will be fixed in 4% buffered formaldehyde solution.

Tissue sampling for choline determination
Tissue portions of the liver of all cohort 1 B males and females per group will be sampled.
As soon as possible after dissection of the animals, the livers will be weighed in toto. Immediately thereafter, the lobus sinister medialis of the liver will be separated from the remaining parts of the liver and weighed. The weight of the lobus sinister medialis will be recorded online in the pathology data system but will not be reported within the pathology report. Immediately after weighing, the liver lobe samples will be frozen in liquid nitrogen. Until analysis, the samples will be stored at -80°C.
For the remaining parts of the liver the standard procedures as described above will be followed.

Histopathology
All organs listed above will be preserved in adequate fixative. Histopathological processing and examination by light microscopy will only be conducted if results from cohort 1A are equivocal or in case the test compound is, from the results obtained so far, suspected to be a reproductive or endocrine toxicant.

Specifically, fixation of reproductive organs will be followed by histotechnical processing and examination by light microscopy and assessment of findings.

COHORT 2A
On postnatal day 77, cohort 2A animals will be weighed, counterbalanced, subjected to deep anesthesia (pentobarbital) and sacrificed by perfusion fixation.
SOERENSEN phosphate buffer will be used as the rinsing solution, and a fixation solution according to KARNOVSKY will be used as a fixative.
The perfusion fixed animals will be necropsied with regard to the question of neuropathology, and the visible organs will be assessed by gross pathology as accurately as is possible after a perfusion fixation. The cranial vault and the spinal cord will be opened and the skin will be removed from both hind extremities. In this state, the perfused animals will be stored in a fixation solution according to KARNOVSKY for at least 48 hours.
Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology. These animals will be sacrificed under isoflurane anesthesia with C02.

Organ weights
The following weights will be determined (the brain will be weighed after its removal but before further preparation):
1. Brain (including olfactory bulb)

The terminal body weights will be recorded to calculate the relative organ weights.

Length and width of brain
The length and maximum width of the brain will be measured in all animals (length: on a line extending from the rostral end of the frontal lobe to the caudal medulla oblongata of the cerebellum; width: pituitary region).

Organ/Tissue fixation
The following organs/tissue specimens will be carefully removed, processed histotechnically in accordance with the data given in the sections of this part of the study plan and examined:
1. All gross lesions
2. Brain with olfactory bulb
3. Eyes with retina and optic nerve
4. M. gastrocnemius
5. Nose (nasal cavity)
6. Pituitary gland
7. Sciatic nerve, proximal section
8. Spinal cord, cervical part (C1-C5)
9. Spinal cord, thoracic part (Th5-8)
10. Spinal cord, lumbar part (L1-L4)
11. Spinal ganglia (C1-C5 [3x])
12. Spinal ganglia (L1-L4 [3x])
13. Tibial nerve (on the knee), proximal section
14. Tibial nerve (nerve branch in the lower leg muscles), distal section
15. Trigeminal ganglia (s. Gasserian)
16. Root fibers, dorsal (C1-C6 and L1-L4)
17. Root fibers, ventral (C1-C6 and L1-L4)

The remaining animal body after trimming will be stored in neutrally buffered, 4% formaldehyde solution.

Neurohistopathology
The histotechnical processing, examination by light microscopy and assessment of findings is performed on organs/tissues mentioned above.
Tissues demonstrating potential treatment–related changes will also be examined in the lower dose groups.
Further examination procedures (e.g. special stains) will depend on the results of the study, especially on the presence of neurofunctional or neuropathological changes.

Morphometry
Thickness measurements of major brain layers (neocortex: frontal and parietal cortices, caudate nucleus/putamen, hippocampus, corpus callosum, cerebellum) will be performed. Measurements will be carried out bilaterally in the left and right halves of the brain with the exception of the corpus callosum and the cerebellum.

Selection of the planes:
• Measurements for the thickness of the neocortex, corpus callosum and caudate nucleus/putamen will be carried out in a cross section which approximates the plane of section on page 88 in Sherwood and Timiras (1970).
• Measurements for the thickness of the hippocampus will be carried out in a cross section which approximates the plane of section on page 110 in Sherwood and Timiras (1970).
• Measurements for the thickness of select folia of the cerebellum will be carried out in a midsagittal section through the vermis of the cerebellum which approximates the plane of section on page 134 in Sherwood and Timiras (1970).
Conduct of the measurements:
• Neocortex (frontal and parietal cortices): The width of the total cortical mantle (layers I-VI – from the surface of the pia mater to the white substance) will be measured vertically to a tangent over a region of the frontal and parietal cortices determined beforehand.
• Caudate nucleus/putamen: The largest lateral extension will be measured.
• Corpus callosum: The width will be measured at the middle line of the cross section.
• Hippocampus: The largest dorsoventral extension will be measured.
• Cerebellum: The width of a select folium (e.g. folium pyramis) will be measured in the middle of a line which runs vertically to a tangent from the tip to the base of the folium.

COHORT 2B
On postnatal day 22, cohort 2B animals will be weighed, counterbalanced, subjected to deep anesthesia (isoflurane) and sacrificed by perfusion fixation.
SOERENSEN phosphate buffer will be used as the rinsing solution, and neutrally buffered, 4% formaldehyde solution will be used as a fixative.
The perfusion fixed animals will be necropsied with regard to the question of neuropathology, and the visible organs will be assessed by gross pathology as accurately as is possible after a perfusion fixation. The cranial vault and the spinal cord will be opened and the skin will be removed from both hind extremities. In this state, the perfused animals will be stored in neutrally buffered, 4% formaldehyde solution for at least 48 hours.
Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology. These animals will be sacrificed under isoflurane anesthesia with C02.

Organ weights
The following weights will be determined (the brain will be weighed after its removal but before further preparation):
1. Brain (including olfactory bulb)

The terminal body weights will be recorded to calculate the relative organ weights.

Length and width of brain
The length and maximum width of the brain will be measured in all animals (length: on a line extending from the rostral end of the frontal lobe to the caudal medulla oblongata of the cerebellum; width: pituitary region).

Organ/Tissue fixation
The following organs/tissue specimens will be carefully removed, processed histotechnically in accordance with the data given in the sections of this part of the study plan and examined:

The following organs or tissues will be fixed in 4% formaldehyde solution:
1. All gross lesions
2. Brain with olfactory bulb
3. Eyes with retina and optic nerve
4. Nose (nasal cavity)
5. Pituitary gland
6. Trigeminal ganglia (s. Gasserian)

The animals and the tissue or organ material remaining after trimming will be stored in neutrally buffered, 4% formaldehyde solution.

Neurohistopathology
The histotechnical processing, examination by light microscopy and assessment of findings is performed on organs/tissues mentioned above.

COHORT 3
All Cohort 3 animals will be sacrificed by decapitation under isoflurane anesthesia. The exsanguinated animals will be necropsied and assessed by gross pathology. Animals which die intercurrently or are sacrificed in a moribund state will be necropsied as soon as possible after their death and assessed by gross pathology.

Organ weights
The following weights will be determined in all animals sacrificed on schedule:
1. Anesthetized animals
2. Spleen
3. Thymus

Organ/ tissue fixation
The following organs or tissues will be fixed in 4% buffered formaldehyde solution:
1. All gross lesions
2. Spleen
3. Thymus

Histopathology
Histotechnical processing and examination will be performed at the request of the sponsor only.

SURPLUS F1 GENERATION PUPS
All surplus F1 generation pups will be sacrificed by decapitation under isoflurane anesthesia with CO2. The exsanguinated animals will be necropsied and assessed by gross pathology with special emphasis on the reproductive organs.

Organ weights
The following weights will be determined in up to 10 animals per sex per group sacrificed on schedule:
1. Anesthetized animals
2. Brain
3. Spleen
4. Thymus

Organ/ Tissue fixation
The following organs or tissues of up to 10 animals per sex per group will be fixed in 4% buffered formaldehyde solution:
1. All gross lesions
2. Target organs
3. Brain
4. Mammary gland (male and female)
5. Spleen
6. Thymus
7. Thyroid glands

Histopathology
Histotechnical processing and examination will be performed at the request of the sponsor only.

Liver sampling for choline determination
Livers of the blood-sampled surplus PND 22 offspring (10 male and 10 female pups per group) will be sampled.
As soon as possible after dissection of the animals, the livers will be weighed in toto. The weight of the livers will be recorded but will not be reported. Immediately after weighing, the liver samples will be frozen in liquid nitrogen. Until analysis, the samples will be stored at -80°C.
Statistics:
Means and standard deviations will be calculated. In addition, the following statistical analyses will be carried out:
- Water consumption, food consumption, body weight and body weight change (parental animals, rearing animals and pups); estrous cycle length; mating days; duration of gestation; number of delivered pups per litter; developmental landmarks (days up to preputial separation or opening of the vagina), anogenital distance and index; implantation sites; postimplantation loss: DUNNETT test (two-sided)
- Number of live and dead pups and different indices (e.g. mating index, fertility index and gestation index) and number of litters with necropsy findings in pups; developmental landmarks (preputial separation or opening of the vagina): FISHER's exact test
- Absolute and relative pup organ weights; feces, rearing, grip strength forelimbs, grip strength hind-limbs, landing foot-splay test, motor activity, startle response: KRUSKAL-WALLIS and WILCOXON test (two-sided)
- Proportion of pups with necropsy findings per litter, presence of areolas/nipples: WILCOXON test (one-sided)
- Clinical pathology and sperm parameters: KRUSKAL-WALLIS and WILCOXON test
- Weight of the anesthetized animals and absolute and relative organ weights: KRUSKAL-WALLIS and WILCOXON test
- DOFC: WILCOXON test (one-sided)
- Brain weights: KRUSKAL-WALLIS and WILCOXON test
- Brain morphometry: linear measurements of selected brain regions: WILCOXON test
Clinical signs:
no effects observed
Description (incidence and severity):
Clinical observations for males and females (except gestation and lactation period)
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the male and female F0 parental animals in any of the groups.

Detailed clinical observations (DCO)
Male and female animals of all dose groups (100, 300 and 1000 ppm) did not show any abnormalities.

Clinical observations for females during gestation of F1 litters (Tab. IA-003)
There were no test substance-related clinical findings in any F0 parental females of all dose groups during the gestation period for F1 litter. One sperm positive and one sperm negative female of test group 02 (Nos. 262 and 275 - 300 ppm) and one sperm positive female of test group 03 (No. 291 - 1000 ppm) did not deliver F1 pups and had no implants in the uterus. One sperm negative female of test group 02 (No. 266) did not deliver F1 pups, but had implants in the uterus. These observations were not considered to be associated with the test compound.

Clinical observations for females and offspring during lactation of F1 litters (Tab. IA-004)
There were no test substance-related clinical observations observed in all F0 parental females of all dosed groups during the lactation period.
Two high-dose females (Nos. 292 and 311 - 1000 ppm) had lost their complete litters by PND 4. One female pup in control (dam No. 219, pup No. 7 - 0 ppm) showed chromodacryorrhea (both eyes) during PND 20 - 21 and one female pup in test group 03 (dam No. 306, pup No. 9 - 1000 ppm) showed microphthalmia (both eyes) during PND 18 - 21. These finding were not considered to be treatment-related.
Mortality:
no mortality observed
Description (incidence):
There were no test substance-related or spontaneous mortalities in any of the groups.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
The body weights of the high-dose F0 parental males (1000 ppm) were statistically significantly below the concurrent control from study day 7 onwards (up to 9%), as were the body weights of the mid-dose F0 parental males (300 ppm) from study day 28 onwards (up to 7%), until the end of the study. The body weights of the low-dose F0 parental male rats (100 ppm) were comparable to the concurrent control values throughout the entire study. The body weights of the high-dose F0 parental females were statistically significantly below the concurrent control from premating day 7 onwards, throughout gestation and lactation (up to 5%, 15% and 13%, respectively).
The body weights of the mid-dose F0 parental females were statistically significantly below the concurrent control values during PND 4 - 7 (up to 5%). There were no effects on body weights during premating and gestation in this dose group. The body weights of the low-dose females were comparable to the concurrent control during the entire study.
A consistently lower body weight gain was noted in the high-dose F0 parental males (1000 ppm) which became statistically significant during several study sections. The overall average decrease of weight gain was about 25%. The mid-dose F0 parental males (300 ppm) still gained less weight than the controls (overall about 23% less), however the difference to the control was statistically significant in fewer sections than in the high-dose group. A lower body weight gain was also noted for the low-dose F0 parental males (100 ppm) during the first week of treatment (14% below control). However, this was the only study section where this was seen and there was no effect on body weight during this study section and the remaining study. Thus, this is considered of no toxicological relevance.
The body weight gain of the high-dose F0 parental females (1000 ppm) was consistently below the concurrent control during gestation (overall about 33%) through early lactation (about 65%). In the other study sections periods of lower weight gain alternated with periods of comparable or higher weight gains, displaying an inconsistent course. The body weight gain of the low- and mid-dose F0 parental females (100 and 300 ppm) was widely comparable to the concurrent control, with some brief episodes of statistically significantly lower (GD 0-7) or higher (premating days 7-14) weight gains. Those minor changes were considered as spurious findings.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
Food consumption of the high-dose F0 parental males (1000 ppm) was statistically significantly below the concurrent control values during premating (up to 9%), while food consumption of all low- (100 ppm) and mid-dose (300 ppm) males was comparable to the concurrent control values.
Food consumption of the high-dose F0 females was statistically significantly below the concurrent control values throughout premating, gestation and lactation periods (up to 17%, 13% and 29%, respectively). For the mid-dose females (300 ppm) food consumption was statistically significantly below the concurrent control values during PND 4 - 7 (about 7%). There were no effects on food consumption during premating and gestation in this dose group. Food consumption of the low-dose females was comparable to the concurrent control during the entire study.
Water consumption and compound intake (if drinking water study):
effects observed, treatment-related
Description (incidence and severity):
Water consumption of all F0 parental male rats of all test groups was comparable to the concurrent control throughout the entire study. The statistically significantly increased water consumption in the low-dose males during study days 18 - 21 and in the mid- and high-dose males during study days 42 - 46 was considered as spontaneous in nature.
In the high-dose F0 females (1000 ppm) water consumption was statistically significantly below concurrent control during GD 14 – 18 (up to 18%) and during the entire lactation period (up to 45%). The overall mean water consumption during lactation was about 35% below control.
There was no effect on water consumption during premating in this dose group. For the mid-dose females (300 ppm) water consumption was statistically significantly below the concurrent control values during PND 1 - 2, 7 - 8 and 20 - 21 (about 11%, 11% and 15%, respectively). The overall mean water consumption during lactation was about 10% below control. There was no effect on water consumption during premating and gestation in this dose group. Water consumption of the low-dose females (100 ppm) was comparable to the concurrent control during the entire study.
Ophthalmological findings:
no effects observed
Haematological findings:
effects observed, treatment-related
Description (incidence and severity):
In the F0 generation rats of both sexes in test group 3 (1000 ppm) red blood cell (RBC) counts, hemoglobin and hematocrit values as well as the calculated red blood cell indices mean corpuscular volume (MCV) and mean corpuscular hemoglobin content (MCH) were decreased. Additionally, in the mentioned test group in males mean corpuscular hemoglobin concentration (MCHC) and in females relative reticulocyte counts were lower compared to controls, but MCHC was within the historical control range (MCHC 20.36-21.48 mmol/L) and in females mean values of the reticulocyte counts were not different to the controls. Therefore, both latter parameter changes were regarded as incidental and not treatment-related. In rats of both sexes of test group 2 (300 ppm) hemoglobin values were decreased. In males of this test group MCV and MCH and in females hematocrit values were decreased. MCV was already decreased in males of test group 1 (100 ppm). In males of test group 2 the measured parameter hemoglobin was within the historical control range (males: hemoglobin 8.6-9.3 mmol/L). In these individuals also the the calculated red blood cell parameters MCV and MCH were within or marginally below (MCV in test group 2) historical control ranges (males: MCH 1.00-1.10 fmol; MCV 48.4-51.9 fl). In females of test group 2 (300 ppm) as well as in female study controls hemoglobin and hematocrit values were not below, but above historical control ranges (females hemoglobin 8.7-9.3 mmol/L; hematocrit 0.410-0.435 L/L). Measured red blood cell parameters, hemoglobin in both sexes and hematocrit in females of test group 2 were only slightly lower compared to study controls (hemoglobin males mean – 6%; females mean -3%, hematocrit females mean -4% compared to respective control means, Mueller et al., 2006). Therefore, the alterations of the red blood cell parameters in test group 2 and test group 1 were regarded as treatment-related, but not adverse.
In rats of both sexes of test groups 2 and 3 (300 and 1000 ppm) prothrombin time (Hepatoquick’s test HQT) was reduced and in males of both test groups platelet counts were increased.
Regarding differential blood cell counts in test group 3 (1000 ppm), in rats of both sexes absolute basophil counts were decreased. Additionally, in males absolute neutrophil counts and in females absolute and relative monocyte and eosinophil counts and relative basophil counts were lower compared to controls. All absolute cell counts were within historical control ranges (males absolute neutrophils 0.70-1.35 Giga/L; absolute basophils 0.00-0.02 Giga/L; females absolute basophils 0.00-0.02 Giga/L; absolute monocyte 0.06-0.08 Giga/L; absolute eosinophils 0.08-0.11 Giga/L). Relative monocyte and eosinophil counts in females of test group 3 were marginally below the historical control ranges (relative monocytes 1.7-2.8 %, relative eosinophils 2.2-3.7 %). However, because no relevant change above historical control ranges occurred in the total and absolute differential cell counts all alterations in the differential blood cell counts in males and females of test group 3 (1000 ppm) were regarded as incidental and not treatment-related.
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
In male F0 generation rats of test group 3 (1000 ppm) aspartate aminotransferase (AST) and alkaline phosphatase (ALP) activities were increased. ALP activities were already higher in males of test group 2 (300 ppm), but in this test group it was the only relevantly changed clinical pathology parameter. ALP means in males of both mentioned test groups was less than 2fold higher compared to controls. Therefore, the ALP change in males of test group 2 was regarded as treatment-related, but not adverse (ECETOC Technical Report No. 85, 2002). Alanine aminotransferase (ALT) activities were lower in males of test group 2 (300 ppm) compared to controls, but the change was not dose-dependent and therefore it was regarded as incidental and not treatment-related. In male and female rats of test group 3 (1000 ppm) urea and albumin levels were increased. Albumin levels were already higher in females of test group 2 (300 ppm), but in these individuals, this was the only relevantly changed clinical pathology parameter (total calcium levels were higher in these individuals as consequence of higher albumin levels). Therefore, the albumin increase in females of test group 2 (300 ppm) was regarded as treatment-related, but not adverse (ECETOC Technical Report No. 85, 2002). Calcium levels were increased in rats of both sexes of test groups 2 and 3 (300 and 1000 ppm). Unbound and protein-bound calcium levels were measured. In order to maintain a constant free calcium level, total calcium level has to increase with higher albumin levels. Therefore, calcium changes were secondary to the albumin level increases and were regarded as treatment-related, but not adverse (ECETOC Technical Report No. 85, 2002). Calcium levels in males were within, those of females slightly above the historical control range (calcium males 2.45-2.67 mmol/L, females 2.49-2.61 mmol/L). The following clinical chemistry values were within historical control ranges and therefore, the alterations were regarded as incidental and not treatment-related: increased sodium and chloride values in rats of both sexes of test groups 2 and 3 (300 and 1000 ppm), increased total protein levels in females of test group 2 and 3, Decreased cholesterol levels in females of test group 3, increased triglyceride levels in males of test group 2 and 3 and in females of test group 2 (males: sodium 141.1-146.7 mmol/L, chloride 98.8-105.9 mmol/L, triglycerides 0.61-1.29 mmol/L; females: sodium 140.3-145.8 mmol/L, chloride 98.7-103.2 mmol/L, total protein 60.65-67.01 g/L, cholesterol 0.99-1.70 mmol/L). Triglyceride levels in females of test group 2 (300 ppm) were higher compared to controls, but the change was not dose-dependent and therefore, it was regarded as incidental and not treatment-related.
Urinalysis findings:
effects observed, treatment-related
Description (incidence and severity):
No treatment-related changes among urinalysis parameters were observed. In males of test groups 2 and 3 (300 and 1000 ppm) urine specific gravity was lower and urine volume was higher (not statistically significantly) compared to controls. In conjunction with histopathological alterations in the kidneys, this change was regarded as treatment-related and adverse.
Behaviour (functional findings):
no effects observed
Immunological findings:
no effects observed
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Histopathological findings: non-neoplastic:
effects observed, treatment-related
Description (incidence and severity):
Please see tables (Tables IC 10 – IC 14) of the attachment.
In the proximal tubules of the kidneys, there were signs of degeneration and regeneration characterized by apoptotic cell death, debris in the tubular lumen, increased numbers of large vesicular nuclei, increased basophilic staining and nuclear crowding. These findings were regarded to be treatment-related. Within the papilla, females of test group 02 and 03 (300 and 1000 ppm) showed an increase of multifocal mineral depositions. Nine males of test group 03 (1000 ppm) showed multifocal mineral depositions at the transition between the outer and inner medulla. The mineralization in males and females was regarded to be treatment-related. The liver revealed a minimal to mild centrilobular hypertrophy in single males of test group 03 (1000 ppm) and females of test groups 02 and 03 (300 and 1000 ppm). This finding was regarded to be treatment-related.

The submucosa of the glandular stomach of females was distended by an edema with infiltration of inflammatory cells. Furthermore, some females of test group 02 and 03 (300 and 1000 ppm) revealed erosion/ulcers in the mucosa of the glandular stomach. These findings in test groups 02 and 03 (300 and 1000 ppm) were regarded to be treatment-related. All other findings occurred either individually or were biologically equally distributed over control and treatment groups. They were considered to be incidental or spontaneous in origin and without any relation to treatment.

Fertility:
The female animals (Nos. 262, 275), which were not pregnant as well as their male mating partners (Nos. 62, 75) did not show relevant histopathological findings consistent with impaired fertility.
Histopathological findings: neoplastic:
no effects observed
Other effects:
effects observed, treatment-related
Description (incidence and severity):
CHOLINE CONCENTRATION IN BLOOD AND LIVER SAMPLES
The analytical results demonstrated the clear presence of choline in all plasma samples from the animals dosed with the test substance 2,2’-iminodiethanol (100 ppm, 300 ppm and 1000 ppm dosed animals) and in those from control, non-dosed animals. In general, it can be stated that the presence of the test substance 2,2’-iminodiethanol led to a reduction in the content of choline in the plasma samples analyzed. This effect appears to be dose-dependent, in that higher dose levels were associated with greater choline reduction. This effect is most clearly visible at lower dose levels (100 ppm and 300 ppm), at which dramatic plasma choline levels could be seen. At higher dosing levels, although further minor plasma choline content reduction was observed, this was by no means as drastic. The analytical results demonstrated the clear presence of choline in all liver samples from the animals dosed with the test substance 2,2’-iminodiethanol (100 ppm,300 ppm and 1000 ppm dosed animals) and in those from control, non-dosed animals. This was true from all time points investigated (4-day old pups, 22-day old pups and ~90-day old adolescents). In general, it can be stated that the presence of the test substance 2,2’-iminodiethanol led to a reduction in the content of choline in the liver samples analyzed. This effect appears to be dose-dependent, in that higher dose levels were associated with greater choline reduction, but only up to moderate dosing levels (300 ppm and 100 ppm, depending on the sampling day). At higher dosing levels, no further dramatic liver choline content reduction was observed. This effect was however, not observed in 4-day old animals, in which no clearly definable dosedependent trend is evident. In 22-day old animals this effect could be clearly observed, although the choline levels of the 100 ppm dosed animals have not yet attained minimal concentrations. In ~90-day old animals the effect is dramatic in that the liver choline levels of all non-control animals have reached an approximate minimum. Only a relatively minor further dose-dependency can be observed at this time point.
Reproductive function: oestrous cycle:
no effects observed
Description (incidence and severity):
Estrous cycle data, generated during the last 2 weeks prior to mating for the F1 litter, revealed regular cycles in the females of all test groups including the control. The mean estrous cycle duration in the different test groups was similar: 3.9 days in control and in the low- and mid-dose groups and 4.0 days in the high-dose group.

Differential ovarian follicle count - F0 generation parental animals
The results of the differential ovarian follicle count (DOFC) – comprising the numbers of primordial and growing follicles, as well as the combined incidence of primordial plus growing follicles – did not reveal significant differences between the control group 00 and animals of test group 03.
Reproductive function: sperm measures:
effects observed, non-treatment-related
Description (incidence and severity):
Concerning motility of the sperms and sperm head counts in the testis and in the cauda epididymidis no treatment-related effects were observed. In males of test group 3 (1000 ppm) incidence of abnormal sperms was slightly higher compared to controls. This was mainly due to missing heads, shortened heads, an abnormal hook of the head and in some samples combined morphology changes of head and tail. However, at least the first three mentioned morphological changes occurred also in the controls and the incidences were still in the range of historical controls (abnormal sperms 6.0-7.5 %). Therefore, these changes were regarded as incidental and not treatment-related.
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
Male reproduction data
For nearly all F0 parental males, which were placed with females to generate F1 pups, copulation was confirmed. Copulation was not confirmed for test group 02 male No. 75 paired with test group 02 female No. 275. Thus, the male mating index was 100% in the control and test groups 01 and 03 and 97% in test group 02. Fertility was proven for most of the F0 parental males within the scheduled mating interval for F1 litter. Two males of test group 02 (Nos. 62 and 75 - 300 ppm) did not generate F1 pups. One male of test group 02 (No. 66) and one male of test group 03 (No. 91) did not generate F1 pups, but implants were found in the uterus of the corresponding females (No. 266 - test group 02 and No. 291 - test group 03). Thus, the male fertility index ranged between 93% and 100% without showing any relation to dosing. This reflects the normal range of biological variation inherent in the strain of rats used for this study. The apparently infertile male rats did not show relevant gross or microscopic lesions. Thus, the test substance 2,2’-iminodiethanol did not adversely affect fertility and reproduction of the F0 generation parental males.

Female reproduction and delivery data
The female mating index calculated after the mating period for F1 litter was 100% in the control and test groups 01 and 03 and 97% in test group 02. The mean duration until sperm was detected (GD 0) varied between 2.2 and 3.1 days without any relation to the dose level. All female rats delivered pups or had implants in utero with the following exceptions:
 Test group 02
female No. 262 (mated with male No. 62) did not become pregnant
female No. 275 (mated with male No. 75) did not become pregnant
The fertility index varied between 97% and 100% without showing any relation to the dose level. This reflects the normal range of biological variation inherent in the strain of rats used for this study. The non-pregnant females had no relevant gross lesions or microscopic findings. The mean duration of gestation was 22.0 / 22.0 / 22.1 and 22.5** (**:p<=0.01) days in the control, low-, mid- and high-dose groups (0, 100, 300 and 1000 ppm), respectively. The gestation index was 100% in the control and test groups 01, 96% in test group 02 and 97% in test group 03. These values reflect the normal range of biological variation inherent in the strain of rats used for this study. The mean number of implantation sites was 12.3 / 12.2 / 11.4 and 7.8** (**:p<=0.01) implants/dam in test groups 00 - 03, respectively. The post-implantation loss did not show any statistically significant differences between the groups and all values were well within the historical control range. Corresponding to the lower number of implants the mean number of F1 pups delivered per dam was lower in the high-dose group as well, the numbers were 11.9 / 11.8 / 11.1 and 7.3** (**:p<=0.01) pups/dam in test groups 00 - 03, respectively. The rate of liveborn pups was not affected by the test substance, as indicated by live birth indices of 98% / 99% / 98% and 96% in test groups 00 - 03, respectively. Despite that the number of litters with stillborn pups was higher at the top dose (2 / 2 / 4 and 8** (**:p<=0.01) in test groups 00 - 03, respectively), the total number of stillborn pups was comparable between the groups (7 / 2 / 5 and 8 in test groups 00 - 03, respectively) and does not indicate an association to treatment. The number of cannibalized pups was 0 / 2 / 2 and 10** (**:p<=0.01) in test groups 00 - 03, respectively.
Key result
Dose descriptor:
NOAEL
Remarks:
based on a lower number of implants, prolonged/irregular estrous cycles as well as pathological changes in sexual organs, pituitary and mammary glands of both genders
Effect level:
100 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
gross pathology
Key result
Dose descriptor:
NOAEL
Remarks:
fertility and reproductive performance
Effect level:
300 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
reproductive function (oestrous cycle)
reproductive performance
Clinical signs:
not examined
Mortality:
mortality observed, treatment-related
Description (incidence):
Pup number and status at delivery
Whereas the litter size of the F1 litters at the top dose (1000 ppm) was lower (11.9 / 11.8 / 11.1 and 7.3** (**:p<=0.01) pups/dam in test groups 00 - 03, respectively) the postimplantation loss as well as the rates of liveborn and stillborn F1 pups were evenly distributed about the groups. Thus the lower litter size in the high-dose group was a consequence of a lower number of implants and not due to prenatal or perinatal mortality. Mortality are based on stillborn pups, dead pups, pups sacrificed moribund and cannibalized pups.

Pup viability/mortality
The viability index indicating pup mortality during early lactation (PND 0 - 4) varied between 100% / 99% / 99% and 93% in test groups 00 - 03. The lower index in the high-dose group was slightly below the historical control range. This slightly higher pup mortality at the high dose came from a higher number of dead (4 vs. 1 in control) and cannibalized (10** (**:p<=0.01) vs. 0 in control) pups which were distributed across 8 litters. In 2 of those 8 litters (292, 311) none of the pups survived. The absolute number of pups surviving early lactation (PND 0 - 4) varied between 350 / 349 / 291* (*:p<=0.05) and 191** (**:p<=0.01) in test groups 00 - 03. The significantly lower number in the mid-dose group reflects the incidentally lower number of liveborn litters in this group (27 vs. 30 in the control) and not an effect on pup survival. The lactation index indicating pup mortality on PND 4 - 21 was 100% in all test groups.
Body weight and weight changes:
not examined
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
not examined
Gross pathological findings:
not examined
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Reproductive function: oestrous cycle:
effects observed, treatment-related
Description (incidence and severity):
please compare with results for the F1 generation details
Reproductive function: sperm measures:
effects observed, non-treatment-related
Description (incidence and severity):
Sperm parameters
Concerning motility of the sperms and sperm head counts in the testis and in the cauda epididymidis no treatment-related effects were observed. In males of test group 13 (1000 ppm) incidence of abnormal sperms was slightly higher compared to controls, although not statistically significant. This was due to two individuals (no. 474 and 477) having sperm with missing heads, abnormal hook of the head, bent heads and combined morphological changes of head and tail. These morphological changes occurred also in controls. The mean incidence of the morphological changes on the whole was still in the range of historical controls (abnormal sperms 6.0-7.5 %). Therefore, this change was regarded as incidental and not treatment-related.
Reproductive performance:
not examined
Description (incidence and severity):
No F2 generatio nas been perfomed according to the agreed study protocol.
Clinical signs:
effects observed, non-treatment-related
Description (incidence and severity):
There were no test substance-related adverse clinical signs observed in any of the F1 generation pups of the different test groups.

F1 rearing animals, Cohort 1A (14R0781A)
Clinical observations for males and females
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the low- and mid-dose males and in any female animal of all dose groups. Two high-dose (1000 ppm) males (No. 476 and 478) showed high-stepping gait and piloerection recurring in several study sections. In addition, both animals had small testes which were detected in study week 5 for the first time.

F1 rearing animals, Cohort 1B (14R0781B)
Clinical observations for males and females
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the low- and mid-dose male and female animals. Several high-dose male and female animals (Nos. 693, 694, 697, 785, 797, 798) showed highstepping gait and piloerection recurring during several study sections. Affected male (No. 697) had small testes in addition, which were detected for the first time in test week 5. These findings were considered as treatment-related and adverse. Female No. 693 had also a malocclusion, which is considered as a spurious finding.

Detailed clinical observations (DCO)
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the low- and mid-dose male and female animals (100 and 300 ppm). A number of high-dose male and female animals (1000 ppm) exhibited high-stepping gait and piloerection on seveal occasions during the study, as follows:
- Male animal No. 693: high-stepping gait during study weeks 6 - 8 and piloerection on study week 2 and during weeks 6 - 8
- Male animal No. 697: high-stepping gait during study weeks 5 - 6 and piloerection on week 6
- Female animal No. 785: high-stepping gait and piloerection during study weeks 6 - 8
- Female animal No. 797: high-stepping gait on study week 8
- Female animal No. 798: high-stepping gait on study week 1 and during study weeks 5 - 8 as well as piloerection during study weeks 1 - 2 and study weeks 5 – 8.
These clinical observations were considered to be treatment-related and adverse.


F1 rearing animals, Cohort 2A (14R0782A)
Clinical observations for males and females
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the test substance-treated male as well as in low- and mid-dose female animals. One high-dose (1000 ppm) female (No. 937) showed high-stepping gait during study weeks 6 – 7 and piloerection during study weeks 5 - 7. These findings were considered as treatment-related and adverse. A malocclusion observed in high-dose male animal No. 833 is considered as a spurious finding.


F1 rearing animals, Cohort 2A (14R0782A)
Detailed clinical observations (DCO)
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the test substance treated male and in any low- and mid-dose female animal. One high-dose (1000 ppm) female (No. 937) showed high-stepping gait and piloerection during study week 6.

F1 rearing animals, Cohort 3 (14R0783)
Clinical observations for males and females
One high-dose (1000 ppm) male (No. 1240) showed high-stepping gait and piloerection recurring in several study sections. In addition, the animal had small testes which were detected in study week 4 for the first time. A malocclusion observed in this male was considered as a spurious finding. High-dose female animal No. 1336 showed high-stepping gait on study week 4 and piloerection during study weeks 3 – 4.

Detailed clinical observations (DCO)
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the low- and mid- dose male and female animals. One high-dose (1000 ppm) male (No. 1240) showed high-stepping gait on study week 2 and piloerection during study weeks 1 - 2. High-dose female animal No. 1336 showed high-stepping gait and piloerection on study week 4. These findings were considered as treatment-related and adverse.
Mortality / viability:
mortality observed, treatment-related
Description (incidence and severity):
Pup number and status at delivery
Whereas the litter size of the F1 litters at the top dose (1000 ppm) was lower (11.9 / 11.8 / 11.1 and 7.3** (**:p<=0.01) pups/dam in test groups 00 - 03, respectively) the postimplantation loss as well as the rates of liveborn and stillborn F1 pups were evenly distributed about the groups. Thus the lower litter size in the high-dose group was a consequence of a lower number of implants and not due to prenatal or perinatal mortality. Mortality are based on stillborn pups, dead pups, pups sacrificed moribund and cannibalized pups.

Pup viability/mortality
The viability index indicating pup mortality during early lactation (PND 0 - 4) varied between 100% / 99% / 99% and 93% in test groups 00 - 03. The lower index in the high-dose group was slightly below the historical control range. This slightly higher pup mortality at the high dose came from a higher number of dead (4 vs. 1 in control) and cannibalized (10** (**:p<=0.01) vs. 0 in control) pups which were distributed across 8 litters. In 2 of those 8 litters (292, 311) none of the pups survived.mThe absolute number of pups surviving early lactation (PND 0 - 4) varied between 350 / 349 / 291* (*:p<=0.05) and 191** (**:p<=0.01) in test groups 00 - 03. The significantly lower number in the mid-dose group reflects the incidentally lower number of liveborn litters in this group (27 vs. 30 in the control) and not an effect on pup survival. The lactation index indicating pup mortality on PND 4 - 21 was 100% in all test groups.

F1 rearing animals, Cohort 1A (14R0781A)
Mortality
There were no test substance-related mortalities in any of the groups up to a dose of 300 ppm. One high-dose male (No. 462, 1000 ppm) was sacrificed moribund in study week 0, after showing lateral position, apathy, hypothermia and diarrhea.

F1 rearing animals, Cohort 1B (14R0781B)
There were no test substance-related mortalities in any of the groups up to 300 ppm. One high-dose male (No. 687) and one high-dose female (No. 781) were found dead on study week 8, respectively.

F1 rearing animals, Cohort 2A (14R0782A)
Mortality
There were no test substance-related or spontaneous mortalities in any of the groups.

F1 rearing animals, Cohort 3 (14R0783)
Mortality
There were no test substance-related or spontaneous mortalities in any of the groups.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
Mean birth weights of the pups were comparable across all test groups including control. Mean body weights of the high-dose pups (1000 ppm) were statistically significant below the concurrent control values from PND 4 until weaning, the difference was about 13% at maximum. A slight decrease of pup weights was still noted in the mid-dose group beginning on PND 14 (male pups only) and definitive on PND 21 (about 6% below control). Mean body weight gain of the high-dose pups was statistically significantly below the concurrent control values during the entire lactation period (about 21 - 25% below control). A similar decrease though less pronounced (7 – 12%) was still noted for the mid-dose pups (300 ppm); the difference to the control became statistically significant during PND 4 - 7, 14 - 21 and 4 - 21. No test compound-related influence on F1 pup body weights/body weight gain were noted in the low-dose group (100 ppm).

F1 rearing animals, Cohort 1A (14R0781A)
The body weights of the high-dose males were statistically significantly below the concurrent control values during the entire study period, the final weight was about 24% below control. The body weights of the mid-dose males were statistically significantly below the concurrent control during the first week after weaning, and stayed below control until the end of the study (final weight about 6% below control) albeit the difference was not statistically significant. The body weights of the low-dose male rats were comparable to the concurrent control values throughout the study. The body weights of the high-dose females were statistically significantly below the concurrent control values during the entire study period, the final weight was about 15% below control. The body weights of the mid-dose females were below control throughout the study (final weight about 5% below control), although the difference was small and not statistically significant. The body weights of the low-dose females were comparable to the concurrent control during the entire study. The body weight gain of the high-dose males was statistically significantly below the concurrent control throughout the study except of the last study week, the average reduction of weight gain was about 25%. Mid-dose males gained about 5% less weight, the reduction was statistically significantly below the concurrent control during study days 35 - 49 (up to 28%). The body weight change of the low-dose males was comparable to the concurrent control during the entire study. The high-dose females gained statistically significantly less weight than the concurrent control during the study (14% on days 0 – 63), further their weight gain was statistically significantly
below control on days 0 – 7 and 42 - 49 (about 13% and 42%, respectively). The body weight change of the low and mid-dose females was comparable to the concurrent control during the study. The statistically significantly increased body weight change in the low dose females during study days 21 - 28 was considered to be spontaneous in nature.

F1 rearing animals, Cohort 2A (14R0782A)
Body weight data
The body weights of the high-dose males were statistically significantly below the concurrent control values during the entire study period, the final weight was about 13% below control. The body weights of the low- and mid-dose male rats were comparable to the concurrent control values throughout the study. The body weights of the high-dose females were statistically significantly below the concurrent control values during the entire study period, the final weight was about 16% below control. The body weights of the mid-dose females were below control throughout the study (final weight about 6% below control), although the difference was small and not statistically significant. The body weights of the low-dose females were comparable to the concurrent control during the entire study. The body weight gain of the high-dose males was statistically significantly below the concurrent control during several sections of the study, the average reduction of weight gain was about 11%. The body weight change of the low- and mid-dose males was comparable to the concurrent control during the entire study. There was no consistent effect on the body weight gain of the high-dose females, although they gained overall less weight than the concurrent control during the study (about 12%). The body weight change of the low and mid-dose females was comparable to the concurrent control during the study. The statistically significantly decreased body weight change in the low-dose females during study days 28 - 35 was considered to be spontaneous in nature.

F1 rearing animals, Cohort 3 (14R0783)
Body weight data
The body weights of the high-dose males were statistically significantly below the concurrent control values during the entire study period, the final weight was about 19% below control; as were the body weights of the high-dose females, their final weight was about 14% below control. The body weights of the low- and mid-dose male and female rats were comparable to the concurrent control values throughout the study. The body weight gain of the high-dose males was statistically significantly below the concurrent control during several sections of the study, the average reduction of weight gain was about 19%. The body weight change of the low- and mid-dose males was comparable to the concurrent control during the entire study. There was no consistent effect on the body weight gain of the high-dose females, although they gained overall less weight than the concurrent control during the study (about 8%). The body weight change of the low and mid-dose females was comparable to the concurrent control during the study. The statistically significantly increased body weight change in the mid dose females during study days 0 - 7 was considered to be spontaneous in nature.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
F1 rearing animals, Cohort 1A (14R0781A)
Food consumption
Food consumption of the high-dose males was statistically significantly below the concurrent control values during the entire study period (up to 22%), the average reduction was about 17%. Food consumption of the mid-dose males was below the concurrent control values throughout the study (average 6%), although the difference was statistically significant only during study days 0 – 7. Food consumption of all low-dose males was comparable to the concurrent control values throughout the study. Food consumption of the high-dose females was statistically significantly below the concurrent control occasionally during the study (up to 11%), however, the average food intake throughout the study was comparable to control. Food consumption of the low- and mid-dose females was comparable to the concurrent control throughout the study.

F1 rearing animals, Cohort 1B (14R0781B)
Food consumption of the high-dose males was statistically significantly below the concurrent control during the entire study (up to 18%), the average decrease was about 14%. There was no significant effect on food consumption of the low- and mid-dose males throughout the entire study. Food consumption of the high-dose females was statistically significantly below the concurrent control during major parts of the study period (up to 17%), the average dcrease was about 9%. There was no significant effect on food consumption of the low- and mid-dose females throughout the entire study.

F1 rearing animals, Cohort 2A (14R0782A)
Food consumption
Food consumption of the high-dose males was below the concurrent control during the entire study period (up to 12%), the average reduction was about 11%.
Food consumption of the high-dose females was statistically significantly below the concurrent control during study days 0 - 7 and 14 - 21 (about 15% and 10%, respectively), the average reduction was about 8%. Food consumption of the low- and mid-dose males and females was comparable to the concurrent control throughout the study.

F1 rearing animals, Cohort 3 (14R0783)
Food consumption
Food consumption of the high-dose males was below the concurrent control during the entire study period although the difference was not statistically significant, the average reduction was about 11%. Food consumption of the high-dose females was also below the concurrent control throughout the study, although the difference was slight (about 5%) and not statistically significant. Food consumption of the low- and mid-dose males and females was comparable to the
concurrent control throughout the study.
Water consumption and compound intake (if drinking water study):
effects observed, treatment-related
Description (incidence and severity):
F1 rearing animals, Cohort 1A (14R0781A)
Water consumption of the high-dose males was below the concurrent control values throughout the study (average about 13% lower), the difference became statistically significant on study days 0 – 21, 24 - 28 and 49 - 56 (up to 24%, 14% and 19%, respectively). A statistically significantly reduced water consumption was also noted for the mid-dose males during study days 7 - 10 (about 10%). Water consumption of the high-dose females was below the concurrent control values throughout the study (average about 14% lower), the difference became statistically significant during several parts of the study period (up to 25%). Mid-dose females occasionally had statistically significantly lower water consumption during the study period (up to 18% below control). Water consumption of the low-dose male and female animals was comparable to the concurrent control values during the entire study.

F1 rearing animals, Cohort 1B (14R0781B)
Water consumption of the high-dose males was below the concurrent control values throughout the study (average about 13% lower), the difference became statistically significant during major parts of the study (up to 23%). A statistically significantly reduced water consumption was also noted for the mid-dose males towards the end of treatment (study days 52 – 59; about 10 - 17%). Water consumption of the low-dose males was consistently comparable to the concurrent control during the entire study, except of a decrease on days 52 – 56 which is considered of no toxicological relevance. Water consumption of the high-dose females was below the concurrent control values throughout the study (average about 16% lower), the difference became statistically significant during major parts of the study period (up to 29%). Mid-dose females also had a consistenly lower water consumption than control (average about 10%), the difference becoming statistically significant during study days 24 – 28, 31 – 38 and 49 - 56 (up to 18% below control). Water consumption of the low-dose females was comparable to the concurrent control during the entire study.


F1 rearing animals, Cohort 2A (14R0782A)
Water consumption
Water consumption of the high-dose males was below the concurrent control values during major parts of the study (average about 7% lower), the difference became statistically significant on study days 0 - 11 (up to 22%). Water consumption of the low and mid-dose males was comparable to the concurrent control
during the entire study. Water consumption of the high-dose females was below the concurrent control values throughout the study (average about 13% lower), the difference became statistically significant during several parts of the study period (up to 18%). Mid-dose females occasionally had statistically significantly lower water consumption during the study period (up to 23% below control). Water consumption of the low-dose female animals was comparable to the concurrent control values during the entire study.

F1 rearing animals, Cohort 3 (14R0783)
Water consumption
Water consumption of the high-dose males was below the concurrent control values during major parts of the study (average about 9% lower), the difference became statistically significant on study days 0 - 7 (up to 16%). Water consumption of the low and mid-dose males was comparable to the concurrent control during the entire study. Water consumption of the high-dose females was below the concurrent control values throughout the study (average about 11% lower), the difference became statistically significant during several parts of the study period (up to 18%). Water consumption of the low- and mid-dose female animals was comparable to the concurrent control values during the entire study.
Ophthalmological findings:
not examined
Haematological findings:
effects observed, treatment-related
Description (incidence and severity):
In rats of both sexes of the F1 generation (PND92) of test group 13 (1000 ppm) red blood cell (RBC) counts, hemoglobin and hematocrit values as well as red blood cell indices mean corpuscular volume (MCV) and mean corpuscular hemoglobin content (MCH) were decreased. In rats of both sexes of test group 12 (300 ppm) hemoglobin and hematocrit values were decreased and additionally in females RBC counts were decreased and in males of the mentioned test group MCV and MCH were lower compared to controls. In females of this test group hemoglobin values and RBC counts were within and hematocrit values were marginally below historical control ranges (females: hemoglobin 8.1-8.9 mmol/L, RBC 7.49-7.93 Tera/L, hematocrit 0.383-0.421 L/L). However, hematocrit mean in females of test group 12 was only 9% lower compared to the controls and this was the only relevantly changed red blood cell parameter. In males of test group 12, the measured red blood cell parameters hemoglobin and hematocrit were within historical control range (males hemoglobin 8.4-9.5 mmol/L, hematocrit 0.400-0.446 L/L) and only the calculated red blood cell indices MCV and MCH were below historical control ranges (males MCV 50.1-54.7 fL, MCH 1.05-1.18 fmol). Therefore, the red blood cell parameter changes in rats of both sexes of test group 12 (300 were regarded as treatment-related but not adverse. In females of test group 11 (100 ppm) RBC counts, hemoglobin and hematocrit values and in males of the same test group MCH were still lower compared to study controls, but all values apart from MCH in males were within historical control ranges (females: hemoglobin 8.1-8.9 mmol/L, RBC 7.49-7.93 Tera/L, hematocrit 0.383-0.421 L/L). In males of test group 11 MCH was still lower compared to historical controls, but the measured red blood cell parameters (i.e. RBC counts hemoglobin and hematocrit values) were not changed. Therefore, the mentioned red blood cell alterations in both sexes of test group 11 (100 ppm) were regarded as incidental and not treatment-related. In females of test group 13 (1000 ppm) prothrombin time (Hepatoquick’s test, HQT) was reduced. In males of test groups 12 and 13 (300 and 1000 ppm) platelet counts were increased. In females of test group 12 (300 ppm) platelet counts were increased, but they were not dose-dependently changed. Therefore, these platelet count alterations were regarded as incidental and not treatment-related. Regarding the differential blood cell counts, in females of test group 13 (1000 ppm) absolute and relative monocyte counts were decreased and this is regarded as adverse finding. The following changes in the differential blood cell counts were within the historical control ranges. Therefore, they were regarded as incidental and not treatment-related: decreased absolute and relative monocyte counts in females of test groups 11 and 12 (100 and 300 ppm), decreased relative basophil counts in females of test groups 11, 12 and 13, increased relative lymphocyte counts in males of test group 13, decreased relative monocyte and eosinophil counts in males of test groups 12 and 13, decreased absolute monocyte counts in males of test group 12 (females: relative monocytes 1.2-1.9 %, absolute monocytes 0.05-0.08 Giga/L, relative basophils 0.1-0.5 %; males: relative lymphocytes 77.7-87.0 %, relative monocytes 0.9-2.3 %, relative eosinophils 0.8-2.0 %, absolute monocytes 0.07-0.14 Giga/L).
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
In rats of both sexes of the F1 generation (PND92) of test group 13 (1000 ppm), aspartate aminotransferase (AST) activities were increased. Additionally, in males of test groups 12 and 13 (300 and 1000 ppm) alkaline phosphatase (ALP) activities were higher compared to controls. However, the ALP activity mean in test group 2 was within the historical control range (ALP 1.64-2.80 μkat/L) and therefore, this change was regarded as incidental and not treatment-related. In male and female rats of test group 13 (1000 ppm) urea levels were increased. In females of test groups 11, 12 and 13 (100, 300 and 1000 ppm) creatinine levels were decreased and glucose levels were increased. However, both parameters were not dosedependently changed regarding means as well as medians. Therefore these alterations were regarded as incidental and not treatment-related. In females of test groups 12 and 13 (300 and 1000 ppm) total protein, albumin and calcium levels were higher compared to controls. Albumin levels were within historical control ranges and total protein levels of test group 12 were within and those of test group 13 marginally above the historical control range (albumin 38.12-42.64 g/L, total protein 58.83-65.33 g/L). Calcium values were slightly above the historical control range (calcium 2.38-2.63 mmol/L). As already mentioned in the corresponding paragraph in the F0 generation, unbound and protein-bound calcium levels were measured. In order to maintain a constant free calcium level, total calcium level has to increase with higher albumin levels. Therefore, calcium changes were secondary to the albumin level increases and were regarded as treatment-related, but not adverse (ECETOC Technical Report No. 85, 2002). Albumin and total protein alterations were regarded as incidental and not treatment-related. The following parameter changes were within historical control ranges and therefore they were regarded as incidental and not treatment-related: increased sodium in both sexes of test groups 12 and 13 (300 and 1000 ppm), increased chloride levels in both sexes of test group 13, increased calcium levels in males of test group 13, decreased levels of cholesterol in females of test groups 12 and 13 (in test group 13 not statistically significant) (males: sodium 140.7-145.4 mmol/L, chloride 97.9-103.4 mmol/L, calcium 2.42-2.71 mmol/L; females: sodium 140.5-144.1 mmol/L, chloride 100.1-104.9 mmol/L, cholesterol 1.04-1.65 mmol/L). Inorganic phosphate levels were decreased and triglyceride levels were increased in females of test group 12 (300 ppm), but the changes were not dose-dependent and therefore they were regarded as incidental and not treatment-related. Calcium levels were already increased in females of test group 11 (100 ppm), but this value was at the upper border of the historical controls (females, calcium 2.38-2.63 mmol/L) and this was the only changed clinical chemistry parameter among these individuals. Therefore, calcium level increase in females of test group 11 was regarded as maybe treatment-related, but not adverse (ECETOC Technical Report No. 85, 2002).
Urinalysis findings:
effects observed, treatment-related
Description (incidence and severity):
In F1 rats of both sexes (PND 92) no treatment-related, adverse changes among urinalysis parameters were observed. In females of test groups 11, 12 and 13 (100, 300 and 1000 ppm) urine volume was lower and urine specific gravity was higher compared to controls. However, both parameters were not dose-dependently changed. Statistically significance in this case occurred because of a high urine volume and a low specific gravity in the controls. Therefore, the changes were regarded as incidental and not treatment-related.
Sexual maturation:
effects observed, treatment-related
Description (incidence and severity):
Vaginal opening
Each female F1 pup, which was selected to be raised as F1 female (cohorts 1A, 1B, 2A, 3), was evaluated for commencement of sexual maturity. The first day when vaginal opening was observed was PND 27, the last was PND 38. The mean number of days to reach the criterion in the control and 100, 300 and 1000 ppm test groups amounted to 29.7 / 30.2 / 30.6* (*:p<=0.05) and 32.0** (**:p<=0.01) days. The mean body weight on the day, when vaginal opening was recorded, amounted to 85.1 g / 87.4 g / 84.8 g and 80.4 g in test groups 00-03. The values for pubertal age and weight of control females were both at the lower end of the historical control range; thus the apparent statistical increase in age at attainment in the mid dose group is considered to be due to the very low concurrent control values in this study and not treatment-related. In the high-dose group the pubertal age is just above the upper limit of the historical range while the weight at puberty is below the historical control range. This clearly indicates that the later onset of puberty is a consequence of a general developmental delay and not a specific effect on the timing of puberty.

Preputial separation
Each male F1 pup, which was selected to be raised as F1 male (cohorts 1A, 1B, 2A, 3), was evaluated for commencement of sexual maturity. The first day when preputial separation was observed was PND 38, the last was PND 66. The mean number of days to reach the criterion in the control and 100, 300 and 1000 ppm test groups amounted to 41.1 / 41.2 / 41.8 and 43.3** (**:p<=0.01) days. The mean body weight on the day, when preputial separation was recorded, amounted to 168.5 g / 168.6 g / 165.7 g and 145.2 g** (**:p<=0.01) in test groups 00-03. In the high-dose group the pubertal age is well within of the historical range while the weight at puberty is distinctly below the historical control range. This indicates that the apparent later onset of puberty may be a spurious finding, and if at all, is a consequence of a general developmental delay and not a specific effect on the timing of puberty.

F1 rearing animals, Cohort 1A (14R0781A)
Estrous cycle data
Estrous cycle data, generated during a maximum of 3 weeks, revealed regular cycles in the females of control as well as of the low- and mid-dose groups. The mean estrous cycle duration was prolonged in the high-dose group (4.1 / 4.0 / 4.2 and 5.5* (*:p<=0.05) days in test groups 10-13). There was no particular cycle phase prolonged, some high-dose F1A females with a longer cycle (beyond 5 days) had a prolonged diestrous, others had equally longer estrous and/or metestrous phases. One female (No. 562) stayed exceptionally long in estrous (11 days in 2 cycles). One female (No. 597) was in diestrous for 14 days which is usually considered as a sign of pseudopregnancy, without this animal the average cycle duration would have been 4.9 days.

F1 rearing animals, Cohort 1B (14R0781B)
Estrous cycle data
Estrous cycle data, generated during a maximum of 3 weeks, revealed regular cycles in the females of control as well as of the low- and mid-dose groups. The mean estrous cycle duration was prolonged in the high-dose group (4.2 / 4.0 / 4.1 and 5.0* (*:p<=0.05) days in test groups 10-13). A number of females displayed irregular cycles, however they did not show a unique pattern of change. Specifically, there was no particular cycle phase prolonged in all females, some females had a prolonged diestrous, others had equally longer estrous and/or metestrous phases. Three females (Nos. 777, 785, 797) had no complete cycle at all (estrous missing), one of them (No. 797) was in diestrous during almost the entire observation period. Another female (No. 798) stayed in diestrous for 10 days, had then one regular cycle and afterwards remained in estrous for the remaining observation period.
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
F1 rearing animals, Cohort 1A
Please see tables (Tables IC 16 – IC 23) of the attachment.
The reduction in terminal body weight in males of test group 12 and 13 (300 and 1000 ppm) and females of test group 13 (1000 ppm) was regarded to be treatment-related. The increase in kidney and liver weight (relative) was partly caused by the terminal body weight decrease but as most of the weights were outside the historical control values (see PART III) they were assessed to be treatment-related.
The increased relative adrenal gland weight in test group 13 males was higher than what could be expected due to the terminal body weight decrease. But as there were no histopathologic findings corresponding to the weight increase this finding might be treatment-related but was not assessed to be adverse. The same comes true for the increase in adrenal weight in test group 12 (300 ppm) females. The male reproductive organs in test group 13 (1000 ppm) were mainly decreased by the reduction in terminal body weight but this change would be less pronounced without the three male animals which revealed histopathologic findings in these organs. All other changed weight parameters were regarded to have been caused by the decrease in terminal body weight.

F1 rearing animals, Cohort 1B
Please see tables (Tables IC 33 – IC 36) of the attachment.
The significant reduction in terminal body weight of males and females of test group 12 and 13 (300 and 1000 ppm) was regarded to be treatment-related. The terminal body weight of males and females of test group 12 (300 ppm) was within the range of historical controls (see PART III). The liver weight increase in males and females of all treated groups was regarded to be treatment-related.The significant decrease in absolute weight of male sex organs in test group 13 (1000 ppm) was regarded to have been mainly caused by the reduced terminal body weight but it would have been less pronounced without the three male animals which revealed histopathologic findings in these organs. The same comes true for the significant increase of relative weights of pituitary gland in males of test group 13 (1000 ppm) and testes weight in test group 12 and 13 (300 and 1000 ppm). Whereas, for the significantly decreased prostate weight in test group 13 (1000 ppm) a treatment-related effect cannot be excluded, as the relative weight was still decreased. But as it was decreased less than in F1 generation, cohort 1A animals, it was still regarded to be secondary to the body weight reduction. The significantly changed ovary weights in test group 12 and 13 (300 and 1000 ppm) were regarded to be incidental due to the to a missing dose response relationship and because it cannot be explained by histologic findings. The significantly increased relative uterus weights in test group 12 and 13 (300 and 1000 ppm) were also thought to be caused by the terminal body weight reduction.

F1 rearing animals, Cohort 3 (Immunotoxicity cohort)
Please see tables (Tables IC 40 – IC 47) of the attachment.
Male and female animals of test group 13 (1000 ppm) revealed a significant decrease in terminal body weight which was regarded to be treatment-related. The increase in relative spleen weight in males of this test group was thought to be a consequence to the body weight reduction and therefore secondary.

Cyclophosphamide monohydrate
Absolut and relative organ weights
When compared to the control group 10 (set to 100%), the mean absolute and relative weight parameters of test group 14 (positive control) were significantly decreased A significant decrease in absolute and relative weights of the spleen and thymus occurred in the positive control male and female animals. This result was expected.
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
The light-brown discoloration in the liver of twelve males and two females of test group 13 (1000 ppm) corresponded in many cases to the diagnosis “fatty change, peripheral” and were regarded to be treatment-related. The size reduction of prostate, seminal vesicle, epididymides and testes in three males (animals No. 476, 478, and 479) of test group 13 (1000 ppm) corresponded to histopathological findings. The foci in the glandular stomach were partly diagnosed as erosion/ulcer, but histopathologically there was not a significantly higher incidence compared to control. Therefore, it was regarded to be incidental.

F1 rearing animals, Cohort 1B (14R0781B)
Gross pathology
(Table IC 37)
In test group 13 (1000 ppm) three males (animal Nos. 693, 694, 697) revealed reduced size of the testes, epididymides, prostate and seminal vesicle. Four females (animal Nos. 777, 785, 797, 798) of test group 13 (1000 ppm) had reduced size of the ovaries. All these male and female animals revealed a severely lower body weight when compared to the average of this group or the control group. The weight decrease was therefore regarded to be treatmentrelated but secondary to the reduced terminal body weight. All other findings occurred either individually or were biologically equally distributed over control and treatment groups. They were considered to be incidental or spontaneous in origin and without any relation to treatment.

F1 rearing animals, Cohort 3 (Immunotoxicity cohort)
Gross pathology
(Table IC 48 – IC 49)
One male of test group 13 (1000 ppm) revealed reduced size of testes, epididymides, seminal vesicle and prostate. This was regarded to be caused by the severely reduced terminal body weight and therefore regarded to be treatment-related but secondary. All other findings occurred either individually or were biologically equally distributed over control and treatment groups. They were considered to be incidental or spontaneous in origin and without any relation to treatment.
Histopathological findings:
effects observed, treatment-related
Description (incidence and severity):
F1 rearing animals, Cohort 1A
Please see tables (Tables IC 26 – IC 31) of the attachment.
Similar to the findings described for the F0 generation animals, in the proximal tubules of the kidneys signs of degeneration and regeneration (apoptotic cell death, debris in the tubular lumen, increased numbers of large, vesicular nuclei, increased basophilic staining, nuclear crowding) were seen. These findings were regarded to be treatment-related. Within the papilla, females of test group 13 (1000 ppm) showed an increase of multifocal mineral deposition. Males of test group 12 and 13 (300 and 1000 ppm) showed multifocal mineral depositions at the transition between the outer and inner medulla. The mineralization in males and females was regarded to be treatment-related. Males and females of test group 12 and 13 (300 and 1000 ppm) showed a centrilobular hypertrophy in the liver. Three males of test group 13 (1000 ppm) revealed a peripheral hypertrophy. Furthermore, males of test group 12 and 13 (300 and 1000 ppm) and females of test group 13 (1000 ppm) showed clear, round vacuoles in the peripheral area. Via the ORO stain it could be demonstrated that they representing lipid vacuoles. These findings wereregarded to be treatment-related.

In the male mammary gland of test group 13 (1000 ppm) there was an increase of female-like mammary gland structures (tubule-alveolar structure, less amount of cytoplasm, increase in basophilia). One male revealed a moderate diffuse hyperplasia of the mammary gland tissue in addition.

Females in test group 13 (1000 ppm) revealed a higher incidence of eosinophilic amorphous material which was secreted into the ducts. These findings were regarded to be treatmentrelated. In males a similar secretion was seen, but there was no significant difference between control and treated animals.

Three males (animal Nos. 476, 478, 479) of test group 13 (1000 ppm) showed the histological picture of juvenile and immature testes which were characterized by lower numbers or not fully developed and differentiated germ cell rows. In addition, there were no or only very few sperm present within testicular tubules. Consequently, these three animals did not have sperm within the epididymides (aspermia) and reduced size of the secondary sexual organs (prostate, seminal vesicle, and coagulating gland). One male (animal No. 479) showed multifocal degeneration of the testicular tubules in addition. The immaturity in these animals was regarded to be a consequence to the reduced body weight. The tubular degeneration in animal No. 479 was assumed to be treatment-related. Only in one male (animal No. 462), which was sacrificed in a moribund state, the reduced size of the ductus deferens did not match a finding in the testes. In this animal, the premature sacrifice was regarded to be responsible for the smaller size. The vacuolation in the ductus deferens in four males of test group 12 (300 ppm) might still be treatment-related but due to the missing findings in all other sexual organs it was not assumed to be adverse.

In test group 13 (1000 ppm) one female revealed a diffuse atrophy of the ovary. Six females revealed luteal cysts. These findings were regarded to be treatment related. The one cyst in a test group 12 female (300 ppm) might be treatment-related, but as luteal cysts sometimes also occur in control animals it might also be an incidental finding (McInnes 2012).

In the pars distalis of the pituitary gland of males and females of all test groups eosinophilic cysts were observed. These cysts differed from the cysts that occur sporadically as background lesion in the pituitary gland, also in this study. Spontaneous cysts which were observed in control and treated animals had a ciliated epithelium and a mucinous content. The treatmentrelated very small eosinophilic cysts revealed a non-ciliated, irregular border with an eosinophilic homogenous content and were multifocally distributed within the pars distalis occasionally revealing clear vacuoles at the border. In the thyroid glands of males of test group 11 and 12 (100 and 300 ppm) there was a slight increase in hypertrophy of the follicular epithelium. As in test group 13 (3000 ppm) no such finding was observed it was regarded to be incidental. Branchiogenic cysts occurred in control and treated animals and are regarded to be incidental findings. In these study, there was a slight increase in test group 13 animals (1000 ppm) compared to control animals. Nevertheless, it was regarded to be incidental and not related to treatment. All other findings occurred either individually or were biologically equally distributed about control and treatment groups. They were considered to be incidental or spontaneous in origin and without any relation to treatment.

Differential ovarian follicle count – F1 rearing animals, Cohorts 1A and 1B
(Table IC 32)
The results of the differential ovarian follicle count (DOFC) – comprising the numbers of primordial and growing follicles, as well as the combined incidence of primordial plus growing follicles – revealed significant differences between the control group 10 and animals of test group 13.

F1 rearing animals, Cohort 1B (14R0781B)
Please see tables (Tables IC 38 – IC 39) of the attachment.
Three males (animal Nos. 693, 694, 697) of test group 13 (1000 ppm) showed the histological picture of juvenile and immature testes which were characterized by lower numbers or even none fully developed and differentiated germ cell rows. In addition, there were no or only very single sperm present within testicular tubules. Consequently, these three animals did not have sperm within the epididymides (aspermia) and reduced size of the secondary sexual organs (prostate, seminal vesicle, and coagulating gland). Two males (animal No. 694 and 697) showed a slight to moderate multifocal degeneration of the testicular tubules in addition. Two males revealed a minimal degeneration in the testicle, only. These findings were regarded to be treatment-related. In test group 1 (100 ppm) one male also had an immature testis and tubular degeneration.

Four females (animal Nos. 777, 785, 797, 798) of test group 13 (1000 ppm) revealed a diffuse atrophy of the ovaries, represented by lower numbers of functional corpora and reduction in size. Two of these females did not have any corpora lutea present. Furthermore, in the ovaries of six females of this test group and one female of test group 12 (300 ppm) luteal cysts were observed. These were large cystic cavities surrounded by luteinized and non-luteinized granulosa cells. These findings were regarded to be treatment-related.

NEUROPATHOLOGY
Summary tables of the results are to be found in Part D of PART I; individual tables are to be found in Part D of PART II. Abbreviations and histopathological grading used in pathology report and tables can be found in the appendix.

F1 rearing animals, Cohort 2A (Developmental Neurotoxicity Cohort, adults)
Weight parameters
(Tables ID 1 – ID 4)

Length and width of brain
(Table ID 5)
All length and width measurements were without any findings. Only the length parameter in females of test group 13 was minimally decreased. As the width of the brain in this test group was comparable with the control animals and the absolute brain weight was unchanged, a treatment-related effect seems unlikely.

Gross lesions
(Table ID 6)
Only the moribund sacrificed male animal No 837 revealed a dilation of jejunum, ileum, cecum and colon. These findings were assessed as incidental and not related to treatment. No other gross findings were recorded.

Histopathology
(Table ID 7 – ID 9)
The medulla oblongata and the spinal cord of treated animals revealed a minimal to marked, multifocal degeneration of nerve fibers. The finding was characterized by disintegrated myelin sheaths, vacuolation of myelin sheaths, pyknotic nuclei of oligodendroglia and spheroids as well as gitter cells within the lesions. The lesions were especially visible in longitudinal sections and less visible in cross sections of the spinal cord.

In the pars distalis of the pituitary gland, multifocally distributed very small eosinophilic cysts with a non-ciliated, irregular border and eosinophilic homogenous content were seen.

Morphometry
(Table ID 10)
All morphometric brain measurements were without any findings. Only some single parameters in male (hippocampus right, base of lobus vermis cerebrelli no 8) or female (parietal cortex left) animals of test group 13 showed a statistical significant increase. As no other values were changed, this minimal width increase is assumed as incidental and not related to treatment.

F1 animals, Cohort 2B (Developmental Neurotoxicity Cohort, weanlings)

Weight parameters
(Tables ID 11– ID 14)
The statistically significant decrease of terminal body weight in male and female animals of test group 12 (females only) and 13 is regarded as treatment-related. The decrease of brain weight in test group 13 males is assumed to be secondary to the delayed development of the animals, nevertheless the length and the width of brains are comparable to control animals. The increased relative brain weights of test group 12 (females only) and 13 (both sexes) animals are secondarily linked to the decreased terminal body weight. The increase in absolute brain weight in test group 11 females is regarded as incidental.

Length and width of brain
(Table ID 15)
All length and width measurements were without any findings.

Gross lesions
(Table ID 16)
No gross findings were recorded.

Histopathology
(Table ID 17 - ID 18)
No treatment-related neurohistopathological findings were recorded. In the pars distalis of the pituitary gland, multifocally distributed very small eosinophilic cysts with a non-ciliated, irregular border and an eosinophilic homogenous content were seen in four males and five females (each out of ten) of test group 13 (1000 ppm). This finding is regarded as treatment-related.
Other effects:
effects observed, treatment-related
Description (incidence and severity):









Thyroid hormones
In F1 generation females of test group 11 and 12 (100 and 300 ppm) at PND 4 and in females
at PND 22 of test group 13 (1000 ppm) T4 was higher compared to controls. In test group 13
(1000 ppm) only two males had a sufficient sample volume for the measurement of the thyroid
hormones at PND 4, but no female pup was available.
The T4 means in F1 generation males and females of test group 13 at PND 92 were 19%/18%
higher compared to controls (medians 18%37% higher), although the values were not
statistically significantly changed.

Platelet activating factor (PAF) determination in serum of F0 parents
The platelet activating factor (PAF) concentration in serum of the F0 females is reduced dosedependently
when regarding medians with a decrease of 31% in test group 3 (1000 ppm)
compared to controls. This decrease in females of test group 3 was statistically significant when
the two-sided Jonckheere-Terpstra trend test was applied. The PAF levels decrease could not
been observed among the F0 males.

CHOLINE CONCENTRATION IN BLOOD AND LIVER SAMPLES
The analytical results demonstrated the clear presence of choline in all plasma samples from
the animals dosed with the test substance 2,2’-iminodiethanol (100 ppm, 300 ppm and 1000
ppm dosed animals) and in those from control, non-dosed animals.
In general, it can be stated that the presence of the test substance 2,2’-iminodiethanol led to a
reduction in the content of choline in the plasma samples analyzed. This effect appears to be
dose-dependent, in that higher dose levels were associated with greater choline reduction.
This effect is most clearly visible at lower dose levels (100 ppm and 300 ppm), at which
dramatic plasma choline levels could be seen. At higher dosing levels, although further minor
plasma choline content reduction was observed, this was by no means as drastic.
The analytical results demonstrated the clear presence of choline in all liver samples from the
animals dosed with the test substance 2,2’-iminodiethanol (100 ppm,300 ppm and 1000 ppm
dosed animals) and in those from control, non-dosed animals. This was true from all time points
investigated (4-day old pups, 22-day old pups and ~90-day old adolescents).
In general, it can be stated that the presence of the test substance 2,2’-iminodiethanol led to a
reduction in the content of choline in the liver samples analyzed. This effect appears to be dose-dependent, in that higher dose levels were associated with greater choline reduction, but
only up to moderate dosing levels (300 ppm and 100 ppm, depending on the sampling day).
At higher dosing levels, no further dramatic liver choline content reduction was observed.
This effect was however, not observed in 4-day old animals, in which no clearly definable dosedependent
trend is evident. In 22-day old animals this effect could be clearly observed,
although the choline levels of the 100 ppm dosed animals have not yet attained minimal
concentrations. In ~90-day old animals the effect is dramatic in that the liver choline levels of
all non-control animals have reached an approximate minimum. Only a relatively minor further
dose-dependency can be observed at this time point.
Developmental immunotoxicity:
effects observed, non-treatment-related
Description (incidence and severity):
T-cell dependent antibody response (Anti SRBC IgM antibodies)
Six days after immunization, no changes in the SRBC IgM titers were found in male and female rats of the F1 generation (PND 60) dosed with the test substance. SRBC titers were statistically significantly lower in rats of the positive control group (dosed with Cyclophosphamide).

Splenic lymphocyte subpopulations
In females of the F1 generation (PND 92) of test group 13 (1000 ppm) relative T-helper cell (CD4+/CD3+ lymphocytes in spleen tissue) counts were decreased and relative cytotoxic Tcell (CD8+/CD3+ lymphocytes in spleen tissue) counts were increased compared to controls. This led to a decrease of the CD4/CD8 ratio (not statistically significantly). Absolute T-helper cell counts were not altered in the mentioned test group, but absolute cytotoxic T-cells were also increased (although not statistically significantly).
Auditory Startle Response
The maximum amplitude of the high-dose males and females (1000 ppm) was below the concurrent control during the entire measurement, while latency of reaction to a startle stimulus corresponded to the age of these animals. There was also no habituation to the test environment seen in these animals, males slightly more affected by this than females.The difference in the amplitude was statistically significant in measurement block 1 and 1 - 5 (males) as well as block 5 (females). No influence of the test substance on auditory startle habituation (maximum amplitude and latency) was observed in the low- and mid-dose male and female animals during the measurement. Amplitude and latency of reaction to a startle stimulus and habituation to the test environment corresponded to the age of these animals at PND 24, when normal biological variation inherent in the strain of rats used for this experiment was considered.

Functional observational battery (FOB)
Home cage observations:
No test substance-related or spontaneous findings were observed in male and female animals of all test groups during the home cage observation.
Open field observations:
The open field observations did not reveal any test substance-related findings in male and female animals of all test groups.
Sensorimotor tests/reflexes:
There were no test substance-related findings in male and female animals of all test groups.
Quantitative Parameters:
No test substance-related impaired parameters (number of rearings, grip strength of fore- and hindlimbs and landing foot splay test) were observed in male and female animals of all test groups. This includes a statistically significantly lower grip strength for forelimbs of the highdose females, which is considedered to be an isolated, and thus incidental, finding.

Motor activity measurement (MA)
Motor activity (number of beam interrupts) of male and female animals was not influenced by the test compound at all dose levels (100, 300 and 1000 ppm). Overall activity levels and habituation to the test environment corresponded to the age of these animals at PND 70. Any occasional differences between treated animals of these dose groups and their concurrent control (like lower numbers of beam interrupts in the low- and high-dose females during interval 3), whether statistically significant or not, were regarded as incidental findings and not related to the test compound.

Sex ratio
The sex distribution and sex ratios of live F1 pups on the day of birth and on PND 21 did not show substantial differences between the control and the test substance-treated groups; slight differences were regarded to be spontaneous in nature.

Anogenital distance/anogenital index
Anogenital distance and index of all test substance treated pups (100; 300 and 1000 ppm) were comparable to the concurrent control values.

Nipple/ areola anlagen
The apparent number and percentage of male pups having areolae was not influenced by the test substance when examined on PND 12. However, owing to the high background rate in control animals on this day, we habitually recheck all animals for nipples/areolae on PND 20, one day prior to weaning. During this re-examination no areolae were detected at all in male pups of all test groups.
Key result
Dose descriptor:
NOAEL
Remarks:
reproductive performance
Generation:
F1
Effect level:
300 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: reproductive performance
Key result
Dose descriptor:
NOAEL
Remarks:
developmental toxicity in the F1 progeny
Generation:
F1
Effect level:
100 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: developmental toxicity in the F1 progeny
Key result
Dose descriptor:
NOAEL
Remarks:
developmental neurotoxicity
Generation:
F1
Effect level:
300 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: developmental neurotoxicity
Key result
Dose descriptor:
NOAEL
Remarks:
developmental immunotoxicity
Generation:
F1
Effect level:
300 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: developmental immunotoxicity
Reproductive effects observed:
not specified
Conclusions:
Under the conditions of the present modified extended 1-generation reproduction toxicity study the NOAEL (no observed adverse effect level) for general toxicity is 100 ppm for the F0 parental animals, based on evidence for distinct kidney toxicity and stomach irritation, as well as corresponding effects on water consumption, food consumption, body weights and clinicalpathological parameters, which were observed at the LOAEL (Lowest Observed Adverse Effect Level) of 300 ppm. Similar toxicity was noted in the adolescent F1 animals, which had no stomach irritation but liver toxicity in addition.
The NOAEL for fertility and reproductive performance for the F0 and F1 rats is 300 ppm, based on a lower number of implants, prolonged/irregular estrous cycles as well as pathological changes in sexual organs, pituitary and mammary glands of both genders at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm. However, eosinophilic cysts in the pituitary gland were present in the F1 animals of cohort 1A down to the 100 ppm dose level, but no assessment on adversity of this finding is possible at present. Therefore, no NOEL can be established for this particular effect.
The NOAEL for developmental toxicity in the F1 progeny is 100 ppm, based on impaired pup survival at 1000 ppm as well as reduced pup body weights in the F1 offspring, which were observed at the LOAEL (Lowest Observed Adverse Effect Level) of 300 ppm. As these weight reductions were only observed in the presence of maternal toxicity, including lower weight gain during pregnancy, they are not regarded as independent effect of the treatment.
The NOAEL for developmental neurotoxicity for the F1 progeny is 300 ppm, based on adverse clinical observations, impaired auditory startle response and corresponding neuropathological findings at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm. In addition, increased T4 values were noted in adult and adolescent males at 1000 ppm as well as newborn and weanling females at 100 and 300 ppm.
The NOAEL for developmental immunotoxicity for the F1 progeny is 300 ppm, based on effects on the T-helper cells and cytotoxic T-cells in the spleen in the F1 females at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm. Lower mean and median anti-SRBC IgM antibody titers of the positive control group (4.5 mg/kg bw/d cyclophosphamide, oral) demonstrated that the test system worked properly.
Executive summary:

2,2’-iminodiethanol was administered to groups of 30 male and 30 female healthy young Wistar rats as addition to the drinking water in concentrations of 100, 300 and 1000 ppm. The vehicle control group was given plain drinking water. Analyses confirmed the correct concentration and the stability of the test substance in drinking water.

The overall mean doses of 2,2’-iminodiethanol throughout all study sections and across all cohorts were 12.75 mg/kg body weight/day (mg/kg bw/d) in the 100 ppm group, 37.68 mg/kg bw/d in the 300 ppm group and 128.35 mg/kg bw/d in the 1000 ppm group. There were no test substance-related mortalities or adverse clinical observations noted in the F0 generation parental animals at any dose level as well as in the F1 adolescents at the low- and mid-dose level (100 and 300 ppm).

 

The high-dose of the test substance (1000 ppm) produced mortalities or adverse clinical observations in the F1 adolescents. Three high-dose offspring (2 males, 1 female) in two rearing cohorts (1A, 1B) were either sacrificed moribund or found dead at different timepoints during the study. These casualties went along with adverse clinical observations such as highstepping gait and piloerection recurring across all F1 cohorts in several study sections. Altogether 11 high-dose animals of both sexes in cohorts 1A, 1B, 2A and 3 were affected. Some of these affected high-dose male F1 offspring had, in addition, small testes for which a histopathological correlate was found. As these animals had a severe reduction in terminal body weight, this size reduction was regarded to be secondary to the body weight decrease. Treatment with 2,2’-iminodiethanol produced no effect on water consumption in the F0 males at all dose levels, while F0 females at 300 and 1000 ppm had decreased water consumption beginning during gestation and distinct to severe during lactation. The F1 adolescents showed this effect consistently and distinctly across all cohorts at the 1000 ppm dose level and less pronounced and consistent, but still noticeable, at the 300 ppm dose level. No such effect was noted at 100 ppm. In the affected groups reduced water consumption was associated with reduced urine volume and histopathological findings in the kidneys.

 

In the 1000 ppm F0 parental males and females food consumption was consistently reduced, at the 300 ppm level reduced food consumption was only noted in females during lactation. In the 1000 ppm F1 males and females food consumption was consistently reduced postweaning, although there was some variability in the extent of the reduction across the 1A, 1B, 2A and 3 cohorts. At the 300 ppm level slightly and non-statistically significant reduced food consumption was only noted in the cohort 1A males. In contrast to this, food consumption of all males and females at the 100 ppm level remained unchanged. In the 1000 ppm F0 parental males and females body weights were consistently reduced throughout all study sections beginning on study day 7. At the 300 ppm level reduced body weights were noted in F0 parental males from study day 28 and in females during lactation only. This was caused by a similarly affected body weight gain, though the course of body weight changes was variable in the different study sections, generally for the females being more severe during the gestation/lactation period. Body weights/body weight gain of the high-dose (1000 ppm) Cohort 1A, Cohort 1B, Cohort 2A and Cohort 3 animals were similarly reduced as of the F0 parental animals, however showing some variability across cohorts. At 300 ppm changes of body weights/body weight gain look rather nonuniform and mild, however, there appears to be evidence that this dose group was affected as well.

 

Regarding clinical pathology in F0 generation male and female rats of test group 3 (1000 ppm) as well as in F1 generation male and female rats at PND92 of test group 13 (1000 ppm) a microcytic anemia was present indicated by decreased red blood cell (RBC) counts hematocrit and hemoglobin values as well as decreased mean corpuscular volume (MCV). Increased urea values in male and female rats of the same test groups in the F0 and F1 generation at PND 92 were due to an increased protein metabolism. This is confirmed by higher albumin levels in the rats of both sexes of test group 3 (1000 ppm) in the F0 generation and most probably also by a reduced prothrombin time in females of test group 13 (1000 ppm) of the F1 generation at PND 92; indicating higher synthesis of coagulation factors. Higher activities of alkaline phosphatase in males of the F0 generation of test group 3 and of the F1 generation in test group 13 may have been caused by a reduced food consumption of these animals. Higher aspartate aminotransferase (AST) activities in F0 males of test group 3 and in male and female rats of the F1 generation of test group 13 may be due to a liver cell effect, although other tissues could also have been involved because AST is not a liver-specific enzyme in rats.

 

In males of test groups 2 and 3 (300 and 1000 ppm) urine specific gravity was lower and urine volume was higher (not statistically significantly) compared to controls. In conjunction with histopathological alterations in the kidneys, this change was regarded as treatment-related and adverse.

 

The increase of platelet counts in F0 males of test groups 2 and 3 (300 and 1000 ppm) as well as males of the F1 generation in test groups 12 and 13 (300 and 1000 ppm) as well as the shortened prothrombin time (Hepatoquick’s test) in males and females of test groups 2 and 3 of the F0 generation and in F1 females of test group 13, indicated a dysregulation of the coagulation homeostasis. This correlates with decreased serum platelet activating factor (PAF) values in F0 females of test group 3 (1000 ppm).

 

Regarding pathology, target organs were the kidneys, liver and glandular stomach.

F0 generation parental animals 

The terminal body weight in the F0 generation parental males and females of test group 03 (1000 ppm) was decreased to below historical control values and for test group 02 animals (300 ppm) it was within historical control values (see PART III). This reflects the different extent of body weight effects at 300 and 1000 ppm in-life. In the kidneys of males of test group 02 and test group 03 and in the kidneys of females of test group 03, degeneration/regeneration of the proximal tubules was observed. This was also regarded as treatment-related. The decrease in terminal body weight in males of test group 12 and 13 (300 and 1000 ppm) and females of test group 13 (1000 ppm) reflects the body weight effect at 300 and 1000 ppm in life.

 

Similar to the findings in the kidneys of the F0 generation, males and females of test group 12 and 13 (300 and 1000 ppm) revealed tubular degeneration and regeneration. This was also reflected by the increased kidney weight. In test group 11 (100 ppm) animals the kidney weight was still increased, but was regarded to be non-adverse due to a missing histopathologic correlate. Comparable to the F0 generation, animals of the F1 generation revealed also mineralization: male animals at the transition between inner and outer medulla and females in the papilla. As for the F0 generation this finding was regarded to be treatment-related but not as adverse.  

Males and females of test group 13 (1000 ppm) revealed centrilobular hypertrophy in the liver. Male animals also had a peripheral hypertrophy. This correlated with the increased liver weights. Furthermore, in both sexes an increase of fatty change was observed in the peripheral area. These findings in combination with clinical pathology findings were regarded to be adverse. In males of test group 12 (300 ppm), the relative liver weight was increased, three animals showed centrilobular hypertrophy and fatty change. Due to the additional fatty change in combination with the hypertrophy, it was regarded as adverse. Females of the same test group revealed increased liver weights and one animal a centrilobular hypertrophy, but no increase in fatty change when compared to control and no relevant findings in clinical pathology. It was therefore regarded to be treatment-related but not as adverse. The same comes true for the increased liver weight in test group 11 males and females (100 ppm), in which no relevant histopathologic findings were observed.

 

The mammary gland of males of test group 13 (1000 ppm) revealed a female phenotype (feminization) in four out of 16 animals. One male animal showed in addition a diffuse hyperplasia of the mammary gland. Females of this test group showed an increase in secretion. These findings were regarded to be adverse.

 

In the left testis, three males of test group 13 (1000 ppm) revealed immature testicular tubules which corresponded to the macroscopic finding “size reduced”. In one male, there was focal degeneration in addition. As a consequence, the secondary sexual glands showed also a reduction in size. In the corresponding epididymis of the affected animals aspermia was found. In the ductus deferens size reduction and an increase in macrovesicular vacuolation was observed. The vacuolation in the ductus deferens of test group 12 animals (300 ppm) might be still treatment-related but due to the absence of findings in all other sexual organs this was regarded to be non-adverse. These three males revealed a severe decrease in terminal body weight. Therefore, this size decrease in the above-mentioned organs was most likely due to the body weight reduction and was assessed to be treatment-related but a secondary effect. A delayed maturation of the testicular epithelium is known to occur in animals which have reduced body weights (McInnes, 2012). But as in cohort 1A animals (and in cohort 1B animals) degeneration of the testicular epithelium was observed in addition to the delay in maturation as well as in animals without a delay in maturation in these organs, the degeneration of tubular epithelium in the testis was assumed to be an adverse but secondary effect.

 

Six out of 20 females of test group 13 (1000 ppm) showed an increased incidence of luteal cysts in the ovary. One female revealed a diffuse atrophy of the ovaries. Luteal cysts might develop in case the follicle fails to ovulate. These findings were regarded to be treatment-related and adverse.

 

Males and females of all treated groups revealed cysts in the pars distalis of the pituitary gland which were filled with a homogenous eosinophilic material and differed from the spontaneously observed cysts (e.g. remnants of the Rathke’s pouch). It could not be determined what material was present in these cysts and whether they were functionally active or not. Thus this finding could not be assessed with regard to potential adversity.

 

The results of the differential ovarian follicle count (DOFC) – comprising the numbers of primordial and growing follicles, as well as the combined incidence of primordial and growing follicles – showed significant differences between the control group 10 and animals of test group 13. The DOFC was performed in cohort 1A and cohort 1B animals together in a fully blinded manner. This statistically significant decrease in primordial and growing follicles was regarded to be adverse.  

F1 rearing animals, cohort 1B

Target organs were the ovaries and testes. No other organs were examined histopathologically.

 

The decrease in terminal body weight in males and females of test group 13 (1000 ppm) reflects the body weight effect at 1000 ppm in life.

 

In males and females of all treated groups the liver weight was significantly increased which was regarded to be treatment-related. Although a conclusive interpretation of adversity cannot be made without a histopathological or clinical pathological examination, similar effects as have been detected in cohort 1A livers are the most likely explanation for this finding. The reduced absolute and relative prostate weight in test group 13 (1000 ppm) males could be related to treatment. But as the terminal body weight was also significantly decreased and the cohort 1B animals were treated the same way as the cohort 1A animals and there were no histopathologic findings in the prostate that could explain the weight decrease beside the three affected animals, it was regarded to be secondary to the body weight reduction. Three males of test group 13 (1000 ppm) had reduced testes sizes and corresponding reduced size of the prostate and seminal vesicle that was thought to be secondary to the testes findings. Microscopically, the gross lesion was related to immaturity as described for cohort 1A. Two of these males had in addition tubular degeneration and two males showed minimal tubular degeneration without other findings. In test group 11 (100 ppm), one male also revealed immature testicular epithelium and degeneration. As there was no male affected in test group 12 (300 ppm) this finding was regarded to be incidental. The immaturity and degeneration in test group 13 animals (1000 ppm) was regarded to be treatment-related and adverse, however, as in the cohort 1A animals are most likely a secondary effect of the body weight reduction, a hypothesis supported when the individual animal data is taken into account.

 

Four females of test group 13 (1000 ppm) had macroscopically reduced ovaries that could be related microscopically to a diffuse atrophy of the ovary. As described for cohort 1A animals here were six females showing luteal cysts in the same test group and in two females of test group 13 (1000 ppm) no corpora lutea could be observed. These findings were regarded treatment-related and adverse. The single animal in test group 12 (300 ppm) showing luteal cysts was regarded to be incidental.

 

F1 rearing animals, cohort 3 (Immunotoxicity)

 

There was a reduction in terminal body weight in test group 13 (1000 ppm) in males and females which reflects the body weight effect at 1000 ppm in life.Macroscopically there was one male showing reduced size of testes, epididymides, prostate and seminal vesicle. As no microscopic investigation was performed a detailed diagnosis could not be made. But considering the testis effects in the other cohorts it is most likely the same finding here.

 

There were no indications from clinical examinations as well as gross and histopathology, that 2,2’-iminodiethanol adversely affected the fertility or reproductive performance of the F0 parental animals up to and including the administered dose of 300 ppm. Estrous cycle data, mating behaviour, conception, gestation, parturition, lactation and weaning as well as sexual organ weights and gross and histopathological findings of these organs (specifically the differential ovarian follicle count) were comparable between the rats of these groups including control and ranged within the historical control data of the test facility. The high dose of the test item (1000 ppm) exerted effects on a number of parameters such as number of implants and duration of gestation in the F0 parental animals as well as estrous cyclicity and morphology of pituitary, ovaries, testes (subsequently accessory sexual glands) and mammary glands in the F1 offspring.  

The high-dose F0 generation animals were successfully paired, fertility and gestation indices were comparable to the concurrent control. There was, however, a significantly lower number of implants and subsequently lower litter size noted along with a small but significant increase in duration of gestation. There were no morphological changes detected in tissues related to reproduction which could have explained these effects.  

The high dose F1 generation females had a prolonged estrous cycle and the cycle was irregular in a higher number of females. There was no obvious pattern of change in the affected females, the irregularities consisted likewise of prolonged diestrous, estrous or metestrous. These irregularities corresponded with ovarian atrophy, pituitary and luteal cysts as well as a decrease in primordial and growing ovarian follicles, as described in the pathology/neuropathology sections.

 

Some high-dose F1 males had smaller and lightweight testes which were immature and, in some cases, showed degeneration of tubular epithelia. Cysts in the pars distalis of pituitary were also present. Most of the affected males exhibited distinct clinical symptoms of systemic toxicity as well. In addition, some high-dose F1 males had feminized and/or hyperplastic mammary glands, while high-dose females showed an increase in secretion, as described in the pathology section.

 

For all liveborn male and female pups of the F0 parents, no test substance-induced signs of developmental toxicity were noted at dose levels as high as 100 ppm. Postnatal survival, pup body weight gain as well as post-weaning development of the offspring of this test group until puberty remained unaffected by the test substance. Furthermore, clinical and/or gross necropsy examinations of the F1 pups revealed no adverse findings. The high dose of the test substance (1000 ppm) caused effects on pup survival during early lactation. While the lower litter size in this dose group was a consequence of a lower number of implants and not due to prenatal or perinatal mortality, the slightly lower viability index came from a higher number of dead and cannibalized pups which were distributed across 8 litters. In 2 of those 8 litters none of the pups survived. Altogether, the lower viability index in this group was only slightly below the historical control range. Postnatal survival after PND 4 of the offspring of all test groups until weaning remained unaffected by the test substance. Furthermore, clinical and/or gross necropsy examinations of the weaned F1 pups revealed no adverse findings.

 

Pup body weight development of the mid and high-dose F1 offspring (300 and 1000 ppm) was affected by the treatment. These offspring had similar weights as the control right after birth but gained significantly less weight than control offspring from PND 4 (high-dose) or PND 14 (mid-dose) onwards. The decrease in terminal body weight in the 300 and 1000 ppm male and 1000 ppm female weanlings not selected for cohorts reflects the body weight effect in life. The impairment of body weight gain in the F1 offspring continued after weaning in the offspring selected for cohorts.

 

Measurement of thyroid hormones revealed higher T4 values in F0 males of test group 3 (1000 ppm) as well as in F1 rats of both sexes in test group 13 (1000 ppm) at PND 92 as well as in F1 females of the same test group at PND 22 and in females of test groups 11 and 12 at PND 4. However, no effects were observed on THS levels.

 

Anogenital distance of all test substance treated F1 pups (100, 300 and 1000 ppm) was comparable to the concurrent control values, as were anogenital indices. In addition, the check for the presence of nipples/areolas, also a very sensitive marker of potential endocrine-mediated imbalances, revealed no test substance-related effects at all. Vaginal opening and preputial separation are commonly used developmental markers for onset of puberty in laboratory rats. A statistically significant delay in vaginal opening of about 1-2 days beyond the control was observed in the female F1 offspring of the mid- and high-dose groups (300 and 1000 ppm). The values for pubertal age and weight of mid-dose females were both at the lower end of the historical control range; thus the apparent statistical increase is considered to be due to the very low concurrent control values in this study and not treatment-related.

 

In the high-dose group the pubertal age is just above the upper limit of the historical range while the weight at puberty is below the historical control range. This clearly indicates that the later onset of puberty is a consequence of a general developmental delay and not a specific effect on the timing of puberty. A statistically significant delay in preputial separation of about 2 days beyond the control was observed in the male F1 offspring of the high-dose group (1000 ppm). In the high-dose group the pubertal age is well within of the historical range while the weight at puberty is distinctly below the historical control range. This indicates that the apparent later onset of puberty may be a spurious finding, and if at all, is a consequence of a general developmental delay and not a specific effect on the timing of puberty. No clinical signs of developmental neurotoxicity were evident in male and female F1 offspring at dose levels as high as 300 ppm. There were no compound related effects on motor activity, auditory startle habituation, and in the field observation battery following exposure to the test compound in these animals.

 

Some findings which might be related to an affection of the nervous system were observed in the F1 offspring at 1000 ppm. Clinically, high-stepping gait and piloerection were observed recurring across all F1 cohorts in several study sections. The only notable finding in neurobehavioral testing, however, were lower maximum amplitudes in the auditory startle response test of the high-dose F1 males and females in Cohort 2A, while FOB and motor activity testing remained without findings. In the auditory startle test there was also no habituation to the test environment seen in these animals, males slightly more affected by this than females. Notably, no corresponding effects were recorded for startle response latency. In addition, regarding neuropathology, treatment-related findings were seen in the medulla oblongata, spinal cord and pituitary gland of Cohort 2A animals (adults, PND 77) as well as in the pituitary gland of Cohort 2B animals (weanlings, PND 22).

 

F1 rearing animals, cohort 2A (adults)

The terminal body weight was decreased in male and female test group 13 animals, reflects the body weight effect at 1000 ppm in life.

 

The medulla oblongata and the spinal cord of male and female animals of test group 13 (1000 ppm) revealed a minimal to marked, multifocal degeneration of nerve fibers. This finding was characterized by disintegrated myelin sheaths, vacuolation of myelin sheaths, pyknotic nuclei of oligodendroglia and spheroids as well as gitter cells within the lesions. The lesions were visible especially in longitudinal sections and less in cross sections of the spinal cord. As also in several control animals minimal spontaneous degeneration was seen, only degeneration with a higher severity grade was assessed as treatment-related and adverse. In the pars distalis of the pituitary gland of male and female animals (all test groups), multifocally distributed very small eosinophilic cysts with a non-ciliated, irregular border and an eosinophilic homogenous content were seen. As no functional or mechanistic data are available, this finding could not be assessed with regard to potential adversity. The brain weight determination, brain length and width measurements as well as brain morphometry and neuropathological examination by light microscopy of all other tissues did not reveal further treatment-related findings.

 

F1 rearing animals, cohort 2B (weanlings)

The terminal body weight was decreased in male and female test group 13 animals and in females of test group 12, which reflects the body weight effect at 300 and 1000 ppm in life. In the pars distalis of the pituitary gland of four male and five female animals of test group 13 (1000 ppm), multifocally distributed, very small eosinophilic cysts with a non-ciliated, irregular border and an eosinophilic homogenous content were seen. As no functional or mechanistic data are available, this finding could not be assessed with regard to potential adversity. There was no evidence that the test substance produced any developmental immunotoxicity up to and including a dose of 300 ppm in both sexes and 1000 ppm in males. Neither T-cell dependent anti-SRBC IgM antibody response, nor absolute and relative lymphocyte subpopulation cell counts in the spleen tissue (B-, T-lymphocytes, CD4-, CD8-T-lymphocytes and natural killer (NK) cells) displayed any treatment-related changes. However, a test compound-related effect on the T-helper cells and cytotoxic T-cells in the spleen in the high-dose F1 females (1000 ppm) cannot be excluded.

Endpoint:
screening for reproductive / developmental toxicity
Remarks:
Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
19 Feb 2018 (study initiation) To:
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 422 (Combined Repeated Dose Toxicity Study with the Reproduction / Developmental Toxicity Screening Test)
Version / remarks:
29 Jul 2016
Deviations:
no
GLP compliance:
yes (incl. QA statement)
Remarks:
12. to 14.09.2016
Limit test:
no
Justification for study design:
SPECIFICATION OF STUDY DESIGN FOR EXTENDED ONE-GENERATION REPRODUCTION TOXICITY STUDY WITH JUSTIFICATIONS:

- Premating exposure duration for parental (P0) animals : 13 days
- Basis for dose level selection : selected by request of the sponsor
- Route of administration : via drinking water
- Other considerations: vehicle and number of animals. The test guideline requires the rat to be used as the animal species. This rat strain was selected since extensive historical control data are available for Wistar rats.
Species:
rat
Strain:
Wistar
Remarks:
Crl:WI(Han)
Details on species / strain selection:
The test guideline requires the rat to be used as the animal species. This rat strain was selected since extensive historical control data are available for Wistar rats.
Sex:
male/female
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH,
- Females nulliparous and non-pregnant: yes
- Age at study initiation: (P) 11-12 wks (male), 10 wks (female)
- Weight at study initiation: (P) Males: 364.8 ± 14.9 to 366.8 ± 15.0 g; Females: 222.4 ± 9.4 to 225.8 ± 10.1 g;
- Housing: During pretreatment of the study period, the rats were housed together (up to 5 animals per sex and cage) in Polysulfonate cages Typ 2000P (H-Temp) supplied by TECHNIPLAST, Hohenpeißenberg, Germany. During the study period, the rats were housed individually in Polycarbonate cages type III supplied by TECHNIPLAST, Hohenpeißenberg, Germany and Becker & Co., Castrop-Rauxel, Germany, with the following exceptions: During overnight mating, male and female mating partners were housed together in Polycarbonate cages type III and pregnant animals and their litters were housed together until PND 13 in Polycarbonate cages type III. Pregnant females were provided with nesting material (cellulose wadding) toward the end of gestation. For enrichment wooden gnawing blocks (Typ Lignocel® block large, J. Rettenmaier & Söhne GmbH + Co KG, Rosenberg, Germany) were added. In addition in Polysulfonate cages large play tunnels (Art. 14153; supplied by PLEXX B.V., Elst, Netherlands) were added. The cages with the test animals were arranged on the racks in such a way that uniform experimental conditions (ventilation and light) were ensured.
- Diet (e.g. ad libitum): The food used was ground Kliba maintenance diet mouse/rat “GLP” meal, supplied by Provimi Kliba SA (new name Garanovit AG), Kaiseraugst, Switzerland, which was available to the animals ad libitum throughout the study (from the day of supply to the day before necropsy).
- Water (e.g. ad libitum): Drinking water was supplied from water bottles (ad libitum).
- Acclimation period: 3 wks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24°C
- Humidity (%): 45-65%
- Air changes (per hr): The air change rate was 15 times per hour
- Photoperiod (hrs dark / hrs light): 12 hours light from 6.00 h to 18.00 h and 12 hours darkness from 18.00 h to 6.00 h

IN-LIFE DATES: From: 19 Feb 2018 (study initiation) To: 12 Apr 2018 (Male parental animals) and 09 May 2018 (Female parental animals)
Route of administration:
oral: drinking water
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
The test substance solutions in drinking water were prepared in intervals, which took into account the analytical results of the stability verification. For the preparation of the administration solutions the test substance was weighed in a calibrated beaker depending on the dose group, topped up with deionized water, were then titrated with aqueous HCl until ph7 was reached and intensely mixed with a magnetic stirrer until it was completely homogeneous and dissolved. For supply of the animals with drinking water and aqueous N,N-Dimethylaminoethanol solutions polycarbonate drinking bottles supplied by Bioscape EBECO GmbH., Castrop- Rauxel, FRG, with the capacity of 300 mL were used. The bottles had a stainless steel cap with a silicone sealing ring and a hole of diameter 0.6 mm, and each was placed in a recess in the cage lid. The animals obtained water or aqueous N,N-Dimethylaminoethanol solutions by licking the drops from the hole in the cap. Their special design substantially prevented the drinking bottles from uncontrolled emptying or leakage.

VEHICLE
- Purity: The drinking water is regularly assayed for chemical contaminants as well as for the presence of (pathogenic) microorganisms by the municipal authorities of Frankenthal and the Environmental Analytics Water/Steam Monitoring of BASF SE.
Details on mating procedure:
- M/F ratio per cage: 1:1
- Length of cohabitation: In general, each of the male and female animals was mated overnight in a 1:1 ratio for a maximum of 2 weeks.
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy
- After successful mating each pregnant female was caged: individually
- Any other deviations from standard protocol: Deviations from the specified times were possible on weekends and public holidays and were reported in the raw data.
Analytical verification of doses or concentrations:
yes
Remarks:
HPLC-MS
Details on analytical verification of doses or concentrations:
Based on the analytical results it is concluded that N,N-Dimethylaminoethanol is stable in deionized water adjusted to pH = 7 with hydrochloric acid over a period of 10 days at room temperature. All determined concentrations were in the range of 90 % - 110 % of the nominal concentration.
Duration of treatment / exposure:
2 weeks before mating and male and female animals were sacrificed 31 and 58 days, respectively, after the beginning of the administration
Frequency of treatment:
continuosly
Details on study schedule:
After the acclimatization period, the test substance was administered to the parental animals as addition to the drinking water continuously throughout the entire study. The animals of the control group were treated in the same way, with the vehicle (drinking water only). Males and females from the same dose group were mated, after two weeks of treatment, overnight at a ratio of 1:1.
The females were allowed to deliver and rear their pups until PND 4 (standardization) or PND 13. On study day 27, a functional observational battery and motor activity measurement were carried out in five male animals per group. On study day 55, a functional observational battery and motor activity measurement were carried out in five female animals (with litter) per group. The male and female animals were sacrificed 31 and 58 days, respectively, after the beginning of the administration, and examined.
Dose / conc.:
200 ppm (nominal)
Remarks:
During the lactation period test substance concentrations in the drinking water of the F0 females were reduced to 50 % (= 100 ppm) due to increased water consumption during this period
Dose / conc.:
1 000 ppm (nominal)
Remarks:
During the lactation period test substance concentrations in the drinking water of the F0 females were reduced to 50 % (= 500 ppm) due to increased water consumption during this period
Dose / conc.:
5 000 ppm (nominal)
Remarks:
During the lactation period test substance concentrations in the drinking water of the F0 females were reduced to 50 % (2500 ppm) due to increased water consumption during this period
No. of animals per sex per dose:
10/sex/ dose
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: on sponsors request
- Fasting period before blood sampling for clinical biochemistry: not specified
Positive control:
none
Parental animals: Observations and examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule:
- Cage side observations checked were included: see "any other information on materials & methods"

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: once prior to the first administration (day 0) and at weekly intervals during the administration period

BODY WEIGHT: Yes
- Time schedule for examinations: once a week at the same time of the day (in the morning)

WATER CONSUMPTION AND COMPOUND INTAKE (if drinking water study): Yes
- Time schedule for examinations: daily

OTHER: For detailed information on: Open field observations, Functional observation battery, Sensory motor tests/Reflexes, Motor activity measurement, Estrous cycle determinations, Male reproduction data, Female reproduction and delivery data, please refer to "any other information on material & methods"
Litter observations:
STANDARDISATION OF LITTERS
- Performed on day 4 postpartum: yes
- If yes, maximum of 8 pups/litter (4/sex/litter as nearly as possible); excess pups were killed and discarded.

PARAMETERS EXAMINED
The following parameters were examined in [F1] offspring:
Pup number and status at delivery, Pup viability/mortality, Sex ratio, Pup clinical observations, Pup body weight data, Anogenital distance, Anogenital index, Nipple/areola anlagen, Pup necropsy observations, Choline Determination, Thyroid hormones,

GROSS EXAMINATION OF DEAD PUPS:
yes, all stillborn pups and all pups that died before weaning were examined externally, eviscerated and their organs were assessed macroscopically. All pups without notable findings or abnormalities were discarded after their macroscopic evaluation. Animals with notable findings or abnormalities were evaluated on a case-by-case basis, depending on the type of finding noted.
Postmortem examinations (parental animals):
SACRIFICE
- Maternal animals: All surviving animals. The male and female animals were sacrificed 31 and 58 days, respectively, after the beginning of the administration, and examined.

GROSS NECROPSY
- Gross necropsy consisted of external and internal examinations including the cervical and thoracic viscera

HISTOPATHOLOGY / ORGAN WEIGHTS
The tissues indicated in "any other information on material & methods" were prepared for microscopic examination and weighed, respectively.
Postmortem examinations (offspring):
SACRIFICE
- The F1 offspring were sacrificed at 13 days of age.
On PND 13, one selected male and one female pup per litter was sacrificed under isoflurane anesthesia by decapitation. Blood was sampled for determination of thyroid hormone concentrations. Thyroid glands/parathyroid glands were fixed in neutral buffered 4% formaldehyde solution and were transferred to the Pathology Laboratory for possible further processing. After sacrifice, all pups were examined externally and eviscerated, and their organs were assessed macroscopically. All stillborn pups and all pups that died before weaning were examined externally, eviscerated and their organs were assessed macroscopically. All pups without notable findings or abnormalities were discarded after their macroscopic evaluation. Animals with notable findings or abnormalities were evaluated on a case-by-case basis, depending on the type of finding noted.
Statistics:
DUNNETT-test (two-sided): Water consumption, food consumption (parental animals), body weight and body weight change (parental animals and pups; for the pup weights, the litter means were used), gestation days, anogenital distance, anogenital index
FISHER'S EXACT test (one-sided): Male and female mating indices, male and female fertility indices, females mated, females delivering, gestation index (females with liveborn pups), females with stillborn pups, females with all stillborn pups
WILCOXON test (one-sided+) with BONFERRONI-HOLM: Mating days until day 0 pc, %postimplantation loss, pups stillborn, %perinatal loss, nipple development
WILCOXON test (one-sided-) with BONFERRONI-HOLM: Implantation sites, pups delivered, pups liveborn, live pups day x, viability Index, survival index
WILCOXON test (two-sided): % live male day x, %live female day x
KRUSKAL-WALLIS test (two-sided): Number of cycles and Cycle Length, rearing, grip strength of forelimbs and hindlimbs, landing foot-splay test, motor activity
Reproductive indices:
Female mating index (%), Female fertility index (%), Gestation index (%), Live birth index (%), Postimplantation loss (%), Male fertility index (%), Male mating index (%)
Offspring viability indices:
Male mating index (%), Postimplantation loss (%)
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
No clinical signs or changes of general behavior, which may be attributed to the test substance, were detected in any of the male F0 parental animals in any of the groups and in any of the female F0 parental animals of test groups 1 - 2 during the entire study period. All clinical signs of the high-dose dams were only observed during lactation. Five out of nine high-dose females (Nos. 131, 133, 136, 138 and 139) showed piloerection. Four high-dose females (Nos. 131, 134, 136 and 138) showed tonic-clonic convulsions (grade: slight to severe) during several parts of the lactation period. During detailed clinical observation (DCO, see below), three high-dose females (Nos. 134, 135 and 137) showed tonic-clonic convulsions (grade: slight to moderate) on study days 42 and 56. In the open field observation (FOB, see below), high-dose females Nos. 134 and 135 showed slight tremors and slight to severe convulsions. The above-mentioned findings were assessed as treatment-related and adverse.
Mortality:
mortality observed, treatment-related
Description (incidence):
There were no test substance-related mortalities in test groups 1-2 in females and all test groups in males. One high-dose female (No. 131 - 5000 ppm) was found dead on PND 17. The female showed piloerection and moderate tonic-clonic convulsions beforehand on PND 7. A relation to treatment cannot be excluded.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
Mean body weights of all males and females of test group 1-2 and body weight change of all male parental animals in all test substance-treated groups were comparable to the concurrent control values during the entire study period. A slight decrease of body weight development in high-dose females was apparent during gestation as the body weight change was statistically significantly below the concurrent control values (GD 0- 20: about 15%). It was assessed as treatment-related and adverse. The body weight change of the low- and mid-dose females was comparable to the concurrent control values during the entire study.
Water consumption and compound intake (if drinking water study):
effects observed, treatment-related
Description (incidence and severity):
In the high-dose F0 males (5000 ppm), mean water consumption was statistically significantly below the concurrent control values during premating days 3 - 7 and 10 - 13 (about 24% and 23%, respectively). During mating, water consumption of the high-dose males was decreased without statistical significance (up to 25% below control). In the high-dose F0 females, water consumption was statistically significantly below the concurrent control values during premating days 3 – 13 (up to 32%), during several parts of the gestation period (up to 28%) and during lactation (PND 4 – 5: up to 24% below control, without statistical significance: up to 30%). The decrease in water consumption in high-dose males and females was most probably due to the bad taste/smell of the test substance related to the administration of the test substance in the drinking water. It was assessed as treatment-related.
Water consumption of the mid- and low-dose males and females was comparable to the concurrent control values during the entire study.
Ophthalmological findings:
no effects observed
Haematological findings:
effects observed, non-treatment-related
Description (incidence and severity):
No treatment-related changes among hematological parameters were observed. At the end of the administration period, in males of test group 2 (1000 ppm) absolute reticulocyte counts were significantly decreased, but the change was not dose-dependent. Therefore, this alteration was regarded as incidental and not treatment-related.
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
No treatment-related changes among clinical chemistry parameters were observed. At day 50, in dams of test group 3 (5000 ppm) triglyceride values were significantly decreased. The mean was below the historical control range (dams, triglycerides 1.19-3.20 mmol/L), but this was an isolated changed parameter among these individuals and, therefore, this alteration was regarded as treatment-related but non-adverse.
Urinalysis findings:
not specified
Behaviour (functional findings):
no effects observed
Description (incidence and severity):
No changes of general behavior, which may be attributed to the test substance, were detected in any of the male F0 parental animals in any of the groups and in any of the female F0 parental animals of test groups 1 - 2 during the entire study period.
Immunological findings:
not specified
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
Adverse, neurological deficits may be explained by the findings observed in pathology: the brain, cervical and thoracic spinal cord were the target organs in female animals. Axonal degeneration in the cerebellum, medulla oblongata, cervical and thoracic spinal cord were observed. This was regarded to be treatment-related and adverse.
Other effects:
no effects observed
Description (incidence and severity):
In parental males (test groups 1, 2 and 3; 200, 1000 and 5000 ppm) no treatment-related alterations of T4 and TSH levels were observed.
Reproductive function: oestrous cycle:
no effects observed
Description (incidence and severity):
Estrous cycle data, generated during the last 2 weeks prior to mating for the F1 litter, revealed regular cycles in the females of all test groups 0 - 3. The mean estrous cycle duration was similar: 4.0 / 4.1 / 4.0 and 4.0 days in test groups 0-3, respectively.
Reproductive function: sperm measures:
no effects observed
Description (incidence and severity):
The stages of spermatogenesis in the testes of males of the high dose test group were comparable to those of the controls. In high dose females the different stages of functional bodies in the ovaries were present and comparable to the control animals.
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
The male animal No. 21 revealed severe diffuse degeneration of seminiferous tubules in the testes resulting in aspermia in the epididymides which was regarded to be the cause of the missing offspring. The female animal No. 122 showed a neutrophilic inflammation in the uterus which was regarded to be the reason for not becoming pregnant. The female animals (Nos. 109, 121, 123 and 137), which were not pregnant as well as the male mating partners (Nos. 9, 22, 23 and 37) did not show relevant histopathological findings.
Thus, the male fertility index ranged between 70.0% and 100% without showing any relation to dosing. This reflects the normal range of biological variation inherent in the strain of rats used for this study. Also the fertility index varied between 77.8% and 100% without showing any relation to the dose level. This reflects the normal range of biological variation inherent in the strain of rats used for this study.
Regarding fertility and reproductive performance, no adverse signs of toxicity were observed in male or female parental animals of test groups 1-3 (200, 1000 and 5000 ppm) during the entire study.
Almost all F0 parental animals proved to be fertile. Mating behavior, conception, implantation and gestation were not influenced.
Dose descriptor:
NOAEL
Effect level:
1 000 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
clinical signs
mortality
body weight and weight gain
water consumption and compound intake
Remarks on result:
not determinable due to absence of adverse toxic effects
Dose descriptor:
NOAEL
Effect level:
5 000 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
reproductive performance
Dose descriptor:
LOAEL
Effect level:
200 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
organ weights and organ / body weight ratios
Dose descriptor:
LOAEL
Effect level:
5 000 ppm
Based on:
test mat.
Sex:
male/female
Basis for effect level:
clinical signs
mortality
body weight and weight gain
water consumption and compound intake
Clinical signs:
not specified
Description (incidence and severity):
There were no test substance-related adverse clinical signs observed in any of the F1 generation pups of the different test groups.
Mortality / viability:
mortality observed, treatment-related
Description (incidence and severity):
Viability/mortality was adversely affected by the test substance at the highest test group 3 (5000 ppm). This was based on an increased postimplantation loss (25.5% vs 0.9% in control) leading to a decrease in mean number of F1 pups delivered per dam. A higher rate of stillborn pups (n=25, 28.7% vs. 7.3% in control) led to a decreased rate of liveborn pups (71.3% vs. 92.7% in control). Furthermore, postnatal death was indicated by higher numbers of found dead (16.1% vs 2.7% in control) and cannibalized high-dose pups (n=5) leading to a viability index of 69.6% from PND 0-4.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
The mean body weight (PND 1) and body weight change (PND 4 - 7) of the high-dose male pups were statistically significantly below to the concurrent control values (about 13% and 18%, respectively). The value of the high-dose pup weights (6.3 g) was slightly below the historical control range (Supplement, HCD, pup weights, day 1, males, range: 6.4 – 7.3 g). In females and in both sexes combined, body weights of test group 3 were also decreased (-8.7 and -12%, respectively, without statistical significance) compared to control after birth (PND 1).
Towards PND 13, high-dose F1 males showed a slight recovery in body weight (-10% below control). The mean body weight of the high-dose F1 females recovered to values comparable to control. The decrease in pup body weights of test group 3 was assessed as treatment-related and adverse.
The mean body weights and body weight change of all low- and mid-dose male and female pups were comparable to the concurrent control values throughout the entire study. One male runt was seen in control, two male runts were seen in test group 2 and four male and two female runts were seen in test group 3. The higher number of runts in test group 3 was assessed as treatment-related and adverse.
Anogenital distance (AGD):
no effects observed
Description (incidence and severity):
The anogenital distance and anogenital index of all test substance treated male pups was comparable to the concurrent control values. The anogenital index of the high-dose female pups was statistically significantly above the concurrent control values (0.87 vs. 0.80 in control). However, the value was within the historical control range (AG index, range 0.72 – 0.87) and the parameter AG distance was not affected. Therefore, it was not assessed as treatment-related, adverse finding. The anogenital distance of all test substance treated female pups and anogenital index of all female pups of test groups 1 - 2 was comparable to the concurrent control values.
Nipple retention in male pups:
no effects observed
Description (incidence and severity):
The apparent number and percentage of male pups having areolae was not influenced by the test substance when examined on PND 13.
Gross pathological findings:
effects observed, non-treatment-related
Description (incidence and severity):
A few pups showed spontaneous findings at gross necropsy, such as discolored testis (red), dilated renal pelvis, post mortem autolysis and discolored thymus (red). These findings occurred without any relation to dosing and/or can be found in the historical control data at comparable or even higher incidences. Thus, all these findings were not considered to be associated to the test substance.
Regarding developmental toxicity, pup number, status at delivery, viability/mortality and pup body weight was adversely affected by the test substance at the highest test group 3 (5000°ppm). This was based on an increased postimplantation loss (25.5% vs 0.9% in control) leading to a decrease in mean number of F1 pups delivered per dam. A higher rate of stillborn pups (n=25, 28.7% vs. 7.3% in control) led to a decreased rate of liveborn pups (71.3% vs. 92.7% in control). Furthermore, postnatal death was indicated by higher numbers of found dead (16.1% vs 2.7% in control) and cannibalized high-dose pups (n=5) leading to a viability index of 69.6% from PND 0-4. Consequently, the effect on sex ratio/distribution at birth (females: 31.2%, males: 68.8%) and on PND13 (females: 23.9%, males 76.1%) was assessed as secondary. A decrease in F1 high-dose pup body weight was observed after birth (PND1) which recovered partially towards PND13.
Dose descriptor:
LOAEL
Generation:
F1
Effect level:
>= 5 000 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
viability
mortality
body weight and weight gain
other: developmental toxicity
Dose descriptor:
NOAEL
Generation:
F1
Effect level:
2 500 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
viability
mortality
body weight and weight gain
other: developmental toxicity
Reproductive effects observed:
no
Lowest effective dose / conc.:
5 000 ppm (nominal)
Treatment related:
no

Female fetility:

The gestation index was 100% in control and test groups 1 - 2 and 88.9% in test group 3. The value of 88.9% is within the historical control range (gestation index: 87.5 – 100 %) and is, therefore, not assessed as treatment-related. Implantation was not affected by the treatment since the mean number of implantation sites was comparable between all test substance-treated groups and the control, taking normal biological variation into account (12.3 / 13.4 / 13.1 and 13.0 implants/dam in test groups 0 - 3, respectively). The postimplantation loss was 0.9% / 4.4% / 3.2% and 25.5%** (**:p0.01) in test groups 0-3, respectively. The mean value of test group 3 was outside the historical control range (postimplantation loss, range of 0.0 – 18.12 %). The finding was assessed as treatment-related and adverse. The mean number of F1 pups delivered per dam was 12.2 / 12.8 / 12.7 and 9.7* (*:p0.05) pups/dam in test groups 0-3, respectively. The value of test group 3 was within the historical control range (pups delivered, mean: 9.3 – 13.9). The low number of delivered pups is caused by the high post-implantation loss in the high-dose dams and is, therefore, assessed as treatment-related and adverse. The number of females with stillborn pups was increased in test group 3 (5 vs. 1 in control). The value was outside the historical control range (females with stillborn pups, range of 0-3). Furthermore, the percentage of stillborn pups was 7.3%, 0.8%, 1.1%, 28.7% in test groups 0-3, respectively. The value of test group 3 was outside the historical control range (pups stillborn, 0.0 - 8.3%). Consequently, the rate of liveborn pups indicated by live birth indices was 92.7% / 99.2% / 98.9% and 71.3%** (**:p0.01) in test groups 0 -3, respectively. The value of test group 3 was outside the historical control range (pups liveborn, range of 91.7 – 100 %). The changes in all three parameters were assessed as treatment-related and adverse. One high-dose female (No. 138) had 6 pups and all pups were stillborn. One further high-dose female (No. 139) had a complete litter loss on PND 2 (9 stillborns, 1 found dead, 2 cannibalized). Since higher numbers of stillborn pups and postnatal death occurred in the highdose group, the findings in the two high-dose dams were assessed as treatment-related and adverse.

Regarding developmental toxicity, pup number, status at delivery, viability/mortality and pup body weight was adversely affected by the test substance at the highest test group 3 (5000°ppm). This was based on an increased postimplantation loss (25.5% vs 0.9% in control) leading to a decrease in mean number of F1 pups delivered per dam. A higher rate of stillborn pups (n=25, 28.7% vs. 7.3% in control) led to a decreased rate of liveborn pups (71.3% vs. 92.7% in control). Furthermore, postnatal death was indicated by higher numbers of found dead (16.1% vs 2.7% in control) and cannibalized high-dose pups (n=5) leading to a viability index of 69.6% from PND 0 -4. Consequently, the effect on sex ratio/distribution at birth (females: 31.2%, males: 68.8%) and on PND13 (females: 23.9%, males 76.1%) was assessed as secondary.

A decrease in F1 high-dose pup body weight was observed after birth (PND1) which recovered partially towards PND13.

Conclusions:
Under the conditions of the present OECD 422 combined repeated dose toxicity study with the reproductive/developmental screening test in Wistar rats N,N-Dimethylaminoethanol caused signs of systemic toxicity in females consisting of decreased food consumption and body weight, clinically apparent signs (mortality, tremors and convulsions) together with axonal degeneration in cerebellum, medulla oblongata, cervical and thoracic spinal cord at a concentration of 5000 ppm in drinking water. Males of the same concentration group showed no adverse findings. Thus, the no observed adverse effect level (NOAEL) for general systemic toxicity was 5000 ppm (approx. 257 mg/kg bw/d) for males and 1000 ppm (approx. 89 mg/kg bw/d) for female Wistar rats. The NOAEL for fertility and reproductive performance was 5000 ppm (approx. 257 and 355 mg/kg bw/d, respectively) for male and female rats, the highest concentration tested. The NOAEL for developmental toxicity in the offspring was 1000 ppm based on adverse effects on pup number, status at delivery, viability/mortality and pup body weight at 5000 ppm.
Executive summary:

In the OECD 422 study, the test compound N,N-Dimethylaminoethanol was administered to groups of 10 male and 10 female healthy young Wistar rats (F0 animals) as a solution to the drinking water in different concentrations, i.e. 0 ppm (test group 0), 200 ppm (test group 1), 1000 ppm (test group 2) and 5000 ppm (test group 3). The duration of treatment covered a 2-weeks premating period and mating in both sexes (mating pairs were from the same test group), 3 days postmating in males and approximately 4-weeks postmating in two females (for sperm negative females) as well as the entire gestation and approximately 3 weeks of lactation period in females up to the day of scheduled sacrifice of the animals.

After 2 weeks of premating treatment the F0 animals were mated to produce F1 generation pups. Mating pairs were from the same test group. Mating was discontinued as soon as sperm were detected in the vaginal smear. F0 animals were examined for their reproductive performance including determination of the number of implantation sites and the calculation of postimplantation loss for all F0 females. A detailed clinical observation (DCO) was performed in all animals before the start of the administration period and, as a rule, thereafter at weekly intervals. Water consumption of the F0 parents was determined twice a week. However, during gestation and lactation water consumption of the F0 females were determined on gestation days (GD) 0-1, 4-5, 7-8, 10-11, 14-15, 17-18 and 19-20 and on postnatal days (PND) 1-2, 4-5, 7-8, 10-11 and 12-13. Food consumption of the F0 parents was determined once a week during premating. In dams food consumption was determined for GD 0-7, 7-14, 14-20 and PND 1-4, 4-7, 7-10 and 10-13. Body weights of F0 parents were determined once a week, in males throughout the study and in females during premating. During gestation and lactation period, F0 females were weighed on GD 0, 4, 7, 10, 14, 17 and 20, on the day after parturition (PND 1) and on PND 4, 7, 10 and 13. Estrous cycle data were evaluated for F0 generation females over a two weeks period prior to mating until evidence of mating occurred. Moreover, the estrous stage of each female was determined on the day of scheduled sacrifice.

The pups were sexed and examined for macroscopically evident changes on PND 0. They were weighed on PND 1, 4, 7 and 13. Their viability was recorded. At necropsy on PND 13, all pups were sacrificed with CO2, under isoflurane anesthesia, and examined macroscopically for external and visceral findings. Anogenital distance (defined as the distance from the anus [center of the anal opening] to the base of the genital tubercle) measurements were conducted in a blind randomized fashion, using a measuring ocular on all live male and female pups on PND 1. All surviving pups were examined for the presence or absence of nipple/areola anlagen on PND 13. The number of nipple/areola anlagen were counted. Blood samples were taken from all surplus pups at PND 4 as well as one male and one female pup per litter at PND 13 by decapitation under isoflurane anesthesia for hormone measurement. Blood samples for choline determination were withdrawn from all parental males and females (with litter). Clinico-chemical and hematological examinations were performed in 5 animals per sex and group towards the end of the administration period. Blood samples from all dams at PND 14 and all males at termination were taken by puncturing the retrobulbar venous plexus under isoflurane anesthesia for hormone measurement. At the end of the administration period a functional observational battery was performed and motor activity was measured in 5 parental males and females per group. All F0 parental animals were sacrificed by decapitation, under isoflurane anesthesia, and were assessed by gross pathology. Weights of selected organs were recorded and a histopathological examination was performed.

Under the conditions of the present OECD 422 combined repeated dose toxicity study with the reproductive/developmental screening test in Wistar rats N,N-Dimethylaminoethanol caused signs of systemic toxicity in females consisting of decreased food consumption and body weight, clinically apparent signs (mortality, tremors and convulsions) together with axonal degeneration in cerebellum, medulla oblongata, cervical and thoracic spinal cord at a concentration of 5000 ppm in drinking water. Males of the same concentration group showed no adverse findings. Thus, the no observed adverse effect level (NOAEL) for general systemic toxicity was 5000 ppm (approx. 257 mg/kg bw/d) for males and 1000 ppm (approx. 89 mg/kg bw/d) for female Wistar rats. The NOAEL for fertility and reproductive performance was 5000 ppm (approx. 257 and 355 mg/kg bw/d, respectively) for male and female rats, the highest concentration tested. The NOAEL for developmental toxicity in the offspring was 1000 ppm based on adverse effects on pup number, status at delivery, viability/mortality and pup body weight at 5000 ppm.

Endpoint:
screening for reproductive / developmental toxicity
Remarks:
based on test type
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
12 OCT 2009 to 29 OCT 2010
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 421 (Reproduction / Developmental Toxicity Screening Test)
Qualifier:
according to guideline
Guideline:
other: EPA OPPTS 870.3550 (Reproduction/ Developmental Toxicity Screening test)
GLP compliance:
yes (incl. QA statement)
Limit test:
no
Species:
rat
Strain:
Wistar
Details on species / strain selection:
The test guideline requires the rat to be used as the animal species. Wistar rats were selected since extensive historical control data were available for this strain.
Sex:
male/female
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH (males) and Charles River Laboratories, France (females)
- Females (if applicable) nulliparous and non-pregnant: yes
- Age at study initiation: 11 - 13 weeks
- Weight at study initiation: male animals: 279.9 g - 315.9 g, female animals: 173.2 g - 202.8 g
- Housing:
- During the study period, the rats were housed individually
- During overnight matings, male and female mating partners were housed together
- Pregnant animals and their litters were housed together until PND 4.
- Diet: ad libitum
- Water: ad libitum
- Acclimation period: 5-6 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Air changes (per hr): 15
- Photoperiod (hrs dark / hrs light): 12/12

IN-LIFE DATES: From: 15 Dec 2009 To: 19 Aug 2010
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
The test substance solutions in drinking water were prepared at the beginning of the administration period and thereafter in intervals, which took into account the analytical results of the stability verification. The maximum period for which each preparation was used was 7 days.

For the preparation of the administration solutions the test substance was weighed in a graduated measuring flask depending on the dose group, topped up with drinking water and subsequently thoroughly shaken until completely dissolved.


VEHICLE
- Concentration in vehicle: 0, 1, 3, 10 g/100mL
- Amount of vehicle (if gavage): 10 mL/kg bw
Details on mating procedure:
Males and females from the same dose group were mated 13 days after the beginning of treatment, overnight in a ratio of 1:1.
- M/F ratio per cage: 1:1
- Length of cohabitation: 2 weeks
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Concentration control analyses of the test substance preparations:
The method of analysis was Capilary electrophoresis (CE). All measured values for N-Methyldiethanolamin were in the expected range of the target concentrations (90-110%).
Duration of treatment / exposure:
The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females.
Frequency of treatment:
once daily
Details on study schedule:
N-Methyldiethanolamin was given daily as an aqueous solution to groups of 10 male and 10 female Wistar rats (F0 animals) by stomach tube at doses of 100, 300 and 1000 mg/kg body weight/day (mg/kg bw/day). Control animals were dosed daily with the vehicle only (drinking water). The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females. The females were allowed to deliver and rear their pups until day 4 after parturition. Four days after PND 4 of the female, which delivered last, all parental females were sacrificed and examined. Pups were sacrificed on PND 4 and gross necropsied. The male animals were sacrificed 28 days after the beginning of the administration and examined.
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Remarks:
Control
Dose / conc.:
100 mg/kg bw/day (actual dose received)
Remarks:
actual ingested
Dose / conc.:
300 mg/kg bw/day (actual dose received)
Remarks:
actual ingested
Dose / conc.:
1 000 mg/kg bw/day (actual dose received)
Remarks:
actual ingested
No. of animals per sex per dose:
10
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale:
The dose levels were selected based on the results of a preceding study (Project No. 06R0087/01248, BASF SE). In this study N-Methyldiethanolamin was given daily as an aqueous solution to groups of 3 male and 3 female Wistar rats (F0 animals) by stomach tube at doses of 100, 300 and 1000 mg/kg body weight/day (mg/kg bw/d) for 2 weeks. Control animals were dosed daily with the vehicle only (drinking water). Clinical signs, food consumption and body weights were determined. The only clearly substance-related effect was “salivation after treatment” in all males and females at the high dose level. Food consumption and body weights did not show significant differences.
Positive control:
no
Parental animals: Observations and examinations:
CAGE SIDE OBSERVATIONS: Yes
A check for moribund or dead animals was made twice daily on working days or once daily (weekends or public holidays).

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: daily

BODY WEIGHT: Yes
- Time schedule for examinations: once a week in males throughout the study and in females during premating and mating. During gestation and lactation, F0 females were weighed on gestation days (GD) 0, 7, 14 and 20, on the parturition day and postnatal day (PND) 4.

FOOD CONSUMPTION:
Food consumption of the F0 parents was determined once weekly during premating. In dams food consumption was determined for gestation days 0 - 7, 7 - 14, 14 - 20 and lactation days 1 - 4.
Oestrous cyclicity (parental animals):
The parturition and lactation behavior of the dams was generally evaluated in the mornings in combination with the daily clinical inspection of the dams. Only particular findings (e.g. inability to deliver) were documented on an individual dam basis. On weekdays (except public holidays) the parturition behavior of the dams was inspected in the afternoons in addition to the evaluations in the mornings. The day of parturition was considered the 24-hour period from about 15.00 h of one day until about 15.00 h of the following day.
The pairing partners, the number of mating days until vaginal sperm were detected, and gestational status were recorded for F0 females.
For the females, mating, fertility and gestation indices were calculated .
Sperm parameters (parental animals):
Parameters examined in P male parental generations:
testis weight and epididymis weight, and mating and fertility indices (male mating index and male fertility index) were calculated.
Litter observations:
STANDARDISATION OF LITTERS
- Performed on day 4 postpartum: no

PARAMETERS EXAMINED
The following parameters were examined in [F1] offspring:
Sex, liveborn or stillborn, number of all delivered pups determined as soon as possible on the day of birth. On the day of birth (PND 0) the sex of the pups was determined by observing the distance between the anus and the base of the genital tubercle; normally, the anogenital distance is considerably greater in male than in female pups (the sex of the pups was finally confirmed at necropsy). In general, a check was made for any dead or moribund pups twice daily on workdays (once in the morning and once in the afternoon) or as a rule, only in the morning on Saturdays, Sundays or public holidays. The live pups were examined daily for clinical symptoms (including gross-morphological findings) during the clinical inspection of the dams. If pups showed particular findings, these were documented with the dam concerned. The pups were weighed on the day after birth (PND 1) and on PND 4.

GROSS EXAMINATION OF DEAD PUPS:
Pups that die before this initial examination are defined as stillborn pups.
All stillborn pups and all pups that died before PND 4 were examined externally, eviscerated and their organs were assessed macroscopically.
Postmortem examinations (parental animals):
SACRIFICE
- Male animals: All surviving animals 28 days after the beginning of the administration
- Maternal animals: All surviving animals. Four days after PND 4 of the female, which delivered last, all parental females were sacrificed and examined.

GROSS NECROPSY
- Gross necropsy consisted of external and internal examinations including the cervical, thoracic, and abdominal viscera.

HISTOPATHOLOGY / ORGAN WEIGHTS
The following weights were determined in all animals sacrificed on schedule:

1. Anesthetized animals
2. Epididymides
3. Testes
4. Ovaries
5. Liver

The following organs or tissues of parental animals were fixed in 4% buffered formaldehyde or in modified Davidson’s solution:

1. Adrenal glands
2. All gross lesions
3. Testes (fixed in modified Davidson’s solution)
4. Epididymides (fixed in modified Davidson’s solution)
5. Pituitary gland
6. Prostate gland, seminal vesicles, coagulation glands
7. Ovaries (fixed in modified Davidson’s solution)
8. Uterus, oviducts, vagina
9. Parts of the liver (lobus dexter lateralis, processus caudatus, processus papillaris) of all male and female animals were deeply frozen and stored at -80°C for possible analysis of choline. The remaining parts of the liver were fixed in 4% buffered formaldehyde solution.

Histopathology was performed on the following organs: All gross lesions, Testes, Epididymides, Ovaries.
Postmortem examinations (offspring):
SACRIFICE
- All pups with scheduled sacrifice on PND 4 were sacrificed under Isoflurane anesthesia by means of CO2.
- These animals were subjected to postmortem examinations (macroscopic and/or microscopic examination) as follows:

GROSS NECROPSY
- All pups were examined externally and eviscerated; their organs were assessed macroscopically. All pups without notable findings or abnormalities were discarded after their macroscopic evaluation. Animals with notable findings or abnormalities were evaluated on a case-by-case basis, depending on the type of finding

Statistics:
Food consumption (parental animals), body weight and body weight change (parental animals and pups; for the pup weights, the litter means were used), number of mating days, duration of gestation, number of implantation sites, postimplantation loss and % postimplantation loss, number of pups delivered per litter: simultaneous com-parison of all dose groups with the control group using the DUNNETT-test (two-sided) for the hypothesis of equal means.

Male and female mating indices, male and female fertility indices, gestation index, females with liveborn pups, females with stillborn pups, females with all stillborn pups, live birth index, pups stillborn, pups died, pups cannibalized, pups sacrificed moribund, viability index, number of litters with affected pups at necropsy: Pairwise comparison of each dose group with the control group using FISHER'S EXACT test for the hypothesis of equal proportions.

Proportions of affected pups per litter with necropsy observations: Pairwise comparison of each dose group with the control group using the WILCOXON-test (one-sided) for the hypothesis of equal medians.

Weight parameters: Non-parametric one-way analysis using KRUSKAL-WALLIS test (two-sided). If the resulting p-value was equal or less than 0.05, a pairwise comparison of each dose group with the control group was performed using the WILCOXON test for the hypothesis of equal medians.
Reproductive indices:
Male mating index (%)=number of males with confirmed mating/number of males placed with females x 100%

Male fertility index (%)= number of males proving their fertility/ number of males placed with females x 100%

Female mating index (%)= number of females mated/ number of females placed with males x 100%

Female fertility index (%)= number of females pregnant/number of females mated x 100%

Gestation index (%)= number of females with live pups on the day of birth/number of females pregnant x 100%

Postimplantation loss (%)=number of implantations – number of pups delivered/number of implantations x 100%

Live birth index (%)= number of liveborn pups at birth/total number of pups born x 100%

Viability index (%) = number of live pups on day 4 after birth/number of live pups on the day of birth x 100%
Offspring viability indices:
Postimplantation loss (%)=number of implantations – number of pups delivered/number of implantations x 100%

Live birth index (%)= number of liveborn pups at birth/total number of pups born x 100%

Viability index (%) = number of live pups on day 4 after birth/number of live pups on the day of birth x 100%
Clinical signs:
no effects observed
Description (incidence and severity):
- At 1000 mg/kg bw/day:
- Nine Males and felames showed transient salivation for a few min. immediately after each treatment. It was likely to be induced by the taste of the test substance or by local irritation of the upper digestive tract. It is not considered to be a sign of systemic toxicity.
- One female (No. 135) with unsteady gait, poor general state and oblique head position. These findings were assessed as being incidental and not test substance related.
- Four females (Nos. 133, 137, 138 and 139) lost all pups (PND 1 or 2). In 2 of this dams undelivered pups were palpable in their abdomen before. The other two femalesdid not properly nurse their pups (pups had no or less milk in the stomach). This was assessed as being test substance-related.

- At 100 mg/kg bw/day: One female lost all pups (PND 2) and did not properly nurse its pups before (pups had no or less milk in the stomach). Due to the isolated occurrence and the lack of a dose-response relationship, this was assessed as being incidental.
Mortality:
no mortality observed
Description (incidence):
There were no mortalities in any of the male and female F0 parental animals in any of the groups.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
- At 1000 mg/kg bw/day:
- Decreased bw gain in males in treatment weeks 2-3 (57% below control) and 0-3 (29% below control)
- Decreased bw in females on gestation days 14 and 20 (up to 14% below control) and lactation day 4 (about 5% below control)
- Decreased bw gain in females between gestation days 7-20 (up to about 46% below control) and body weight loss (-3.9 g) between lactation days 0-4
- At 300 mg/kg bw/day:
- Decreased bw gain in males in treatment weeks 1-2 (45% below control)
- Decreased terminal bw in males and females (4 and 5 % below controls, respectively).
- At 100 mg/kg bw/day: no test substance-related adverse findings
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
- At 1000 mg/kg bw/day: Decreased food consumption in females (-37%) during lactation days 1-4 but might be due to the lower pup number in this group and consequently a lower caloric demand of the F0 females.
- At 300 and 100 mg/kg bw/d: Females id not show any test substance-related changes of food consumption during the whole treatment period.

Males did not show any test substance-related changes of food consumption during the whole treatment period.
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Histopathological findings: non-neoplastic:
no effects observed
Description (incidence and severity):
All lesions noted are regarded to be incidental or spontaneous in nature and not related to treatment. Especially, there were no pathomorphological patterns in the liver that did correlate to enlarged size and weight increase.

Non pregnant pairs:
- At 300 mg/kg bw/d:
- One male (No. 25) showed macroscopically slightly reduced size of epididymides and testes. Histopathologically, diffuse seminiferous tubule atrophy (grade 4) in the testes and aspermia in the epididymides were observed and are correlating to the macroscopic findings.
- One female (No. 125) did not show any macroscopic findings and was therefore not examined histopathologically.
Reproductive function: oestrous cycle:
no effects observed
Reproductive function: sperm measures:
no effects observed
Description (incidence and severity):
The male fertility index ranged between 90% and 100% without showing any relation to dosing. This reflects the normal range of biological variation inherent in the strain of rats used for this study.
For all males except one (No. 25, 300 mg/kg bw/day) copulation was confirmed. Thus, the male mating index varied in the different test groups between 90% and 100%.
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
- The mean duration until sperm was detected (GD 0) varied between 1.6 and 2.6 days without any relation to dosing
- The female mating index varied between 90%- 100%
- All sperm positive rats delivered pups or had implants in utero. Thus, the fertility index was 100% in all test groups.
- At 1000 mg/kg bw/day: the mean duration of gestation was statistically significantly increased (22.8 days).
- At 0, 100, 300 mg/kg bw/day: the mean duration of gestation varied between 21.9 and 22.1 days
- At 1000 mg/kg bw/day: the number of implantation sites was distinctly reduced ( 6.7 vs. 12.9 in control). In this group, both the number of resorptions as well as the % postimplantation loss was distinctly increased (21 and 31.0% vs. 8 and 6.0% in control).
- The gestation index was 100% in all groups
The non pregnant female rat of test group 300 mg/kg bw/day, animal No. 125, mated with male No. 25 did not show any macroscopic findings to explain these infertility. However, as the male animal No. 25 sowed reduced size of epididymides and testes, accompanied by seminiferous tubule atrophy and aspermia, the infertilily of this pair can clearly be attributed to the male animal. However, due to the lack of a dose response relationship, this was assessed as being incidental.
Dose descriptor:
NOAEL
Remarks:
(for systemic toxicity)
Effect level:
100 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
body weight and weight gain
food consumption and compound intake
Dose descriptor:
NOAEL
Remarks:
(for reproductive performance and fertility)
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Sex:
female
Basis for effect level:
reproductive performance
Clinical signs:
no effects observed
Description (incidence and severity):
There were no test substance-related adverse clinical signs observed in any pups of the different test groups
Mortality / viability:
mortality observed, treatment-related
Description (incidence and severity):
- At 1000 mg/kg bw/day: Reduced viability index (62% vs 99% in control), resulting from significantly higher numbers of died (5 vs. 0 in control) and cannibalized pups (12 vs. 1 in control).
- At 300 and 100 mg/kg bw/day: No test substance-related adverse findings
- At 100 mg/kg bw/d; 87% vs. 99% in cotrol), reduced viablility index resulting from significantly higher numbers of cannibalized pups (11 vs. 1 in control), mainly caused by dam No. 112, which cannibalized 11 of its 14 pups. This can therefore be seen as a spontaneous event.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
- At 1000 mg/kg bw/day:
- Decreased pup body weights on PND 4, average difference to the control 20%
- Decreased pup body weight gain during PND 1 – 4, average difference to the control 52%

- At 300 and 100 mg/kg bw/day: No test substance-related adverse findings
Gross pathological findings:
no effects observed
Description (incidence and severity):
A few pups showed spontaneous findings at gross necropsy, such as post mortem autolysis and empty stomach. Due to the low incidence and the lack of a clear dose-response relationship, these findings were assessed as being indicental.
Dose descriptor:
NOAEL
Remarks:
(for developmental toxicity)
Generation:
F1
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
mortality
body weight and weight gain
Reproductive effects observed:
not specified

Clinical signs of toxicity were seen at the high dose level (1000 mg/kg bw/day) such as decreased food consumption in females during lactation and decreased body weight gain in males and females, resulting in reduced terminal body weights in both sexes. Reduced terminal body weights were also seen at the mid dose level (300 mg/kg bw/day).

Concerning reproductive parameters,total litter loss in 4 females, undelivered pups, insufficient lactation behavior, increased duration of gestation, decreased number of implantation sites, increased postimplantation loss and decreased number of delivered pups, reduced pup viability, decreased pup body weights and weight gains were seen exclusivelyat the high dose level (1000 mg/kg bw/day), i.e. only in the presence of parental toxicity.

Pathology revealed increased liver weights at all dose levels, however without any morphological correlate. These findings were therefore assessed as being an adaptive phenomenon, but not an adverse effect. There were no further treatment-related lesions detected, especially, there were no weight or substance-related pathomorphological effects on testes, epididymides, and ovaries present.

Conclusions:
In conclusion, the administration of N-Methyldiethanolamin at dose levels of 1000 and 300 mg/kg bw/d caused toxic effects on body weight. The NOAEL (no observed adverse effect level) for general, systemic toxicity was therefore 100 mg/kg body weight/day for the F0 parental males and females.
The NOAEL for reproductive performance and fertility was 300 mg/kg body weight/day for the F0 parental rats based upon findings such as litter loss, insufficient lactation behavior, and increased duration of gestation.
The NOAEL for developmental toxicity was 300 mg/kg body weight/day, based on findings such as reduced viability index and reduced postnatal offspring weight gain.
Thus, reproductive parameters were affected only in the presence of parental toxicity.
Executive summary:

This reproduction/developmental toxicity screening study was conducted according to OECD 421 and in accordance with GLP.

N-Methyldiethanolamin was given daily as an aqueous solution to groups of 10 male and 10 female Wistar rats (parental generation) by stomach tube at doses of 100, 300 and 1000 mg/kg bw/day. Control animals were dosed daily with the vehicle only (drinking water). The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females.

At the dose level of 1000 mg/kg bw/day, decreased food consumption in females, and decreased body weight gain and terminal body weights in males and females were observed. Furthermore, total litter loss, undelivered pups palpable, insufficient lactation behaviour (pups had no or less milk in stomach), increased duration of gestation, decreased number of implantation sites, increased post-implantation loss, decreased number of delivered pups, decrease of terminal body weights in males and females were recorded. In F1 pups a reduced viability index, decreased body weights and body weight gain was observed.

At the dose level of 300 mg/kg bw/day, decreased terminal body weights in males and females were observed. In pups, there were no test substance-related adverse findings.

At the dose level 100 mg/kg bw/day, no test substance-related adverse findings neither in parental generation nor in pups.

 

In conclusion, the administration of N-Methyldiethanolamin at dose levels of 1000 and 300 mg/kg bw/d caused toxic effects on body weight. The NOAEL (no observed adverse effect level) for general, systemic toxicity was therefore 100 mg/kg body weight/day for the F0 parental males and females.

The NOAEL for reproductive performance and fertility was 300 mg/kg body weight/day for the F0 parental rats based upon findings such as litter loss, insufficient lactation behavior, and increased duration of gestation.

The NOAEL for developmental toxicity was 300 mg/kg body weight/day, based on findings such as reduced viability index and reduced postnatal offspring weight gain.

Thus, reproductive parameters were affected only in the presence of parental toxicity.

Endpoint:
two-generation reproductive toxicity
Remarks:
based on test type
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
Published 2009
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
other: GLP Guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 416 (Two-Generation Reproduction Toxicity Study)
Deviations:
yes
Remarks:
Food consumption was not determined between days 14 and 21 after parturition
GLP compliance:
yes
Limit test:
no
Species:
rat
Strain:
other: Crl:WI (Han)
Sex:
male/female
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services GmbH, Germany
- Age at study initiation: (P) 16 days
- Weight at study initiation: (P) Males: 162.1 (142.5 – 186.5) g ; Females: 126.2 (110.6 – 145.1) g;
- Fasting period before study: none
- Housing: housed individually in type DK III stainless steel wire mesh cages
- Diet: ground Kliba maintenance diet mouse/rat “GLP” meal, supplied by Provimi Kliba SA, Kaiseraugst, Switzerland ad libitum
- Water: ad libitum
- Acclimation period: 16 days


ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Air changes (per hr): 10-15
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
oral: feed
Vehicle:
unchanged (no vehicle)
Details on exposure:
DIET PREPARATION
The test substance (ethanolamine hydrochloride, EAH) was weighed and thoroughly mixed with a small amount of food. Then corresponding amounts of food, depending on the dose group, were added to this premix in order to obtain the desired concentrations. Mixing was carried out for about 10 minutes in a laboratory mixer. Test diets were prepared at intervals, which guaranteed that the test substance in the diet remained stable throughout the feeding period.
Details on mating procedure:
- M/F ratio per cage: 1/1
- Length of cohabitation: over night
- Proof of pregnancy: [sperm in vaginal smear] referred to as [day 0] of pregnancy
- After 14 days of unsuccessful pairing replacement of first male by another male with proven fertility.
- After successful mating each pregnant female was caged (how): individual
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The stability of EAH in the diet over 32 days at room temperature was investigated analytically before the beginning of the study. Homogeneity and concentration control analyses were carried out at the beginning and toward the end of the premating periods. At least one analysis of test substance preparations for female animals was carried out during the gestation and lactation periods.

The analyses were carried out at the Analytical Chemistry Laboratory of Experimental Toxicology and Ecology of BASF SE, Ludwigshafen, Germany.
Duration of treatment / exposure:
Test material exposure was continuous. At least 75 days after the beginning of treatment, males and females from the same dose group were mated. The day on which sperm were detected was denoted "Day 0" and the following day "Day 1" post coitum (p.c.). Females were allowed to litter and rear their pups (F1 generation pups) until Day 4 (standardization) or 21 post partum (p.p.). After weaning of F1 pups the F0 generation parental animals were terminated.
Frequency of treatment:
daily
Details on study schedule:
After standardization (on PND 4), all pups were killed (with the exception of 25 male and 25 female F1 pups from each dose group which were chosen to be F1 generation parental animals). These animals were examined for sexual maturity prior to mating. If fewer than 25 litters were available in a group or if one sex was missing in a litter, more animals were taken from the other litters of the respective test group to obtain the full number. All selected animals were treated with the test substance at the same dose level as their parents from their growth into adulthood up to about one day before they were terminated. At least 75 days after assignment of the F1 generation parental animals, the males and females were generally mated at a ratio of 1 : 1. Partners were randomly assigned, with mating of siblings excluded. The females were allowed to litter and rear their pups (F2 generation pups) until Day 4 (standardization) or 21 p.p. Shortly after the F2 generation pups had been weaned, the F1 generation parental animals were terminated.
Dose / conc.:
100 mg/kg bw/day (nominal)
Remarks:
nominal in diet
Dose / conc.:
300 mg/kg bw/day (nominal)
Remarks:
nominal in diet
Dose / conc.:
1 000 mg/kg bw/day (nominal)
Remarks:
nominal in diet
No. of animals per sex per dose:
25
Control animals:
yes, plain diet
Positive control:
not done
Parental animals: Observations and examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: twice daily on working days and once daily on weekends

DETAILED CLINICAL OBSERVATIONS: Yes

BODY WEIGHT: Yes
- Time schedule for examinations: body weights of F0 and F1 parents were determined once weekly; during gestation and lactation F0 and F1 females were weighed on days 0, 7, 14 and 20 of gestation, and on days 1, 4, 7, 14 and 21 after birth.

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study):
- Time schedule: once weekly (over a period of at least 6 days each) and weekly during gestation (days 0-7, 7-14, 14-20 post coitum; p.c.) and lactation periods (days 1-4, 4-7, 7-14 post partum; p.p.).

Food consumption was not determined between days 14 and 21 after parturition as required in the test guidelines. Food consumption also was not determined for females without positive evidence of sperm and for females without litter.

OTHER:
The F1 and F2 pups were sexed on the day of birth (day 0 p.p.) and weighed on days 1, 4, 7, 14, and 21 p.p. Their viability was recorded. At necropsy, all pups were examined macroscopically (including weight determinations of brain, spleen and thymus in one pup/sex/litter).

Serum concentrations of the test substance:
Blood samples were taken from all F0 and F1 parental animals of each sex and test group during week 10 of premating treatment and the plasma was analyzed for the concentration of Ethanolamine hydrochloride
Oestrous cyclicity (parental animals):
Estrous cycle data were evaluated for F0 and F1 generation females over a three week period prior to mating until evidence of mating occurred. Moreover, the estrous stage of each female was determined on the day of scheduled sacrifice.
Sperm parameters (parental animals):
Immediately after necropsy and organ weight determination, the right testis and cauda epididymidis were taken from the F0 and F1 males of all dose groups. Sperm head counts in the testis and cauda epididymis, sperm morphology and sperm motility were determined according to established methods. Tissues from the control and highest dose group (1000 mg/kg bw/d) were evaluated. Only in case of significant differences between these groups, additional sperm head counts were made for the mid-dose (300 mg/kg bw/d) and, if necessary, for the low-dose group (100 mg/kg bw/d) to reveal the dose level affecting these parameters.
Litter observations:
STANDARDISATION OF LITTERS
- Performed on day 4 postpartum: yes
- Individual litters were standardized in such a way that, where possible, each litter contained 4 male and 4 female pups (always the first 4 pups/sex and litter were taken for further rearing). If it was not possible for individual litters to have 4 pups/sex, it was proceeded in such a way that 8 pups per litter were present for further rearing (e.g. 5 male and 3 female pups). Standardization of litters was not performed in litters with 8 pups.

On the day of birth, all pups derived from the F0 parents (F1 litter) and the F1 parents (F2 litter) were examined as soon as possible to determine the total number of pups, and the number of liveborn as well as stillborn pups of each litter.
On the day of birth (day 0 p.p.), the sex of the pups was determined by observing the distance between the anus and the base of the genital tubercle; normally, the anogenital distance is considerably greater in male than in female pups. Subsequently (on Day 21 p.p.), the sex of the pups was assessed by the external appearance of the ano­genital region and/or the mammary line and was finally confirmed at necropsy. The live pups were examined daily for clinical symptoms (including gross-morphological findings) during the clinical inspection of the dams. If pups showed any special findings, these were documented with the dam concerned. The pups were weighed on the day after birth (day 1 p.p.) and on days 4 (before standardization), 7, 14 and 21 after birth.
Pups, which died before examination, were designated as stillborn pups. In general, a check was made for dead or moribund pups twice daily on workdays (once in the morning and once in the afternoon) and once in the morning on Saturdays, Sundays or public holidays. The number and percentage of dead pups on the day of birth (day 0) and of pups dying between Days 1-4, 5-7, 8-14 and 15-21 of the lactation period were determined; however, pups, which died accidentally or had to be sacrificed due to maternal death, were not included in these calculations. The number of live pups/litter was calculated on the day of birth, and on lactation days 4, 7, 14, and 21.
Postmortem examinations (parental animals):
All F0 and F1 parental animals were sacrificed by decapitation under Isoflurane anesthesia. The exsanguinated animals were necropsied and assessed by gross pathology, special attention was given to the reproductive organs. The liver, kidneys, adrenal glands, testes, epididmides. Cauda epididymis, prostate, seminal vesicles, ovaries, uterus, spleen, brain, pituitary gland and thyroid glands (with parathyroids) were weighed and the vagina, cervix uterie, uterus, ovaries, oviducts, left testis, left epididymis, seminal vesicles, coagulation glands, prostate, pituitary gland, adrenal glands, liver, kidneys, spleen, brain, thyroids (with parathyroids)and all gross lesions were fixed in an appropriate fixative, histologically processed and examined by light microscopy. From both ovaries (”ovary 1” and “ovary 2”) of F1 female animals (control and top dose), five sections were taken from the proximal and the distal part of the ovaries, at least 100 µm apart from the inner third of the ovary. All ovarian sections were prepared and evaluated for numbers of primordial and growing follicles.
As soon as possible after termination, one portion of the liver (lobus medialis) of each 10 dams per group was sampled to be analyzed for choline concentration.
Postmortem examinations (offspring):
All pups with scheduled sacrifice (i.e. pups, which were culled on day 4 p.p., and pups, which were sacrificed on day 21 p.p. or subsequent days) were killed by means of CO2. The spleen and thymus of 1 pup/sex and litter from the F1 and F2 pups were weighed. All stillborn pups and all pups that died up to weaning were examined externally, eviscerated and their organs were assessed macroscopically. All pups without any notable findings or abnormalities were discarded after their macroscopic evaluation.
Statistics:
Please see 'any other information'
Reproductive indices:
For the males, mating and fertility indices were calculated for F1 and F2 litters.
For F0 and F1 females, following parameters were noted: mating partners, number of mating days until positive evidence of sperm, and gestational status. The total number of delivered pups/dam was recorded and the number of liveborn and stillborn pups were noted. The live birth index(in %) was calculated for F1 and F2 litters. Implantation sites were stained and counted and the postimplantation loss (in %) was calculated
Offspring viability indices:
- Viability and lactation indices were calculated.
Clinical signs:
no effects observed
Mortality:
no mortality observed
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Statistically significantly decreased body weight gain of the dams during gestation, body weight 8% below control on gestation day 20
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Statistically significantly decreased food consumption in parental females during lactation
Urinalysis findings:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d (male/female): Yellow discoloured urine
Organ weight findings including organ / body weight ratios:
effects observed, non-treatment-related
Histopathological findings: non-neoplastic:
no effects observed
Reproductive function: sperm measures:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d:
- Statistically significantly decreased sperm head count in the cauda epididymidis of males.
- Statistically significantly decreased absolute and relative weight of epididymides, cauda epididymidis and prostate in males
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Statistically significantly less implantation sites, statistically significantly increased post-implantation loss, statistically significantly smaller litters
300 mg/kg bw/d
F0 or F1 parental animals: No test substance-related adverse effects

100 mg/kg bw/d
F0 or F1 parental animals: No test substance-related adverse effects
Dose descriptor:
NOAEL
Effect level:
300 mg/kg bw/day (nominal)
Sex:
male/female
Basis for effect level:
body weight and weight gain
food consumption and compound intake
reproductive function (sperm measures)
reproductive performance
other: fertility
Clinical signs:
no effects observed
Mortality:
no mortality observed
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Statistically significantly decreased body weight gain of the dams during gestation
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Decreased food consumption in parental females during lactation
Urinalysis findings:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Yellow discoloured urine for male and female parental animals
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d :The decrease of absolute weights of cauda epididymis and epididymides in male top-dose animals were considered to be treatment-related.

The increase of absolute kidney weights of male and female animals in mid- (300 mg/kg bw/d) and top-dose (1000 mg/kg bw/d) groups, respectively, was statistically significant. Because no histomorphological correlate was detected, a treatment-related weight increase was less likely.
The decrease of spleen weights in top-dose males as well as the increase of thyroid glands in top-dose males and mid- and top-dose females, respectively, is considered incidental and not treatment-related due to a missing dose-response relationship

(please refer to 'any other information on results')
Gross pathological findings:
no effects observed
Histopathological findings: non-neoplastic:
no effects observed
Reproductive function: sperm measures:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d: Statistically significantly decreased absolute and relative weight of epididymides and cauda epididymidis in males
Reproductive performance:
effects observed, treatment-related
Description (incidence and severity):
1000 mg/kg bw/d:
Statistically significantly less implantation sites
Statistically significantly increased post-implantation loss
Statistically significantly smaller litters
300 mg/kg bw/d
F0 or F1 parental animals: No test substance-related adverse effects

100 mg/kg bw/d
F0 or F1 parental animals: No test substance-related adverse effects
Dose descriptor:
NOAEL
Effect level:
300 mg/kg bw/day (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
body weight and weight gain
food consumption and compound intake
ophthalmological examination
urinalysis
reproductive function (sperm measures)
reproductive performance
other: Fertility
Mortality / viability:
no mortality observed
Body weight and weight changes:
no effects observed
Sexual maturation:
no effects observed
Anogenital distance (AGD):
no effects observed
The following test substance-related effects/findings were recorded:

1000 mg/kg bw/d
F1 pups: No test substance-related adverse effects

300 mg/kg bw/d
F1 pups: No test substance-related adverse effects

100 mg/kg bw/d
F1 pups: No test substance-related adverse effects
Dose descriptor:
NOAEL
Generation:
F1
Effect level:
1 000 mg/kg bw/day (nominal)
Sex:
male/female
Basis for effect level:
other: no pre-and postnatal developmental toxicity; no altered pup viability, no changes in body weight, sex ratio and sexual maturation.
Mortality / viability:
no mortality observed
Body weight and weight changes:
no effects observed
Sexual maturation:
no effects observed
Anogenital distance (AGD):
no effects observed
The following test substance-related effects/findings were recorded:

1000 mg/kg bw/d
F2 pups: No test substance-related adverse effects

300 mg/kg bw/d
F2 pups: No test substance-related adverse effects

100 mg/kg bw/d
F2 pups: No test substance-related adverse effects
Dose descriptor:
NOAEL
Generation:
F2
Effect level:
1 000 mg/kg bw/day (nominal)
Sex:
male/female
Basis for effect level:
other: no pre-and postnatal developmental toxicity; no altered pup viability, no changes in body weight, sex ratio and sexual maturation.
Reproductive effects observed:
not specified

Test substance stability:

The stability of test substance in rat diet was demonstrated for a period of 32 days at room temperature in a different batch of comparable quality, which was not used for the study. The homogeneity of the mixtures was verified. The concentration control analyses of the samples taken revealed that the values were within a range of 90-110% of the nominal concentration in all analyses at all time points, with the exception of one concentration in the feed of the high-dose group (88%).

Plasma concentrations of 2 -aminoethanol were below 3 mg/kg for all control animals, <3 - 4 mg/kg for the low dose animals, 8 - 11 mg/kg for the mid dose animals and 60 – 81 mg/kg for the high dose animals.

Toxicokinetic data of 2 -aminoethanol (calculated as 2 -aminoethanol hydrochloride) from this two-generation reproduction toxicity study show a dose dependency of the plasma levels of 2 -aminoethanol in the experimental animals and there with prove the bioavailability of 2 -aminoethanol hydrochloride in principle.

 

Under these conditions, no test substance-related findings from clinical examinations or gross and histopathology were observed, which indicate that the administration of the test compound via the diet adversely affected the fertility or reproductive performance of the F0 or F1 parental animals up to and including a nominal dose of 300 mg/kg bw/d. Estrous cycle data, mating behavior, conception, gestation, parturition, lactation and weaning as well as sperm parameters, sexual organ weights and gross and histopathological findings of these organs (including differential ovarian follicle counts in the F1 females) were comparable between the rats of all test groups.

At the high-dose level (1000 mg/kg bw/d), absolute and relative weights of epididymides and cauda epididymidis were decreased and, in the F0 generation only, the number of homogenization resistant caudal epididymal sperm was slightly, but significantly reduced. However, histomorphological correlates for these findings were missing.

 

In the high-dose F0 and F1 generation females (1000 mg/kg bw/d), decreased numbers of implants and increased resorption rates resulted in significantly smaller litters, giving evidence for an adverse effect of the test compound on fertility and/or reproductive performance at high doses. It has to be noted that a dose of 1000 mg/kg bw/d also caused beginning systemic toxicity in these females, as was indicated by reduced food consumption and/or body weight gain during gestation/lactation.

 

All data recorded during gestation and lactation in terms of embryo-/fetal and pup development gave no indications for any developmental toxicity in the F1 and F2 offspring up to a dose level of 1000 mg/kg bw/d. The test substance did not adversely influence pup viability, body weight, sex ratio and sexual maturation.

 

Thus, under the conditions of the present two-generation reproduction toxicity study, the NOAEL (no observed adverse effect level) for fertility, reproductive performance and systemic toxicity in parental F0 and F1 Wistar rats is 300 mg/kg bw/d.

 

The NOAEL for pre-and postnatal developmental toxicity in their offspring is 1000 mg/kg bw/d.

Tables

Mean test substance intake (mg/kg bw/d; minimum value / maximum value)

 

Test group 01
(100 mg/kg bw/d)

Test group 02
(300 mg/kg bw/d)

Test group 03
(1000 mg/kg bw/d)

F0 males

94.3 (72.4 / 102.5)

283.2 (218.4 / 309.4)

943.3 (716.7 / 1032.6)

F0 females (premating)

96.7 (80.5 / 100.7)

289.6 (241.2 / 304.9)

964.4 (792.4 / 1017.8)

F0 females (F1 litter)
- gestation period
- lactation period*


103.5 (92.6 / 111.6)
99.2 (81.6 / 120.2)


315.2 (284.8 / 337.9)
306.7 (249.7 / 370.3)


1043.2 (989.4 / 1084.7)
866.0 (668.6 / 1053.9)

* = Days 1–14 p.p. only

Absolute organ weights (P-generation)

Compared to the controls (= 100%), the following values (in %) were significantly changed (printed in bold):

 

Male animals

Female animals

Group

01

100 mg/kg bw/d

02

300 mg/kg bw/d

03

1000 mg/kg bw/d

01

100 mg/kg bw/d

02

300 mg/kg bw/d

03

1000 mg/kg bw/d

Brain

99%

100%

97%*

 

 

 

Cauda epididymis

99%

102%

88%**

 

 

 

Epididymides

100%

101%

92%**

 

 

 

Prostate

92%

99%

86%**

 

 

 

Spleen

 

 

 

105%*

107%

97%

 

*: p≤0.05; **: p≤0.01

 

All other mean absolute weight parameters did not show significant differences compared to the control groups.

The decrease of absolute weights of cauda epididymis, epididymides, and prostate in male top-dose animals (1000 mg/kg bw/d) were considered as treatment-related effects.

The decrease of brain weights in top-dose males (1000 mg/kg bw/d) as well as the increase of spleen weights in low-dose females (100 mg/kg bw/d) was considered as incidental and not treatment-related due to a missing dose-response relationship.

Absolute organ weights (F1 generation)

Compared to the controls (= 100%), the following values (in %)were significantly changed (printed in bold):

 

Male animals

Female animals

Group

11

100 mg/kg bw/d

12

300 mg/kg bw/d

13

1000 mg/kg bw day

11

100 mg/kg bw/d

12

300 mg/kg bw/d

13

1000 mg/kg bw/d

Cauda epididymis

96%

99%

88%**

 

 

 

Epididymides

100%

101%

91%**

 

 

 

Kidneys

99%

106%*

111%**

103%

106%**

115%**

Spleen

99%

103%

92%*

 

 

 

Thyroid glands

106%

99%

109%*

110%

118%**

111%*

 

*: p≤0.05; **: p≤0.01

All other mean absolute weight parameters did not show significant differences compared to the control groups.

The decrease of absolute weights of cauda epididymis and epididymides in male top-dose animals (1000 mg/kg bw/d) were considered to be treatment-related.

The increase of absolute kidney weights of male and female animals in mid- (300 mg/kg bw/d) and top-dose (1000 mg/kg bw/d) groups, respectively, was statistically significant. Because no histomorphological correlate was detected, a treatment-related weight increase was less likely.

The decrease of spleen weights in top-dose males as well as the increase of thyroid glands in top-dose males and mid- and top-dose females, respectively, is considered incidental and not treatment-related due to a missing dose-response relationship.

Conclusions:
Under the conditions of the present two-generation reproduction toxicity study, the NOAEL for fertility, reproductive performance and systemic toxicity in parental F0 and F1 Wistar rats is 300 mg/kg bw/d. The NOAEL for pre-and postnatal developmental toxicity in their offspring is 1000 mg/kg bw/d.
Executive summary:

The present two-generation reproduction toxicity study was performed accoding to OECD 416 and in compliance with GLP.

Two generations of Wistar rats were exposed to ethanolamine hydrochloride via diet at dose levels of 0, 100, 300, 1000 mg/kg bw/day (nominal in diet).

In the high-dose F0 and F1 generation females (1000 mg/kg bw/d), decreased numbers of implants and increased resorption rates resulted in significantly smaller litters, giving evidence for an adverse effect of the test compound on fertility and/or reproductive performance at high doses. It has to be noted that a dose of 1000 mg/kg bw/d also caused beginning systemic toxicity in these females, as was indicated by reduced food consumption and/or body weight gain during gestation/lactation. All data recorded during gestation and lactation in terms of embryo-/fetal and pup development gave no indications for any developmental toxicity in the F1 and F2 offspring up to a dose level of 1000 mg/kg bw/d. The test substance did not adversely influence pup viability, body weight, sex ratio and sexual maturation.

 

Thus, under the conditions of the present two-generation reproduction toxicity study, the NOAEL for fertility, reproductive performance and systemic toxicity in parental F0 and F1 Wistar rats is 300 mg/kg bw/d.

 

The NOAEL for pre-and postnatal developmental toxicity in their offspring is 1000 mg/kg bw/d.

Effect on fertility: via oral route
Endpoint conclusion:
no adverse effect observed
Dose descriptor:
NOAEL
50 mg/kg bw/day
Study duration:
subacute
Species:
rat
Quality of whole database:
High quality (GLP guideline study)
Effect on fertility: via inhalation route
Endpoint conclusion:
no study available
Effect on fertility: via dermal route
Endpoint conclusion:
no study available
Additional information

A weight-of-evidence approach with structurally related substances is applied to address the endpoint ‘toxicity to reproduction’ of the registered substance Methylmonoethanolamine (MMEA). The substances included in this approach are shown in table 1.

Table1: Substances included in the weight-of-evidence approach

 

Registered substance

Analogue 1

Analogue 2

Analogue 3

Analogue 4

Chemical name

Methylmonoethanolamine

MMEA

2-Aminoethanol

MEA

Diethanolamine

DEA

Methyldiethanolamine

MDEA

Dimethylaminoethanol

DMAE

CAS no

109-83-1

141-43-5

111-42-2

105-59-9

108-01-0

EC no

203-710-0

205-483-3

203-868-0

203-312-7

203-542-8

Molecular formula

C3H9NO

C2H7NO

C4H11NO2

C5H13NO2

C4H11NO

Chemical class

Category:

“aliphatic amines”

Subcategory:

“secondary amine”

Category:

“aliphatic amines”

Subcategory:

“primary amine”

Category:

“aliphatic amines”

Subcategory:

“secondary amine”

Category:

“aliphatic amines”

Subcategory: “tertiary amine”

Category:

“aliphatic amines”

Subcategory:

“tertiary amine”

Molecular weight [g/mole]

75

61

105

119

89

Smiles code

CNCCO

NCCO

OCCNCCO

CN(CCO)CCO

CN(C)CCO

Toxicity to reproduction

OECD 422, GLP, rat (2010)

NOAEL = 50 mg/kg bw/day (oral)

 

OECD 416, GLP, rat (2009)

NOAEL (parental animals) = 300 mg/kg bw/day (oral)

 

NOAEL (F1/F2 gen) 1000 mg/kg bw/day (oral)

OECD 443, GLP, rat (2018)

NOAEL (systemic toxicity) = 100 ppm (12.75 mg/kg bw/d) (oral)

 

NOAEL = 300 ppm (37.68 mg/kg bw/d (reproductive performance, developmental neurotoxicity and developmental immunotoxicity) (oral)

 

NOAEL = 100 ppm (developmental toxicity in the F1 progeny)

OECD 421, GLP, rat (2010)

NOAEL (systemic toxicity) 100 mg/kg bw/day (oral)

 

NOAEL (parental reproductive toxicity; F1 toxicity) 300 mg/kg bw/day (oral)

OECD 422, GLP, rat (2019)

NOAEL (fertility) = 5000 ppm (257 mg/kg bw/d) (oral)

 

NOEL (offspring) = 1000 ppm (oral)

 

The results of repeated-dose and reproductive toxicity studies differ between the substances. In a study according to OECD Guideline 422 available for the registered substance MMEA (2010) the NOAEL for general toxicity in males was < 50 mg/kg bw/d and 50 mg/kg bw/d for females. Basis for the general toxicity NOAEL were reduction of body weight in females, tubular degeneration in kidneys and testes and cysts and vacuolization of sex cord stroma in ovaries. Fertility was severely impaired by test-substance administration at dose levels of 150 and 450 mg/kg bw/d. Although mating (male and female mating indices) was not influenced, no lifeborn pups were delivered for both test groups. 6 females of test group 2 (150 mg/kg bw/d), and 9 females of test group 3 (450 mg/kgbw/d) did not become pregnant. In test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) a significant reduction with only 5 and 1 implantation sites was found. In test group 3 (450 mg/kg bw/d), the infertility was linked to the reduced number of sperms (oligospermia) caused by tubular degeneration in testes. In addition, the occurrence of ovarian cysts and vacuolization of the sex cord stroma in females may have influenced the fertility. In test group 2 (150 mg/kg bw/d), the severity of the findings in testes or ovaries was only minimal or slight and the findings did not occur in all infertile animals. Nevertheless, these lesions may have affected fertility.

A study according to OECD Guideline 422 is also available for DMAE (2019). Under the conditions of this study DMAE caused signs of systemic toxicity in females consisting of decreased food consumption and body weight, clinically apparent signs (mortality, tremors and convulsions) together with axonal degeneration in cerebellum, medulla oblongata, cervical and thoracic spinal cord at a concentration of 5000 ppm in drinking water. Males of the same concentration group showed no adverse findings. Thus, the no observed adverse effect level (NOAEL) for general systemic toxicity was 5000 ppm (approx. 257 mg/kg bw/d) for males and 1000 ppm (approx. 89 mg/kg bw/d) for female Wistar rats. Regarding fertility and reproductive performance, no adverse signs of toxicity were observed in male or female parental animals of test groups 1-3 (200, 1000 and 5000 ppm) during the entire study. Almost all F0 parental animals proved to be fertile. Mating behaviour, conception, implantation and gestation were not influenced. Thus, the NOAEL for fertility and reproductive performance was 5000 ppm (approx. 257 and 355 mg/kg bw/d, respectively) for male and female rats, the highest concentration tested. The NOAEL for developmental toxicity in the offspring was 1000 ppm based on adverse effects on pup number, status at delivery, viability/mortality and pup body weight at 5000 ppm.

The study results of the OECD 422 for MMEA and DMAE show that the two substances elicit a deviant toxicity with regard to general toxicity, as well as reproductive toxicity. While for MMEA tubular degeneration in kidneys and testes and cysts and vacuolization of sex cord stroma in ovaries were the main effects leading to a NOAEL of < 50 mg/kg bw/d, in DMAE axonal degeneration in cerebellum, medulla oblongata, cervical and thoracic spinal in females was the basis for the NOAEL of 89 mg/kg bw/d. Thus, the affected target organs are different between MMEA and DMAE. In addition, while MMEA strongly reduces the fertility in males and females at doses of 150 and 450 mg/kg bw/d, fertility was not impaired by the administration of DMAE at doses of up to 257 and 355 mg/kg bw/d, for male and female rats, respectively. DMAE, however, showed some developmental effects in the highest dose groups. This could not be examined in the study with MMEA based on the absence of viable offspring in the mid- and high-dose groups.

For MEA a two-generation reproductive toxicity study according to OECD Guideline 416 is available (2019). The NOAEL for general toxicity was 300 mg/kg bw/d based on minor effects of body weight and food consumption and a decrease of absolute and relative organ weights of cauda epididymidis and epididymides in the top-dose F0 and F1 males. Furthermore, prostate weight and the number of homogenization resistant caudal epididymal sperm was slightly, but significantly decreased in the F0 males. These findings were considered to be treatment-related effects, whereas histomorphological correlates were missing. No test substance-related findings from clinical examinations or gross and histopathology were observed, which indicate that the administration of the test compound via the diet adversely affected the fertility or reproductive performance of the F0 or F1 parental animals up to and including a nominal dose of 300 mg/kg bw/d. Estrous cycle data, mating behavior, conception, gestation, parturition, lactation and weaning as well as sperm parameters, sexual organ weights and gross and histopathological findings of these organs (including differential ovarian follicle counts in the F1 females) were comparable between the rats of all test groups. At the high-dose level (1000 mg/kg bw/d), absolute and relative weights of epididymides and cauda epididymidis were decreased and, in the F0 generation only, the number of homogenization resistant caudal epididymal sperm was slightly, but significantly reduced. However, histomorphological correlates for these findings were missing. In the high-dose F0 and F1 generation females (1000 mg/kg bw/d), decreased numbers of implants and increased resorption rates resulted in significantly smaller litters, giving evidence for an adverse effect of the test compound on fertility and/or reproductive performance at high doses. It has to be noted that a dose of 1000 mg/kg bw/d also caused beginning systemic toxicity in these females, as was indicated by reduced food consumption and/or body weight gain during gestation/lactation. All data recorded during gestation and lactation in terms of embryo-/fetal and pup development gave no indications for any developmental toxicity in the F1 and F2 offspring up to a dose level of 1000 mg/kg bw/d. The test substance did not adversely influence pup viability, body weight, sex ratio and sexual maturation. Thus, under the conditions this two-generation reproduction toxicity study, the NOAEL (no observed adverse effect level) for fertility, reproductive performance and systemic toxicity in parental F0 and F1 Wistar rats is 300 mg/kg bw/d. The NOAEL for pre-and postnatal developmental toxicity in their offspring is 1000 mg/kg bw/d.

In contrast to MMEA, no histopathological effects on the kidney, testes and ovaries were observed in this study. Also, clinical signs indicating neurotoxicity, as found for DMAE were not observed in this study. In addition, the fertility of the males and females was not impaired up to a concentration of 1000 mg/kg bw/d, while for MMEA infertility was identified in the 150 and 450 mg/kg bw/d dose-groups.

In the EOGRTS available for DEA (OECD 443, 2018). Males and females of test group 12 and 13 (300 ppm (37.68 mg/kg bw/d) and 1000 ppm (128.35 mg/kg bw/d)) revealed tubular degeneration and regeneration. However, although some effects on reproductive organs were observed, DEA did not adversely affect fertility and reproduction. Observed effects were an increased incidence of luteal cysts in the ovary and immature testicular tubules, which corresponded to the macroscopic finding “size reduced” in the females and males of the highest dose group. This size decrease testicular tubules were most likely due to the body weight reduction and was assessed to be treatment-related but a secondary effect. But as in cohort 1A animals (and in cohort 1B animals) degeneration of the testicular epithelium was observed in addition to the delay in maturation as well as in animals without a delay in maturation in these organs, the degeneration of tubular epithelium in the testis was assumed to be an adverse but secondary effect. Under the conditions of this study the NOAEL (no observed adverse effect level) for general toxicity is 100 ppm (12.75 mg/kg bw/d) for the F0 parental animals, based on evidence for distinct kidney toxicity and stomach irritation, as well as corresponding effects on water consumption, food consumption, body weights and clinical pathological parameters, which were observed at the LOAEL (Lowest Observed Adverse Effect Level) of 300 ppm (37.68 mg/kg bw/d). Similar toxicity was noted in the adolescent F1 animals, which had no stomach irritation but liver toxicity in addition. The NOAEL for fertility and reproductive performance for the F0 and F1 rats is 300 ppm (37.68 mg/kg bw/d), based on a lower number of implants, prolonged/irregular estrous cycles as well as pathological changes in sexual organs, pituitary and mammary glands of both genders at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm (128.35 mg/kg bw/d). However, eosinophilic cysts in the pituitary gland were present in the F1 animals of cohort 1A down to the 100 ppm dose level, but no assessment on adversity of this finding is possible at present. Therefore, no NOEL can be established for this particular effect. The NOAEL for developmental toxicity in the F1 progeny is 100 ppm (12.75 mg/kg bw/d), based on impaired pup survival at 1000 ppm (128.35 mg/kg bw/d) as well as reduced pup body weights in the F1 offspring, which were observed at the LOAEL (Lowest Observed Adverse Effect Level) of 300 ppm (37.68 mg/kg bw/d). As these weight reductions were only observed in the presence of maternal toxicity, including lower weight gain during pregnancy, they are not regarded as independent effect of the treatment. The NOAEL for developmental neurotoxicity for the F1 progeny is 300 ppm (37.68 mg/kg bw/d), based on adverse clinical observations, impaired auditory startle response and corresponding neuropathological findings at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm. In addition, increased T4 values were noted in adult and adolescent males at 1000 ppm (128.35 mg/kg bw/d) as well as newborn and weanling females at 100 and 300 ppm (12.75 mg/kg bw/d and 37.68 mg/kg bw/d). The NOAEL for developmental immunotoxicity for the F1 progeny is 300 ppm (37.68 mg/kg bw/d), based on effects on the T-helper cells and cytotoxic T-cells in the spleen in the F1 females at the LOAEL (Lowest Observed Adverse Effect Level) of 1000 ppm (128.35 mg/kg bw/d). Lower mean and median anti-SRBC IgM antibody titres of the positive control group (4.5 mg/kg bw/d cyclophosphamide, oral) demonstrated that the test system worked properly.

For MDEA a study according to OECD Guideline 421 is available (2010). In this test, the fertility was not reduced. Fertility indices for male and female animals lay in the normal range of biological variation. The NOAEL for general toxicity is 100 mg/kg bw/d based on decreased terminal body weights in males and females (5 % below controls in both sexes) and the NOAEL for reproductive performance and fertility is 300 mg/kg bw/d based on the following effects observed at the highest dose of 1000 mg/kg bw/d: Total litter loss in 4 females, undelivered pups palpable in 2 females, insufficient lactation behaviour in 2 females (pups had no or less milk in stomach), increased duration of gestation (22.8 days vs. 21.9 days in control), decreased number of implantation sites (6.7 vs. 12.9 in control), increased post-implantation loss (31.0 % vs. 6.0 % in control) and mean post-implantation loss, decreased number of delivered pups (4.6 vs. 12.1 in control).

Conclusion

As a result of this evaluation, it becomes evident that the substance which is the closest analogue based on similar target organs in repeated dose tests and effect levels within the same range, is DEA. The classification of DEA regarding reproductive toxicity (cat. 2; H361fd) was adopted accordingly for MMEA. 

 

Effects on developmental toxicity

Description of key information

Nelson et al., 1984. Comparative Inhalation Teratogenicity of Four Glycol Ether Solvents and an Amino derivative in Rats. Environmental Health perspectives, Vol., 57, pp.261-271, 1984.

Weight-of-evidence approach (please refer to Section 'Additional information')

Link to relevant study records

Referenceopen allclose all

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
key study
Study period:
no information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
study well documented, meets generally accepted scientific principles, acceptable for assessment
Qualifier:
no guideline followed
Principles of method if other than guideline:
The chemical N-Methylethanolamine was vaporized and administered to approximately 15 pregnant rats in one to three concentrations for 7 hr/day on gestation days 7 to 15, and dams were sacrificed on day 20. Fetuses were individually weighed, and two-thirds of them were fixed in Bouin's solution and examined for soft-tissue anomalies. The other one-third were fixed in alcohol, stained with Alizarin Red and examined for skeletal defects.
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Sprague-Dawley
Remarks:
Virgin female and male Sprague-Dawley rats specified to be free of mycoplasma and Sendai virus and of internal and external parasites
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Breeding Laboratories, Wilmington, MA
- Weight at study initiation: males: > 300 g, females: 200 to 300 g
- Housing: Females were placed alone in 38 x 33 x 17-cm polycarbonate cages with filter tops. Bedding consisted of cleaned, heat-treated sawdust from a local supplier (Absorb-Dri, Tasty Foods, Cincinnati, OH)
- Diet (e.g. ad libitum): Purina Lab Chow was available ad libitum except when pregnant animals were in exposure chambers
- Water (e.g. ad libitum): Tap water was available ad libitum except when pregnant animals were in exposure chambers
- Acclimation period: Acclimatisation to a 12-hr light-dark cycle (lights on at 6 am) and to a temperature of 24 + 2°C for 2 weeks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 24 + 2°C
- Humidity (%): not controlled, typically was in the range of 40 + 20%
- Air changes: Air flow through the chambers provided approximately four air changes per minute.
- Photoperiod (hrs dark / hrs light): 2-hr light-dark cycle (lights on at 6 am)
Route of administration:
inhalation: vapour
Type of inhalation exposure (if applicable):
whole body
Vehicle:
unchanged (no vehicle)
Details on exposure:
GENERATION OF TEST ATMOSPHERE / CHAMBER DESCRIPTION
- Exposures were conducted sequentially in one or two chambers, with a third chamber for sham exposure of control subjects.
- Pregnant females were transported from the animal quarters to the inhalation chambers in their home cages with filter tops (Hazleton Systems, Aberdeen, MD). They were placed individually in 13 x 25 x 189-cm stainless steel wire mesh cages within exposure chambers.
- Control animals were placed in similar chambers for the same hours as the exposed animals; a pooled group of controls (N = 34) served as the comparison group for the first three chemicals examined. Another group of 15 controls served as the comparison group for the last two chemicals examined, as these groups were exposed at a later time (approximately 6 months later) than the first three.
- Air flow through the chambers provided approximately four air changes per minute.
- Exposures, as outlined above, were conducted 7 hr/day, and the animals were left in the chamber for at least one additional hour blow-off time after vapor generation terminated.
- Vaporization was achieved by bubbling air through the test item which was heated to approximately 40°C (for the higher concentrations). This vapor was then mixed with filtered room air to obtain the desired concentration and introduced into 0.5 m3 exposure chambers (Charles Spengler and Associates, Cincinnati, OH).

TEST ATMOSPHERE
- The concentration was continually monitored by a Miran I infrared spectrophotometer (Foxboro Analytical, South Norwalk, CT) with the concentration recorded hourly.
- Typically, two or three charcoal tube samples (silica gel tubes for 2- methylaminoethanol) were taken weekly from the chamber air for independent verification of the concentration by gas chromatograph.
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The concentrations within the exposure chambers as measured by the infrared analyser were relatively close to those obtained from gas chromatography (please refer to corresponding table).
Details on mating procedure:
Males weighing over 300 g were placed individually into a cage with three females weighing 200 to 300 g. Vaginal smears were taken each morning, and the presence of sperm marked day zero of gestation.
Duration of treatment / exposure:
7 hours (animals were left in the chamber for at least one additional hour blow-off time after vapour generation terminated)
Frequency of treatment:
Exposures, as outlined above, were conducted 7 hr/day, and the animals were left in the chamber for at least one additional hour blow-off time after vapour generation terminated. They were then returned in their individual housing cages to the animal quarters, where water bottles were replaced. Exposures were conducted on gestation days 7-15.
Duration of test:
Exposures were conducted on gestation days 7-15.
15 days of gestation.
On day 20 of gestation, dams were sacrificed.
Dose / conc.:
150 ppm
Remarks:
Doses / Concentrations: 150.0 +/- 15.2 ppm
Basis: other: vapour generated, by gas chromatography
No. of animals per sex per dose:
approximately 15 pregnant rats
Control animals:
yes
Details on study design:
Controls: three solvents were compared with a pooled group (N = 34) of sham-exposed controls, and the remaining two were compared with a group of 15 controls.
Maternal examinations:
BODY WEIGHT: Yes
- Time schedule for examinations: weekly (i.e., on days 7, 14, and 21)

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): Yes
- Time schedule for examinations: weekly (i.e., on days 7, 14, and 21)

WATER CONSUMPTION AND COMPOUND INTAKE (if drinking water study): Yes
- Time schedule for examinations: weekly (i.e., on days 7, 14, and 21)

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 20


OTHER: any other signs of maternal toxicity were noted daily.
Ovaries and uterine content:
The entire uterus was removed and numbers of resorption sites (classified as early, middle or late) and live fetuses were determined.
Blood sampling:
Not examined
Fetal examinations:
Fetuses were serially removed, blotted of excess fluids, weighed, examined for external malformations and external sex determined.
One third of the fetuses were randomly selected and placed in 95% ethanol, and the remaining fetuses were placed in Bouin's solution. After being in the Bouin's solution for at least 1 week, these fetuses were examined for visceral abnormalities using Wilson's razor blade sectioning technique. The viscera were examined with the aid of a dissecting microscope. A representative sample of sections with malformations was identified by dam number and saved in 70% alcohol.

Fetuses were examined for skeletal defects by using a modified Staples technique. They were fixed in 95% alcohol, eviscerated and macerated in 2% KOH/Alizarin Red S solution. The fetuses were further macerated and cleared in the appropriate solutions of 2% KOH/glycerin (60:40, 40:60, 20:80) and stored in 100% glycerin. A crystal of thymol was added to each storage vial to retard fungal growth. Storage vials were individually identified by dam number.
Statistics:
Numbers of implants and proportions of resorptions were independently analysed by using a Kruskal-Wallis test corrected for ties, with subsequent multiple comparisons to determine where the differences occurred. Analysis of pup weights involved a mixed model analysis of covariance (with the number of live pups in the litter as the covariate) using maximum likelihood estimation. The model was mixed, since there was both within-litter and between-litter variation. Subsequently, pairwise comparisons between the pooled control group and each treatment group were performed. Incidence of total defects and of total variants were compared using a Kruskal-Wallis test with multiple comparisons with the litter as the experimental unit and the level of significance at p< 0.05.
Indices:
no information given
Historical control data:
no information given
Clinical signs:
not specified
Dermal irritation (if dermal study):
not examined
Mortality:
not specified
Body weight and weight changes:
not specified
Food consumption and compound intake (if feeding study):
not specified
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not specified
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
not examined
Gross pathological findings:
not examined
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
no effects observed
Description (incidence and severity):
see attachment
Details on results:
At 150 ppm N-Methylethanolamine (mean concentration from 28 silica gel tubes, one per day, analyzed in duplicate = 150.0 ppm), no maternal toxicity was observed.
Number of abortions:
not examined
Pre- and post-implantation loss:
not examined
Total litter losses by resorption:
not specified
Early or late resorptions:
no effects observed
Description (incidence and severity):
Litters with resorptions (%): 10(56)
Resorptions/litter: 0.7
Dead fetuses:
no effects observed
Description (incidence and severity):
Total live fetuses: 212
Live fetuses/litter: 11.8
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
not examined
Other effects:
no effects observed
Description (incidence and severity):
Number pregnant/number mated: 18/24
Implants/dam: 11.9
Number (%) of litters with abnormal fetuses (Litter with at least one fetus having either skeletal or visceral anomalies): 7(39)
Details on maternal toxic effects:
At 150 ppm N-Methylethanolamine (mean concentration from 28 silica gel tubes, one per day, analyzed in duplicate = 150.0 ppm), no maternal toxicity was observed.
Dose descriptor:
NOAEC
Effect level:
>= 150 ppm (nominal)
Based on:
test mat.
Basis for effect level:
other: maternal toxicity
Remarks on result:
other: no adverse effects observed
Fetal body weight changes:
no effects observed
Description (incidence and severity):
(see attachment)
Reduction in number of live offspring:
not specified
Changes in sex ratio:
not specified
Changes in litter size and weights:
not specified
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Skeletal malformations:
no effects observed
Description (incidence and severity):
(see attachment)
Visceral malformations:
no effects observed
Description (incidence and severity):
(see attachment)
Other effects:
no effects observed
Description (incidence and severity):
(see attachment)
Details on embryotoxic / teratogenic effects:
At 150 ppm N-Methylethanolamine (mean concentration from 28 silica gel tubes, one per day, analyzed in duplicate = 150.0 ppm), no fetal toxicity was observed.
Dose descriptor:
NOAEC
Effect level:
>= 150 ppm (nominal)
Based on:
test mat.
Basis for effect level:
other: fetotoxicity
Abnormalities:
not specified
Developmental effects observed:
not specified

Low vapor pressure also prevented our generating high concentrations of 2-MAE. At 150 ppm 2-MAE (mean concentration from 28 silica gel tubes, one per day, analyzed in duplicate = 150.0 ppm), no maternal or fetal toxicity was observed (see attachment).

Finally, the lack of teratogenic response of 2-methylaminoethanol was interesting and from a mechanistic or theoretical point of view, would merit follow up using a different route of exposure. At first glance, one might expect that its biotransformation would be similar to that of 2-ME. However, our results of no maternal or fetal toxicity at 150 ppm 2-MAE suggest that this may not be the case; since the amine is likely more lipidsoluble and less water-soluble than the methoxy portion, the absorption and excretion of the 2-MAE is likely quite different from that of 2-ME. Thus it would be of interest to see if a higher dose of 2-MAE would be teratogenic, though a route other than inhalation would be required, since the vapor concentration we used was near the saturation point.' This lack of teratogenicity at three times the concentration of a teratogenic level of its structurally similar glycol ether, points to a relatively strict structural requirement to produce teratogenic effects.

We observed that embryotoxicity decreases as alkyl chain length increases, similar to observations with testicular atrophy.

Conclusions:
In this study N-Methylethanolamine showed neither maternal nor fetal toxicity effects.
Executive summary:

MMEA was vaporized and administered (near the saturation point) to 18 pregnant rats in one to three concentrations for 7 h/day on gestation days 7 to 15. The dams were sacrificed on day 20 of gestation. Fetuses were individually weighed, and two-thirds of them were fixed in Bouin's solution and examined for soft-tissue anomalies. The other one-third were fixed in alcohol, stained with Alizarin Red and examined for skeletal defects. As overall result for the substance MMEA, neither maternal nor fetal toxicity effects were found. No teratogenicity was observed. The NOAECs were 460 mg/m³, the highest dose tested for both, dams and fetuses.

Endpoint:
developmental toxicity
Remarks:
Dose-range finder
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
other: Range-finding study, acceptable, well-documented study report which meets basic scientific principles
Reason / purpose for cross-reference:
reference to same study
Remarks:
Main study
Qualifier:
no guideline followed
Principles of method if other than guideline:
Range-finding study
GLP compliance:
yes
Limit test:
no
Species:
rat
Strain:
Wistar
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Dr. K Thomae GmbH, Biberach Germany
- Age at study initiation: 68-70 days
- Weight at study initiation: ca. 214 g
- Housing: individual in wire mesh cages
- Diet: Kliba rat/mouse laboratory diet, Klingenthalmühle AG Kaiseraugst, Switzerland ad libitum
- Water: Tap water ad libitum
- Acclimation period: 5 days
- Adaption period: 5 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
inhalation
Type of inhalation exposure (if applicable):
nose/head only
Vehicle:
unchanged (no vehicle)
Details on exposure:
GENERATION OF TEST ATMOSPHERE / CHAMBER DESCRIPTION
The test substance was supplied to a two-component atomizer at a constant rate by means of a metric pump.
An aerosol was generated by means of compressed air and passed into the inhalation system.
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Monitoring of the inhalation atmosphere: Vacuum compressed air pump (Millipore) Sampling equipment with probe (Millipore), 7 mm diameter, filter MN 85/90 Bf, 1.25 m/s sampling velocity, hourly sampling frequency Gravimetrical determination of concentration by means of balance (Mettler AE 249) - Particle size analysis: EACD 50% MMAD Geometric standard deviation (GSD) Total dust Respirable dust
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 1/1-4
- Length of cohabitation: 4 pm - 7.30 am
- Verification of same strain and source of both sexes: yes
- Proof of pregnancy:sperm in vaginal smear referred to as day 0 of pregnancy
Duration of treatment / exposure:
gestation days 6 - 15
Frequency of treatment:
6h/day
Dose / conc.:
0 mg/L air
Remarks:
Control
Dose / conc.:
0.1 mg/L air (nominal)
Remarks:
see corresponding analytical concentration
Dose / conc.:
0.2 mg/L air (nominal)
Remarks:
see corresponding analytical concentration
Dose / conc.:
0.4 mg/L air (nominal)
Remarks:
see corresponding analytical concentration
Dose / conc.:
0.11 mg/L air (analytical)
Dose / conc.:
0.21 mg/L air (analytical)
Dose / conc.:
0.4 mg/L air (analytical)
No. of animals per sex per dose:
10 females
Control animals:
yes, concurrent vehicle
Details on study design:
Duration of test: 20 days
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes

BODY WEIGHT: Yes
body weight/body weight gain and corrected body weight

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day #
- Organs examined: uterus weight, weight of placenta

OTHER: necropsy finding of the dams, clinical pathology and hematology in the dams
Ovaries and uterine content:
Reproduction data of the dam, uterus weight, weight of fetus, weight of placenta
Blood sampling:
Yes
Fetal examinations:
Weight of fetus
Statistics:
no information given
Indices:
no information given
Historical control data:
no information given
Clinical signs:
no effects observed
Dermal irritation (if dermal study):
not examined
Mortality:
no mortality observed
Body weight and weight changes:
no effects observed
Food consumption and compound intake (if feeding study):
not examined
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
Test group 0.4 mg/l: increase in sodium and creatinine serum concentrations decrease in cholesterol and triglycerides serum concentrations
Test group 0.2 mg/l: increase in sodium and creatinine serum concentrations decrease in cholesterol and triglycerides serum concentrations
Test group 0.1 mg/l: No treatment related effects
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
Test group 0.4 mg/l: absolute and relative liver weight increased
Test group 0.2 mg/l: relative liver weight increased
Test group 0.1 mg/l: No treatment related effects
Gross pathological findings:
no effects observed
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
effects observed, treatment-related
Description (incidence and severity):
Test group 0.4 mg/L: significantly increased mean placental weights
Test group 0.2 mg/L: significantly increased mean placental weights
No substance related effects were observed. The marginal,however statistically significant  decrease in the mean placental weights in test groups 2 and 3 is without any biological relevance.
Number of abortions:
not examined
Pre- and post-implantation loss:
not examined
Total litter losses by resorption:
not examined
Early or late resorptions:
not examined
Dead fetuses:
not examined
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
not examined
Other effects:
no effects observed
Description (incidence and severity):
No effects observed on reproduction data of the dam or uterus weight

Details on maternal toxic effects:
Maternal toxic effects: yes
Dose descriptor:
NOAEC
Remarks:
general toxicity
Effect level:
0.2 mg/L air
Based on:
test mat.
Basis for effect level:
clinical biochemistry
organ weights and organ / body weight ratios
Dose descriptor:
NOAEC
Remarks:
developmental toxicity
Based on:
test mat.
Basis for effect level:
other: developmental toxicity parameter
Remarks on result:
not determinable
Remarks:
no NOAEC identified
Abnormalities:
not examined
Fetal body weight changes:
no effects observed
Reduction in number of live offspring:
not examined
Changes in sex ratio:
not examined
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not specified
External malformations:
not examined
Skeletal malformations:
not examined
Visceral malformations:
not examined
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: no effects

Remarks on result:
not determinable due to absence of adverse toxic effects
Abnormalities:
not specified
Developmental effects observed:
not specified

Detailed results: Concentrations:
Target
       Measured        MMAD (GSD)
0.1 mg/l,
      0.11 mg/l        0.6 µm (4.5)
0.2 mg/l
       0.21 mg/l        0.1.2 µm (3.6)
0.4 mg/l
       0.40 mg/l        0.0.6 µm (4.5)

Conclusions:
At 0.4 mg/L a marked indication of hepatotoxic effects were observed (increase of absolute and relative liver weights, decrease in cholesterol and triglyceride serum concentration, increase in transaminase AST). At 0.2 mg/L relative liver weight was still increased and cholesterol and triglycerides decreased. These findings were considered as clear indications for maternal toxicity.
Executive summary:

In the present dose-range finding study pregnant female Wistar rats were exposed to an aerosol of Diethanolamine in a head/nose exposure systems for 6 h/day on day 6 through day 15 post coitum at concentrations of 0; 0.1; 0.2; 0.4 mg/L. At 0.4 mg/L a marked indication of hepatotoxic effects were observed (increase of absolute and relative liver weights, decrease in cholesterol and triglyceride serum concentration, increase in transaminase AST). At 0.2 mg/L relative liver weight was still increased and cholesterol and triglycerides decreased.

These findings were considered as clear indications for maternal toxicity.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
GLP compliance:
yes
Limit test:
no
Species:
rat
Strain:
Wistar
Details on test animals or test system and environmental conditions:
- Source: Dr. K Thomae GmbH, Biberach Germany
- Age at study initiation: 68-70 days
- Weight at study initiation: ca. 214 g
- Housing: individual in wire mesh cages
- Diet: Kliba rat/mouse laboratory diet, Klingenthalmuehle AG Kaiseraugst, Switzerland ad libitum
- Water: Tap water ad libitum
- Acclimation period: 5 days
- Adaption period: 5 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
inhalation
Type of inhalation exposure (if applicable):
nose/head only
Vehicle:
unchanged (no vehicle)
Details on exposure:
Test group 1
The substance to be tested was supplied to a two component atomizer by means of a continuous infusion pump and was atomized with compressed air. Having passed a glass separator, the liquid aerosol was diluted with blast air (about 50% rel. humidity; 22°C) and part of the air stream was supplied to the exposure apparatus (INA 60). The remainder of the aerosol generated was supplied to the exhaust air equipment (exhaust air 2).

Test group 2
The substance to be tested was supplied to a two component atomizer by means of a continuous infusion pump and was atomized with compressed air. Having passed a glass separator, the liquid aerosol was diluted with blast air (about 50% rel. humidity; 22°C) and the air stream was supplied to the exposure apparatus (INA 60).

Test group 3
The substance to be tested was supplied to a two component atomizer out of a storage vessel by means of a diaphragm metering pump and was atomized with compressed air . Having passed a glass separator, the liquid aerosol was diluted with blast air (about 50% rel. humidity; 22°C) and the air stream was supplied to the exposure apparatus (INA 60).

As the test substance is sensitive to oxygen, the open storage vessel of the diaphragm pump was kept covered with a nitrogen cushion (5 l/h).

TEST ATMOSPHERE
- Brief description of analytical method used: The concentration of the atmospheres in test groups 1 and 2 was monitored by means of light scattering photometers. The light scattering photometers were calibrated with mixtures of the test substance and air that were generated in the exposure apparatus not containing animals according to the test group concentrations.
The concentration in test group 3 was analyzed by a corrected gravimetrical determination which was calibrated with an indirect photometric method.
- Samples taken from breathing zone: yes
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
- Monitoring of the inhalation atmosphere:
Vacuum compressed air pump (Millipore) Sampling equipment with probe (Millipore), 7 mm diameter, filter MN 85/90  Bf, 1.25 m/s sampling velocity, hourly sampling frequency Gravimetrical determination of concentration by means of balance (Mettler  AE 249) - Particle size analysis: EACD 50% MMAD Geometric standard deviation (GSD) Total dust Respirable dust
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 1/1-4
- Length of cohabitation: 4 pm - 7.30 am
- Verification of same strain and source of both sexes: yes
- Proof of pregnancy:sperm in vaginal smear referred to as day 0 of pregnancy
Duration of treatment / exposure:
gestation day 6 - 15
Frequency of treatment:
6h/day
Duration of test:
gestation day 6-20 (gd 16-20: post exposure observation period)
Dose / conc.:
0.01 mg/L air (nominal)
Dose / conc.:
0.05 mg/L air (nominal)
Dose / conc.:
0.2 mg/L air (nominal)
Dose / conc.:
10 mg/m³ air (analytical)
Dose / conc.:
50.2 mg/m³ air (analytical)
Dose / conc.:
202 mg/m³ air (analytical)
No. of animals per sex per dose:
25 females
Control animals:
yes, concurrent vehicle
Details on study design:
no further information given
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: once daily during the preflow and the observation period, 3 times per day during the exposure period

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: once daily during the preflow and the observation period, 3 times per day during the exposure period

BODY WEIGHT: Yes
- Time schedule for examinations: 0, 3, 6, 9, 12, 15, 18, 20 p.c.

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 20 (gross pathology)
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
- Other:

- Weight of uterus before it was opened
- Number of corpora lutea
- Number and distribution of implantation sites classified as :
• live fetuses
• dead implantations:
a) early resorptions (only decidual or placental tissues visible or according to SALEWSKI from uteri from apparently non-pregnant animals and the empty uterus horn in the case of single-horn pregnancy)
b) late resorptions (embryonic or fetal tissue in addition to placental tissue visible)
c) dead fetuses (hypoxemic fetuses which did not breathe spontaneously after the uterus had been opened)

Furthermore calculations of conception rate and pre- and postimplantation losses were carried out :
- The conception rate (in %) was calculated according to the following formula:
(number of pregnant animals/ number of corpora lutea) x100

-The postimplantation loss (in %) was calculated* from the following formula:
(number of implantations - number of live fetuses)/ number of implantations x100

* Calculation on the basis of each individual pregnant animal with scheduled sacrifice
Fetal examinations:
- External examinations: Yes: all per litter
- Soft tissue examinations: Yes: half per litter
- Skeletal examinations: Yes: half per litter
- Head examinations: No data
Statistics:
- Dunnett's Test: body weight, body weight change, corrected body weight  gain (net maternal body weight change), weight of the uterus before it  was opened, weight of fetuses, weight of placentae, corpora lutea,  implantations, pre and postimplantation losses, resorptions and live  fetuses.
- Fisher's Exact Test: conception rate, mortality (of the dams) and all  fetal findings.
Indices:
no information given
Historical control data:
yes
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
During the preflow period the animals of all showed no clinical signs and findings but during the exposure period eight animals of the highest concentration group (0.2 mg/L) showed bloody discharge from the vagina on day 14 p.c. after exposure
Dermal irritation (if dermal study):
not examined
Mortality:
no mortality observed
Body weight and weight changes:
no effects observed
Description (incidence and severity):
No treatment related finding based on body weight or corrected body weight gain.
Food consumption and compound intake (if feeding study):
not examined
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
No treatment related finding based on uterus weight.
Gross pathological findings:
no effects observed
Description (incidence and severity):
No treatment related finding based on necropsy findings and uterus weight.
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Number of abortions:
not examined
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
No significant and dose-respronse related implantation loss was observed.
Total litter losses by resorption:
not specified
Early or late resorptions:
no effects observed
Dead fetuses:
no effects observed
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
no effects observed
Other effects:
no effects observed
Description (incidence and severity):
Placenta and gavid uterus weights, mean corproa lutea, mean implantation: Not affected in any group.
Details on maternal toxic effects:
No treatment related finding based on reproduction data of the dams.
Dose descriptor:
NOAEC
Effect level:
0.05 mg/L air
Based on:
test mat.
Basis for effect level:
other: maternal toxicity
Fetal body weight changes:
no effects observed
Description (incidence and severity):
Not affected in any group.
Reduction in number of live offspring:
not examined
Changes in sex ratio:
no effects observed
Description (incidence and severity):
Not affected in any group.
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
All differences between the control group and the substance-treated groups concerning fetal external observations were without biological relevance and/or appeared to about the same extent as in the historical control data.
Skeletal malformations:
effects observed, treatment-related
Description (incidence and severity):
The only substance-related finding was the increased occurrence of high dose fetuses with rudimentary cervical rib(s). Due to the high frequency of this finding in the 0.2 mg/l fetuses, the incidence of skeletal variations was statistically significantly increased. All other differences between the control group and the substance-treated groups concerning skeletal observations were without biological relevance and/or appeared to about the same extent as in the historical control data.
Visceral malformations:
no effects observed
Description (incidence and severity):
All other differences between the control group and the substance-treated groups concerning soft tissue observations were without biological relevance and/or appeared to about the same extent as in the historical control data.
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: yes
Dose descriptor:
NOAEC
Effect level:
>= 0.2 mg/L air
Based on:
test mat.
Sex:
male/female
Basis for effect level:
skeletal malformations
Remarks on result:
other: signs of prenatal developmental toxicity did only occur at a maternal toxic concentration
Abnormalities:
not specified
Developmental effects observed:
not specified

Detailed results:

Concentrations:                    

Target

Measured

MMAD      

% aerosol

0.01 mg/l        

10.0 mg/m³        

1.2 µm        

100%

0.05 mg/l        

50.2 mg/m³ 

0.4 µm        

98%

0.2 mg/l

202 mg/m³        

0.6 µm        

98%


Maternal, litter, fetal data - prenatal developmental toxicity  (inhalation) study in Wistar rats

mg/l

0

0.01

0.05

0.2

n (females)

25

25

25

25

Pregnant

21

21

23

23

Mortality

0

0

0

0

Body weight gain GD6-15 (g)

71

71

77

70

NWC from GD 6

38

43

39

41

Gravid uterus (g)

71

68

76

67

Corpora lutea (mean)                

14.5

15.3

15.2

14.6

Implantation  (mean)                

13.2

13.2

13.9

12.5

Post implantation loss

5.5

10.0

4.9

10.2

Resorption  (mean) 

0.8

1.4

0.7

1.3

Live fetuses (mean)                

12.5

11.8

13.2

11.2

Placental weight (g)

0.44

0.44

0.43

0.43

Fetus evaluated

262

248

303

258

Litter evaluated

21

21

23

23

Fetal weight (g) (males/females)        

4.0/3.8        

4.0/3.8

4.0/3.8        

4.1/3.9

Total external malformations (fetus/litter)

0/0        

1/1        

0/0        

0/0        

Total external variations (fetus/litter)

0/0        

0/0        

0/0        

0/0        

Total soft tissue malformations (fetus/litter)        

1/1        

2/2        

2/2        

0/0

Total soft tissue variations (fetus/litter)        

20/9

20/15        

25/16        

16/10

Total skeletal malformations (fetus/litter)        

5/5

5/4        

7/6        

5/5

Total skeletal variations (fetus/litter)        

59/19        

58/20        

69/22        

78/22*

GD = gestation day, NWC = net body weight change, 
* p<0.05 

**p<0.01

Conclusions:
At the highest concentration (0.2 mg/L) signs of maternal toxicity were observed was substantiated by adverse clinical symptoms (vaginal hemorrhages) in 8 of the 21 pregnant rats on day 14 p.c. At this dose level a markedly increased number of fetuses with skeletal variations (mainly cervical rib(s)) were also recorded but substance-related teratogenic effects were not detected in any fetus. Thus, signs of prenatal developmental toxicity did only occur at a maternal toxic concentration. There were no adverse effects on dams or fetuses at the low or mid concentrations (0.01 or 0.05 mg/L).
The NOAEC for maternal and prenatal developmental toxicity was 0.05 mg/l (50 mg/m³), the NOAEC for teratogenicity was ≥ 0.2 mg/l (200 mg/m³).
Executive summary:

This prenatal developmental toxicity study was conduced according to OECD TG 414 and in accordance with GLP. In the study pregnant female Wistar were exposed to an aerosol of Diethanolamine in a head/nose exposure systems for 6 h/day on day 6 through day 15 post coitum at concentrations of 0; 0.01; 0.05; 0.2 mg/L (0; 10, 50, 200 mg/m³).

At the highest concentration (0.2 mg/L) signs of maternal toxicity were observed and were substantiated by adverse clinical symptoms (vaginal hemorrhages) in 8 of the 21 pregnant rats on day 14 p.c. At this dose level a markedly increased number of fetuses with skeletal variations (mainly cervical rib(s)) were also recorded but substance-related teratogenic effects were not detected in any fetus. Thus, signs of prenatal developmental toxicity did only occur at a maternal toxic concentration. There were no adverse effects on dams or fetuses at the low or mid concentrations (0.01 or 0.05 mg/L).

The NOAEC for maternal and prenatal developmental toxicity was 0.05 mg/l (50 mg/m³), the NOAEC for teratogenicity was ≥ 0.2 mg/l (200 mg/m³).

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Reason / purpose for cross-reference:
reference to same study
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Principles of method if other than guideline:
Prenatal developmental toxicity study with dermal application in rats
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Crj: CD(SD)
Details on test animals or test system and environmental conditions:
- Source: Charles River Laboratories, Portage, MI
- Weight at study initiation: 209-251g
- Housing: individual
- Diet Ground certified rodent diet (RMH 3200, Agway. Inc. Waverly, NY) ad libitum
- Water ad libitum
- Acclimation period: 2 weeks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 16-21
- Humidity (%): 40-70
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
dermal
Vehicle:
unchanged (no vehicle)
Details on exposure:
- Area of exposure: clipped back skin
- % coverage: 100
- Type of wrap if used: steril gauze and further occluded with polyvenyl film attached to a specially designed Lycra-Spandex jacket  with Velcro closures
- Time intervals for shavings or clipplings: no data

REMOVAL OF TEST SUBSTANCE
- Washing (if done): warm water
- Time after start of exposure: 6 hours

TEST MATERIAL
- Amount(s) applied (volume or weight with unit): 4 mL
- Constant volume or concentration used: yes

USE OF RESTRAINERS FOR PREVENTING INGESTION: no
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
gas chromatography
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 1/1
- Length of cohabitation: over night
- Proof of pregnancy: vaginal plug referred to as day 0 of pregnancy
Duration of treatment / exposure:
gestation day 6 -15
Frequency of treatment:
6 h/day
Duration of test:
gestation day 0-21
Dose / conc.:
0 mg/kg bw/day
Dose / conc.:
150 mg/kg bw/day
Dose / conc.:
380 mg/kg bw/day
Remarks:
due to a preparation error the mid dose had to be reduced from 500 to 380 mg/kg bw/day
Dose / conc.:
1 500 mg/kg bw/day
No. of animals per sex per dose:
25 females
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: Dose selection was based on the results of developmental toxicity range finding studies using dermally applied DEA
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: daily

DETAILED CLINICAL OBSERVATIONS: Yes (skin)
- Time schedule: twice daily during the dosing period, once daily during the post-treatment period

BODY WEIGHT: Yes
- Time schedule for examinations: GD 0, 6, 9, 12, 15, 18

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): Yes
- Time schedule for examinations: 3-day intervals from GD 0-21

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 21
- Organs examined: liver, kidney

BLOOD:
- Prior to sacrifice
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
Blood sampling:
BLOOD:
- Prior to sacrifice
Fetal examinations:
- External examinations: Yes: all per litter (variation/malformation) / half per litter (thoracic, abdominal visceral abnormalities)
- Soft tissue examinations: No data
- Skeletal examinations: Yes: half per litter
- Head examinations: Yes: half per litter
Statistics:
Continuous variables were compared for homogeneity of variance using  Levene's test for equal variances (Levene, 1960). A parametric or nonparametric analysis of variance (ANOVA) was performed , f parametric ANOVA analyses were significant, pooled T-tests were used for pairwise comparisons. If results from a nonparametric ANOVA were significant, separate variance T tests for pairwise comparisons were performed. Data  from nongravid females and females delivering early were not included in  the statistical analyses. Nonparametric data were statistically evaluated  using the Kruskal-Wallis test, followed by the Mann Whitney U test. Incidence data were compared using the Fisher's exact test (Sokal and  Rohlf, 1969), with the exception of frequency data for fetal malformations and variations, statistical analyses were performed using BMDP Statistical Software (Dixon, 1990). For all statistical tests, the critical level of significance was set a priori at a = 0.05 (two-tailed).
Indices:
no information given
Historical control data:
no information given
Clinical signs:
not specified
Dermal irritation (if dermal study):
effects observed, treatment-related
Description (incidence and severity):
At 380 (500) and 1500 mg/kg bw signs of skin irritation, which were dose-dependent in incidence and severity. At the high dose, this condition persisted into the post-treatment period. No significant effects on skin irritation were observed at 150 mg/kg bw. No skin irritation was observed in control animals.
Mortality:
not specified
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
Dams at 1500 mg/kg bw showed significant declines in body weight gains at the end of the dosing period and during the post-dosing period (GD 15-21). This corresponded with a 4.5% decrease in body weight, when corrected for gravid uterine weight. Despite significant differences in body weight gain at discrete time points, there was no significant difference in body weight gains across dose groups over the entire duration of the study (GD 0-21). At 150 and 380 (500) mg/kg/day, there were no treatment-related declines in maternal weight gain at any time interval.
Food consumption and compound intake (if feeding study):
no effects observed
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
effects observed, treatment-related
Description (incidence and severity):
DEA slightly decreased red blood cell parameters, including hematocrit, MCV, MCH, hemoglobin concentration, and erythrocyte number at all concentrations. The highest dose level of DEA produced increased numbers of leukocytes and lymphocytes, but decreased platelet numbers. Changes in the profile of red cell morphology (poikilocytosis, anisocytosis, polychromasia) were observed in treated animals from all dose groups.
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
Dose dependent increases in both absolute and relative kidney weights at the 380 (500) and 1500 mg/kg/day dose levels. No significant changes in absolute or relative liver weights were observed
Gross pathological findings:
not examined
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Details on results:
Maternal toxic effects: yes
Number of abortions:
not specified
Pre- and post-implantation loss:
not specified
Total litter losses by resorption:
not specified
Early or late resorptions:
no effects observed
Description (incidence and severity):
DEA administration had no effect on resorption rate.
Dead fetuses:
no effects observed
Description (incidence and severity):
DEA administration had no effect on number of dead fetuses.
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
no effects observed
Description (incidence and severity):
DEA administration had no effect on pregnancy rate.
Other effects:
no effects observed
Description (incidence and severity):
DEA administration had no effect on corpora lutea number, implantation number, litter size and gravid uterine weight.
Details on maternal toxic effects:
Maternal toxic effects: yes at the lowest test concentration
Maternal developmental effects: No. DEA administration had no effect on pregnancy rate, corpora lutea number, implantation number, litter size, resorption rate, number of dead fetuses, fetal body weight, fetal sex ratio, or gravid uterine weight at any of the doses tested.
Dose descriptor:
LOAEL
Remarks:
general toxicity
Effect level:
150 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
dermal irritation
haematology
organ weights and organ / body weight ratios
Dose descriptor:
NOEL
Remarks:
Prenatal developmental toxicity
Remarks on result:
not determinable due to absence of adverse toxic effects
Fetal body weight changes:
no effects observed
Description (incidence and severity):
DEA administration had no effect on fetal body weight.
Reduction in number of live offspring:
not specified
Changes in sex ratio:
no effects observed
Description (incidence and severity):
DEA administration had no effect on fetal sex ratio
Changes in litter size and weights:
not specified
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not specified
External malformations:
no effects observed
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rat fetuses
Skeletal malformations:
effects observed, non-treatment-related
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rat fetuses. Although there was no difference in the overall incidence, litters from the 1500 mg/kg bw group had significantly increased incidences of six specific skeletal alterations. These alterations consisted primarily of delays in ossification. An elevated incidence of "all proximal hindlimb phalanges unossified corresponded with significant reciprocal reductions in litter frequency for "some proximal hindlimb phalanges poorly ossified." In the 1500 mg / kg body weight dose group there was also an increase in the incidence of "some foreleg metacarpal bones with weak ossification". At 150 or 380 (500) mg / kg body weight there was no impact on any of the development parameters.
Visceral malformations:
no effects observed
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rat fetuses.
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: no effects
Dose descriptor:
NOAEL
Effect level:
>= 1 500 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: overall teratogenicity effects
Remarks on result:
other: No adverse effects observed
Abnormalities:
not specified
Developmental effects observed:
not specified

Detailed results:

Maternal, litter, fetal data - prenatal developmental toxicity (dermal) study in CD rats

Dose (mg/kg bw)

0

150

380 (500)

1500

n (females)

25

25

25

25

pregnant

23

24

25

22

mortality

0

0

0

0

body weight gain GD 6-15 (g)

49

51

50

45

corrected body weight (g)

307.6

308.3

307.5

293.8

skin irritation

0

0

1

10

blood parameters

RBC

6.22

6.01        

5.95        

5.69**

HGB

11.7        

11.1        

11.0*        

10.3**

HCT

34.2        

32.4*        

32.2*        

30.1**

MCV

55.1        

54.0*        

54.2*        

53.1**

MCH

18.7        

18.3*        

18.4        

18.0**

kidney weight absolute (g)

1.96        

2.01        

2.13**        

2.24**

% kidney weight (g)    

0.64        

0.65        

0.69**        

0.77**

gravid uterus (g)

116

119

114

118

corpora lutea  (mean)    

18.8        

17.1        

17.5        

17.4

implantation  (mean) 

16.2        

16.3        

15.6        

16.1

% resorption        

5.4

5.4        

4.6        

4.2

live fetuses  (mean)                

15.3        

15.4        

14.8        

15.4

fetal weight (g)

5. 4

5.5        

5.5        

5.4

Total external malformations (fetus/litter)        

0/0        

0/0        

0/0        

0/0        

Total external variations (fetus/litter)        

50/21        

57/22        

61/22        

47/19

Total soft tissue malformations (fetus/litter)      

5/4        

3/3        

9/7        

10/8

Total soft tissue variations (fetus/litter)

95/22

97/24        

110/25        

114/20

Total skeletal malformations (fetus/litter)

1/1

0/0        

0/0        

0/0        

Total skeletal variations (fetus/litter)

171/23

179/24      

180/25

163/22

Incidences for key skeletal variations expressed as No. of fetus (litter)

Cervical centrum (No.5) poorly ossified:

25(13)      

35(16)

26(14)

38(19)*

Thoracic centrum (No.10) poorly ossified:

1(1)

1(1)

6(5)

7(7)*

Parietal, reduced ossification:

7(3)

5(4)

10(6)

44(13)**

all proximal phalanges (hindlimb) poorly ossified:

71(17)

84(19)

104(23)

105(22)*

some metacarpals (forelimb) poorly ossified:

0(0)        

0(0)        

2(2)

4(4)

BWG = body weight gain, * p0.05 **p0.01

Conclusions:
The LOAEL for maternal toxicity was set at 150 mg/kg bw. No effects on reproductive parameters were recorded. The NOAEL for teratogenicity was set to ≥ 1500 mg/kg bw.
Executive summary:

This prenatal developmental toxicity study was conducted comparable the the OECD TG 414. In this study DEA was administered to pregnant CD rats from gestation day 6 through day 15 at concentration of 0, 150, 500 and 1500 mg/kg bw. At 500 and 1500 mg/kg bw moderate and severe skin irritation was caused, respectively. Maternal body weight gain was decreased in the 1500 mg/kg bw group. Absolute and relative kidney weights were increased at 500 and 1500 mg/kg bw. Hematological effects including anemia, abnormal red cell morphology (poikilocytosis, anisocytosis, polychromasia), and decreased platelet count were observed in all treatment groups. The 1500 mg/kg bw group also had increased lymphocytes and total leukocytes.

In the fetuses, there were no effects of treatment on body weight or on incidence of external, visceral, or skeletal malformations/abnormalities. Increased incidences of six skeletal variations involving the axial skeleton and distal appendages were observed in litters from the 1500 mg/kg bw group. The skeletal variations included poor ossification in the parietal bones, cervical centrum, and thoracic centrum, lack of ossification in all proximal hindlimb phalanges and some forelimb metacarpals, and callused ribs.

Consequently, the LOAEL for maternal toxicity was 150 mg/kg bw. No effects on reproductive parameters were recorded. The NOAEL for teratogenicity was ≥ 1500 mg/kg bw.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Reason / purpose for cross-reference:
reference to same study
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Principles of method if other than guideline:
Prenatal developmental toxicity study with dermal application in rabbits
GLP compliance:
not specified
Limit test:
no
Species:
rabbit
Strain:
New Zealand White
Details on test animals or test system and environmental conditions:
- Source: Hazleton research Products Inc., Denver, PA
- Weight at study initiation: 2959-4414g
- Housing: individual
- Diet Ground certified rodent diet (RMH 3200, Agway. Inc. Waverly, NY) ad libitum
- Water ad libitum
- Acclimation period: 2 weeks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 16-21
- Humidity (%): 40-60
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
dermal
Vehicle:
unchanged (no vehicle)
Details on exposure:
TEST SITE
- Area of exposure: no data (clipped back skin)
- % coverage: 100
- Type of wrap if used: steril gauze and further occluded with polyvenyl film attached to a specially designed Lycra-Spandex jacket  with Velcro closures
- Time intervals for shavings or clipplings: no data


REMOVAL OF TEST SUBSTANCE
- Washing (if done): warm water
- Time after start of exposure: 6 hours


TEST MATERIAL
- Amount(s) applied (volume or weight with unit): 4 ml
- Constant volume or concentration used: yes

USE OF RESTRAINERS FOR PREVENTING INGESTION: no
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
gas chromatography
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 2/1
- Length of cohabitation: overnight
- Verification of same strain and source of both sexes: yes
- Proof of pregnancy: day of coitus referred to as day 0 of pregnancy
Duration of treatment / exposure:
gestation days 6 - 18
Frequency of treatment:
6 h/day
Duration of test:
gestation days 0-29
Dose / conc.:
0 mg/kg bw/day
Dose / conc.:
35 mg/kg bw/day
Dose / conc.:
100 mg/kg bw/day
Dose / conc.:
350 mg/kg bw/day
No. of animals per sex per dose:
15
Control animals:
other: water
Details on study design:
- Dose selection rationale: based on preliminary results of developmental toxicity range finding studies

In these studies, DEA concentrations of 500 mg/kg/day or greater produced excessive maternal toxicity, which included severe skin irritation (i .e ., necrosis, eccbymosis, edema, and severe erythema), increased relative and absolute liver weights, and inkidney weights. Minor skin irritation and increased liver weights were observed for some rabbit dams exposed to 250 mg DEHA/day.
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: daily


DETAILED CLINICAL OBSERVATIONS: Yes (skin)
- Time schedule: twice daily during the dosing period, once daily during the post-treatment period


BODY WEIGHT: Yes
- Time schedule for examinations: GD 0, 6, 9, 12, 15, 24, 29


FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): Yes
- Time schedule for examinations: daily

WATER CONSUMPTION AND COMPOUND INTAKE (if drinking water study): Yes / No / No data
- Time schedule for examinations:


POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 29
- Organs examined: liver, kidney
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
Blood sampling:
BLOOD:
- Prior to sacrifice on GD 29
Fetal examinations:
- External examinations: Yes: all per litter (variation/malformation) / half per litter (thoracic, abdominal visceral abnormalities)
- Soft tissue examinations: No data
- Skeletal examinations: Yes: all per litter
- Head examinations: Yes: half per litter
Statistics:
Continuous variables were compared for homogeneity of variance using  Levene's test for equal variances (Levene, 1960). A parametric or nonparametric analysis of variance (ANOVA) was performed , f parametric ANOVA analyses were significant, pooled T-tests were used for pairwise comparisons. If results from a nonparametric ANOVA were significant, separate variance T tests for pairwise comparisons were performed. Data  from nongravid females and females delivering early were not included in  the statistical analyses. Nonparametric data were statistically evaluated  using the Kruskal-Wallis test, followed by the Mann Whitney U test. Incidence data were compared using the Fisher's exact test (Sokal and  Rohlf, 1969), with the exception of frequency data for fetal malformations and variations, statistical analyses were performed using BMDP Statistical Software (Dixon, 1990). For all statistical tests, the critical level of significance was set a priori at a = 0.05 (two-tailed).
Indices:
no information given
Historical control data:
no information given
Clinical signs:
not specified
Dermal irritation (if dermal study):
effects observed, treatment-related
Description (incidence and severity):
Skin lesions and irritation were observed in the 350 mg/kg bw group during both the DEA treatment period and the subsequent recovery period. There were no perceptible changes in skin irritation or lesion frequency in rabbits administered 35 or 100 mg/kg/day. Skin irritation was not observed in the water control group
Mortality:
no mortality observed
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
There was an overall decrease in gestational body weight gains in the 100 and 350 mg/kg bw dose groups during the treatment period. At the highest dose, weight gain was decreased during the post-treatment period.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
On GD 17 and 18 of the treatment period and for 4 days subsequent to treatment, food consumption was reduced in the 350 mg/kg bw group.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not specified
Ophthalmological findings:
not examined
Haematological findings:
no effects observed
Description (incidence and severity):
There were no significant changes in hematological parameters at any of the DEA doses tested.
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, non-treatment-related
Description (incidence and severity):
Although not statistically significant, absolute and relative liver weights and relative kidney weights were increased.
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
At the high dose, 50% of treated rabbit dams exhibited color changes in the kidneys compared with 17% in control rabbits.
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Number of abortions:
no effects observed
Description (incidence and severity):
There were no significant effects on reproductive parameters. One dam from the 350 mg/kg bw group aborted her litter on GD 27, an outcome which followed a sharp decline in food consumption beginning on GD 14-15. At necropsy, this animal had color changes in the kidneys and a reticular pattern in all hepatic lobes
Pre- and post-implantation loss:
not examined
Total litter losses by resorption:
no effects observed
Description (incidence and severity):
One dam from the 100 mg/kg/day group had a single implantation site with a fetus that had undergone early resorption.
Early or late resorptions:
no effects observed
Dead fetuses:
no effects observed
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
not examined
Other effects:
not examined
Details on maternal toxic effects:
Maternal toxic effects: yes
Dose descriptor:
NOEL
Remarks:
general toxicity
Effect level:
35 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
Fetal body weight changes:
no effects observed
Reduction in number of live offspring:
not examined
Changes in sex ratio:
not examined
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rabbit fetuses.
Although there was a greater variety of malformations in the 350 mg/kg bw group, many of these malformations (ovoid lenses; common truncus; ventricular septal defect; herniated diaphragm; extra lumbar centrum No. 8; extra bilateral lumbar arch No. 8; bone island at caudal segment No. 2; and duplicated, misshaped, and fused sternebrae) occurred in the same fetus. Furthermore, the overall incidence of malformations in the high dose DEA group (4.5% of fetuses, 30.8% of litters) was similar to the incidence seen in control animals (6.7% of fetuses, 25.0% of litters). Of the 53 different fetal skeletal variations observed, only the occurrence of poorly ossified interparietal was statistically increased in the 350 mg/kg/day dose group. However, a consistent, dose-dependent profile of delayed ossification in the head region was not observed
Skeletal malformations:
no effects observed
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rabbit fetuses.
Although there was a greater variety of malformations in the 350 mg/kg bw group, many of these malformations (ovoid lenses; common truncus; ventricular septal defect; herniated diaphragm; extra lumbar centrum No. 8; extra bilateral lumbar arch No. 8; bone island at caudal segment No. 2; and duplicated, misshaped, and fused sternebrae) occurred in the same fetus. Furthermore, the overall incidence of malformations in the high dose DEA group (4.5% of fetuses, 30.8% of litters) was similar to the incidence seen in control animals (6.7% of fetuses, 25.0% of litters). Of the 53 different fetal skeletal variations observed, only the occurrence of poorly ossified interparietal was statistically increased in the 350 mg/kg/day dose group. However, a consistent, dose-dependent profile of delayed ossification in the head region was not observed
Visceral malformations:
no effects observed
Description (incidence and severity):
No effect on the overall incidence of external, visceral, or skeletal malformations or variations observed in rabbit fetuses.
Although there was a greater variety of malformations in the 350 mg/kg bw group, many of these malformations (ovoid lenses; common truncus; ventricular septal defect; herniated diaphragm; extra lumbar centrum No. 8; extra bilateral lumbar arch No. 8; bone island at caudal segment No. 2; and duplicated, misshaped, and fused sternebrae) occurred in the same fetus. Furthermore, the overall incidence of malformations in the high dose DEA group (4.5% of fetuses, 30.8% of litters) was similar to the incidence seen in control animals (6.7% of fetuses, 25.0% of litters). Of the 53 different fetal skeletal variations observed, only the occurrence of poorly ossified interparietal was statistically increased in the 350 mg/kg/day dose group. However, a consistent, dose-dependent profile of delayed ossification in the head region was not observed
Other effects:
not examined
Dose descriptor:
NOEL
Remarks:
prenatal development and teratogenicity
Effect level:
>= 350 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: overall teratogenicity effects
Remarks on result:
other: No adverse effects observed
Abnormalities:
not specified
Developmental effects observed:
not specified

Detailed results:

Maternal, litter, fetal data - prenatal developmental toxicity data

mg/kg bw        

0

35

100

350

femals used

15

15

15

15

pregnant

12

14

14

14

Mortality

0

0

0

0

BWG GD 6-18 (g)

59

98

12

-5.6

FC GD 6-18 (g)

164

175

160

152

Skin irritation        

0

0

0

10

Gravid uterus (g)

528

562

513

542

Corpora lutea 
(mean)                

9.6

9.8        

9.1        

9.5

Implantation 
(mean)                

8.0        

8.7        

8.0        

9.1

% Resorption

16.7        

7.1        

21.4        

23.1

Live fetuses 
(mean)                

7.5        

8.6        

7.7        

8.5

Fetal weight (g)

44

44

43

40

Total external malformations (fetus/litter)        

1/1

0/0

0/0

1/1

Total external variations (fetus/litter)        

0/0

0/0

0/0

0/0

Total soft tissue malformations (fetus/litter)        

4/1

9/4

5/3

2/2

Total soft tissue variations (fetus/litter)        

19/12        

30/9        

28/10

35/13

Total skeletal malformations (fetus/litter)        

1/1

1/1

2/2

3/3

Total skeletal variations (fetus/litter)        

90/12        

120/14        

108/13        

111/13

BWG = body weight gain, FC = food consumption, * p<0.05;  **p<0.01

Conclusions:
Maternal toxicity was observed in dams at dose levels of ≥100 mg/kg bw. There was no impairment of gestational parameters. No evidence of developmental toxicity was observed at any dose level, especially, there were no apparent effects of treatment on the incidences of external, visceral, or skeletal abnormalities.
Consequently, the NOEL for maternal toxicity was 35 mg/kg bw, the NOEL for prenatal developmental toxicity including teratogenicity was ≥ 350 mg/kg bw
Executive summary:

This prenatal developmental toxicity study was conducted comparable the the OECD TG 414. In the study DEA was administered to pregnant New Zealand White rabbits from gestation day 6 through day 18 at concentration of 0, 35, 100 and 350 mg/kg bw. Rabbit dams at 350 mg/kg bw showed several signs of marked skin irritation, reduced food consumption, and color changes in the kidneys but no hematological changes. Body weight gain was reduced at ≥ 100 mg/kg bw. There was no impairment of gestational parameters. No evidence of developmental toxicity was observed at any dose level, especially, there were no apparent effects of treatment on the incidences of external, visceral, or skeletal abnormalities.

Consequently, the NOEL for maternal toxicity was 35 mg/kg bw, the NOEL for prenatal developmental toxicity including teratogenicity was 350 mg/kg bw, the highest dose of DEA tested in rabbits.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
study well documented, meets generally accepted scientific principles, acceptable for assessment
Qualifier:
no guideline followed
Principles of method if other than guideline:
No guideline for this screening study available.
Postnatal developmental study of rat pups after maternal exposure from gestation day 6 - 19
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Sprague-Dawley
Details on test animals or test system and environmental conditions:
- Source: Charles River Laboratories
- Weight at study initiation: 221-275 g
- Housing: individually
- Diet Purina certified rodent chow ad libitum
- Water ad libitum
- Acclimation period: 10 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 18-23
- Humidity (%): 40-70
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
Diethanolamine was formulated in distilled (Pico) water at nominal concentrations of 10–60 mg/mL, and pH was adjusted to 7.470.2 with hydrochloric acid. Formulations were stored in sealed, amber glass bottles at room temperature and administered within the period of proven stability. One set of dose formulations was used for the entire dosing period.

VEHICLE
- Amount of vehicle (if gavage): 5 mL/kg
- Concentration in vehicle: 10-60 mg/mL
- pH adjustment to 7.4
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Each nominal concentration of DEA in water was sampled before use and analyzed by G.C. to verify concentration of the test chemical. Found concentrations ranged from 97.8–101.2% of their respective nominal concentration.
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 1/1
- Length of cohabitation: overnight
- Verification of same strain and source of both sexes: yes
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy
Duration of treatment / exposure:
gestation day 6 - postnatal day 19
Frequency of treatment:
daily
Duration of test:
40 days
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Dose / conc.:
50 mg/kg bw/day (actual dose received)
Dose / conc.:
125 mg/kg bw/day (actual dose received)
Dose / conc.:
200 mg/kg bw/day (actual dose received)
Dose / conc.:
300 mg/kg bw/day (actual dose received)
No. of animals per sex per dose:
12 pregnant females plus 10 sentinels
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: lowest dose was was within the range associated with mild hematotoxicity, the highest dose was expected to cause significant maternal toxicity including morbidity or mortality
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: at least once daily

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: GD 0-5 daily, 6-19 daily before dosing and 1.5 h after dosing, and on GD 20 and PND 0, 4, 7, 14, 21 once daily

FOOD CONSUMPTION: Yes
- Time schedule for examinations: GD 0, 6, 9, 12, 15, 18, and 20 for all timed-mated females, as well as PND 0, 4, 7, 14, and 21 for dams with litters.

WATER CONSUMPTION: Yes
- Time schedule for examinations: GD 0, 6, 9, 12, 15, 18, and 20 for all timed-mated females, as well as PND 0, 4, 7, 14, and 21 for dams with litters.

BODY WEIGHT: Yes
- Time schedule for examinations: GD0, 6-20, and PND 0, 4, 7, 14, 21 for dams with litters

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 20 or on PND21
- Organs examined: liver, kidney, and Gross pathology
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: No data
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
Fetal examinations:
- External examinations: Yes (pups were examined for malformations or variations on PND 0)
- Soft tissue examinations: No data
- Skeletal examinations: No data
- Head examinations: No data

OTHER:
Naturally delivered litters were evaluated for clincal signs, litter size, pub sex and individual pub weight on PND 0, 4, 7, 14, 21. Gross necropsy on PND 7 (culling) or PND21 (scheduled termination)
Statistics:
Statistical procedures were based on SAS software (Version 6.12; SAS  Institute, Inc., Cary, NC), the alpha level was 0.05. Nonparametric tests applied to continuous variables included the  Kruskal-Wallis one-way analysis of variance by ranks for among-group  differences and, if significant (p < 0.05), the Mann- Whitney U-test for  pairwise comparisons to the vehicle control group. A one-tailed Mann-Whitney U-test was used for all parameters, except that  maternal and pup body weight parameters, and maternal feed and water  consumption were examined using a two-tailed test. Jonckheere's test for k-independent samples was used to identify  significant dose-response trends. Nominal scale measures were analyzed by Chi-Square test for independence  for differences among treatment groups and by the Cochran-Armitage test  for linear trend on proportions. When Chi-Square test showed significant (p < 0.05) differences among  groups, then a one-tailed Fisher's exact probability test, with  appropriate adjustments for multiple comparisons, was used for pairwise  comparisons between control and DEA treated groups. A probit analysis was used to determine the maternal LD10 in this study.
Indices:
no information given
Historical control data:
no information given
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
During treatment, clinical signs of toxicity were unremarkable in the control, 50 and 125 mg/kg/day groups. At >= 200 mg/kg bw, the clinical signs noted most frequently during the treatment period were piloerection, and lethargy.
During lactation, treatment-related clinical signs were limited to an apparent increase in the incidence of piloerection in the 125, 200, and 250 mg/kg bw groups on PND 0 and 1. Only one female in the 250 mg/kg/day group exhibited piloerection on PND 4 and 5 and no treatment-related clinical signs were observed thereafter.
Dermal irritation (if dermal study):
not examined
Mortality:
mortality observed, treatment-related
Description (incidence):
At the highest dose (300 mg/kg bw), two females were terminated in extremis on GD 11. Due to this excessive toxicity, all remaining females in that group were humanely terminated before scheduled necropsy and excluded from further summary of results.
At 200 mg/kg bw, one female was terminated in extremis on GD 22 while attempting to deliver a litter of 15 pups, all of which were found dead in utero at necropsy.
At 250 mg/kg bw, one female was found dead on GD 15 and another was terminated in extremis on GD 21 with 12 dead pups in utero.
Twelve females each from the control, 50 and 125 mg/kg/day groups, as well as 11 females from the 200 mg/kg/day group delivered a live litter and survived until scheduled necropsy on PND 21.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
There were no statistically significant effects of DEA on maternal body weight or weight change at 50 or 125 mg/kg/day relative to the vehicle control group. Reduced maternal body weight was notable at >=200 mg/kg/day during the first 6 days of the treatment period, with significantly reduced body weight change for the first two measurement periods (GD 6–9 and GD 9–12; weight change data not shown for these measurement periods). At 200 mg/kg/day, maternal body weight was 10% lower than controls on GD 12 and 7% lower than controls on PND 4 (both significant). At 250 mg/kg/day, maternal body weight was significantly decreased, as follows: 16–20% reduction on GD 12, 15, 18, and 20, and 6% reduction on PND 4. For the treatment (GD 6–20) and gestational periods (GD 0–20), maternal body weight change was significantly reduced at 200 and 250 mg/kg/ day. During lactation, maternal body weight change showed a decreasing trend from PND 0–4, but individual groups did not differ significantly from controls. All DEA-treated groups showed body weight change comparable to or greater than controls and the pattern for some measurement periods suggested compensatory rebound after cessation of exposure.
At scheduled necropsy (PND 21), maternal body weight was unaffected.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
During treatment, maternal relative feed intake was decreased at 200 and 250mg/kg bw from GD 6-9, 9-12, and 12-15. At 250 mg/kg/ day, feed intake was also transiently decreased from PND 0-4. Otherwise, maternal relative feed intake was comparable to or greater than controls for specific measurement periods.
Averaged across the treatment period, maternal relative feed intake was reduced at >= 200 mg/kg bw.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
effects observed, treatment-related
Description (incidence and severity):
Maternal relative water intake was reduced from GD 9-12 at 125 and 250 mg/kg bw. Otherwise, maternal relative water intake was comparable to or greater than controls for specific measurement periods.
Averaged across the treatment period, maternal relative water intake was not affected.
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
At scheduled necropsy (PND 21), maternal body weight and liver weight were unaffected. Maternal kidney weight showed an increasing trend, and absolute kidney weight was significantly elevated at 125, 200, and 250 mg/kg bw.
Gross pathological findings:
not specified
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Details on results:
Mortality: Twelve females each from the control, 50 and 125mg/ kg/day groups, as well as 11 females from the 200 mg/kg/day group delivered a live litter.
At 250 mg/kg on GD 15 one female was terminated in extremis on GD 21 with 12 dead pups in utero. Among the remaining females in the 250 mg/kg/day group, five delivered live litters and survived to scheduled termination on PND 21, one delivered a litter in which all pups were dead (PND 0).
At 200 mg/kg bw, one female was terminated in extremis on GD 22 while attempting to deliver a litter of 15 pups, all of which were found dead in utero at necropsy.
Number of abortions:
not specified
Pre- and post-implantation loss:
effects observed, treatment-related
Description (incidence and severity):
Postimplantation loss was significantly higher than controls in the 200 and 250 mg/kg bw groups and for 2, 6, 3, 17, and 51% of implantation sites/dam in the control - 250 mg/kg bw group. At 250 mg/kg bw day, four of nine (44%) females had 100% postimplantation loss.
Total litter losses by resorption:
effects observed, treatment-related
Description (incidence and severity):
At 250 mg/kg three females had completely resorbed litters at uterine examination on GD 24.
Early or late resorptions:
not examined
Dead fetuses:
effects observed, treatment-related
Description (incidence and severity):
Exposure was associated with a decreasing trend for the average number of live pups per litter on PND 0; at 50, 125, 200 and 250 mg/kg bw, the mean number of live pups per litter was 91, 97, 85, and 55% of the control group mean, respectively, but the average number of live pups/live litter showed no statistically significant difference among groups.
Changes in pregnancy duration:
effects observed, treatment-related
Description (incidence and severity):
Dams in the control group delivered on GD 21 or 22, whereas dams in DEA-exposed groups delivered on GD 22 or 23.
Changes in number of pregnant:
effects observed, treatment-related
Description (incidence and severity):
Twelve females each from the control, 50 and 125 mg/kg bw groups, as well as 11 females from the 200 mg/kg bw group delivered a live litter and survived until scheduled necropsy on PND 21. At 250 mg/kg bw, one female was found dead on GD 15 and another was terminated in extremis on GD 21 with 12 dead pups in utero. Among the remaining females in the 250 mg/kg bw group, five delivered live litters and survived to scheduled termination on PND 21, one delivered a litter in which all pups were dead (PND 0), three had completely resorbed litters at uterine examination on CD 24, and one was not pregnant.
Other effects:
effects observed, treatment-related
Description (incidence and severity):
Probit analysis yielded a calculated LD10 of 218 mg/kg/day under the exposure conditions for this study.
Details on maternal toxic effects:
Maternal toxic effects: yes

For further details please refer to 'any other information on results'
Dose descriptor:
NOAEL
Effect level:
50 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
clinical signs
food consumption and compound intake
mortality
organ weights and organ / body weight ratios
water consumption and compound intake
Dose descriptor:
NOAEL
Effect level:
50 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: postimplantation mortality, early postnatal mortality, pup body weight was reduced
Remarks on result:
other: signs of developmental toxicity did only occur at maternal toxic dose levels
Fetal body weight changes:
effects observed, treatment-related
Description (incidence and severity):
Pup body weight was not affected at 50 mg/kg bw, at 125mg/kg/day, there was a 7-8% increase in average pup body weight/litter on PND 14. At 200 mg/kg bw, female pup body weight was significantly reduced by 8% on PND 0 and 4 and by 10% on PND 21. At the same dose, male pup body weight was significantly reduced by 11% on PND 21. At 250 mg/kg bw, female pup body weight/litter was significantly reduced by 16-17% on PND 0, 4, 7, and by 10% on PND 21 and male pup body weight/litter was significantly reduced by 11% on PND 0, 15% on PND 4 and 14% on PND 21
Reduction in number of live offspring:
no effects observed
Description (incidence and severity):
Survival, average live litter size was comparable across treatment groups at the beginning of the postnatal period.
Changes in sex ratio:
not specified
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
effects observed, treatment-related
Description (incidence and severity):
At >= 125 mg/kg bw postnatal deaths primarily during the early postnatal period (PND 0–4) occured. Significant decreasing trends for the number of live pups/live litter were noted on PND 4 and 7 but after culling (PND 7), there were no differences among groups for the number of live pups/live litter on PND 14 or 21.
From PND 0-4, postnatal mortality occurred in 0, 0.6, 1.8, 2.8, and 13.4% of pups /litter in the control - 250 mg/kg bw, respectively, being statistically significant at >= 125 mg/kg bw.
There was no biological relevant difference in the proportion of male and female pups.
External malformations:
effects observed, non-treatment-related
Description (incidence and severity):
At scheduled necropsy on PND 7 (culling) and PND 21 (study termination), morphological findings were limited to one pup with distended ureter (PND 7, 200 mg/ kg/ day) and one with unilateral hydronephrosis (PND 21, 50 mg/kg/ day).
Skeletal malformations:
not examined
Visceral malformations:
not examined
Other effects:
effects observed, non-treatment-related
Description (incidence and severity):
At 250 mg/kg/ day, five dead pups displayed hydroureter and/or hydronephrosis but these findings are common to this species/strain. For most pups that died postnatally, there was no milk found in the stomach, indicating unsuccessful nursing.
At 200 mg/kg bw, there was a non-dose-related increase in the incidence of alopecia.
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: yes
Dose descriptor:
NOAEL
Effect level:
50 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
fetal/pup body weight changes
changes in postnatal survival
Remarks on result:
other: Developmental effects only occure at concentrations of maternal toxicity
Abnormalities:
not specified
Developmental effects observed:
not specified

Detailed results:

Maternal data

mg/kg bw/day

0

50

125

200

250

no of females        

12

12

12

12

12

deaths

0

0

0

1

2

pregnant

12

12

12

12

11

live litters

12

12

12

11

5

Dams with litters

PND 0

12

12

12

11

6

PND 4-21   

12

12

12

11

5

Dams on PND 21

12

12

12

11

5

BW PND 21 (g)

308

318

310

327

325

liver weight (g)    

17.38   

17.69   

18.23 

17.17   

17.82

% liver weight   

5.65   

5.57   

5.89   

5.22   

5.48

kidney weight (g) 

2.64   

2.77

2.91*   

3.01*   

3.10*

% kidney weight   

0.86   

0.87   

0.94   

0.92   

0.95

* p <0.05, ** p <0.01



Litter data

mg/kg bw/day

0

50

125

200

250

dams

12

12

12

12

12

pregnant

12

12

12

12

11

live litters   

12

12

12

11

5

Gestation day

21.9   

22.2   

22.3   

22.3   

22.2

Implant./litter   

14.25   

13.75   

14.33   

14.42   

14.91

Postimplant.
loss %       

2.48   

5.83   

3.41   

17.31**   

50.9**

Dead pups                    

PND 0

1

0

5

0

0

PND 1

0

0

0

0

1

PND 4

0

1

3

4

8

PND 14           

0

0

2

1

2

PND 0-21   

1

1

10

5

11

% mortality/litter

PND 0-4       

0.6   

0.6   

1.8*   

2.8*   

13.4**

PND 7-14

0.0   

0.0   

2.1   

1.1   

5.0

* p< 0.05, ** p< 0.01

Pup body weight gain (mean)

mg/kg bw/day

0

50

125

200

250

Live litter   

12

12

12

11

5

PND 0

6.1   

6.34   

6.33   

5.65   

5.25*

PND 4

9.72   

10.17   

9.96   

8.81*   

8.15*

PND 7

13.86   

14.71   

14.54   

12.74   

11.99*

PND 14       

33.1   

33.88   

35.53   

30.72   

30.18

PND 21       

53.8   

55.26   

55.96   

48.02*   

47.46*

* p< 0.05

Conclusions:
In conclusion, this study demonstrates reduced postnatal growth and survival in a second species after gestational exposure to DEA, persistence of toxic effects through the end of lactation, possibly due to long elimination half-life, and maternal and developmental toxicity no-observed-adverse-effect level (NOAELs) (50 mg/kg/day) and lowest-observed-adverse-effect level (LOAELs) (125 mg/kg/day) for oral DEA exposure during embryo/fetal development in the rat.
Executive summary:

This study investigated DEA-induced postnatal toxicity in Sprague-Dawley rats. In the study, timed-mated Sprague-Dawley rats were dosed (0, 50, 125, 200, 250, or 300 mg DEA/kg/day, p.o.) on gestational days (GD) 6–19. Dams and pups were monitored for body weight, feed/water intake, clinical signs, litter size, and sex ratio. At necropsy (PND 21), maternal liver and kidney weights and number of uterine implantation sites were recorded.

The high-dose group was terminated early due to excessive toxicity. The estimated maternal LD10 was 218 mg/kg/day. Maternal effects included decreased body weight and relative feed intake (200 mg/kg/day), transiently reduced relative water intake (125 and 250  mg/kg/day), and increased absolute kidney weight (125  mg/kg/day). Postimplantation loss (PND 0) and pup mortality (PND 0–4) were increased (200 and125  mg/kg/day, respectively). Pup body weight was reduced (200  mg/kg/day) as late as PND 21.

In conclusion, this study demonstrates reduced postnatal growth and survival after gestational exposure to DEA, persistence of toxic effects through the end of lactation, possibly due to long elimination half-life. The maternal and developmental toxicity no-observed-adverse-effect level (NOAEL) is 50  mg/kg/day and the lowest-observed-adverse-effect level (LOAEL) is 125 mg/kg/day for oral DEA exposure during embryo/fetal development in the rat.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
27 MAY 2008 (Study plan) to Jun/July 2008 (end of experimental phase); 14 MAR 2019 (Report date of summary of results)
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
comparable to guideline study
Qualifier:
equivalent or similar to guideline
Guideline:
other: Prenatal part: Proposal for updating Guideline 414: Prenatal Developmental Toxicity Study
Version / remarks:
22 Jan 2001
Qualifier:
equivalent or similar to guideline
Guideline:
other: Postnatal part: OECD guideline for the Testing of Chemicals; No.421 (SIDS): reproduction/Developmental Toxicity Screening Test
Version / remarks:
July 1995
Deviations:
yes
Remarks:
no pairing/fertility part
Qualifier:
equivalent or similar to guideline
Guideline:
other: EPA Health Effects Test Guideline, OPPTS 870.3550: Reproduction/Developmental Toxicity Screening Test (July 2000)
Version / remarks:
Jul 2000
Principles of method if other than guideline:
The aim of this screening study was to obtain initial information on the effect of the test substance after repeated oral administration (gavage) to pregnant female Wistar rats from gestation day (GD) 6 to GD 19 (prenatal study) and from GD 6 to postnatal day (PND) 3 (postnatal study). For the prenatal study part, selected dams of each group (5 animals of the control group, 5 animals of dose group 1 (300 mg/kg bw/day) and 10 animals of dose group 2 (600 mg/kg bw/day)) were sacrificed on GD 20; dams and fetuses were examined.This endpoint study record refers mainly to the prenatal study.
GLP compliance:
yes
Limit test:
no
Species:
rat
Strain:
Wistar
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH
- Age at study initiation: about 10-15 weeks
- Housing: 1 animal / cage; from delivery to sacrifice (rearing) - 1 dam with its litter / cage, Makrolon cages type M III, Wooden gnawing blocks (Typ NGM E-022), Type Lignocel FS 14 fibres, dustfree bedding
- Diet (e.g. ad libitum): ad libitum
- Water (e.g. ad libitum): ad libitum
- Acclimation period: During the acclimatization period, the animals were accustomed to the environmental conditions and to the diet.

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24°C
- Humidity (%): 30-70%
- Air changes (per hr): 15 air changes per hour
- Photoperiod (hrs dark / hrs light):12 hours light (6:00 a.m. - 6:00 p.m.), 12 hours darkness (6:00 p.m. - 6:00 a.m.)


IN-LIFE DATES: From: 29 MAY TO: 18 JUN 2008 (GD 20/Sacrifice of selected animals)
Route of administration:
oral: gavage
Vehicle:
olive oil
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
For the test substance preparation, the specific amount of test substance was weighed, topped up with olive oil in a volumetric flask and intensely shaken until it was completely dissolved

VEHICLE
- Amount of vehicle (if gavage): 4 mL/kg body weight
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The stability of the test substance in olive oil over a period of up to 7 days at room temperature was verified analytically before the start of the study (Analytical report: 01Y0262/078001)
Details on mating procedure:
The animals paired by the breeder (time-mated animals) were supplied on the day of evidence of mating; this day is referred to as GD 0 and the following day as GD 1.
Duration of treatment / exposure:
from GD 6 through GD 19
Frequency of treatment:
Once daily
Duration of test:
25 days
Dose / conc.:
0 mg/kg bw/day
Remarks:
Group 0
Dose / conc.:
300 mg/kg bw/day
Remarks:
Group 1
Dose / conc.:
600 mg/kg bw/day
Remarks:
Group 2
No. of animals per sex per dose:
Control: 10 (5 in prenatal study)
300 mg/kg bw: 10 (5 in prenatal study)
600 mg/kg bw: 20 (10 in prenatal study)
Control animals:
yes
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- A check for moribund and dead animals was made twice daily from Monday to Friday and once daily on Saturday, Sunday and public holidays.
- A cageside examination was conducted at least once daily for any signs of morbidity, pertinent behavioral changes and signs of overt toxicity, further abnormalities, changes, littering and lactation behavior of the dams.

BODY WEIGHT: Yes
- Time schedule for examinations: Body weights were recorded on GD 0 and on GD 1, 3, 6, 8,10, 13, 15, 17, 19, 20

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): Yes
- Food consumption was recorded on GD 1, 3, 6, 8,10, 13, 15, 17, 19, 20

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 20
- Gross-pathological examination
- Weight of the unopened uterus
- Weight of liver (livers will be retained in 4% buffered formaldehyde solution and transferred to Pathology Laboratory for potential histopathological processing and evaluation)
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Weight of the unopened uterus
- Number of corpora lutea
- Number of implantations (differentiated according to live and dead fetuses and early or late resorptions)
- Early resorptions in animals that do not appear to be pregnant and animals with single-horn pregnancy
- Site of implantations in the uterus
Fetal examinations:
After the fetuses have been removed from the uterus, the following examinations or weight determinations were carried out:
- Weight of each fetus
- Sex
- Weight of the placentas
- Gross-pathological examination of the fetuses after dissection from the uterus (including abnormalities of the fetal membranes, placentas, amniotic fluid and umbilical cord); all fetuses are sacrificed by subcutaneous injection of pentobarbital.
- About half of the fetuses of each dam: Skeletons/cartilage examination
- About half of the fetuses of each dam: Soft tissue examination
Statistics:
Means and standard deviations will be calculated.
- DUNNETT test (two-sided): Food consumption, body weight and body weight change; duration of gestation
- KRUSKAL-WALLIS and WILCOXON test: Weight of liver
Indices:
Female fertility index
Historical control data:
no information given
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day: Salivation (20 out of 20 animals), respiratory sounds (7 out of 20 animals) were observed.
300 mg/kg bw/day: Salivation after treatment (10 out of 10 animals).
Dermal irritation (if dermal study):
not examined
Mortality:
mortality observed, treatment-related
Description (incidence):
600 mg/kg bw/day:
- One animal sacrificed moribund on GD 14 (gross pathological examination revealed stomach erosions and no feces in intestine).
- One animal found dead on GD 20 (gross pathological examination revealed stomach ulcerations).

300 mg/kg bw/day: no effects observed
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day:
- Transient statistically significantly reduced mean body weight compared to the control group (set to 100 %), i.e. on GD 13 (93%). No statistical relevant difference on GD 20.
- Transient statistically significantly reduced mean body weight change compared to the control group (set to 100 %), i.e. between GD 8-10 (52 %). No statistically significantly difference between GD 0-20.

300 mg/kg bw/day: no effects observed
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day: Transient statistically significantly reduced mean food consumption compared to the control group (set to 100 %), i.e. between GD 6-8 (81 %) and GD 8-10 (78 %). No statistically significantly difference between GD 0-20.

300 mg/kg bw/day: no effects observed.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day - Prenatal study part (10 dams sacrificed on GD 20): Statistically significantly increased mean liver weight (118 %) compared to the control group (set to 100 %). Since no macroscopic or microscopic examinations were made, no conclusion on adversity is possible.

300 mg/kg bw/day: no effects observed.
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day - Prenatal study part (10 dams sacrificed on GD 20): Stomach erosions/ulcera (8 out of 10 animals)
300 mg/kg bw/day - Prenatal study part (5 dams sacrificed on GD 20): Stomach erosions/ulcera (4 out of 5 animals)
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not specified
Number of abortions:
not specified
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
600 mg/kg bw/day - Prenatal study part (10 dams sacrificed on GD 20): Slightly increased post-implantation loss compared to the control group and the historical control data (11.8 % vs. 5.2 % in control), not statistically significant.
300 mg/kg bw/day - Prenatal study part (5 dams sacrificed on GD 20): Slightly increased post-implantation loss compared to the control group and the historical control data (15.2 % vs. 5.2 % in control), not statistically significant.
Total litter losses by resorption:
not specified
Early or late resorptions:
no effects observed
Description (incidence and severity):
600 mg/kg bw/day - Prenatal study part (5 dams sacrificed on GD 20): Slightly increased resorptions (mean/litter) compared to the control group (0.8 % vs. 0.6 % in control), not statistically significant and without dose-response relation.
300 mg/kg bw/day - Prenatal study part (5 dams sacrificed on GD 20): Slightly increased resorptions (mean/litter) compared to the control group and the historical control data (1.4 % vs. 0.6 % in control), not statistically significant and without dose-response relationship.
Dead fetuses:
no effects observed
Description (incidence and severity):
600 and 300 mg/kg bw/day: no effects observed
Changes in pregnancy duration:
no effects observed
Description (incidence and severity):
600 and 300 mg/kg bw/day: no effects observed
Changes in number of pregnant:
effects observed, treatment-related
Description (incidence and severity):
600 mg/kg bw/day - Postnatal study part (8 dams): One out of 8 animals did not deliver (7 animals left for further assessment)

300 mg/kg bw/day: no effects observed.
Other effects:
not specified
Details on maternal toxic effects:
Maternal toxicity: yes
Dose descriptor:
LOAEL
Remarks:
local effects
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
gross pathology
Dose descriptor:
NOAEL
Remarks:
general toxicity
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
clinical signs
food consumption and compound intake
mortality
Fetal body weight changes:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Reduction in number of live offspring:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Changes in sex ratio:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Changes in litter size and weights:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
External malformations:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Skeletal malformations:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Visceral malformations:
no effects observed
Description (incidence and severity):
600 mg/kg body weight/day - Prenatal study part (10 dams sacrificed on GD 20): Fetuses: No test substance-related findings
300 mg/kg body weight/day - Prenatal study part (5 dams): Fetuses: No test substance-related findings
Other effects:
not specified
Dose descriptor:
NOAEL
Basis for effect level:
other: No test item-related findings
Remarks on result:
not determinable
Remarks:
No NOAEL identified
Abnormalities:
not specified
Developmental effects observed:
no

Gestation

Test group (mg/kg bw/d)

0 (0; control)

1 (300)

2 (600)

Mortality

-

-

1 animal sacrificed moribund on GD 14

1 animal found dead on GD 20

Clinical observation

NAD

Salivation after treatment (10/10)

Salivation after treatment (20/20)

Respiratory sounds (7/20)

Labored respiration (1/20)

Fur smeared with urine (1/20)

FC (day 6-8)

FC (day 8-10)

FC (day 0-6)

16.1 g

17.4 g

15.1 g

16.0 g (99 %)

16.9 g (97 %)

15.4 g (102 %)

13.1 g (81 %)**

13.5 g (78 %)**

15.6 g (103 %)

FC (day 6-20)

18.1 g

17.9 g (99 %)

16.7 g (92 %)

FC (day 0-20)

17.2 g

17.1 g (99 %)

16.4 g (95 %)

BW (day 0)

162.6 g

157.7 g (97 %)

160.5 g (99 %)

BW (day 6)

BW (day 13)

190.8 g

223.1 g

187.8 g (98%) 215.3 g (96 %)

189.8 g (99 %)

208.5 g (93 %)*

BW (day 20)

281.4 g

268.5 g (95 %)

263.5 g (94 %)

BWC (day 8-10)

BWC (day 0-6)

11.8 g

28.2 g

8.7 g (74 %)

30.1 g (107 %)

6.2 g (52 %)**

29.3 g (104 %)

BWC (day 6-20)

90.5 g

80.7 g (89 %)

73.8 g (81 %)

BWC (day 0-20)

118.7 g

110.8 g (93 %)

103.3 g (87 %)

Duration of Gestation

21.8

22.0

22.3

Cesarean section

Test group

(mg/kg bw/d)

0 (0; control)

1 (300)

2 (600)

Uterus weight

52.8 g

43.5 g (82 %)

49.7 g (94 %)

Carcass weight

228.8 g

221.0 g (97 %)

216.1 g (94 %)

Corrected body weight gain

40.3 g

36.6 g (91 %)

28.7 g (71 %)

Implantation sites (mean/litter)

10.4

9.4

10.2

Postimplantation loss

5.2

15.2

11.8

Resorptions (mean/litter)

0.6

1.4

0.8

Live fetuses/dam

9.8

8.0

10.4

Placental weights

0.43 g

0.41 g (95 %)

0.39 g (91 %)

Fetal weights

3.5 g

3.7 g (104 %)

3.6 g (101 %)

Total external malformations

Fetuses:

Litter:

Affected fetuses/litter:

 

 

0/49 (0.0 %)

0/5 (0.0 %)

0.0 %

 

 

0/40 (0.0 %)

 0/5 (0.0 %)

0.0 %

 

 

0/94 (0.0 %)

0/9 (0.0 %)

0.0 %

Total visceral malformations

Fetuses:

Litter:

Affected fetuses/litter:

 

 

0/24 (0.0 %)

0/5 (0.0 %)

0.0 %

 

 

0/19 (0.0 %)

0/5 (0.0 %)

0.0 %

 

 

0/44 (0.0 %)

0/9 (0.0 %)

0.0 %

Total skeletal malformations

Fetuses:

Litter:

Affected fetuses/litter:

 

 

0/25 (0.0 %)

0/5 (0.0 %)

0.0 %

 

 

0/21(0.0 %)

0/5 (0.0 %)

0.0 %

 

 

0/50(0.0 %)

0/9 (0.0 %)

0.0 %

Cesarean section: Pathology (Dams)

Test group

(mg/kg bw/d)

0 (0; control)

1 (300)

2 (600)

Incidence of gross lesions

NAD

Forestomach: Erosion/Ulcer (4/5)

Forestomach: Erosion/Ulcer (8/10)

Liver weights (absolute)

10.886 g

11.106 g (102 %)

12.203 g (112 %)

Liver weights (relative)

4.761 g

5.018 g (105 %)

5.632 g (118 %)*

Conclusions:
In conclusion, signs of maternal toxicity occurred at 300 mg/kg bw/day, comprising local effects (strong ulcera in the stomach) and at 600 mg/kg bw/day, as evident by systemic effects. In the postnatal part of the study, in the dose group of 600 mg/kg bw/day the live birth index was reduced (91 %  vs. control 100 %). Pups of the high dose group were less viable compared to the control group (viability index 43 % and 100 %, respectively); and reduced body weights were recorded.
No substance related findings were observed in foetuses.

Based on these observations the following effect levels were determined:
Dams (F0):
- LOAEL for local effects: 300 mg/kg bw/day
- NOAEL for systemic effects: 300 mg/kg bw/day
- LOAEL for reproductive performance: 600 mg/kg bw/day
Offspring (F1)
- NOAEL for foetuses: not determinable; no effects observed
- NOAEL for pups: 300 mg/kg bw/day
Executive summary:

In the Pre- and Postnatal Developmental Toxicity Screening test the test compound 2-Dimethylaminoethanol was administered via oral gavage to time-mated Wistar rats from GD 6 through GD 19 (prenatal study part) and from GD 6 through PND 3 (postnatal study part). The following concentrations were administered: i.e. 0 mg/kg bw/day (test group 0, 10 animals), 300 mg/kg bw/day (test group 1, 10 animals), 600 mg/kg bw/day (test group 2, 20 animals). The duration of treatment covered a 2-weeks in gestation and up to the 3rd day after parturition up to the day of scheduled sacrifice of the animals.

Animals dosed with the high dose of 600 mg/kg bw/day showed after treatment the following symptoms: salivation (20/20); respiratory sounds (7/20), statistically significantly reduced mean food consumption (i.e. between GD 6-8 (81 %) and GD 8-10 (78 %) compared to control group (100 %)), statistically significantly reduced mean body weight (i.e. on GD 13 (93 %) compared to the control group (100 %), statistically significantly reduced mean body weight change (i.e. between GD 8-10 (52 %) compared to the control group (100 %). Moreover, one animal was found dead on GD20 (gross pathological examination revealed stomach ulcerations) and one animal was sacrificed moribund on GD14 (gross pathological examination revealed stomach erosions and no feces in intestine).

For the prenatal study part, 10 dams were sacrificed on GD 20 and subsequently examined. In the dams the following observation were made: stomach erosions/ulcera (8 /10) and statistically significantly increased mean liver weight (118 %) compared to the control group (100 %). The following effects on reproduction performance were observed, but assessed statistically not significant and without a dose-response relationship: Slightly increased post-implantation loss (11.8 % vs. 5.2 % in control (compared to the control group and the historical control data)).

For the postnatal part, the remaining 8 animals were sacrificed on PND 4 and subsequently examined. In these dams the following observations were made: stomach erosions/ulcera (8/8), 1/8 dams did not deliver (7 animals left for further assessment), salivation after treatment (7/7), statistically significantly reduced mean food consumption (81 % between PND 0 – 4) compared to the control group (100 %) and a live birth index of 91 % compared to the control group (100 %). A slightly increased post-implantation loss (9.9 % vs. 2.0% in control) was noted, but assessed as statistically not significant.

Animals dosed with the low dose of 300 mg/kg bw/day showed after treatment the following symptoms: salivation (10/10).

For the prenatal study part, 5 dams were sacrificed on GD 20 and subsequently examined, in 4/5 dams stomach erosions/ulcera was recorded. The following effects on reproduction performance were observed, but assessed statistically not significant and without a dose-response relationship: Slightly increased post-implantation loss(15.2 % vs. 5.2 % in control (compared to the control croup and the historical control data)) and slightly increased resorptions (mean/litter) (1.4% vs. 0.6% in control (compared to the control group and the historical control data)).

For the postnatal part, the remaining 5 animals were sacrificed on PND 4 and subsequently examined. In these dams the following local effects were observed: stomach erosions/ulcera (5/5).A slightly increased post-implantation loss (5.5 % vs. 2.0% in control) was noted, but assessed as statistically not significant.

Concerning the effects and observations on foetuses of dams treated with 600 mg/kg bw/day, no test-substances-related findings were reported (prenatal part).

For thepostnatal study part,pups derived of dams dosed with 600 mg/kg bw/day, thefollowing is reported (as1/8 dams did not deliver, only 7 litters left for further assessment):six stillborn pups in 7 litters (64 pups in toto, 58 liveborn), 24/58 pups died ahead of schedule, 9/58 pups were cannibalized, no more pups alive in 4 out of 7 litters (2 litters on PND 1, 1 on PND 2, 1 on PND 3), viability index of 43 % (control: 100%), statistically significantly reduced mean body weight (i.e. on PND 1 (76 %) and on PND 4 (71 %) compared to the control group (100 %)), statistically significantly reduced mean body weight change ((57 % between PND 1-4) compared to the control group (100 %)) and 12 runts (no runts in the control).

In conclusion, signs of maternal toxicity occurred at 300 mg/kg bw/day, comprising local effects (strong ulcera in the stomach) and at 600 mg/kg bw/day, as evident by systemic effects. In the postnatal part of the study, in the dose group of 600 mg/kg bw/day the live birth index was reduced (91 %  vs. control 100 %). Pups of the high dose group were less viable compared to the control group (viability index 43 % and 100 %, respectively); and reduced body weights were recorded.

No substance related findings were observed in foetuses.

Based on these observations the following effect levels were determined:

Dams (F0):

- LOAEL for local effects: 300 mg/kg bw/day

- NOAEL for systemic effects: 300 mg/kg bw/day

- LOAEL for reproductive performance: 600 mg/kg bw/day

Offspring (F1)

- NOAEL for foetuses: not determinable; no effects observed

- NOAEL for pups: 300 mg/kg bw/day

Endpoint:
developmental toxicity
Remarks:
Prenatal Developmental Toxicity Study
Type of information:
experimental study
Adequacy of study:
key study
Study period:
30 JAN 2020 to 23 MAR 2021
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Version / remarks:
June 2018
Deviations:
no
Qualifier:
according to guideline
Guideline:
EPA OPPTS 870.3700 (Prenatal Developmental Toxicity Study)
Version / remarks:
August 1998
Deviations:
no
GLP compliance:
yes
Limit test:
no
Specific details on test material used for the study:
- Physical Description: Clear, colorless liquid
- Storage Conditions: 18 °C to 24 °C, under nitrogen
- Density: 0.89 g/mL
Species:
rabbit
Strain:
New Zealand White
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Envigo, Inc., Denver, PA
- Age at study initiation: approximately 6 to 7 months old
- Weight at study initiation: 2964 and 3924 g
- Housing: Single/Individual in stainless steel cages with perforated flooring elevated above ground corncob bedding
- Diet (e.g. ad libitum): ad libitum
- Water (e.g. ad libitum): ad libitum
- Acclimation period: yes

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 16 °C to 22 °C
- Humidity (%): 30 % to 70 %
- Photoperiod (hrs dark / hrs light): 12 hours light and 12 hours dark.

IN-LIFE DATES: From: 30 Jan 2020 To: 11 Mar 2021
Route of administration:
oral: gavage
Vehicle:
water
Remarks:
Deionized (DI) water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
Dose formulations were divided into aliquots where required to allow them to be dispensed on each dosing occasion. Dosing formulations were prepared at appropriate concentrations to meet dose level requirements. The dosing formulations were stirred continuously during dosing.
Frequency of Preparation: At least weekly

VEHICLE
- Concentration in vehicle: 0, 6, 20, 50 mg/mL
- Amount of vehicle (if gavage): 5 mL/kg
- Purity: Not corrected for salt, purity, and water content.
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Analyses described below were performed by a gas chromatography method using flame ionization detection using a validated analytical procedure.
Concentration Analysis:
Sample Allocation: 2 x 1 mL (primary samples), 2 x 1 mL (backup samples)
Storage Conditions: Temperature set to maintain a target of 5 °C
Acceptance Criteria: Mean sample concentration within 100 % ± 10 % of theoretical concentration.

Stability Analysis: Test substance formulations have been previously shown to be stable over the range of concentrations used on this study for at least 24 hours of room temperature storage or 10 days of refrigerated (target of 5 °C) storage.
Details on mating procedure:
- Impregnation procedure: time-mated
Duration of treatment / exposure:
during Gestation Days 7 through 28
Frequency of treatment:
single daily oral gavage
Duration of test:
Gestation Days 5 through 29
Dose / conc.:
0 mg/kg bw/day
Remarks:
Control
Dose / conc.:
30 mg/kg bw/day
Dose / conc.:
100 mg/kg bw/day
Dose / conc.:
250 mg/kg bw/day
No. of animals per sex per dose:
25 females/control and 24 females/dose group (to obtain a sample size of 20 females/group at termination)
Control animals:
yes, concurrent vehicle
Details on study design:
Dose selection rationale: Based on previous studies (7-day tolerability study and dose range-finding study).
- In a previous 7-day tolerability study, 2-Dimethylaminoethanol was administered once daily by oral gavage to 4 groups of nonpregnant female rabbits at dosage levels of 35, 75, 150, and 300 mg/kg/day. Lower food consumption was noted at the high-dose group (300 mg/kg/day) generally throughout the study, but absolute body weights were unaffected across groups (approximately 2.5 %). At necropsy, no remarkable macroscopic findings were observed at any dosage level. Following microscopic examination of the stomachs from animals in the 150 and 300 mg/kg/day groups, gastric mucosal hemorrhage associated with increased mitoses and sometimes with minimal erosion and epithelial regeneration was detected at 300 mg/kg/day; no microscopic findings were noted in the stomach at 150 mg/kg/day.

- In a previous dose range-finding prenatal developmental toxicity study, 2-Dimethylaminoethanol was administered once daily by oral gavage to 3 groups of time-mated female rabbits at dosage levels
of 50, 150, and 450 mg/kg/day during Gestation Days 7–28. Severe body weight loss and reduced food consumption, with corresponding clinical observations, resulted in moribundity and abortion at 450 mg/kg/day. In addition, macroscopic (dark red areas, depressed areas, and thickening of the stomach) and microscopic (necrosis and/or heterophilic infiltration with or without congestion) findings were noted in the stomach at 150 and 450 mg/kg/day. Based on these results (body weight loss, reduced food consumption, moribundity and abortion at 450 mg/kg/day and microscopic findings noted at 300 and 450 mg/kg/day), dosage levels of 30, 100, and 250 mg/kg/day were selected for the current study.
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: At least twice daily
- Cage side observations checked in table [No.?] were included.

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: Once daily, beginning with the day of animal arrival and continuing through (and including) the day of euthanasia.

BODY WEIGHT: Yes
- Time schedule for examinations: Gestation Days 0 (by supplier), 5, 7–29 (daily)

FOOD CONSUMPTION AND COMPOUND INTAKE: Yes
- Gestation Days 5–29 (daily)
- Reported as g/animal/day for each corresponding body weight interval during gestation.

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 29
- Organs examined:
Histopathology: Stomach, gross lesions;
Macroscopic Examination: The thoracic, abdominal, and pelvic cavities
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
- Other:
- Number of viable and nonviable fetuses
- The placentae were examined
Fetal examinations:
- External examinations: Yes [all]
- Soft tissue examinations: Yes [all]
- Skeletal examinations: Yes [each eviscerated fetus]
- Head examinations: Yes: [all ]
Statistics:
Numerical data and clinical and necropsy observations data were summarized by sex and occasion or by litter.
All statistical tests were conducted at the 5 % significance level. All pairwise comparisons were conducted using two sided tests and were reported at the 1 % and 5 % levels, unless otherwise noted.
The pairwise comparisons of interest are listed below:
Group 2 vs. Group 1
Group 3 vs. Group 1
Group 4 vs. Group 1

Parametric/Non-parametric:
Variables: Ovarian and Uterine Content, Litter % of Fetuses with Gross/External/Visceral/Skeletal Abnormalities
Levene’s test was used to assess the homogeneity of group variances.
The groups were compared using an overall one-way ANOVA F-test if Levene’s test was not significant or the Kruskal-Wallis test if it was significant. If the overall F-test or Kruskal-Wallis test was found to be significant, then pairwise comparisons were conducted using Dunnett’s or Dunn’s test, respectively.

Non-Parametric:
Variables: Body Weight, Body Weight Gains, Food Consumption, Gravid Uterine Weight and Corrected Maternal Body Weights, Litter Means
The groups were compared using an overall Kruskal-Wallis test. If the overall Kruskal-Wallis test was found to be significant, then the above pairwise comparisons were conducted using Dunn’s test.

Incidence:
A Fisher’s exact test was used to conduct pairwise group comparisons of interest
Indices:
No information given
Historical control data:
yes
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
In the 250 mg/kg/day group, 6 females (Nos. 4504, 4507, 4512, 4601, 4513, 4517) were noted with clinical observations (a thin body and/or excreta-related findings) noted at the daily examinations prior to death, abortion, or delivery.
Test substance-related increased incidences of decreased fecal output, which corresponded to reduced mean food consumption, were noted for surviving females in the 250 mg/kg/day group at the daily examinations.
No test substance-related clinical observations were noted at the daily examinations at 30 and 100 mg/kg/day or 1–2 hours postdosing at any dosage level.
Other observations noted in the test substance-treated groups, including scabbing and brown, yellow, or red staining on various body surfaces, occurred infrequently, at similar frequencies in the control group, and/or in a manner that was not dose-related.
Dermal irritation (if dermal study):
not examined
Mortality:
mortality observed, treatment-related
Description (incidence):
In the 250 mg/kg/day group, 1 female (No. 4513) was found dead on Gestation Day 23. The unscheduled death was considered secondary to the test substance-related effects on body weight and food consumption in this group.
In the 30 mg/kg/day group, Female No. 2501 was found dead on Gestation Day 25; at necropsy this female was noted with a perforated esophagus and fluid accumulation in the thoracic cavity, which were indicative of a dosing error. Therefore, this death was not attributed to the test substance.
In the control group, Female No. 1501 was euthanized on Gestation Day 8 due limited usage of a hindlimb, which was confirmed to be a tibial fracture at necropsy. All other females in the control, 30, 100, and 250 mg/kg/day groups survived to the scheduled necropsy on Gestation Day 29.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
In the 250 mg/kg/day group, 6 females (Nos. 4504, 4507, 4512, 4601, 4513, 4517) were noted with severe body weight losses (11.4 % to 21.7 % of Gestation Day 7 body weight).
Test substance-related mean body weight losses and lower mean body weight gains, with corresponding lower mean food consumption, were noted in the 250 mg/kg/day group throughout the treatment period (Gestation Days 7–29) when compared to the control group. As a result, absolute mean body weights in this group were 5.0 % to 11.2 % lower than the control group during Gestation Days 12–29.
In addition, lower mean corrected body weight, corrected mean body weight gain, and mean gravid uterine weight were noted in the 250 mg/kg/day group compared to the control group. Mean maternal body weights, body weight gains, corrected body weights, corrected body weight changes, gravid uterine weights, and food consumption in the 30 and 100 mg/kg/day groups were unaffected by test substance administration.

Mean body weights, body weight gains, corrected body weights, corrected body weight changes, and gravid uterine weights in the 30 and 100 mg/kg/day groups were unaffected by test substance administration. Differences from the control group were slight and not statistically significant.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
In the 250 mg/kg/day group, 6 females (Nos. 4504, 4507, 4512, 4601, 4513, 4517) were noted with reduced food consumption (0 to 86 g/day for 6 to 21 days).
In the 250 mg/kg/day group, lower mean food consumption, evaluated as g/animal/day, was noted throughout the treatment period (Gestation Days 7–10 [-50.7 %], 10–13 [54.5 %], 13–20 [54.9 %], and 20–29 [34.8 %]) and when the entire treatment period (Gestation Days 7–29; 42.5 %) was evaluated; differences from the control group were statistically significant.
The decrements in mean food consumption in this group corresponded to the mean body weight losses/lower mean body weight gains, and were considered test substance-related and adverse.
Mean maternal food consumption in the 30 and 100 mg/kg/day groups was unaffected by test substance administration. Any statistically significant differences from the control group did not impact mean body weights or body weights gains, and therefore were considered unrelated to the test substance.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not specified
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not specified
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
not examined
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
Test substance-related gross pathology findings were limited to the stomach of the 250 mg/kg/day group females and are summarized 'Any other information on results'.
A single dark red focus of the pyloric region was observed for 1 female in the 30 mg/kg/day group. This finding did not have a clear dose-response, lacked a histologic correlate, and was considered unlikely to be test substance-related.
Other gross findings observed were considered incidental, of the nature commonly observed in this strain and age of rabbits, and/or were of similar incidence in control and treated animals and, therefore, were considered unrelated to the test item.
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
effects observed, treatment-related
Description (incidence and severity):
Microscopic findings in the stomach were observed in all test substance-administered groups and included hemorrhage, necrosis, edema, fibrosis, and heterophilic infiltrates. Based on the absence of other signs of toxicity, the microscopic findings in the 30 and 100 mg/kg/day groups were considered nonadverse. Fore more details please refer to 'Details on results'
Histopathological findings: neoplastic:
not examined
Other effects:
effects observed, treatment-related
Description (incidence and severity):
Test substance-related lower mean fetal body weights noted in 250 mg/kg bw/day resulted in a lower mean gravid uterine weight in this group.
Details on results:
Histopathology: Test substance-related changes in the stomach noted at terminal euthanasia in all dose groups were similar in character to those observed in the 250 mg/kg/day group females that did not survive to terminal euthanasia, aborted, or delivered, although a clear dose-response was not apparent. Hemorrhage ranged from minimal, which was observed primarily as small foci in the glandular mucosa, to moderate, extending from the mucosa and dissecting through the muscularis. Minimal to mild necrosis was noted in all dose groups and was characterized by foci of necrotic epithelium along the mucosa of the glandular stomach and rarely in the cardia. Necrosis and hemorrhage were variably accompanied by a minimal to moderate infiltrate of heterophils and edema. Fibrosis along the superficial muscularis was rarely observed but was considered likely secondary to hemorrhage and necrosis. Based on the absence of other signs of toxicity, the microscopic findings in the 30 and 100 mg/kg/day groups were considered nonadverse.
Number of abortions:
effects observed, treatment-related
Description (incidence and severity):
In the 250 mg/kg/day group, 4 females (Nos. 4504, 4507, 4512, and 4601) aborted during Gestation Days 20–28
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
Mean litter proportions of postimplantation loss, mean number of live fetuses, and mean fetal body weights were evaluated. Differences from the control group were slight, not statistically significant, and/or occurred in a manner that was not dose related.
Total litter losses by resorption:
not examined
Early or late resorptions:
not specified
Dead fetuses:
no effects observed
Description (incidence and severity):
Intrauterine survival in the 30, 100, and 250 mg/kg/day groups were unaffected by test substance administration.
Changes in pregnancy duration:
effects observed, treatment-related
Description (incidence and severity):
In the 250 mg/kg/day group, 1 female delivered on Gestation Day 29, prior to the scheduled necropsy.
Changes in number of pregnant:
not specified
Other effects:
no effects observed
Description (incidence and severity):
Mean numbers of corpora lutea and implantation sites and the mean litter proportions of pre-implantation loss were similar across all groups.
Details on maternal toxic effects:
Adverse effects on maternal survival, maternal body weight losses and/or lower body weight gain, reduced food consumption, and corresponding clinical observations at 250 mg/kg/day
Dose descriptor:
NOAEL
Remarks:
maternal toxicity
Effect level:
100 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
clinical signs
food consumption and compound intake
mortality
Fetal body weight changes:
effects observed, non-treatment-related
Description (incidence and severity):
Test substance-related lower (12.16 % to 13.83 %) mean fetal body weights (male, female, and sexes combined) were noted in the 250 mg/kg/day group. However, the mean fetal weight values in this group were within the respective ranges of historical control data and the differences were considered secondary to the maternal toxicity noted at this dosage level, and nonadverse data.
Intrauterine growth in the 30 and 100 mg/kg/day groups was unaffected by test substance administration.
Reduction in number of live offspring:
no effects observed
Description (incidence and severity):
Mean litter proportions of postimplantation loss, mean number of live fetuses, and mean fetal body weights were evaluated. Differences from the control group were slight, not statistically significant, and/or occurred in a manner that was not dose related.
Changes in sex ratio:
not specified
Changes in litter size and weights:
no effects observed
Description (incidence and severity):
Mean litter proportions of postimplantation loss, mean number of live fetuses, and mean fetal body weights were evaluated. Differences from the control group were slight, not statistically significant, and/or occurred in a manner that was not dose related.
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
There were no test substance-related fetal malformations or developmental variations noted at any dosage level.
Skeletal malformations:
no effects observed
Description (incidence and severity):
There were no test substance-related fetal malformations or developmental variations noted at any dosage level.
Visceral malformations:
no effects observed
Description (incidence and severity):
There were no test substance-related fetal malformations or developmental variations noted at any dosage level.
Other effects:
not specified
Dose descriptor:
NOAEL
Remarks:
Prenatal development
Effect level:
250 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: General developmental parameters
Remarks on result:
other: No substance-related effects observed
Abnormalities:
no effects observed
Developmental effects observed:
no

Results of Concentration Analyses

Mean Concentration, mg/mL (% of Target)

Date of Preparation

Group 2

(6 mg/mL)

Group 3

(20 mg/mL)

Group 4

50 mg/mL)

31 Jan 2020

6.30 (105)

22.0 (110)

55.2 (110)*

20 Feb 2020

6.56 (109)

22.2 (111)*

55.0 (110)

* = Back-up samples included in calculations.

Summary of Gross Pathology Findings – All Animals

Group

1

2

3

4

Dose (mg/kg/day)

0

30

100

250

No. Animals per Group

24

24

24

24

Stomach (No. Examined)

(24)

(24)

(24)

(24)

Focus, dark, red

0

1

0

7

Thick

0

0

0

3

Focus, raised

0

0

0

3

Discoloration, dark, red

0

0

0

2

Conclusions:
In conclusion, based on adverse effects on maternal survival, maternal body weight losses and/or lower body weight gain, reduced food consumption, and corresponding clinical observations at 250 mg/kg/day, considered secondary to local corrosion of the gastric mucosa, a dosage level of 100 mg/kg/day was considered to be the NOAEL for maternal toxicity. Lower mean fetal body weights observed at 250 mg/kg/day were considered secondary to the adverse maternal effects; therefore, a dosage level of 250 mg/kg/day was considered to be the NOAEL for prenatal development.
Executive summary:

The Prenatal Developmental Toxicity Study was conduced according to OECD 414 and in accordance with GLP.

2-Dimethylaminoethanol was administered orally by gavage to time-mated New Zealand White rabbits at doses of 0, 30, 100 and 250 mg/kg bw/day; animals were dosed once daily during Gestation Days 7–28. The following parameters and end points were evaluated in this study: clinical signs, body weights, body weight gains, gravid uterine weights, food consumption, gross necropsy findings, stomach histopathology, intrauterine growth and survival, and fetal morphology.

In the 250 mg/kg/day group, 1 female was found dead on Gestation Day 23, 4 females aborted during Gestation Days 20–28, and 1 female delivered on Gestation Day 29, prior to the scheduled necropsy. All 6 of these females were noted with severe body weight losses (11.4 % to 21.7 %), reduced food consumption (0 to 86 g/day), and corresponding clinical observations (a thin body and/or excreta-related findings) noted at the daily examinations prior to death, abortion, or delivery. At necropsy, macroscopic findings including dark red discoloration, dark red foci, thickened, and raised foci in the stomach with microscopic correlates (hemorrhage, necrosis, edema, fibrosis, and heterophilic infiltrates) were noted for the majority of these females. The effects on survival to the scheduled necropsy in the 250 mg/kg/day group were considered secondary to the test substance-related effects on body weight and food consumption in this group. One female in the 30 mg/kg/day group was found dead on Gestation Day 25; necropsy findings for this female were indicative of a dosing error, and therefore this death was not attributed to the test substance. In the control group, 1 female was euthanized in extremis on Gestation Day 8 due to limited usage of a hindlimb (tibial fracture). All other females survived to the scheduled necropsy on Gestation Day 29. Test substance-related increased incidences of decreased fecal output, which corresponded to reduced mean food consumption, were noted for surviving females in the 250 mg/kg/day group at the daily examinations. No test substance-related clinical observations were noted at the daily examinations at 30 and 100 mg/kg/day or 1–2 hours postdosing at any dosage level.

Test substance-related mean body weight losses and lower mean body weight gains, with corresponding lower mean food consumption, were noted in the 250 mg/kg/day group throughout the treatment period (Gestation Days 7–29) when compared to the control group. As a result, absolute mean body weights in this group were 5.0 % to 11.2 % lower than the control group during Gestation Days 12–29. In addition, lower mean corrected body weight, corrected mean body weight gain, and mean gravid uterine weight were noted in the 250 mg/kg/day group compared to the control group. Mean maternal body weights, body weight gains, corrected body weights, corrected body weight changes, gravid uterine weights, and food consumption in the 30 and 100 mg/kg/day groups were unaffected by test substance administration. Test substance-related gross observations were observed in the stomach of the 250 mg/kg/day group females at the scheduled necropsy, including dark red discoloration, dark red foci, thickened, and raised foci; comparable findings were also noted for females that did not survive to the scheduled necropsy. Microscopic findings in the stomach were observed in all test substance-administered groups and included hemorrhage, necrosis, edema, fibrosis, and heterophilic infiltrates. Based on the absence of other signs of toxicity, the microscopic findings in the 30 and 100 mg/kg/day groups were considered nonadverse.

Test substance-related lower (12.16 % to 13.83 %) mean fetal body weights (male, female, and sexes combined) were noted in the 250 mg/kg/day group, which resulted in a lower mean gravid uterine weight in this group. However, the mean fetal weight values in this group were within the respective ranges of values noted in historical control data, and the differences were considered secondary to the maternal toxicity noted at this dosage level, and nonadverse. Intrauterine growth in the 30 and 100 mg/kg/day groups and intrauterine survival in the 30, 100, and 250 mg/kg/day groups were unaffected by test substance administration. There were no test substance-related fetal malformations or developmental variations noted at any dosage level.

In conclusion, based on adverse effects on maternal survival, maternal body weight losses and/or lower body weight gain, reduced food consumption, and corresponding clinical observations at 250 mg/kg/day, considered secondary to local corrosion of the gastric mucosa, a dosage level of 100 mg/kg/day was considered to be the NOAEL for maternal toxicity. Lower mean fetal body weights observed at 250 mg/kg/day were considered secondary to the adverse maternal effects; therefore, a dosage level of 250 mg/kg/day was considered to be the NOAEL for prenatal development.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
key study
Study period:
not reported, published in 1996
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
other: Acceptable, well documented publication which meets basic scientific principles.
Reason / purpose for cross-reference:
reference to same study
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Deviations:
no
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Fischer 344
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Harlan Sprague-Dawley, Inc., Indianapolis, IN.
- Age at study initiation: 67-79 days old on arrival
- Weight at study initiation: not reported
- Fasting period before study: no
- Housing: in stainless-steel wire-meshcages
- Diet (e.g. ad libitum): ad libitum except during exposure
- Water (e.g. ad libitum): ad libitum except during exposure
- Acclimation period: 2 weeks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): not reported
- Humidity (%): not reported
- Air changes (per hr): not reported
- Photoperiod (hrs dark / hrs light):12/12
Route of administration:
inhalation: vapour
Type of inhalation exposure (if applicable):
whole body
Vehicle:
unchanged (no vehicle)
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
Liquid DMEA was metered from a piston pump into a heated glass maintained at the lowest temperature to vaporize the liquid. The resultant vapor was carried into the exposure chamber by a countercurrent flow of conditioned air through the evaporator. Exposure was conducted in 4320-litre stainless-steel and glass chambers at an airflow of 1000 L/min.

Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Chamber atmosphere was analyzed for DMEA concentrations once every 32 min during each 6-h exposure, using a Perkin-Elmer 3920B gas chromatograph equipped with a flame ionization detector. Nominal concentrations were calculated daily based on the amount of DMEA used and the chamber tube air flow during the exposure period.
Details on mating procedure:
- Impregnation procedure:[cohoused]
- If cohoused:
- M/F ratio per cage: 1/1
- Length of cohabitation: not reported

- Further matings after two unsuccessful attempts: [no]
- Verification of same strain and source of both sexes: [yes / no (explain)]
- Proof of pregnancy: [vaginal plug] referred to as [day 0] of pregnancy
- Any other deviations from standard protocol:
Duration of treatment / exposure:
6 h per day
Frequency of treatment:
each day
Duration of test:
on gestational days 6-15
Dose / conc.:
0 ppm (nominal)
Dose / conc.:
10 ppm (nominal)
Dose / conc.:
30 ppm (nominal)
Dose / conc.:
100 ppm (nominal)
No. of animals per sex per dose:
In a range-finding study, eight plug-positive females each were assigned to four DMEA-exposed groups (target DMEA concentrations 8, 25, 75 and 100 ppm) and an air-exposed control group.
In the definitive study, 25 plug-positive females each were assigned to three DMEA-exposed groups (10, 30 and 100 ppm) and a control group.
Control animals:
yes, sham-exposed
Details on study design:
- Dose selection rationale: Based on the results of the range-finding study.
The highest exposure concentration in the range-finding study, 100 ppm, was retained in the definitive study since it produced maternal toxicity (reduced body weights and weight gain, and clinical signs) and possible embryotoxicity (increased preimplantation loss) but no apparent fetotoxicity. The middle exposure concentration of 30 ppm chosen for the definitive study was slightly above the 25 ppm in the range-finding study which produced maternal toxicity (transient weight gain depression and clinical signs limited to the eyes) and possible embryotoxicity (reduced implantations, increased preimplantation loss and reduced number of viable fetuses per litter). The lowest exposure concentration, 10 ppm, was chosen as essentially the same as the 8 ppm in the range-finding study which produced no effects on maternal weights and only transient ocular changes and no evidence of embryofetal toxicity.
- Rationale for animal assignment (if not random): randomized
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes - Time schedule: daily
DETAILED CLINICAL OBSERVATIONS: Yes - Time schedule: daily
BODY WEIGHT: Yes - Time schedule for examinations:Maternal body weights were measured on gd 0, 6, 12, 15, 18 and 21.
POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 21
- Organs examined: The gravid uterus, ovaries (including corpora lutea), cervix, vagina and peritoneal and thoracic cavities were examined grossly. Ovarian corpora lutea of pregnancy were counted. Maternal liver and uterine weights were measured.
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
- Other: Uteri were examined externally for signs of hemorrhage. All live and dead fetuses were recorded-
Fetal examinations:
- External examinations: Yes: [all per litter] including cleft palate
- Soft tissue examinations: Yes: [half per litter]
- Skeletal examinations: Yes: [half per litter]
- Head examinations: Yes: [half per litter]
Statistics:
The unit of comparison was the pregnant female or the litter. Continuous quantitative data were compared between the DMEA-exposed groups and air-exposed control group by the use of Levene's test for equal variances analysis of variance (ANOVA) and t-tests with Bonferroni probabilities. The t-tests were used when the F value from the ANOVA was significant. When Levene's test indicated homogeneous variances, and the ANOVA was significant, the pooled t-test was used. When Levene's test indicated heterogeneous variances, all groups were compared by an ANOVA for unequal variances," followed by the separate variance t-test when necessary. Non-parametric data obtained following laparohysterectomy were statistically treated using the Kruskal- Wallis test followed by the Mann-Whitney U test when appropriate. Incidence data were compared using Fisher's exact test. For all statistical tests, the fiducial limit of 0.05 (two-tailed) was used as the criterion for significance.
Indices:
listed in the table 4 in "Remarks on results"
Historical control data:
no information given
Clinical signs:
effects observed, treatment-related
Description (incidence and severity):
Clinical examination showed that dams at 100 ppm only exhibited perinasal fur discoloration, presumably from chromodacryorrhea. At 30 and 100 ppm there were darkened (maroon), cloudy and hazy eyes, slight corneal vascularization and pupils dilated and fixed. Cloudy and hazy eyes were observed only during the exposure period. Darkened eyes were also observed at 10 ppm during the exposure period.
Dermal irritation (if dermal study):
not examined
Mortality:
no mortality observed
Description (incidence):
There were no maternal deaths or abortions.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
Reduced body weight and reduced body weight gain were observed at 100 ppm. Body weight reduced on gd 12 and 15 (during the exposure period) and on gd 15 and 21 (postexposure period). Body weight gain was reduced for all intervals except gd 6- 9 (pre-exposure) and gd 15-21 (post-exposure). There were no effects on body weight or body weight gain for the 10 or 30 ppm groups.
Food consumption and compound intake (if feeding study):
not examined
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
effects observed, treatment-related
Description (incidence and severity):
At 30 and 100 ppm there were darkened (maroon), cloudy and hazy eyes, slight corneal vascularization and pupils dilated and fixed. Cloudy and hazy eyes were observed only during the exposure period. Darkened eyes were also observed at 10 ppm during the exposure period.
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
There were no statistically significant differences in gravid uterine weight and absolute or relative liver weights between the DMEA-exposed groups and the controls.
Gross pathological findings:
no effects observed
Description (incidence and severity):
There were no effects of treatment on gonads.
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Number of abortions:
no effects observed
Description (incidence and severity):
There were no maternal deaths or abortions.
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
There were no effects of treatment on pre- and post implantation loss.
Total litter losses by resorption:
no effects observed
Description (incidence and severity):
Pregnancy rate ranged from 88 to 96% and all pregnant females had one or more live fetuses at scheduled sacrifice, except one dam at 100 ppm, which had a totally resorbed litter.
Early or late resorptions:
no effects observed
Description (incidence and severity):
Pregnancy rate ranged from 88 to 96% and all pregnant females had one or more live fetuses at scheduled sacrifice, except one dam at 100 ppm, which had a totally resorbed litter.
Dead fetuses:
not specified
Changes in pregnancy duration:
no effects observed
Description (incidence and severity):
There were no effects of treatment on any gestational parameters.
Changes in number of pregnant:
no effects observed
Description (incidence and severity):
Pregnancy rate ranged from 88 to 96% and all pregnant females had one or more live fetuses at scheduled sacrifice, except one dam at 100 ppm, which had a totally resorbed litter.
Other effects:
not examined
Details on maternal toxic effects:
Maternal toxic effects:yes

Details on maternal toxic effects:
Table 2 (in "Remarks on results") shows the pregnancy and litter data of all plug-positive females on study. There were no maternal deaths or abortions. Pregnancy rate ranged from 88 to 96% and all pregnant females had one or more live fetuses at scheduled sacrifice, except one dam at 100 ppm, which had a totally resorbed litter. Reduced body weight and reduced body weight gain were observed at 100 ppm. Body weight reduced on gd 12 and 15 (during the exposure period) and on gd 15 and 21 (postexposure period). Body weight gain was reduced for all intervals except gd 6- 9 (pre-exposure) and gd 15-2 1 (post-exposure). There were no effects on body weight or body weight gain for the 10 or 30 ppm groups. Clinical examination showed that dams at 100 ppm only exhibited perinasal fur discoloration, presumably from chromodacryorrhea. At 30 and 100 ppm there were darkened (maroon), cloudy and hazy eyes, slight corneal vascularization and pupils dilated and fixed. Cloudy and hazy eyes were observed only during the exposure period. Darkened eyes were also observed at 10ppm during the exposure period. There were no statistically significant differences in gravid uterine weight and absolute or relative liver weights between the DMEA-exposed groups and the controls.
Dose descriptor:
NOAEL
Remarks:
general toxicity
Effect level:
10 ppm (nominal)
Based on:
test mat.
Basis for effect level:
body weight and weight gain
clinical signs
ophthalmological examination
Fetal body weight changes:
no effects observed
Description (incidence and severity):
Fetal body weights were elevated at 100 ppm relative to those in controls.
Reduction in number of live offspring:
no effects observed
Description (incidence and severity):
The present developmental toxicity evaluation revealed no treatment-related embryotoxicity at any exposure concentration employed. No consistent pattern of fetotoxicity was observed.
Changes in sex ratio:
no effects observed
Description (incidence and severity):
The present developmental toxicity evaluation revealed no treatment-related embryotoxicity at any exposure concentration employed. No consistent pattern of fetotoxicity was observed. There were no effects of treatment on sex ratio.
Changes in litter size and weights:
no effects observed
Description (incidence and severity):
The present developmental toxicity evaluation revealed no treatment-related embryotoxicity at any exposure concentration employed. No consistent pattern of fetotoxicity was observed.
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
no effects observed
Description (incidence and severity):
The present developmental toxicity evaluation revealed no treatment-related embryotoxicity at any exposure concentration employed. No consistent pattern of fetotoxicity was observed.
External malformations:
no effects observed
Description (incidence and severity):
No increases in malformations were observed at any concentration of DMEA employed, including those which produced maternal toxicity.
Skeletal malformations:
no effects observed
Description (incidence and severity):
One skeletal district, the cervical centra, exhibited evidence of reduced ossification at 100 ppm, an exposure concentration which also produced maternal toxicity. This finding is the only one which could be consistent with indicating possible minimal fetotoxicity; however, there were no other indications of fetotoxicity, such as reduced fetal body weight. No other skeletal districts, of the number identified as sensitive indicators of delayed development in rat fetuses,” exhibited a delay in ossification. No increases in malformations were observed at any concentration of DMEA employed, including those which produced maternal toxicity.
Visceral malformations:
no effects observed
Description (incidence and severity):
No increases in malformations were observed at any concentration of DMEA employed, including those which produced maternal toxicity.
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects:no effects

Details on embryotoxic / teratogenic effects:
The present developmental toxicity evaluation revealed no treatment-related embryotoxicity at any exposure concentration employed. No consistent pattern of fetotoxicity was observed. Fetal body weights were elevated at 100 ppm relative to those in controls (Table 3), and only one skeletal district, the cervical centra, exhibited evidence of reduced ossification at 100 ppm (Table 4), an exposure concentration which also produced maternal toxicity. This finding is the only one which could be consistent with indicating possible minimal fetotoxicity; however, there were no other indications of fetotoxicity, such as reduced fetal body weight. No other skeletal districts, of the number identified as sensitive indicators of delayed development in rat fetuses,” exhibited a delay in ossification. No increases in malformations were observed at any concentration of DMEA employed, including those which produced maternal toxicity.
Dose descriptor:
NOAEL
Effect level:
>= 100 ppm (nominal)
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: see "Remarks"
Abnormalities:
not specified
Developmental effects observed:
not specified

Table 2. Pregnancy and litter data for Fischer 344 rats exposed whole body to N,N-dimethylethanolamine vapor

Exposure concentration (ppm)

0

10

30

100

Number in study

25

25

25

25

Number of early delivery

1

0

0

0

Number aborted

0

0

0

0

Number (%) pregnant at scheduled

22

23

22

24

sacrifice

(91.7)

(92.0)

(88.0)

(96.0)

Number of litters examined

22

23

22

23a

aOne dam carried a totally resorbed litter.

Table 3. Gestational parameters and fetal body weights in Fischer 344 rats exposed whole body to N,N-dimethylethanolamine vapor*

Exposure conc. (ppm)

0

10

30

100

Number of dams

22

23

22

24

Corpora lutea/dam

11.4 ± 1.2

11.1 ± 1.2b

11.3 ± 1.1

11.8 ± 1.2

Total implants/litter

9.6 ± 1.7

7.9 ± 3.0

8.6 ± 3.3

8.5 ± 3.0

Preimplantation loss (%)

15.5 ± 14.6

26.9 ± 25.4b

25.9 ± 26.8

28.1 ± 24.3

Viable implants/litter

9.5 ± 1.7

7.6 ± 3.1*

8.5 ± 3.4

8.0 ± 3.3

Non-viable implants/litter

0.0 ± 0.2

0.3 ± 0.6

0.1 ± 0.4

0.5 ± 1.5

Early resorptions

0.0 ± 0.0

0.2 ± 0.5

0.0 ± 0.2

0.4 ± 1.4

Late resorptions

0.0 ± 0.2

0.1 ± 0.3

0.1 ± 0.3

0.0 ± 0.0

Dead fetuses

0.0 ± 0.0

0.0 ± 0.2

0.0 ± 0.0

0.0 ± 0.2

Live fetuses/litter (%/litter)

99.5 ± 2.4

95.6 ± 9.0*

97.9 ± 6.1

94.4 ± 20.6

Sex ratio (% males)

56.0 ± 16.0

60.0 ± 21.0

41.6 ± 18.4*

49.0 ± 16.9c

Live litter size

9.5 ± 1.7

7.6 ± 3.1*

8.5 ± 3.4

8.4 ± 2.9c

Fetal body weight/litter (g)

 

 

 

 

All fetuses

4.47 ± 0.15

4.56 ± 0.26

4.53 ± 0.24

4.66 ± 0.27c

Male fetuses

4.63 ± 0.16

4.67 ± 0.24

4.61 ± 0.26d

4.82 ± 0.26*c

Female fetuses

4.28 ± 0.13

4.38 ± 0.26e

4.43 ± 0.26

4.52 ± 0.30**c

aValues presented as mean ± standard deviations; *p < 0.05 and **p < 0.01 versus control.
bn = 22 because the corpora lutea count from one dam was inadvertently not recorded.
cn = 23 because one dam carried a totally resorbed litter.
dn = 20 because two litters consisted of only female fetuses.
en = 22 because one litter consisted of only male fetuses.

Table 4. Skeletal variations in the fetuses of Fischer 344 rats exposed whole body to N,N-dimethylethanolamine vapor

 

Fetuses

Litters

Exposure concentration (ppm)

0

10

30

100

0

10

30

100

Number examined skeletally

102

82

89

91

22

23

20

23

Cervical centrum 6 poorly ossified

43

46

48

33

22

21

18

15**

Cervical centra 1, 2, 3 and/or 4 split

3

2

6

13

3

2

5

12*

Thoracic centrum 1 bilobed

8

14

15

11

6

14*

12

10

Thoracic centrum 9 bilobed

14

5

6

6

12

5*

6

6

Some proximal phalanges (forelimb) unossified

7

0

5

5

6

0*

5

5

Sternebra 5 bilobed

16

10

6

14

12

5*

5

11

*p < 0.05 and **p < 0.01 versus control.

Conclusions:
In summary, whole-body exposure to DMEA vapor of timed-pregnant Fischer 344 rats during organogenesis at 0, 10, 30 or 100 ppm resulted in maternal toxicity at 30 and 100 ppm (with transient minor ocular changes at 10 ppm). There was no evidence of embryonic or fetal toxicity, including teratogenicity, at any exposure concentration employed. Therefore, the no-observed-adverse-effect level (NOAEL) is around 10 ppm for maternal toxicity and ≥ 100 ppm for embryofetal toxicity and teratogenicity in this study.
Executive summary:

The study was conducted similar to the OECD TG 414. In the study timed-pregnant Fischer 344 rats were exposed whole body to N,N-dimethylethanolamine vapor for 6 h per day on gestational days 6-15 at mean (±SD) analytically measured concentrations of 10.4 ± 0.86, 29.8 ± 2.14 and 100 ± 4.9 ppm. Dams were sacrificed on gestational day 21. There was no maternal mortality in any exposed groups. Maternal toxicity observed in the 100 ppm group included reduced body weight during and after exposures, reduced weight gain during exposure and ocular changes (darkened, cloudy and hazy eyes, slight corneal vascularization and fixed, dilated pupils). Ocular effects were also noted in the other two exposure groups; the effects were quite marked at 30 ppm but only minimal and transient at 10 ppm. There were no effects of treatment on any gestational parameters, including pre- and postimplantation loss or sex ratio. Fetal body weights per litter were statistically significantly increased at 100 ppm relative to controls. There were no increases in the incidences of total malformations by category (external, visceral or skeletal) or individually. The incidence of six skeletal variations out of 120 noted differed in exposed groups relative to that of control. Four of these variations were decreases in incidence; only one fetal variation, the split (bipartite) cervical centrum, was elevated at 100 ppm relative to controls. In the absence of any other indications of delayed ossification or fetal body weights, the observed fetal variation does not suggest a consistent pattern of fetal toxicity. Hence, the no-observed-adverse-effect level (NOAEL) is around 10 ppm for maternal toxicity and at or above 100 ppm for embryofetal toxicity and teratogenicity.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
12 OCT 2009 to 29 OCT 2010
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
other: OECD Guideline 421 (Reproduction / Developmental Toxicity Screening Test)
Qualifier:
according to guideline
Guideline:
other: EPA OPPTS 870.3550 (Reproduction/Developmental Toxicity Screening Test)
GLP compliance:
yes (incl. QA statement)
Limit test:
no
Species:
rat
Strain:
Wistar
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Research Models and Services, Germany GmbH (males) and Charles River Laboratories, France (females)
- Females (if applicable) nulliparous and non-pregnant: yes
- Age at study initiation: 11 - 13 weeks
- Weight at study initiation: male animals: 279.9 g - 315.9 g, female animals: 173.2 g - 202.8 g
- Housing:
- During the study period, the rats were housed individually
- During overnight matings, male and female mating partners were housed together
- Pregnant animals and their litters were housed together until PND 4.
- Diet: ad libitum
- Water: ad libitum
- Acclimation period: 5-6 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Air changes (per hr): 15
- Photoperiod (hrs dark / hrs light): 12/12

IN-LIFE DATES: From: 15 Dec 2009 To: 19 Aug 2010
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
The test substance solutions in drinking water were prepared at the beginning of the administration period and thereafter in intervals, which took into account the analytical results of the stability verification. The maximum period for which each preparation was used was 7 days.

For the preparation of the administration solutions the test substance was weighed in a graduated measuring flask depending on the dose group, topped up with drinking water and subsequently thoroughly shaken until completely dissolved.


VEHICLE
- Concentration in vehicle: 0, 1, 3, 10 g/100mL
- Amount of vehicle (if gavage): 10 mL/kg bw
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Concentration control analyses of the test substance preparations:
The method of analysis was Capilary electrophoresis (CE). All measured values for N-Methyldiethanolamin were in the expected range of the target concentrations (90-110%).
Details on mating procedure:
Males and females from the same dose group were mated 13 days after the beginning of treatment, overnight in a ratio of 1:1.
- M/F ratio per cage: 1:1
- Length of cohabitation: 2 weeks
- Proof of pregnancy: sperm in vaginal smear referred to as day 0 of pregnancy
Duration of treatment / exposure:
The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females.
Frequency of treatment:
once daily
Duration of test:
The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females. The females were allowed to deliver and rear their pups until day 4 after parturition. Four days after PND 4 of the female, which delivered last, all parental females were sacrificed and examined. Pups were sacrificed on PND 4 and gross necropsied. The male animals were sacrificed 28 days after the beginning of the administration and examined.
Dose / conc.:
0 mg/kg bw/day
Remarks:
Control
Dose / conc.:
100 mg/kg bw/day
Dose / conc.:
300 mg/kg bw/day
Dose / conc.:
1 000 mg/kg bw/day
No. of animals per sex per dose:
10
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale:
The dose levels were selected based on the results of a preceding study (Project No. 06R0087/01248, BASF SE). In this study N-Methyldiethanolamin was given daily as an aqueous solution to groups of 3 male and 3 female Wistar rats (F0 animals) by stomach tube at doses of 100, 300 and 1000 mg/kg body weight/day (mg/kg bw/d) for 2 weeks. Control animals were dosed daily with the vehicle only (drinking water). Clinical signs, food consumption and body weights were determined. The only clearly substance-related effect was “salivation after treatment” in all males and females at the high dose level. Food consumption and body weights did not show significant differences.
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
A check for moribund or dead animals was made twice daily on working days or once daily (weekends or public holidays).

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: daily

BODY WEIGHT: Yes
- Time schedule for examinations: once a week in males throughout the study and in females during premating and mating. During gestation and lactation, F0 females were weighed on gestation days (GD) 0, 7, 14 and 20, on the parturition day and postnatal day (PND) 4.

FOOD CONSUMPTION:
Food consumption of the F0 parents was determined once weekly during premating. In dams food consumption was determined for gestation days 0 - 7, 7 - 14, 14 - 20 and lactation days 1 - 4.
Ovaries and uterine content:
The parturition and lactation behavior of the dams was generally evaluated in the mornings in combination with the daily clinical inspection of the dams. Only particular findings (e.g. inability to deliver) were documented on an individual dam basis. On weekdays (except public holidays) the parturition behavior of the dams was inspected in the afternoons in addition to the evaluations in the mornings. The day of parturition was considered the 24-hour period from about 15.00 h of one day until about 15.00 h of the following day.
The pairing partners, the number of mating days until vaginal sperm were detected, and gestational status were recorded for F0 females.
For the females, mating, fertility and gestation indices were calculated .
Fetal examinations:
The status (sex, liveborn or stillborn) and number of all delivered pups were determined as soon as possible on the day of birth. At the same time, the pups were also examined for macroscopically evident changes. Pups that die before this initial examination are defined as stillborn pups.

In general, a check was made for any dead or moribund pups twice daily on workdays (once in the morning and once in the afternoon) or as a rule, only in the morning on Saturdays, Sundays or public holidays.

On the day of birth (PND 0) the sex of the pups was determined by observing the distance between the anus and the base of the genital tubercle; normally, the anogenital distance is considerably greater in male than in female pups. The sex of the pups was finally confirmed at necropsy.

The live pups were examined daily for clinical symptoms (including gross-morphological findings) during the clinical inspection of the dams. If pups showed particular findings, these were documented with the dam concerned.

The pups were weighed on the day after birth (PND 1) and on PND 4.

All pups with scheduled sacrifice on PND 4 were sacrificed under Isoflurane anesthesia by means of CO2. All pups were examined externally and eviscerated; their organs were assessed macroscopically. All stillborn pups and all pups that died before PND 4 were examined externally, eviscerated
and their organs were assessed macroscopically. All pups without notable findings or abnormalities were discarded after their macroscopic
evaluation. Animals with notable findings or abnormalities were evaluated on a case-by-case basis, depending on the type of finding.
Statistics:
Food consumption (parental animals), body weight and body weight change (parental animals and pups; for the pup weights, the litter means were used), number of mating days, duration of gestation, number of implantation sites, postimplantation loss and % postimplantation loss, number of pups delivered per litter: simultaneous com-parison of all dose groups with the control group using the DUNNETT-test (two-sided) for the hypothesis of equal means.

Male and female mating indices, male and female fertility indices, gestation index, females with liveborn pups, females with stillborn pups, females with all stillborn pups, live birth index, pups stillborn, pups died, pups cannibalized, pups sacrificed moribund, viability index, number of litters with affected pups at necropsy: Pairwise comparison of each dose group with the control group using FISHER'S EXACT test for the hypothesis of equal proportions.

Proportions of affected pups per litter with necropsy observations: Pairwise comparison of each dose group with the control group using the WILCOXON-test (one-sided) for the hypothesis of equal medians.

Weight parameters: Non-parametric one-way analysis using KRUSKAL-WALLIS test (two-sided). If the resulting p-value was equal or less than 0.05, a pairwise comparison of each dose group with the control group was performed using the WILCOXON test for the hypothesis of equal medians.
Indices:
Male mating index (%)=number of males with confirmed mating/number of males placed with females x 100%
Male fertility index (%)= number of males proving their fertility/ number of males placed with females x 100%
Female mating index (%)= number of females mated/ number of females placed with males x 100%
Female fertility index (%)= number of females pregnant/number of females mated x 100%
Gestation index (%)= number of females with live pups on the day of birth/number of females pregnant x 100%
Postimplantation loss (%)=number of implantations – number of pups delivered/number of implantations x 100%
Live birth index (%)= number of liveborn pups at birth/total number of pups born x 100%
Viability index (%) = number of live pups on day 4 after birth/number of live pups on the day of birth x 100%
Clinical signs:
no effects observed
Description (incidence and severity):
At 1000 mg/kg bw/day:
- Nine Males and felames showed transient salivation for a few min. immediately after each treatment. It was likely to be induced by the taste of the test substance or by local irritation of the upper digestive tract. It is not considered to be a sign of systemic toxicity.
- One female (No. 135) with unsteady gait, poor general state and oblique head position. These findings were assessed as being incidental and not test substance related.
- Four females (Nos. 133, 137, 138 and 139) lost all pups (PND 1 or 2). In 2 of this dams undelivered pups were palpable in their abdomen before. The other two femalesdid not properly nurse their pups (pups had no or less milk in the stomach). This was assessed as being test substance-related.

At 100 mg/kg bw/day: One female lost all pups (PND 2) and did not properly nurse its pups before (pups had no or less milk in the stomach). Due to the isolated occurrence and the lack of a dose-response relationship, this was assessed as being incidental.
Dermal irritation (if dermal study):
not examined
Mortality:
no mortality observed
Description (incidence):
There were no mortalities in any of the male and female F0 parental animals in any of the groups.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day:
- Decreased bw gain in males in treatment weeks 2-3 (57% below control) and 0-3 (29% below control)
- Decreased bw in females on gestation days 14 and 20 (up to 14% below control) and lactation day 4 (about 5% below control)
- Decreased bw gain in females between gestation days 7-20 (up to about 46% below control) and body weight loss (-3.9 g) between lactation days 0-4
At 300 mg/kg bw/day:
- Decreased bw gain in males in treatment weeks 1-2 (45% below control)
- Decreased terminal bw in males and females (4 and 5 % below controls, respectively).
At 100 mg/kg bw/day: no test substance-related adverse findings
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: Decreased food consumption in females (-37%) during lactation days 1-4 but might be due to the lower pup number in this group and consequently a lower caloric demand of the F0 females.
At 300 and 100 mg/kg bw/d: Females id not show any test substance-related changes of food consumption during the whole treatment period.

Males did not show any test substance-related changes of food consumption during the whole treatment period.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
Absolute weights:
Males: The terminal bw decrease in the 300 and 1000 mg/kg test groups and the liver weight increase in the 300 and 1000 mg/kg test groups are regarded to be a treatment-related effect.
Females: The terminal bw decrease in the 300 and 1000 mg/kg test groups and the liver weight increase in the 100, 300 and 1000 mg/kg test groups are regarded to be a treatment-related effect.

Relative organ weights:
- Males: the liver weight increase in the 300 and 1000 mg/kg test groups is regarded to be a treatment-related effect.
- Females. the liver weight increase in the 100, 300 and 1000 mg/kg test groups is regarded to be a treatment-related effect.
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
The "enlarged" liver does correlate with the weight increase for the males in the 300 and 1000 mg/kg test groups and for the females in the 100, 300, and 1000 mg/kg test groups and is therefore considered a treatment-related effect.

All other findings are regarded to be incidental or spontaneous in nature and not related to treatment
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
no effects observed
Description (incidence and severity):
All lesions noted are regarded to be incidental or spontaneous in nature and not related to treatment. Especially, there were no pathomorphological patterns in the liver that did correlate to enlarged size and weight increase.

Non pregnant pairs:
At 300 mg/kg bw/d:
- One male (No. 25) showed macroscopically slightly reduced size of epididymides and testes. Histopathologically, diffuse seminiferous tubule atrophy (grade 4) in the testes and aspermia in the epididymides were observed and are correlating to the macroscopic findings.
- One female (No. 125) did not show any macroscopic findings and was therefore not examined histopathologically.
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Number of abortions:
not specified
Pre- and post-implantation loss:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: the number of implantation sites was distinctly reduced ( 6.7 vs. 12.9 in control).
Total litter losses by resorption:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: total litter loss in 4 females
Early or late resorptions:
not specified
Description (incidence and severity):
At 1000 mg/kg bw/day the number of resorptions as well as the % postimplantation loss was distinctly increased (21 and 31.0% vs. 8 and 6.0% in control).
Dead fetuses:
not specified
Changes in pregnancy duration:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: the mean duration of gestation was statistically significantly increased (22.8 days).
At 0, 100, 300 mg/kg bw/day: the mean duration of gestation varied between 21.9 and 22.1 days
Changes in number of pregnant:
not examined
Other effects:
no effects observed
Description (incidence and severity):
The mean duration until sperm was detected (GD 0) varied between 1.6 and 2.6 days without any relation to dosing
The female mating index varied between 90%- 100%
All sperm positive rats delivered pups or had implants in utero. Thus, the fertility index was 100% in all test groups.
Details on maternal toxic effects:
Maternal toxic effects:yes

Details on maternal toxic effects:
1000 mg/kg bw/day:
- Decreased food consumption in females (-37 %) during lactation days 1-4
- Decreased body weights in females on gestation days 14 and 20 (up to 14% below control) and lactation day 4 (about 5 % below control)
- Decreased body weight gain in females between gestation days 7-20 (up to about 46 % below control)
- Total litter loss in 4 females
-Undelivered pups palpable in 2 females
- Insufficient lactation behavior in 2 females (pups had no or less milk in stomach)
- Increased duration of gestation (22.8 days vs. 21.9 days in control)
- Decreased number of implantation sites (6.7 vs. 12.9 in control)
- Increased postimplantation loss (31.0 % vs. 6.0 % in control) and mean postimplantation loss
- Decreased number of delivered pups (4.6 vs. 12.1 in control)
- Decrease of terminal body weights in females

300 mg/kg bw/day:
- Decreased terminal body weights in females (5 % below controls ).

100 mg/kg bw/day:
- No test substance-related adverse findings.
Dose descriptor:
NOAEL
Remarks:
maternal toxicity
Effect level:
100 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
Dose descriptor:
NOAEL
Remarks:
Developmental toxicity
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
effects on pregnancy duration
pre and post implantation loss
other: Insufficient lactation behavior in 2 females at higher dose level and Decreased number of delivered pups
Fetal body weight changes:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day:
- Decreased pup body weights on PND 4, average difference to the control 20 %
- Decreased pup body weight gain during PND 1 – 4, average difference to the control 52 %

At 300 and 100 mg/kg bw/day: No test substance-related adverse findings
Reduction in number of live offspring:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: reduced pup viability
Changes in sex ratio:
no effects observed
Description (incidence and severity):
The sex distribution and sex ratios of live F1 pups on the day of birth and PND 4 did not show substantial differences between the control and the test substance-treated groups; slight differences were regarded to be spontaneous in nature.
Changes in litter size and weights:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: decreased pup body weights and weight gains
Changes in postnatal survival:
effects observed, treatment-related
Description (incidence and severity):
At 1000 mg/kg bw/day: Reduced viability index (62% vs 99% in control), resulting from significantly higher numbers of died (5 vs. 0 in control) and cannibalized pups (12 vs. 1 in control).
At 300 and 100 mg/kg bw/day: No test substance-related adverse findings
At 100 mg/kg bw/d; 87% vs. 99% in cotrol), reduced viablility index resulting from significantly higher numbers of cannibalized pups (11 vs. 1 in control), mainly caused by dam No. 112, which cannibalized 11 of its 14 pups. This can therefore be seen as a spontaneous event.
External malformations:
not examined
Skeletal malformations:
not examined
Visceral malformations:
not examined
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Toxic effects on offspring: yes

Details on effects:
1000 mg/kg bw/day:
-Reduced viability index (62%), resulting from significantly higher numbers of died and cannibalized pups
- Decreased pup body weights on PND 4, average difference to the control 20%
- Decreased pup body weight gain during PND 1 – 4, average difference to the control 52%

300 and 100 mg/kg bw/day:
-No test substance-related adverse findings
Dose descriptor:
NOAEL
Remarks:
F1 Development
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: Reduced viability index (62%), at higher dose level, decreased bw on PND 4 and decreased pup body weight gain during PND 1 - 4, at higher dose level
Abnormalities:
not specified
Developmental effects observed:
not specified

Details on the results:

Clinical signs of toxicity were seen at the high dose level (1000 mg/kg bw/day) such as decreased food consumption in females during lactation and decreased body weight gain in males and females, resulting in reduced terminal body weights in both sexes. Reduced terminal body weights were also seen at the mid dose level (300 mg/kg bw/day).

Concerning reproductive parameters, total litter loss in 4 females, undelivered pups, insufficient lactation behavior, increased duration of gestation, decreased number of implantation sites, increased postimplantation loss and decreased number of delivered pups, reduced pup viability, decreased pup body weights and weight gains were seen exclusivelyat the high dose level (1000 mg/kg bw/day), i.e. only in the presence of parental toxicity.

Pathology revealed increased liver weights at all dose levels, however without any morphological correlate. These findings were therefore assessed as being an adaptive phenomenon, but not an adverse effect. There were no further treatment-related lesions detected, especially, there were no weight or substance-related pathomorphological effects on testes, epididymides, and ovaries present.

Conclusions:
In conclusion, the administration of N-Methyldiethanolamine at dose levels of 1000 and 300 mg/kg bw/d caused toxic effects on body weight. The NOAEL (no observed adverse effect level) for general, systemic toxicity was therefore 100 mg/kg body weight/day for the F0 parental males and females.
The NOAEL for reproductive performance and fertility was 300 mg/kg body weight/day for the F0 parental rats based upon findings such as litter loss, insufficient lactation behavior, and increased duration of gestation.
The NOAEL for developmental toxicity was 300 mg/kg body weight/day, based on findings such as reduced viability index and reduced postnatal offspring weight gain.
Thus, reproductive parameters were affected only in the presence of parental toxicity.
Executive summary:

This reproduction/developmental toxicity screening study was conducted according to OECD 421 and in accordance with GLP.

N-Methyldiethanolamine was given daily as an aqueous solution to groups of 10 male and 10 female Wistar rats (parental generation) by stomach tube at doses of 100, 300 and 1000 mg/kg bw/day. Control animals were dosed daily with the vehicle only (drinking water). The duration of treatment covered premating period of 2 weeks and a mating period (max. of 2 weeks) in both sexes and the entire gestation period as well as 4 days of lactation in females.

At the dose level of 1000 mg/kg bw/day, decreased food consumption in females, and decreased body weight gain and terminal body weights in males and females were observed. Furthermore, total litter loss, undelivered pups palpable, insufficient lactation behaviour (pups had no or less milk in stomach), increased duration of gestation, decreased number of implantation sites, increased post-implantation loss, decreased number of delivered pups, decrease of terminal body weights in males and females were recorded. In F1 pups a reduced viability index, decreased body weights and body weight gain was observed.

At the dose level of 300 mg/kg bw/day, decreased terminal body weights in males and females were observed. In pups, there were no test substance-related adverse findings.

At the dose level 100 mg/kg bw/day, no test substance-related adverse findings neither in parental generation nor in pups.

 

In conclusion, the administration of N-Methyldiethanolamine at dose levels of 1000 and 300 mg/kg bw/d caused toxic effects on body weight. The NOAEL (no observed adverse effect level) for general, systemic toxicity was therefore 100 mg/kg body weight/day for the F0 parental males and females.

The NOAEL for reproductive performance and fertility was 300 mg/kg body weight/day for the F0 parental rats based upon findings such as litter loss, insufficient lactation behavior, and increased duration of gestation.

The NOAEL for developmental toxicity was 300 mg/kg body weight/day, based on findings such as reduced viability index and reduced postnatal offspring weight gain.

Thus, reproductive parameters were affected only in the presence of parental toxicity.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
other: CD
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Laboratories, Portage, MI
- Age at study initiation: male: 77 days; females: 70 days;
- Weight at study initiation: Female rats weighed at least 200 g at the time of mating.
Route of administration:
dermal
Vehicle:
water
Details on exposure:
TEST SITE
- Area of exposure: dorsal trunk
- Type of wrap if used: refolded eight-ply sterilized gauze square and a 2.75 x 2.25 inch area of polyvinyl film.
- Time intervals for shavings or clipplings: 3 days prior to the first dose and subsequently as needed throughout the study.

REMOVAL OF TEST SUBSTANCE
- Washing (if done): the dosing site was wiped gently with a paper towel dampened with warm water and blotted dry.
- Time after start of exposure: 6 ha fter the application.

TEST MATERIAL
- Amount(s) applied (volume or weight with unit): 4 mL
- Concentration (if solution): Range finding study: Definitive study: 250, 500, and 1000 mg/kg/day (Range finding study: 100, 500, 750, and 1000 mg/kg/day).

VEHICLE
- Amount(s) applied (volume or weight with unit): 4 mL
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
MDEA concentrations in dosing solutions were verified using a gas chromatograph equipped with a flame ionization detector.
Details on mating procedure:
- Impregnation procedure: cohoused
- If cohoused:
- M/F ratio per cage: 1/1
- Proof of pregnancy: vaginal plug referred to as day 0 of pregnancy
Duration of treatment / exposure:
days 6 - 15 of gestation
Frequency of treatment:
6 hours per day
Duration of test:
21 days
Dose / conc.:
0 mg/kg bw/day
Remarks:
definitive study
Dose / conc.:
250 mg/kg bw/day
Remarks:
definitive study
Dose / conc.:
500 mg/kg bw/day
Remarks:
definitive study
Dose / conc.:
1 000 mg/kg bw/day
Remarks:
definitive study
Dose / conc.:
0 mg/kg bw/day
Remarks:
range finding study
Dose / conc.:
100 mg/kg bw/day
Remarks:
range finding study
Dose / conc.:
500 mg/kg bw/day
Remarks:
range finding study
Dose / conc.:
750 mg/kg bw/day
Remarks:
range finding study
Dose / conc.:
1 000 mg/kg bw/day
Remarks:
range finding study
No. of animals per sex per dose:
Twenty-five plug-positive females were assigned to each group (range finding test: 8 plug-positive females)
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: based on results of the range finding study
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: daily

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: daily

BODY WEIGHT: Yes
- Time schedule for examinations: on gd 0, 6, 9, 12,15, 18, and 21

FOOD CONSUMPTION AND COMPOUND INTAKE (if feeding study): Yes

WATER CONSUMPTION AND COMPOUND INTAKE (if drinking water study): No data

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day # 21
- Organs examined: gravid uterus, ovaries including corpora lutea, cervix, vagina, and peritoneal and thoracic cavities were examined grossly

Other: blood was used for erythrocyte, platelet, total and differential leukocyte counts, hematocrit, hemoglobin, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration.
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
Blood sampling:
yes
Fetal examinations:
- External examinations: Yes: all per litter
- Soft tissue examinations: Yes: half per litter
- Skeletal examinations: Yes: half per litter
- Head examinations: Yes: half per litter
Statistics:
Data from quantitative, continuous variables were intercompared for the dosed groups and the control group by use of Levene's test for equality of variances, analysis of variance, (ANOVA), and t-tests. Nonparametric data were evaluated using the Kruskal- Wallis test, followed by the Mann-Whitney U test when appropriate incidence data were compared using the Fisher's Exact Test. The probability value of p < 0.05 (two-tailed) was used as the critical level of significance.
Indices:
No information given
Historical control data:
No information given
Dermal irritation (if dermal study):
no effects observed
Description (incidence and severity):
Skin reactions at the dosing site were observed in the 500 and 1000 mg/kg bw/day groups. Effects included exfoliation, excoriation, crusting, ecchymoses, and necrosis.
Body weight and weight changes:
no effects observed
Description (incidence and severity):
There were no effects of dosing with MDEA on maternal body weight or body weight gain.
Food consumption and compound intake (if feeding study):
no effects observed
Description (incidence and severity):
There were no effects of dosing with MDEA on food consumption.
Haematological findings:
effects observed, treatment-related
Description (incidence and severity):
The erythrocyte count and hematocrit were reduced in the 1000 mg/kg bw/day group. Also, while not statistically significant, hemoglobin was reduced by about 6% in the 1000 mg/kg bw/day group. However, there were no effects on mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, platelet count, and total or differential leukocyte counts.
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
There were no effects of dosing with MDEA gravid uterine weight, or liver and kidney weights.
Number of abortions:
not specified
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
There were no effects on the number of ovarian corpora lutea, the number of total, viable or non-viable implantations, preimplantation loss.
Total litter losses by resorption:
not examined
Early or late resorptions:
not examined
Dead fetuses:
not examined
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
not examined
Other effects:
not examined
Details on maternal toxic effects:
Maternal toxic effects: yes
Dose descriptor:
NOAEL
Effect level:
250 mg/kg bw/day
Basis for effect level:
other: maternal toxicity
Fetal body weight changes:
no effects observed
Description (incidence and severity):
There were no effects fetal body weights.
Reduction in number of live offspring:
not examined
Changes in sex ratio:
not examined
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
There were no increased incidences of total external, visceral, and skeletal malformations or variations or individual malformations compared to controls. Among individual variations, there was a statistically significant decrease in the incidence of a soft-tissue variation (dilated bilateral renal pelvis) and a skeletal variation (split number 1, 2, 3, and/or 4 cervical centra) at 1000 mg/kg bw/day. These decreases are not considered characteristic of a developmental delay.
Skeletal malformations:
no effects observed
Description (incidence and severity):
There were no increased incidences of total external, visceral, and skeletal malformations or variations or individual malformations compared to controls. Among individual variations, there was a statistically significant decrease in the incidence of a soft-tissue variation (dilated bilateral renal pelvis) and a skeletal variation (split number 1, 2, 3, and/or 4 cervical centra) at 1000 mg/kg bw/day. These decreases are not considered characteristic of a developmental delay.
Visceral malformations:
no effects observed
Description (incidence and severity):
There were no increased incidences of total external, visceral, and skeletal malformations or variations or individual malformations compared to controls. Among individual variations, there was a statistically significant decrease in the incidence of a soft-tissue variation (dilated bilateral renal pelvis) and a skeletal variation (split number 1, 2, 3, and/or 4 cervical centra) at 1000 mg/kg bw/day. These decreases are not considered characteristic of a developmental delay.
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: no effects
Dose descriptor:
NOAEL
Effect level:
>= 1 000 mg/kg bw/day
Basis for effect level:
other: teratogenicity
Remarks on result:
other: no effects were reported
Abnormalities:
not specified
Developmental effects observed:
not specified
Conclusions:
In conclusion, occluded cutaneous applicatation of methydiethanolamine to pregnant rats during organogenesis resulted in maternal toxicity as indicated by skin irritation and mild anaemia from 500 mg/kd/day onwards, but no treatment-related developmental effects were observed.
Executive summary:

The Prenatal Developmental toxicity study was conduced similar to the OECD TG 414. In the study, timed pregnant CD rats were dosed cutaneously with aqueous N-methyldiethanolamine (MDEA) daily from gestational day (gd) 6 to 15, inclusive. Dosages employed were 0, 250, 500, and 1000 mg/kg/day and were selected on the basis of maternal toxicity responses determined from a range-finding study. Prior to the first dose, the hair was removed from the animal's dorsal trunk skin by clipping with veterinary clippers. The dosing site was covered and the test material was left in contact with the skin for 6 hours per day. Rats were sacrificed on the 21st day of gestation, and the foetuses were examined for malformations and variations. There were no effects of dosing with MDEA on maternal body weight, body weight gain, food consumption, gravid uterine weight, or liver and kidney weights. Skin reactions at the dosing site were observed in the 500 and 1000 mg/kg bw/day groups. Effects included exfoliation, excoriation, crusting, ecchymoses, and necrosis. The erythrocyte count and hematocrit were reduced in the 1000 mg/kg bw/day group. Also, while not statistically significant, hemoglobin was reduced by about 6% in the 1000 mg/kg bw/day group. However, there were no effects on mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, platelet count, and total or differential leukocyte counts. There were no effects on the number of ovarian corpora lutea, the number of total, viable or nonviable implantations, preimplantation loss, and fetal body weights.

There were no increased incidences of total external, visceral, and skeletal malformations or variations or individual malformations compared to controls. Among individual variations, there was a statistically significant decrease in the incidence of a soft-tissue variation (dilated bilateral renal pelvis) and a skeletal variation (split number 1, 2, 3, and/or 4 cervical centra) at 1000 mg/kg bw/day. These decreases are not considered characteristic of a developmental delay.

In conclusion, occluded cutaneous application of methydiethanolamine to pregnant rats during organogenesis resulted in maternal toxicity as indicated by skin irritation and mild anaemia from 500 mg/kd/day onwards, but no treatment-related developmental effects were observed.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
GLP compliance:
yes
Limit test:
no
Species:
rat
Strain:
not specified
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:

VEHICLE
- Concentration in vehicle: 0, 500, 1500, 3000, 5000 mg/100 mL
- Amount of vehicle (if gavage): 10 mL/kg bw
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
Recharacterization was carried out after the end of all studies.
Details on mating procedure:
no information given
Duration of treatment / exposure:
day 6-15 of gestation
Frequency of treatment:
daily
Duration of test:
no information given
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Dose / conc.:
50 mg/kg bw/day (actual dose received)
Dose / conc.:
150 mg/kg bw/day (actual dose received)
Dose / conc.:
300 mg/kg bw/day (actual dose received)
Dose / conc.:
500 mg/kg bw/day (actual dose received)
No. of animals per sex per dose:
10 females
Control animals:
yes
Details on study design:
Sex: female
Duration of test: until day 16 of gestation
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes

DETAILED CLINICAL OBSERVATIONS: Yes

BODY WEIGHT: Yes

FOOD CONSUMPTION: Yes

WATER CONSUMPTION: No

POST-MORTEM EXAMINATIONS: Yes
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
Blood sampling:
no information given
Fetal examinations:
- External examinations: Yes
- Soft tissue examinations: Yes
- Skeletal examinations: Yes
- Head examinations: Yes
Statistics:
no information given
Indices:
no information given
Historical control data:
no information given
Clinical signs:
no effects observed
Description (incidence and severity):
During the whole study period the animals showed no abnormal clinical signs or findings.
Dermal irritation (if dermal study):
not examined
Mortality:
no mortality observed
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
The mean body weights of the high dose animals were statistically significantly lower on days 10, 13 and 15 p.c. when compared with the control group. Furthermore body weight gain of the 500 mg/kg rats was clearly reduced on days 6 - 15 p.c. During this period the relevant animals gained about 25 % less weight than the controls.

The results of the corrected body weight gain (terminal body weight on day 15 p.c. minus weight of the unopened uterus minus body weight on day 6 p .c .) was clearly reduced (to about 65% of the actual control value) in the 500 mg/kg group.
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
The food consumption of the high dose females (500 mg/kg body weight/day) was statistically significantly reduced during days 8-15 p.c. The reduced food intake of the 300 mg/kg females on days 6-8 p.c. might be a spontaneous effect due to the missing dose-response relationship.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not specified
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
effects observed, treatment-related
Description (incidence and severity):
Total protein and albumin values were statistically significantly reduced in the highest dose group (500 mg/kg body weight/day), which has to be related to the substance-induced impairment of the body weight gain of the dams.
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
effects observed, treatment-related
Description (incidence and severity):
The weights of the gravid uteri were not clearly influenced by the administration of the test substance.
No substance-related effects were observed on the placentae weight. The marginal, however statistically significant decrease in the mean placental weights in test group 3 (300 mg /kg body weight/day) is without any biological relevance.
Gross pathological findings:
effects observed, treatment-related
Description (incidence and severity):
According to the pathology report besides the reduced mean body weights of the high dose dams, the only necropsy finding which was assessed as substance -related was a thickening of the wall of the forestomach in 3 high dose animals.
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Number of abortions:
not specified
Description (incidence and severity):
See details on maternal maternal toxic effects
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
There were no substance-related and/or statistically significant differences between the groups in the values calculated for the pre- and post-implantation losses.
Total litter losses by resorption:
not examined
Early or late resorptions:
no effects observed
Description (incidence and severity):
There were no substance-related and/or statistically significant differences between the groups in the number of resorptions
Dead fetuses:
no effects observed
Description (incidence and severity):
There were no substance-related and/or statistically significant differences between the groups in the number of viable fetuses
Changes in pregnancy duration:
no effects observed
Description (incidence and severity):
See details on maternal maternal toxic effects
Changes in number of pregnant:
no effects observed
Description (incidence and severity):
The conception rate varied between 90 % (high dose group) and 100 % (all other groups). There were no substance-related and/or statistically significant differences between the groups in conception rate.
Other effects:
no effects observed
Description (incidence and severity):
There were no substance-related and/or statistically significant differences between the groups in the mean number of corpora lutea and implantation sites.
Details on maternal toxic effects:
All differences evident in the examined parameters are considered to be incidental and within the normal range of deviations for animals of this strain and age.
Dose descriptor:
NOAEL
Remarks:
general toxicity
Effect level:
300 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: maternal toxicity
Fetal body weight changes:
no effects observed
Description (incidence and severity):
No substance-related effects were observed.
Reduction in number of live offspring:
not examined
Changes in sex ratio:
not examined
Changes in litter size and weights:
not examined
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
The external examination of the fetuses, which was limited due to the stage of development of the fetuses at this part of the gestation period, revealed no malformation and variation in any test group. Only one so-called unclassified observation (placentae fused) was recorded for one fetus of test group 3 (300 mg/kg body weight/day).
Skeletal malformations:
not examined
Visceral malformations:
not examined
Other effects:
not examined
Dose descriptor:
NOAEL
Effect level:
>= 500 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: overall teratogenic effects
Remarks on result:
other: no adverse effects observed
Abnormalities:
not specified
Developmental effects observed:
not specified
Conclusions:
From the results of this preliminary study it can be seen, that monoethanolamine caused clear signs of maternal toxicity at 500 mg/kg body weight substantiated by impairments of food consumption and body weight gain, reductions of total protein and albumin values and substance-induced reactions of the forestomach wall. Concerning the other substance-treated test groups, no clear signs of maternal toxicity appeared. Due to the stage of development of the implants on the day when the study was terminated the examination of the uterus content was very limited; taking this into consideration, no adverse effects on the fetuses occurred.
Executive summary:

This preliminary study was conduced to prevaluate the prenatal toxicity of monoethanolamine (MEA) in rats after oral administration. MEA was administered by gavage, on day 6-15 of gestation at concentrations of 0, 50, 150, 300 and 500 mg/kg bw/day to pregnant rats.

The oral administration of MEA by gavage to pregnant rats probably caused the following substance-induced changes:

At 500 mg/kg body weight/day the following observations were made:

- clearly reduced food consumption (days 8 - 15 p.c.)

- lower mean body weights and distinctly impaired weight gain; reduced corrected body weight gain

- statistically significantly reduced total protein and albumin values

- thickened wall of the forestomach in 3 dams

At 300, 150 and 50 mg/kg body weight/day no clear substance-related effects on dams or fetuses were recorded.

From the results of this preliminary study it can be seen, that MEA caused clear signs of maternal toxicity at 500 mg/kg body weight substantiated by impairments of food consumption and body weight gain, reductions of total protein and albumin values and substance-induced reactions of the forestomach wall. Concerning the other substance-treated test groups, no clear signs of maternal toxicity appeared. Due to the stage of development of the implants on the day when the study was terminated the examination of the uterus content was very limited; taking this into consideration, no adverse effects on the fetuses occurred.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
1 (reliable without restriction)
Rationale for reliability incl. deficiencies:
guideline study
Qualifier:
according to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Wistar
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Sexually mature, virgin Wistar rats (Chbb :THOM (SPF)) supplied by Karl THOMAE, Biberach an der Riss, Germany,
- Age at study initiation: 60 days
- Weight at study initiation: mean weight approx. 223.7 g
- Fasting period before study: none
- Housing: singly in type DK III stainless steel wire mesh cages supplied by Becker & Co., Castrop-Rauxel, Germany (floor area about 800 cm²)
- Diet: ground Kliba 343 feed rat/mouse/hamster supplied by Klingentalermuehle AG, Kaiseraugst, Switzerland ad libitum
- Water: tap water ad libitum
- Acclimation period: 5 days

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20-24
- Humidity (%): 30-70
- Photoperiod (hrs dark / hrs light): 12/12
Route of administration:
oral: gavage
Vehicle:
water
Details on exposure:
PREPARATION OF DOSING SOLUTIONS:
Each day the test substance solutions were freshly prepared shortly before the test substance was administered. For the preparation of the solutions, an appropriate amount of the test substance was weighed in a volumetric flask and subsequently topped up with doubly distilled water and intensively shaken.

VEHICLE
- Amount of vehicle (if gavage): 10 ml/ kg bw.
Analytical verification of doses or concentrations:
yes
Details on analytical verification of doses or concentrations:
The homogeneity of the test substance was proven by visual inspection. The content of active ingredient was 99 .4% before the beginning of the study. The reanalysis of the test substance proved its stability (content: 99 .5%)
Details on mating procedure:
- Impregnation procedure: [cohoused]
- If cohoused:
- M/F ratio per cage: 1/1
- Verification of same strain and source of both sexes: [yes]
- Proof of pregnancy: [sperm in vaginal smear] referred to as [day 0] of pregnancy
Duration of treatment / exposure:
day 6 - 15 of gestation
Frequency of treatment:
daily, once per day
Duration of test:
up to day 21 of gestation
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Remarks:
Control
Dose / conc.:
40 mg/kg bw/day (actual dose received)
Dose / conc.:
120 mg/kg bw/day (actual dose received)
Dose / conc.:
450 mg/kg bw/day (actual dose received)
No. of animals per sex per dose:
40 dams per dosing group
Control animals:
yes, concurrent vehicle
Details on study design:
On day 0, the animals were assigned to the different test groups according to a randomization plan. The test substance was administered to the animals orally (by gavage) once a day during the period of major organogenesis (day 6 to day 15 p .c .) always at approx. the same time of day (in the morning). The animals of the control group were treated in the same way with the vehicle (doubly distilled water). The volume administered each day was 10 mL/kg body weight. The calculation of the volume administered was based on the individual body weight determined at the beginning of the administration period (day 6 p.c.). On day 20 p.c., the first 25 animals/group were sacrificed in a randomized order and examined macroscopically. The fetuses were dissected from the uterus and further investigated with different methods. The other animals (15/group) were allowed to litter and rear their pups up to day 21 p.p. (post partum). On day 21 post partum (p.p.) or one of the following days the relevant dams and pups were sacrificed and examined macroscopically.
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Mortality
A check was made twice a day on working days or once a day (Saturday, Sunday or on public holidays).

- Clinical symptoms
All animals were examined for clinical symptoms at least once a day, or more often when clinical signs of toxicity were elicited. The nesting, littering, and lactation behavior of the dams with terminal sacrifice day 21 p.p. was generally evaluated in the mornings in connection with the daily clinical inspection of the dams. Only if there were any special findings (e.g., animal could not litter, umbilical cord not cut), these specific findings were documented with the dam concerned.

The littering behavior of the relevant dams was also inspected on weekdays (except holidays) in the afternoons in addition to the evaluations in the mornings. These reevaluation were documented separately, but, as before, findings were only recorded with the dams concerned. Moreover, the duration of gestation, the number of live and dead pups at birth and litter size were recorded for the animals with terminal sacrifice on day 21 p.p. For these animals the fertility and the gestation indices were calculated.

BODY WEIGHT: Yes
All animals were weighed on days 0, 1, 3, 6, 8, 10, 13, 15, 17 and 20 p.c. Body weights of the animals with terminal sacrifice on day 21 p.p. were additionally determined on the day of birth and on days 4, 7, 14 and 21 p.p. The body weight change of the animals was calculated from these results. Body weights of the animals without litter were not determined during the lactation period of the dams used in parallel.

Corrected body weight gain (net maternal body weight change). Furthermore, the corrected body weight gain was calculated for all animals with terminal sacrifice on day 20 p.c. (terminal body weight on day 20 p.c. minus weight of the uterus before it was opened minus body

FOOD CONSUMPTION AND COMPOUND INTAKE: Yes
With the exception of day 0 p.c. (all animals) and days 0 p.p. and 21 p.p. (for animals with terminal sacrifice on day 21 p.p. only), the consumption of food was determined on the same days as was body weight. Food consumption was not determined for the females without litter during the lactation period of the dams used in parallel.weight on day 6 p.c.).
Ovaries and uterine content:
Examinations of the dams at termination
Dams with terminal sacrifice on day 20 p.c. On day 20 p.c., the dams were sacrificed in randomized order by cervical dislocation and the fetuses dissected from the uterus. After the dams had been sacrificed , they were necropsied and assessed by gross pathology. The uterus and the ovaries were removed and the following data were recorded :

Weight of uterus before it was opened
- Number of corpora lutea: Yes
- Number and distribution of implantation sites classified as: live fetuses, dead implantations:
- Number of early resorptions: Yes (only decidual or placental tissues visible or according to Salewski from uteri from apparently non-pregnant animals and the empty uterus horn in the case of single-horn pregnancy)
- Number of late resorptions: Yes (embryonic or fetal tissue in addition to placental tissue visible)

OTHER:
Dead fetuses (hypoxemic fetuses which did not breathe spontaneously after the uterus had been
opened)

Calculations of conception rate and pre- and postimplantation losses were carried out:
- The conception rate (in %) was calculated according to the following formula: conception rate = (number of pregnant animals/ number of fertilized animals) x 100
- The preimplantation loss (in % ) was calculated according to the following formula: ((number of corpora lutea - number of implantations)/number of corpora lutea) x 100
- The post implantation loss (in % ) was calculated from the following formula: ((number of implantations - number of live fetuses)/number of implantations) x 100


Dams with terminal sacrifice on day 21 p.p. On day 21 post partum (p. p. ) the relevant dams were sacrificed by cervical dislocation. After the dams had been sacrificed, the following examinations were carried out:
- gross - pathological examination
- staining of uterus according to Salewski for determination of the number of implantations

Calculations of conception rate and postimplantation loss were carried out:
- The conception rate (in %) was calculated according to the following formula: conception rate = (number of pregnant animals/ number of fertilized animals) x 100
- The post implantation loss (in % ) was calculated from the following formula: ((number of implantations - number of live fetuses)/number of implantations) x 100
Blood sampling:
- Plasma: No
- Serum: No
Fetal examinations:
Examination of the fetuses: Examination of the fetuses after dissection from the uterus.

- External examinations: Yes
At necropsy each fetus was weighed, sexed and examined macroscopically for any external findings. The sex was determined by observing the distance between the anus and the base of the genital tubercle and was later confirmed in all fetuses fixed in Bouin's solution by internal examination. If there were discrepancies between the "external" and the "internal" sex of a fetus, the fetus was finally sexed according to the appearance of its gonads. Furthermore, the viability of the fetuses and the condition of the placentae, the umbilical cords, the fetal membranes and fluids were examined. Individual placental weights were recorded. After these examinations, approximately one half of the fetuses per dam was placed in ethyl alcohol and the other half was placed in Bouin's solution for fixation and further evaluation.

- Soft tissue examinations: Yes: [half per litter]
After fixation in Bouin's solution, approximately one half of the fetuses of the dams of all groups was examined for any findings in the organs according to the method of Barrow and Taylor with special attention being paid to the kidneys and the ureters. After the examination, these fetuses were discarded with the exception of the kidneys, which were placed into cassettes separately for each fetus and kept in 4% formaldehyde solution for possible further examination by light microscopy. Moreover, after fixation of the fetuses placed in ethyl alcohol for further evaluation of the fetal skeletons the organs of these fetuses were examined macroscopically. Thereafter, the kidneys of each fetus were placed into cassettes and kept in 4% formaldehyde solution for a possible further examination by light microscopy, while the other organs were discarded. Afterwards the carcasses of these fetuses were stained according to a modified method (Dawson) for the presentation of the skeletons.

- Skeletal examinations: Yes [half per litter]
After fixation in ethyl alcohol and examination of the organs, the skeletons of the fetuses were stained according to a modified method of Dawson. Thereafter, the skeletons of these fetuses were examined under a stereomicroscope. After these examinations the relevant fetuses were retained by litter.

Evaluation criteria for assessing skeletons and organs of the fetuses:
In the present investigations the following terms (definitions) were used for describing a change:
- Malformations (concerning external, soft tissue and skeletal observations). Rare and/or probably lethal changes were classified as malformations (e.g. exencephaly, atresia ani, hernia umbilicalis).

- Variations (concerning external, soft tissue and skeletal observations): Changes which occur regularly also in control groups and have generally no adverse effect on survival were regarded as variations (e.g. dilated renal pelvis).

- Retardations (concerning skeletal observations only): Delays in skeletal development compared with the norm at the time of the examination were considered to be retardations (e.g. sternebra(e) not ossified)

- Unclassified observations (concerning external and soft tissue observations, only): External or soft tissue observations, which could not be classified as malformations or variations (e.g.
blood coagulum around placenta).

Examination of the pubs:
- Pup number and status at delivery: All pups derived from the females were examined as soon as possible on the day of birth to determine the total number of pups and the number of liveborn and stillborn members of each litter. Pups which died before the first determination of their status on the day of birth were designated as stillborn pups.

- Pup viability / mortality: In general, a check was made for any dead or moribund pups twice daily on workdays (once in the morning and once in the afternoon) or as a rule, only in the morning on Saturdays, Sundays or public holidays. Dead pups were evaluated by the methods which will be described in detail before. The number and percentage of dead pups on the day of birth (day 0) and of pups dying between days 1-4, 5-7, 8-14 and 15-21 of the lactation period were determined; however, pups which died accidentally were not included in these calculations. The number of live pups/litter was calculated on the day of birth, and on lactation days 4, 7, 14 and 21.

- Sex ratio: On the day of birth (day 0) the sex of the pups was determined by observing the distance between the anus and the base of the genital tubercle; normally, the anogenital distance is considerably greater in male than in female pups. During the following time the sex of the pups was assessed by the external appearance of the anogenital region and/or the mammary line of the animals and was finally confirmed at necropsy. The sex ratio was calculated for day 0 and day 21 after birth according to the following formula:

sex ratio = (number of live male or female pups on day 0/21 / number of live male and female pups on day 0/21) x 100

- Pup body weight data: The pups were weighed on the day after birth (day 1 p.p.) and on days 4, 7, 14 and 21 after birth. Pups' body weight change was calculated from these results. The individual weights were always determined at about the same time of the day (in the morning). In the relevant summary tables pup body weights and pup body weight gains are listed for males, females and males + females.

- Pup clinical observations: The pups were examined each day for clinical symptoms (including gross-morphological findings).

- Pup necropsy observations: After sacrifice on day 21 p.p. or one of the following days (by means of CO2) or intercurrent death, the pups were examined externally, eviscerated and their organs were assessed macroscopically with special attention being paid to the urinary tract. After the macroscopic examination of the pups, the kidneys of each pup were placed into cassettes and fixed in 4% formaldehyde solution for a possible further examination by light microscopy. If there were notable findings or if abnormalities were found in the daily clinical observation of the animals after their delivery, the affected animals were, if it was deemed necessary, examined additionally using appropriate methods (e.g., skeletal staining according to modified Dawson's method and/or further processing of head according to Wilson's method. The stained skeletons were evaluated under a stereomicroscope or a magnifying glass. All pups without any notable findings or abnormalities were discarded after their macroscopic evaluation (with the exception of the kidneys (see above)).
Statistics:
Dunnett-Test was used for a simultaneous comparison of several dose groups with the control. The hypothesis of equal means was tested. This test was performed two-sided and was used for the statistical evaluation of food consumption, body weights and body weight change (females and pups), corrected body weight gain (net maternal body weight change), weight of the uterus before it was opened, number of corpora lutea, number of implantations, number of resorptions and number of live fetuses; proportion of preimplantation loss, postimplantation loss, resorptions and live fetuses in each litter; litter mean fetal body weight and litter mean placental weight, duration of gestation and number of pups delivered per litter. For the body weight and the body weight change of the pups the mean weight of each litter was used for the statistical analysis (statistical unit = litter).
Fisher' s Exact Test was used for a pairwise comparison of each dose group with the control for the hypothesis of equal proportions. This test was performed one - sided was used for statistical evaluation of the following parameters: female mortality, females pregnant at terminal sacrifice, number of litters with fetal findings, female fertility index, gestation index, females with liveborn, stillborn and with all stillborn pups, live birth index, pups stillborn, pups died, pups cannibalized, pups sacrificed moribund, viability index, lactation index, number of litters with affected pups at necropsy.
The Wilcoxon Test was used for a comparison of each dose group with the control for the hypothesis of equal medians. This test was performed one-sided and was used for the proportion of fetuses with malformations, variations, retardations and/or unclassified observations in each litter and for the proportion of affected pups per litter with necropsy observations. If the results of these tests were significant, labels (*for ≤ 5 0.05, ** for p ≤ 0.01) were printed in the summary tables.
Indices:
Viability index (%) = (number of live pups on day 4 after birth/ number o f liveborn pups on the day of birth) x 100
Lactation index (%) = (number of live pups on day 21 after birth/ number of live pup s on day 4 after birth) x 100
Fertility index = (n pregnant animals/ n mated animals) x 100
Gestation index = (n animals with litters/ n pregnant animals) x 100
Historical control data:
Yes
Clinical signs:
effects observed, non-treatment-related
Description (incidence and severity):
No signs that might be attributed to the test substance administered were detected during gestation and lactation periods. During gestation, piloerection was recorded for one high dose animal on day 13. Without any dose-response relationship insufficient nesting activity was observed for several dams of all groups.
All of these findings are spontaneous in nature and cannot be attributed to the test substance administration
Dermal irritation (if dermal study):
not examined
Mortality:
mortality observed, non-treatment-related
Description (incidence):
One dam of the 40 mg/kg/day group died intercurrently during delivery. Undelivered pups were found in the uterus. The death of this dam is considered spontaneous in nature and cannot be attributed to the test substance administration.
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
Test group 3 (450 mg/kg body weight/day):
- Statistically significantly lower mean dam body weights than the controls on days 15, 17 and 20 p.c. and on days 0, 4, 7 and 21 p.p. ; impaired body weight gain of the dams during posttreatment days 15 - 20 p.c.
- The results of the corrected body weight gain on gestation day 20 of all groups did not show any differences of biological significance.
Test group 2 (120 mg/kg body weight/day): no substance-related effects observed
Test group 1 (40 mg/kg body weight/day): no substance-related effects observed
Food consumption and compound intake (if feeding study):
effects observed, treatment-related
Description (incidence and severity):
Test group 3 (450 mg/kg body weight/day):
- Statistically significantly reduced food consumption at the beginning of the treatment period (days 6 - 8 p.c.), the final days of the gestation period (days 17 - 20 p.c.) and during the first days of the lactation period (days 0- 4 p.p.).
Test group 2 (120 mg/kg body weight/day): no substance-related effects observed
Test group 1 (40 mg/kg body weight/day): no substance-related effects observed
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
The mean placental weights in the test groups were not influenced by the test substance administration.
Effects on total uterine weight: The uterus weights, which were determined for the animals with termination on day 20 of gestation only, were not influenced by the administration of the test substance. The differences between the groups is without biological relevance and do not show any dose-response relationship.
Gross pathological findings:
effects observed, non-treatment-related
Description (incidence and severity):
There were no substance-related observations at necropsy in any of the dams. Hydrometra (a spontaneous finding) was recorded for one female of the control group, for 2 females of the 40 mg/kg/day group, and 3 females of the 120 mg/kg/day group. These animals did not become pregnant. Edema of the lungs which has to be related to the termination of the rats was recorded for several dams of the control, low and intermediate groups without any relation to dosing. For the one low dose female that died intercurrently during parturition, undelivered pups were found in the uterus
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not specified
Details on results:
Maternal toxic effects:yes
Details on maternal toxic effects:
Monoethanolamine caused some signs of maternal toxicity when administered by gavage to pregnant Wistar rats from days 6 - 15 of gestation at the highest dose level tested, 450 mg/kg/day. Maternal toxicity was substantiated by reduced food consumption, lower mean body weights and impaired body weight gain. The oral administration of monoethanolamine at 450 mg/kg/day or doses below this had no influence on resorption rate, number of live fetuses or pups/dam, mean fetal weight or pup body weights.
Number of abortions:
no effects observed
Description (incidence and severity):
No fetuses were aborted or delivered early in any of the groups.
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
The mean % pre-implantation loss was 3.4, 6.8, 9.9 and 11.7 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. The mean % post-implantation loss for the 20 gestational females was 10.3, 7.3, 7.0 and 6.3 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. The mean % post-implantation loss for the 21 p.p. females was 5.7, 3.9, 9.7 and 7.3 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. In the animals euthanized on day 20 of gestation, there were no-substance related and/or statistically significant differences in the values calculated for the pre- and postimplantation losses. The females euthanized on day 21 p.p. showed no substance-associated effects on the postimplantation loss.
Total litter losses by resorption:
effects observed, non-treatment-related
Description (incidence and severity):
One dam of the 120 mg/kg/day group had a total litter loss on day 1 after birth. The total litter loss of one dame is condiered incidental in nature and not test-item related.
Early or late resorptions:
no effects observed
Description (incidence and severity):
Mean early resorptions were 1.4, 0.9, 1.0 and 0.9 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. Mean late resorptions were 0.2, 0.3, 0.1 and 0.0 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. In the animals euthanized on day 20 of gestation, there were no-substance related and/or statistically significant differences in the number of resorptions.
Dead fetuses:
no effects observed
Description (incidence and severity):
Dams with viable fetuses was 21, 20, 20 and 24 and the number of fetuses alive/dead were 293/0, 282/0, 263/0 and 311/0 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively.
Changes in pregnancy duration:
no effects observed
Description (incidence and severity):
The mean duration of gestation for the 21 postpartum (p.p.) females was 21.8 days, 21.6 days, 21.6 days, and 21.4 days for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. In the females euthanized on day 21 p.p., the duration of gestation and the gestation index were substantially similar in all groups.
Changes in number of pregnant:
no effects observed
Description (incidence and severity):
Number pregnant per dose level: The conception rate varied between 85% (450 mg/kg/day group) and 75% (40 mg/kg/day group). The conception per dose level was 33 in the control group, 30 in the 40 mg/kg/day group, 33 in the 120 mg/kg/day group and 34 in the 450 mg/kg/day group. The number pregnant at caesarian-section was 21 in the control, 20 in the 40 and 120 mg/kg/day groups and 24 in the 450 mg/kg/day group. The females euthanized on day 21 p.p. showed no substance-associated effects on the fertility index which was 80%, 67%, 87% and 67% for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively.
Other effects:
no effects observed
Description (incidence and severity):
Number of implantations: The mean number of implantation sites for the 20 females euthanized on day 20 of gestation were 15.6, 15.3, 14.3 and 13.8 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. The mean number of implantation sites for the 21 p.p. females were 14.6, 14.6, 14.1 and 13.4 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. In the animals euthanized on day 20 of gestation and the females euthanized on day 21 p.p., there were no-substance related and/or statistically significant differences in the mean number of implantation sites.

Number of corpora lutea: The mean numbers of corpora lutea were 16.1, 16.3, 15.6 and 15.7 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively. In the animals euthanized on day 20 of gestation, there were no-substance related and/or statistically significant differences in the mean number of corpora lutea.
Details on maternal toxic effects:
Maternal toxic effects:yes

Details on maternal toxic effects:
Monoethanolamine caused some signs of maternal toxicity when administered by gavage to pregnant Wistar rats from days 6 - 15 of gestation at the highest dose level tested, 450 mg/kg/day. Maternal toxicity was substantiated by reduced food consumption, lower mean body weights and impaired body weight gain. The oral administration of monoethanolamine at 450 mg/kg/day or doses below this had no influence on resorption rate, number of live fetuses or pups/dam, mean fetal weight or pup body weights.
Dose descriptor:
NOAEL
Remarks:
general toxicity
Effect level:
120 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: maternal toxicity
Fetal body weight changes:
no effects observed
Description (incidence and severity):
Fetuses: The mean fetal body weights were not influenced by the test substance administration. The mean body weight of viable fetuses was 3.9 grams for all groups.
Pups: Without any clear relation to dosing, pup weights were occasionally statistically significantly lower in the substance-treated groups than in the respective control values. On day 21 p.p. control and high dose pup weights were substantially similar, whereas the mean pup body weights of the 40 and 120 mg/kg/day group were still slightly, but not significantly lower than the control values. On days 1-4 p.p. pup body weight gains were also statistically significantly lower in the substance-treated groups, again without a clear dose-response relationship. Because treatment of the dams took place only until day 15 of gestation and because of no clear dose-response relationship, it seems very unlikely that the differences in pup body weight/body weight gain are substance-related.
Reduction in number of live offspring:
no effects observed
Description (incidence and severity):
Of the females euthanized on day 21 p.p. the number of females with liveborn was 12, 10, 13, and 10 and the number of pups delivered was 165, 132, 170, and 125 for the control, the 40 mg/kg/day, 120 mg/kg/day and the 450 mg/kg/day groups, respectively.
Changes in sex ratio:
no effects observed
Description (incidence and severity):
Sex distribution and ratio: The sex distribution of the fetuses in the test groups was comparable with the control fetuses. Sex ratios (M/F in %) on day 0 were:
control: 54.6/45.4
40 mg/kg/day: 55.7/44.3
120 mg/kg/day: 46.1/53.9
450 mg/kg/day: 53.6/46.4
Sex ratios (M/F in %) on day 21 were:
control: 54.4/45.6
40 mg/kg/day: 54.0/46.0
120 mg/kg/day: 45.2/54.8
450 mg/kg/day: 54.2/45.8
The sex distribution and sex ratios of live pups on the day of birth and on day 21 p.p. did not show any substantial difference between controls and treated test groups.
Changes in litter size and weights:
no effects observed
Description (incidence and severity):
The litter size was not influenced by the test substance administration.
Anogenital distance of all rodent fetuses:
not specified
Changes in postnatal survival:
no effects observed
Description (incidence and severity):
The mean number of delivered pups/dam was not influenced by the administration of test substance. There were no substantial biological relevant differences concerning pup viability/ mortality in any of the groups. Viability and lactation indices were unaffected.
External malformations:
no effects observed
Description (incidence and severity):
The only external malformation which was found was an anasarca in one high-dose fetus. This malformation is also present at a low incidence in the historical control data. The external examination of the fetuses revealed no variations in any group. One unclassifed observation, fused placentae, was recorded in one fetus of the 40 mg/kg/day group and one fetus of the 450 mg/kg/day group.
Skeletal malformations:
effects observed, non-treatment-related
Description (incidence and severity):
Fetuses: All skeletal malformations, variations, or retardations which occurred did not show a clear dose-response relationship, can be found at comparable or even higher rates within the historical control and/or the differences between the groups are without biological significance.

Pups: The only skeletal malformations which occurred were related to the thoracic part of the vertebral column (thoracic vertebral body/bodies dumbbell-shaped (asymmetrical) or bipartite (asymmetrical)). One or both of these malformations were found in a few fetuses of each test group including the controls without any biological relevant differences. The variations elicited were related to the ribs (shortened 13th rib(s), accessory 14th rib(s), rudimentary cervical rib(s), and the sternum (sternebra(e) of irregular shape or bipartite). These variations had no clear dose-response relationship, can be found in a similar frequency in historical control data, and/or the differences between groups are without biological significance. In all groups signs of retardations (incomplete or missing ossification of vertebral bodies/arches and the sternebra(e)) were found. The differences between the groups, however, are not associated with the test substance administration. All of the skeletal retardations are to be found at a comparable frequency in the historical control data and most often a clear dose-response relationship is not present. The only statistically significant difference, an increased rate of total variations in the 120 mg/kg/day group, is without biological relevance because it shows no dose-dependence.
Visceral malformations:
effects observed, non-treatment-related
Description (incidence and severity):
Fetuses: In all groups, including the control, some soft tissue malformations were found. These malformations were related to the eyes (microphthalmia), the heart (dilatation of the right or both ventricles; dextrocardia), the lung (uni-lobular) or the kidneys (hyper-/hypoplasia) and did not show any relation to dose. Two soft tissue variations, which were related to the urinary tract (dilated renal pelvis; hydroureter) occurred in all groups without any dose-response relationship and were fully within the historical control range. One unclassified observation (bloody inhibition of the kidneys) was recorded for 3 control and one high dose fetus.
Other effects:
effects observed, non-treatment-related
Description (incidence and severity):
- Pup necropsy observations: Only a few pups showed findings at necropsy. Post mortem autolysis, incisor sloped and dilated renal pelvis (1 high-dose pup) occurred in single pups of the control, the 40 and 450 mg/kg/day groups.
- Pup clinical observations: None of the pups of the different groups showed any clinical signs until termination.
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects:no effects

Details on embryotoxic / teratogenic effects:
No signs of developmental toxicity occurred up to and including the highest dose level (450 mg/kg/day), especially no substance-induced teratogenic effects were observed neither in the fetuses nor in the pups. Furthermore, there were no indications for any substance-related growth retardations. The urinary tract of the rat fetuses/pups did not show any treatment-related findings. Dilated renal pelvis and/or hydroureter were found in a considerable, but according to historical control data, not unexpected high number of fetuses of all groups including the controls without any relation to dosing, but did not occur at an increased rate in the pups of the substance-related groups. All skeletal malformations, variations, or retardations which occurred did not show a clear dose-response relationship, can be found at comparable or even higher rates within the historical control and/or the differences between the groups are without biological significance.
Dose descriptor:
NOAEL
Effect level:
>= 450 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: Embryotoxicity and teratogenicity
Remarks on result:
other:
Abnormalities:
not specified
Developmental effects observed:
not specified
Conclusions:
In conclusion, under the conditions of this study, Monoethanolamine pure caused some signs of maternal toxicity when administered by gavage to pregnant Wistar rats from days 6 - 15 p.c. at the highest dose level tested (450 mg/kg body weight/day). Maternal toxicity was substantiated in this dose group by a reduced food consumption, lower mean body weights and impaired body weight gain. 120 and 40 mg/kg body weight did not induce any signs of maternal toxicity in the rats.
There occurred no signs of developmental toxicity up to and including the highest dose level (450 mg/kg body weight/day). The reproductive parameters were unaffected and neither the fetuses nor the pups showed an increased malformation rate or any indications for a substance-induced growth retardation; especially, the urinary tract of the rat fetuses/pups did not show any treatment-related findings.
Based on these study results, the no observable adverse effect level (NOAEL) on the maternal organism is 120 mg/kg body weight/day and 450 m g/kg body weight/day for the progeny.
Executive summary:

The study was conducted in accordance with OECD TG 414 and GLP. The test substance was administered to the animals orally (by gavage) once a day during the period of major organogenesis (day 6 to day 15 p .c .). On day 20 p.c., the first 25 animals/group were sacrificed in a randomized order and examined macroscopically. The fetuses were dissected from the uterus and further investigated with different methods. The other animals (15/group) were allowed to litter and rear their pups up to day 21 p.p. (post partum). On day 21 post partum (p.p.) or one of the following days the relevant dams and pups were sacrificed and examined macroscopically. The following findings were obtained and assessed as substance-related: In the test group 3 (450 mg/kg body weight/day) a statistically significantly reduced food consumption at the beginning of the treatment period (days 6 - 8 p.c.), at the final days of the gestation period (days 17 - 20 p.c.) and during the first days of the lactation period (days 0- 4 p.p.) was recorded. Furthermore, statistically significantly lower mean dam body weights than the controls on days 15, 17 and 20 p.c. and on days 0, 4, 7 and 21 p.p. were observed and body weight gain of the dams during posttreatment days 15 - 20 p.c. were impaired. In the test group 2 (120 mg/kg body weight/day) and test group 1 (40 mg/kg body weight/day), no substance-related effects were observed.

No signs of developmental toxicity occurred up to and including the highest dose level (450 mg/kg/day), especially no substance-induced teratogenic effects were observed neither in the fetuses nor in the pups. Furthermore, there were no indications for any substance-related growth retardations. The urinary tract of the rat fetuses/pups did not show any treatment-related findings. Dilated renal pelvis and/or hydroureter were found in a considerable, but according to historical control data, not unexpected high number of fetuses of all groups including the controls without any relation to dosing, but did not occur at an increased rate in the pups of the substance-related groups. All skeletal malformations, variations, or retardations which occurred did not show a clear dose-response relationship, can be found at comparable or even higher rates within the historical control and/or the differences between the groups are without biological significance.

 

In conclusion, under the conditions of this study, Monoethanolamine pure caused some signs of maternal toxicity when administered by gavage to pregnant Wistar rats from days 6 - 15 p.c. at the highest dose level tested (450 mg/kg body weight/day). Maternal toxicity was substantiated in this dose group by a reduced food consumption, lower mean body weights and impaired body weight gain. 120 and 40 mg/kg body weight did not induce any signs of maternal toxicity in the rats.

There occurred no signs of developmental toxicity up to and including the highest dose level (450 mg/kg body weight/day). The reproductive parameters were unaffected and neither the fetuses nor the pups showed an increased malformation rate or any indications for a substance-induced growth retardation; especially, the urinary tract of the rat fetuses/pups did not show any treatment-related findings.

Based on these study results, the no observable adverse effect level (NOAEL) on the maternal organism is 120 mg/kg body weight/day and 450 m g/kg body weight/day for the progeny.

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Reason / purpose for cross-reference:
reference to same study
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Principles of method if other than guideline:
Pregnant New Zealand White rabbits were exposed dermally to 0, 10, 25, and 75 mg/kg/day of monoethanolamine (MEA) for approximately 6 hr/day on Days 6 through 18 of gestation.
GLP compliance:
not specified
Limit test:
no
Species:
rabbit
Strain:
New Zealand White
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Hazelton Research Products, Inc. (Denver, PA), USA
- Weight at study initiation: 3.0 - 4.0 kg
- Fasting period before study: none
- Housing: in wire-bottom cages
- Diet: ad libitum
- Water: ad libitum
- Acclimation period: 2 weeks

ENVIRONMENTAL CONDITIONS
- Temperature (°C): 20
- Humidity (%): 40-60
- Photoperiod (hrs dark / hrs light): 12/12



Route of administration:
dermal
Vehicle:
water
Details on exposure:
TEST SITE
- Area of exposure: shaved skin of the back
- Type of wrap if used: sterile gauze held in place by Lycra-Spandex jacket

REMOVAL OF TEST SUBSTANCE
- Washing (if done): water-dampened towel was used to wipe remaining test material off
- Time after start of exposure: 6 hours

TEST MATERIAL
- Amount(s) applied (volume or weight with unit): 2 mL/kg
- Constant volume or concentration used: no
Analytical verification of doses or concentrations:
not specified
Details on mating procedure:
- Impregnation procedure: [cohoused]
- If cohoused:
- M/F ratio per cage: 2/1
- Length of cohabitation: over night
- Verification of same strain and source of both sexes: [yes]
- Proof of pregnancy: [copulation] referred to as [day 0] of pregnancy
Duration of treatment / exposure:
day 6 - 18 of gestation
Frequency of treatment:
6 hours/day, daily
Duration of test:
up to day 29 of gestation
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
10 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
25 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
75 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
No. of animals per sex per dose:
15 dams/group
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: Dose levels selected for these studies were chosen based upon the results of dermal range-finding and teratology probe studies conducted in rabbits
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: daily

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: daily

BODY WEIGHT: Yes
- Time schedule for examinations: gestation days 0, 3, 6, 9, 12, 15, 18, 24 and 21

FOOD CONSUMPTION: Yes
- Food consumption for each animal determined and mean daily diet consumption calculated as g food/kg body weight/day: Yes

WATER CONSUMPTION: No

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 29
- Organs examined: weights of liver, kidneys

OTHER: Blood analysis prior to caesarian section; skin irritation was evaluated once daily during the postdosing period
Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
- Other: Uteri with no visible implantations were stained with a 10% sulfide solution.
Blood sampling:
Immediately prior to scheduled cesarean section, blood was obtained from the central auricular artery of all surviving rabbits for hematologic determinations.
Fetal examinations:
- External examinations: Yes: [all per litter]
- Soft tissue examinations: Yes: [all per litter]
- Skeletal examinations: Yes: [half per litter]
- Head examinations: Yes: [half per litter ]

All fetuses were weighed and examined for evidence of external alterations and palate closure. All of the rabbit fetuses in each litter were examined for visceral alterations (Staples,1974). The sex of all live fetuses was determined. The heads of one half of the rabbit fetuses not selected for skeletal examination were removed, placed in Bouin's solution, and subsequently sectioned and examined for craniofacial defects (Wilson, 1 965 ; Van Julsingha and B ennet, 1977). All fetuses were eviscerated and stained with alizarin red-S ( Dawson, 1926; Crary, 1962). Skeletal examinations were conducted only on the rat fetuses not selected for Bouin's examination.
Statistics:
Continuous data were evaluated for homogeneity of variance using Levene's test (Levene, 1960). Based upon the outcome of this test, a parametric or nonparametric analysis of variance (ANOVA) was performed. If the ANOVA was significant, analysis by Dunnett's test (Steel and Torrie, 1960), the Wilcoxen Rank-Sum test with Bonferroni's correction (Miller, 1966), or a pooled t test was performed as appropriate. The level of statistical significance was set a priori at a = 0.05. Nonparametric data were compared using Fischer's exact probability test (Siegel, 1956).
Indices:
no information given
Historical control data:
no information given
Clinical signs:
not specified
Dermal irritation (if dermal study):
effects observed, treatment-related
Description (incidence and severity):
Rabbits administered 75 mg/kg/day of MEA exhibited severe skin irritation (erythema, edema, ecchymosis, necrosis, exfoliation, and crusting) at the site of exposure. Subsequent to the dosing period, exfoliation, crusting, and areas of necrosis persisted. The skin of the majority of these rabbits began to heal as evidenced by scab formation late in the gestation period. Crusting, transient erythema, and edema were noted in a few rabbits administered 25 mg MEA/kg/day. No significant dermal irritation or lesions were observed among rabbits administered 10 mg MEA/kg/day.
Mortality:
no mortality observed
Description (incidence):
No females died during the study.
Body weight and weight changes:
no effects observed
Description (incidence and severity):
No statistically identified changes were observed in body weight and body weight gain, the average body weight gain of high-dose rabbits over the course of gestation was decreased when compared to that of the control and other dose groups, mainly due to weight loss or very little weight gain during the treatment period.
Food consumption and compound intake (if feeding study):
no effects observed
Description (incidence and severity):
There were also no significant treatment-related effects observed on feed consumption.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
not examined
Ophthalmological findings:
not examined
Haematological findings:
no effects observed
Description (incidence and severity):
There were also no significant treatment-related effects observed on hematologic parameters of MEA-exposed rabbits at any dose level tested.
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
There were also no significant treatment-related effects observed on kidney and liver weights of MEA-exposed rabbits at any dose level tested.
Gross pathological findings:
not examined
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not specified
Details on results:
Dermal exposure of pregnant rabbits to 75 mg/kg/day resulted in significant increases in the incidence of skin irritation/lesions and maternal body weight effects. In general, the dermal irritation observed at the high dose was progressive, beginning with erythema and leading to necrosis, scabs, and scar formation. Doses of 25 mg/kg/day produced only minor irritation.
Number of abortions:
no effects observed
Description (incidence and severity):
No females aborted during the study
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
No treatment-related effects on number of corpora lutea.
No treatment-related effects on number of implantations.



No treatment-related effects on fetal body weight
No treatment-related effects on gravid uterine weight
Total litter losses by resorption:
no effects observed
Description (incidence and severity):
No treatment-related effects on resorptions
Early or late resorptions:
no effects observed
Description (incidence and severity):
No treatment-related effects on resorptions
Dead fetuses:
no effects observed
Description (incidence and severity):
No treatment-related effects on number of dead fetuses
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
no effects observed
Description (incidence and severity):
No treatment-related effects on pregnancy rate.
Other effects:
no effects observed
Description (incidence and severity):
No treatment-related effects were observed on reproductive parameters.
No treatment-related effects on fetal sex ratio.
Details on maternal toxic effects:
Details on maternal toxic effects:
Rabbits administered 75 mg/kg/day of MEA exhibited severe skin irritation (erythema, edema, ecchymosis, necrosis, exfoliation, and crusting) at the site of exposure. Subsequent to the dosing period, exfoliation, crusting, and areas of necrosis persisted. The skin of the majority of these rabbits began to heal as evidenced by scab formation late in the gestation period. Crusting, transient erythema, and edema were noted in a few rabbits administered 25 mg MEA/kg/day. No significant dermal irritation or lesions were observed among rabbits administered 10 mg MEA/kg/day.

No females died, aborted, delivered early, or were removed from the test during the study. There were also no significant treatment-related effects observed on feed consumption, hematologic parameters, or kidney and liver weights of MEA-exposed rabbits at any dose level tested.

No statistically identified changes were observed in body weight and body weight gain, the average body weight gain of high-dose rabbits over the course of gestation was decreased when compared to that of the control and other dose groups, mainly due to weight loss or very little weight gain during the treatment period.
Dose descriptor:
NOAEL
Remarks:
local
Effect level:
10 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: maternal toxicity
Dose descriptor:
NOAEL
Remarks:
Developmental toxicity
Effect level:
> 75 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: Overall maternal develpmental effects
Remarks on result:
other: No adverse effects observed
Fetal body weight changes:
no effects observed
Reduction in number of live offspring:
not examined
Changes in sex ratio:
no effects observed
Changes in litter size and weights:
no effects observed
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
There were no statistically or biologically significant treatment-related differences in the incidence of any fetal variation or malformation, or in the number of malformed fetuses in any dose group.
Skeletal malformations:
no effects observed
Description (incidence and severity):
There were no statistically or biologically significant treatment-related differences in the incidence of any fetal variation or malformation, or in the number of malformed fetuses in any dose group.
Visceral malformations:
no effects observed
Description (incidence and severity):
There were no statistically or biologically significant treatment-related differences in the incidence of any fetal variation or malformation, or in the number of malformed fetuses in any dose group.
- Among control litters the following types of malformations were noted: ventricular septal defect, common opening at entry of the vessels of the heart, missing lung lobe, missing gallbladder, and extra lumbar centrum and
- Malformations observed in fetuses from the 75 mg/kg/day dose group litters included the following: dilated lateral cerebral ventricle with tissue depression, missing lung lobe, diagonally displaced thoracic centra, missing thoracic arch, and a single missing rib.
- Malformations observed in litters from rabbits given 10 mg/kg/day included dilated lateral cerebral ventricle with tissue depression, missing lung lobe, missing gallbladder, misaligned or fused thoracic centra, extra lumbar centrum and arches, and fused ribs. Limited numbers of malformations noted at 25 mg/kg/day included dilated lateral cerebral ventricle with tissue depression and missing lung lobe.

The classification of the mentioned fetal effects as malformations was atypicalb and no dose-response relationship was observed for any of the "malformations" reported.
Other effects:
not specified
Details on embryotoxic / teratogenic effects:
Dermal exposure of rabbits to MEA via occluded cutaneous application at doses which produced severe maternal dermal irritation, did not produce evidence of embryo toxicity, fetotoxicity, or teratogenicity.
Dose descriptor:
NOAEL
Effect level:
>= 75 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: Overall developmental / fetal toxicity
Remarks on result:
other: no evidence of developmental or fetal toxicity observed
Abnormalities:
not specified
Developmental effects observed:
not specified
Conclusions:
Dermal exposure of pregnant rabbits to 75 mg/kg/day resulted in significant increases in the incidence of skin irritation/lesions and maternal body weight effects. Despite these maternal effects observed, no evidence of developmental or fetal toxicity was observed at any dose level tested. Thus, it was concluded that MEA was not developmentally toxic following dermal application at exposure levels up to and including 75 mg/kg/ day for rabbits. Based on these results the following effect levels were derived:
NOAEL maternal toxicity, local effects: 10 mg/kg bw/day
NOAEL developmental or fetal toxicity: > 75 mg/kg/day
Executive summary:

The present study was conduced equivalent to the OECD TG 414. New Zealand White rabbits were exposed dermally to 0,10,25, and 75 mg/kg/day of monoethanolamine (MEA) for approximately 6 hr/day on Days 6 through 18 of gestation.

Dermal exposure of pregnant rabbits to 75 mg/kg/day resulted in significant increases in the incidence of skin irritation/lesions and maternal body weight effects. Despite these maternal effects observed, no evidence of developmental or fetal toxicity was observed at any dose level tested. Thus, it was concluded that MEA was not developmentally toxic following dermal application at exposure levels up to and including 75 mg/kg/ day for rabbits.

Based on these results the following effect levels were derived:

NOAEL maternal toxicity, local effects: 10 mg/kg bw/day

NOAEL developmental or fetal toxicity: > 75 mg/kg/day

Endpoint:
developmental toxicity
Type of information:
experimental study
Adequacy of study:
weight of evidence
Study period:
No information given
Reliability:
2 (reliable with restrictions)
Rationale for reliability incl. deficiencies:
comparable to guideline study with acceptable restrictions
Reason / purpose for cross-reference:
reference to same study
Qualifier:
equivalent or similar to guideline
Guideline:
OECD Guideline 414 (Prenatal Developmental Toxicity Study)
Principles of method if other than guideline:
Pregnant Sprague-Dawley rats were exposed dermally to 0, 10, 25, 75 and 225 mg/kg/day of monoethanolamine (MEA) for approximately 6 hr/day on Days 6 through 15 of gestation .
GLP compliance:
not specified
Limit test:
no
Species:
rat
Strain:
Sprague-Dawley
Details on test animals or test system and environmental conditions:
TEST ANIMALS
- Source: Charles River Breeding Laboratories, Inc. (Kingston, NY), USA
- Age at study initiation:
- Weight at study initiation: 250-300 g)
- Housing: in wire-bottom cages
- Diet: ad libitum
- Water: ad libitum
- Acclimation period: 2 weeks


ENVIRONMENTAL CONDITIONS
- Temperature (°C): 22
- Humidity (%): 40-60
Route of administration:
dermal
Vehicle:
water
Details on exposure:
TEST SITE
- Area of exposure: shaved skin of the back
- Type of wrap if used: absorbant gauze pad followed by nonabsorbant cotton; an elastic bandage was wrapped securely around the animal to hold the patch in place and to prevent accidental ingestion of the test material via grooming during the exposure.


REMOVAL OF TEST SUBSTANCE
- Washing (if done): water-dampened towel was used to wipe remaining test material off
- Time after start of exposure: 6 hours


TEST MATERIAL
- Amount(s) applied (volume or weight with unit): 1 ml/kg
- Constant volume or concentration used: no
Details on mating procedure:
- Impregnation procedure: [cohoused]
- If cohoused:
- M/F ratio per cage: 1/1
- Length of cohabitation: over night
- Verification of same strain and source of both sexes: [yes]
- Proof of pregnancy: [sperm in vaginal smear] referred to as [day 0] of pregnancy
Duration of treatment / exposure:
day 6 - 15 of gestation
Frequency of treatment:
6 hours/day, daily
Duration of test:
up to day 21 of gestation
Dose / conc.:
0 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
10 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
25 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
75 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
Dose / conc.:
225 mg/kg bw/day (actual dose received)
Remarks:
received dermal dose
No. of animals per sex per dose:
30-45 rats
Control animals:
yes, concurrent vehicle
Details on study design:
- Dose selection rationale: Dose levels selected for these studies were chosen based upon the results of dermal range-finding and teratology probe studies conducted in rats
Maternal examinations:
CAGE SIDE OBSERVATIONS: Yes
- Time schedule: daily

DETAILED CLINICAL OBSERVATIONS: Yes
- Time schedule: daily

BODY WEIGHT: Yes
- Time schedule for examinations: gestation days 0, 3, 6-16, and 21

FOOD CONSUMPTION: Yes
- Food consumption for each animal determined and mean daily diet consumption calculated as g food/kg body weight/day: Yes

WATER CONSUMPTION: Yes

POST-MORTEM EXAMINATIONS: Yes
- Sacrifice on gestation day 21
- Organs examined: weights of liver, kidneys

Ovaries and uterine content:
The ovaries and uterine content was examined after termination: Yes
Examinations included:
- Gravid uterus weight: Yes
- Number of corpora lutea: Yes
- Number of implantations: Yes
- Number of early resorptions: Yes
- Number of late resorptions: Yes
- Other: Uteri with no visible implantations were stained with a 10% sulfide solution.
Blood sampling:
not examined
Fetal examinations:
- External examinations: Yes: [all per litter]
- Soft tissue examinations: Yes: [half per litter]
- Skeletal examinations: Yes: [half per litter]
- Head examinations: Yes: [half per litter ]


All fetuses were weighed and examined for evidence of external alterations and palate closure. At least one-half of the rat fetuses in each litter were examined for visceral alterations (Staples,1974). The sex of all live fetuses was determined. The heads of rat fetuses not selected for skeletal examination were removed, placed in Bouin's solution, and subsequently sectioned and examined for craniofacial defects (Wilson, 1965; Van Julsingha and Bennet, 1977). All fetuses were eviscerated and stained with alizarin red-S ( Dawson, 1926; Crary, 1962). Skeletal examinations were conducted only on the rat fetuses not selected for Bouin's examination.
Statistics:
Continuous data were evaluated for homogeneity of variance using Bartlett's test (Winer, 1971). Based upon the outcome of these tests, a parametric or nonparametric analysis of variance (ANOVA) was performed. If the ANOVA was significant, analysis by Dunnett's test (Steel and Torrie, 1960), the Wilcoxen Rank-Sum test with Bonferroni's correction (Miller, 1966), or a pooled t test was performed as appropriate. The level of statistical significance was set a priori at a = 0.05. Nonparaznetric data were statistically treated using the Kruskal-Wallis test followed by the Mann-Whitney U test (Sokal and Rohlf, 1969), when appropriate. Incidence data for rats were analyzed using the Wilcoxon test as modified by Haseman and Hoel (1974). Fetal sex ratios were analyzed using a binomial distribution test (Steel and Torrie, 1960).
Indices:
No information given
Historical control data:
No information given
Clinical signs:
not specified
Dermal irritation (if dermal study):
effects observed, treatment-related
Description (incidence and severity):
Rats administered 225 mg MEA/kg/day exhibited a treatment-related increased incidence of skin irritation at the site of exposure. In general, the dermal irritation followed a progression, beginning with erythema and leading to necrosis, scabs, and scar formation.
No significant dermal irritation or lesions were observed among rats administered lower doses of MEA.
Mortality:
not specified
Body weight and weight changes:
effects observed, treatment-related
Description (incidence and severity):
The body weight gain of rats given 225 mg MEA/kg/day was significantly decreased during the exposure period. No effect on weight gain was observed in dams treated with lower levels of MEA.
Food consumption and compound intake (if feeding study):
no effects observed
Description (incidence and severity):
No significant differences were observed in the feed or water consumption of MEA exposed rats relative to controls.
Food efficiency:
not examined
Water consumption and compound intake (if drinking water study):
no effects observed
Description (incidence and severity):
No significant differences were observed in the feed or water consumption of MEA exposed rats relative to controls.
Ophthalmological findings:
not examined
Haematological findings:
not examined
Clinical biochemistry findings:
not examined
Endocrine findings:
not examined
Urinalysis findings:
not examined
Behaviour (functional findings):
not examined
Immunological findings:
not examined
Organ weight findings including organ / body weight ratios:
no effects observed
Description (incidence and severity):
No effects on liver or kidney weights were observed at any dose level.
Gross pathological findings:
not examined
Neuropathological findings:
not examined
Histopathological findings: non-neoplastic:
not examined
Histopathological findings: neoplastic:
not examined
Other effects:
not examined
Details on results:
maternal toxic effects: yes
Exposure of pregnant rats to 225 mg MEA/kg/day resulted in significant increases in the incidence of skin irritation/lesions and a significant decrease in body weight gain over the exposure period. There were no post-mortem treatment-related effects observed in any dose group. Despite significant maternal effects observed in rats given 225 mg MEA/kg/day, no embryonal or fetal effects were observed in litters from these dams.
Number of abortions:
no effects observed
Description (incidence and severity):
Reproductive parameters among MEA exposed rats were unaffected
Pre- and post-implantation loss:
no effects observed
Description (incidence and severity):
Reproductive parameters among MEA exposed rats were unaffected
Total litter losses by resorption:
no effects observed
Description (incidence and severity):
Reproductive parameters among MEA exposed rats were unaffected
Early or late resorptions:
no effects observed
Description (incidence and severity):
Reproductive parameters among MEA exposed rats were unaffected
Dead fetuses:
no effects observed
Description (incidence and severity):
Reproductive parameters among MEA exposed rats were unaffected.
Changes in pregnancy duration:
not examined
Changes in number of pregnant:
not examined
Other effects:
not specified
Details on maternal toxic effects:
All examined reproductive parameters among MEA exposed rats were unaffected.
Dose descriptor:
NOAEL
Remarks:
general toxicity
Effect level:
75 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
body weight and weight gain
dermal irritation
Dose descriptor:
NOAEL
Remarks:
developmental toxicity
Effect level:
225 mg/kg bw/day
Based on:
test mat.
Basis for effect level:
other: Overall maternal developmental effects
Remarks on result:
other: No adverse effects observed
Fetal body weight changes:
no effects observed
Reduction in number of live offspring:
no effects observed
Changes in sex ratio:
no effects observed
Changes in litter size and weights:
not specified
Anogenital distance of all rodent fetuses:
not examined
Changes in postnatal survival:
not examined
External malformations:
no effects observed
Description (incidence and severity):
There were no treatment-related increases in the incidence of variations or malformations observed externally, viscerally, or at skeletal examination by individual category, or in total variations or malformations when compared to controls.
Skeletal malformations:
no effects observed
Description (incidence and severity):
There were no treatment-related increases in the incidence of variations or malformations observed externally, viscerally, or at skeletal examination by individual category, or in total variations or malformations when compared to controls.
Visceral malformations:
no effects observed
Description (incidence and severity):
There were no treatment-related increases in the incidence of variations or malformations observed externally, viscerally, or at skeletal examination by individual category, or in total variations or malformations when compared to controls.
Other effects:
not examined
Details on embryotoxic / teratogenic effects:
Embryotoxic / teratogenic effects: no effects

Details on embryotoxic / teratogenic effects:
Despite maternal effects observed among dams given 225 mg/kg/day, reproductive parameters among MEA exposed rats were unaffected at this or lower dose levels. There were no differences in pregnancy rate, number of corpora lutea, number of implantations, resorptions, litter size, number of dead fetuses, fetal sex ratio, fetal body weight, or gravid uterine weight among any of the dose groups when compared to controls.
There were no treatment-related increases in the incidence of variations or malformations observed externally, viscerally or at skeletal examination by individual category, or in total variations or malformations when compared to controls. Among controls, the following types of malformations were observed: microphthalmia, retroesophageal right subclavian artery, and an extra cervical rib. No malformed fetuses were observed in the 10 mg/kg/day dose group. Malformations observed in the 25 mg MEA/kg/day dose group included retroesophageal right subclavian artery and an extra cervical rib. A single fetus was malformed in the 75 mg MEA/kg/day dose group. This fetus had multiple craniofacial malformations consisting of micrognathia, cleft lip and soft palate, and aglossia. No malformations were observed in fetuses from dams administered 225 mg MEA/kg/day.
Dose descriptor:
NOAEL
Effect level:
>= 225 mg/kg bw/day
Based on:
test mat.
Sex:
male/female
Basis for effect level:
other: Overall embryotoxic and teratogenic parameters
Remarks on result:
other:
Abnormalities:
not specified
Developmental effects observed:
not specified
Conclusions:
Exposure of pregnant rats to 225 mg MEA/kg/day resulted in significant increases in the incidence of skin irritation/lesions and a significant decrease in body weight gain over the exposure period. Despite significant maternal effects observed in rats given 225 mg MEA/kg/day, no embryonal or fetal effects were observed in litters from these dams. No significant maternal toxicity, including dermal irritation, or embryonal/fetal toxicity was observed in rats administered doses of 10, 25, or 75 mg MEA/kg/day.
Based on these results the following effect levels were derived:
- NOAEL, maternal toxicity: 75 mg/kg/day (decreased body weight gain, skin irritation)
- NOAEL for develomental, embryonal and fetal toxicity: 225 mg/kg/day (the highest dose level tested)
Executive summary:

The study was conduced similar to the OECD TG 414.

In the study, pregnant Sprague-Dawley rats were exposed dermally to 0,10,25, 75 and 225 mg/kg/day of monoethanolamine (MEA) for approximately 6 hr/day on Days 6 through 15 of gestation.

Exposure of pregnant rats to 225 mg MEA/kg/day resulted in significant increases in the incidence of skin irritation/lesions and a significant decrease in body weight gain over the exposure period. Despite significant maternal effects observed in rats given 225 mg MEA/kg/day, no embryonal or fetal effects were observed in litters from these dams. No significant maternal toxicity, including dermal irritation, or embryonal/fetal toxicity was observed in rats administered doses of 10, 25, or 75 mg MEA/kg/day.

Based on these results the following effect levels were derived:

- NOAEL, maternal toxicity: 75 mg/kg/day (decreased body weight gain, skin irritation)

- NOAEL for develomental, embryonal and fetal toxicity: 225 mg/kg/day (the highest dose level tested)

Endpoint:
developmental toxicity
Data waiving:
study scientifically not necessary / other information available
Justification for data waiving:
other:
Justification for type of information:
According to Regulation EC no 1907/2006 (REACH) Annex X, point 8.7.2. a ‘Developmental toxicity study, one species, most appropriate route of administration, having regard to the likely route of human exposure (OECD 414)’ is a standard requirement. This study needs to be performed in a non-rodent species, in addition to the rodent study already required under Annex IX.
For the registered substance N-Methylethanolamine (MMEA, CAS 109-83-1) an inhalation pre-natal developmental toxicity (PNDT) study is available in rats (Nelson et al. 1984). In this study MMEA was vaporized and administered to 18 pregnant rats in one to three concentrations for 7 h/day on gestation days 7 to 15. The dams were sacrificed on day 20 of gestation. Fetuses were individually weighed, and two-thirds of them were fixed in Bouin's solution and examined for soft-tissue anomalies. The other one-third were fixed in alcohol, stained with Alizarin Red and examined for skeletal defects. As overall result for the substance MMEA, neither maternal nor fetal toxicity effects were found. No teratogenicity was observed. The NOAECs were ≥ 460 mg/m³, the highest dose tested for both, dams and fetuses (Nelson et al. 1984).
Although there is no indication of developmental toxicity, a PNDT study in a second non-rodent species is required according to Annex X. However, this study is not available for MMEA.
To fulfil this requirement, data sets for the analogue aliphatic amine substances monoethanolamine (MEA; CAS 141-43-5), diethanolamine (DEA; CAS 111-42-2; also known as 2,2’iminodiethanol), methyldiethanolamine (MDEA; CAS 105-59-9) and 2-dimethylaminoethanol (DMAE; CAS 108-01-0) have been evaluated within a weight-of-evidence approach in detail. Extensive robust study summaries of the studies evaluated in this approach are also included in the current dossier. A full summary of the weight-of-evidence approach can be found in the endpoint summary.
For MEA, DEA and DMAE, PNDT studies on two species (rat and rabbit) are available for each. These studies were analysed in order to identify any possibly existing differences between rats and rabbits with regard to developmental toxicity. For the registered substance this should show if either species is expected to be more sensitive to developmental effects.
MEA was tested in rats and rabbits in a study according to OECD TG 414 via the dermal route (Liberacki et al. 1996). Doses of 0, 10, 25, 75 mg/kg bw/day were applied to rats and rabbits. In rats, additionally a dose of 225 mg/kg bw/day was used. The local NOAEL for the maternal animals was determined to be 75 mg/kg bw/day and 10 mg/kg bw/day in rats and rabbits, respectively, based on skin irritation effects. In rats of the highest dose group of 225 mg/kg bw/day the body weight gain decreased significantly during the exposure period. No other systemic effects were observed in rats and rabbits. Further, no fetotoxicity or teratogenicity was identified in either study. Thus, the NOAEL for developmental toxicity was in each case the highest dose tested, which was ≥ 225 and ≥ 75 mg/kg bw/day in rats and rabbits, respectively.
These studies show that rabbits were more sensitive to local skin irritation effects of MEA than rats. Thus, the highest exposure concentration was lower for rabbits than for rats. No significant differences with regard to systemic toxicity, fetotoxicity and teratogenicity were observed between rats and rabbits after dermal exposure to MEA.
MEA was also tested in rats by gavage administration in a study according to OECD TG 414. Also in this study, there was no indication of developmental toxicity (BASF 1994).
DEA was also tested in rats and rabbits according to OECD TG 414 via the dermal route (Marty et al. 1999). Doses of 0, 150, 380 (500), 1500 mg/kg bw/day and 0, 35, 100, 350 mg/kg bw/day were applied to rats and rabbits, respectively. The dose of 500 mg/kg bw/day in the study in rats was adjusted to 380 mg/kg bw/day due to dosing discrepancy at the 500 mg/kg bw group. No NOAEL could be determined for the maternal animals in rats due to haematological effects occurring in all treatment groups. The LOAEL for maternal toxicity was 150 mg/kg bw. In the fetuses, there were no effects of treatment on body weight or on the incidence of external, visceral, or skeletal malformations/abnormalities. Thus, the NOAEL for teratogenicity was >1500 mg/kg bw/day.
In rabbits the NOAEL for maternal toxicity was 35 mg/kg bw/day based on an overall decrease in gestational body weight gains in the 100 and 350 mg/kg bw dose groups during the treatment period. In contrast to rats, haematological effects did not occur in rabbits. There was no evidence of developmental toxicity. Consequently, the NOAEL for prenatal developmental toxicity including teratogenicity was 350 mg/kg bw/day. All in all, no significant differences with regard to fetotoxicity and teratogenicity were observed between rats and rabbits after dermal exposure to DEA.
For DEA there are also developmental toxicity studies with gavage and inhalation administration in rats available. In the oral study mortality and decreased body weight occurred in the foetuses at maternal toxic doses (Price 1999). In the inhalation study at the highest concentration (0.2 mg/L) signs of maternal toxicity were observed and were substantiated by adverse clinical symptoms (vaginal hemorrhages) in 8 of the 21 pregnant rats on day 14 p.c. At this dose level a markedly increased number of fetuses with skeletal variations (mainly cervical rib(s)) were also recorded but substance-related teratogenic effects were not detected in any foetus. Thus, signs of prenatal developmental toxicity did only occur at a maternal toxic concentration. There were no adverse effects on dams or fetuses at the low or mid concentrations (0.01 or 0.05 mg/L) (BASF 1993). DMAE was also tested in rats and rabbits in studies similar or according to OECD TG 414 via the oral route (gavage administration). In a modified developmental screening test (BASF 2008) rats were dosed with 0, 300 and 600 mg/kg bw/day once daily from gestational day 6 to 19. The aim of this screening study was to obtain initial information on the effect of the test substance after repeated oral administration (gavage) to pregnant female Wistar rats from gestation day (GD) 6 to GD 19 (prenatal study) and from GD 6 to postnatal day (PND) 3 (postnatal study). For the prenatal study part, selected dams of each group (5 animals of the control group, 5 animals of dose group 1 (300 mg/kg bw/day) and 10 animals of dose group 2 (600 mg/kg bw/day)) were sacrificed on GD 20; dams and fetuses were examined. The following refers to the prenatal study. Maternal effects (some already occurring at 300 and the majority occurring at 600 mg/kg bw/day) included mortality, erosion of the forestomach, reduced food consumption and body weight. Increased resorptions and post-implantation losses were observed at the highest dose group of 600 mg/kg bw/day and in the middle dose group of 300 mg/kg bw/day. However, this was not dose-dependent and not significant. No effects were observed in the two groups receiving 600 or 300 mg/kg bw/day. Based on this 300 mg/kg bw/day was the maternal LOAEL for local effects and the maternal NOAEL for systemic effects. Regarding fetotoxicity, the viability of pups in the high dose group (600 mg/kg bw/day) was lower compared to the control group (viability index 43 % and 100 %, respectively). In addition, reduced body weights were recorded. No effects were observed at 300 mg/kg bw/day. The developmental NOAEL for the pups was 300 mg/kg/day based on reduced viability.
In the PNDT study in rabbits (Millard 2021), doses of 0, 30, 100 and 250 mg/kg bw/day were administered by oral gavage. The maternal NOAEL was determined to be 100 mg/kg bw/day based on mortality, reduced body weight gain/body weight losses and clinical signs at 250 mg/kg bw/day. There were no test substance-related fetal malformations or developmental variations noted at any dosage level. Thus, the NOAEL for developmental toxicity was 250 mg/kg bw/day, the highest dose tested.
In addition, DMAE was tested by inhalation in rats in a developmental toxicity study. No developmental toxic effects were observed up to the highest concentration tested (Leung 1996).
For the substance MDEA a dermal developmental toxicity study in rats is available. No developmental toxic effects were observed up to the highest concentration tested (Leung and Ballantyne 1993).
As evident from all available studies for MEA, DEA and DMAE, no differences between rats and rabbits were identified with regard to developmental toxicity. None of the available studies showed teratogenic effects.
Therefore, also for the registered substance MMEA, it is not expected that the developmental toxicity profile will be different between rats and rabbits. Thus, all in all, the available PNDT in rats is considered to be sufficient to address the endpoint developmental toxicity.
However, since there is evidence that MMEA exerts its toxicity by the same mode-of action like DEA via a choline depletion, the classification of DEA regarding reproductive toxicity (cat. 2; H361fd) was adopted accordingly for MMEA based on a worst-case approach (please refer also to section 7.8.1 Toxicity to reproduction).
Species:
rabbit
Effect on developmental toxicity: via oral route
Endpoint conclusion:
no adverse effect observed
Effect on developmental toxicity: via inhalation route
Endpoint conclusion:
no adverse effect observed
Dose descriptor:
NOAEC
460 mg/m³
Species:
rat
Quality of whole database:
Moderate (2 studies are available: Nelson et al., 1984 and the OECD 422 study).
Effect on developmental toxicity: via dermal route
Endpoint conclusion:
no adverse effect observed
Additional information

Nelson 1984

MMEA was vaporized and administered (test concentration was close to the saturation point) to 18 pregnant rats in one to three concentrations for 7 h/day on gestation days 7 to 15. The dams were sacrificed on day 20 of gestation. Fetuses were individually weighed, and two-thirds of them were fixed in Bouin's solution and examined for soft-tissue anomalies. The other one-third were fixed in alcohol, stained with Alizarin Red and examined for skeletal defects. As overall result for the substance MMEA, neither maternal nor fetal toxicity effects were found. No teratogenicity was observed. The NOAECs were 460 mg/m³, the highest dose tested for both, dams and fetuses.

WoE approach

There is also data available on structural analogues. These are included in this weight-of-evidence approach. The available data for the substances used in the weight-of-evidence approach is described in detail in the waiver. A summary of the available data is shown in table 1.

Table 1: Data included in this weight-of-evidence approach.

substance

species

route

Test method

NOAEL/C P

NOAEL/C F1

effects P

effects F

reference

MMEA

rat

inhal

exposure gd 7- 15

≥ 460 mg/m³

≥ 460 mg/m³

none

none

Nelson 1984

MEA

rat

gavage

acc to 414

120 mg/kg bw/d

≥ 450 mg/kg bw/d

reduced food consumption, lower mean body weights and impaired body weight gain

none

BASF 1994

MEA

rabbit

dermal

similar to 414

10 mg/kg bw/d

≥ 75 mg/kg bw/d

local skin effects

none

Liberacki 1996

MEA

rat

dermal

similar to 414

75 mg/kg bw/d

≥ 225 mg/kg bw/d

local skin effects, decreased bw gain

none

Liberacki 1996

MEA

rat

gavage

similar to 414

300 mg/kg bw/d

≥ 500 mg/kg bw/d

reduced food consumption and bw gain, clinical chemistry

none

BASF 1992

DEA

rat

gavage

exposure gd 6- 19

50 mg/kg bw/d

50 mg/kg bw/d

morbidity, mortality

mortality, decreased bw gain

Price 1999

DEA

rabbit

dermal

similar to 414

35 mg/kg bw/d

≥ 350 mg/kg bw/d

decrease in bw gain

none

Marty 1999

DEA

rat

dermal

similar to 414

< 150 mg/kg bw/d

≥380mg/kg bw/d

hematology

none

Marty 1999

DEA

rat

inhal

acc to 414

0.05 mg/L

0.05 mg/L

bloody vaginal discharge

skeletal variations (mainly cervical rib(s))

BASF 1993

DEA

rat

inhal

DRF

0.2 mg/L

not identified

organ weights, clinical chemistry

not specified

BASF 1991

MDEA

rat

dermal

similar to 414

250 mg/kg bw/d

≥1000 mg/kg bw/d

local skin effects

none

Leung and Ballantyne 1993

MDEA

rat

gavage

acc to OECD 421

100 mg/kg bw/d

300 mg/kg bw/d

decreased bw

decreased bw and viability index

BASF 2010

DMAE

rat

inhal

similar to 414

10 ppm

≥ 100 ppm

reduced body weights, and reduced weight gain, clinical signs of toxicity, involving the eyes with slight, transient ocular changes

none

Leung 1996

DMAE

rat

gavage

modif 421/414

300 mg/kg bw/d

not identified

mortality, food consumption, clinical signs

no findings

BASF 2008

DMAE

rabbit

gavage

acc to OECD 414

100 mg/kg bw/d

250 mg/kg bw/d

mortality, clinical signs, reduced bw gain/food consumption

none

Millard 2021

Since there is evidence that MMEA exerts its toxicity by the same mode-of action like DEA via a choline depletion, the classification of DEA regarding reproductive toxicity (cat. 2; H361fd) was adopted accordingly for MMEA based on a worst-case approach.

Toxicity to reproduction: other studies

Additional information

Zahniser et al. investigated whether or not one could obtain a clear-cut answer to the question whether or not structural and/or metabolic alterations in the brain could be responsible for the death of the N-methylaminoethanol (MMAE) and N,N-dimethylaminoethanol (DMAE) exposed pups, observed in a previous study performed by tha same working group (Katyal and Lombardi, 1978, cited by Zahniser et al., 1978).

Pregnant rats were fed for 15 days predelivery until 15 days postpartum a choline (Ch)-deficient diet (CD diet) or a CD diet supplemented with 0.8 % Ch-CI (CS), 1 % MMAE or 1 % DMAE. Gestation and parturition of the pregnant rats proceeded normally. However, all the pups born of dams fed the MMAE diet, and most of those born of dams fed the DMAE diet, died within 36 h of birth. No histological or cytological alterations were detected in the brain of the pups. Levels of Ch and acetylcholine (ACh) were elevated in the brain of pups born of dams fed the MMEA and DMAE diets, but not the CS diet. It would appear, thus, that presence of MMAE or DMAE in the diet did not stimulate endogenous synthesis of Ch and ACh in adult rats. Moreover, there was no difference in the brain levels of Ch and of ACh between the CS and CD groups of dams. The content of total phospholipids in the brain of the pups was not altered by the diet fed to the dams. However, the phosphatidyl-Ch and phosphatidylaminoethanol (PAE) contents in the brain of the MMAE- and DMAE exposed pups were markedly reduced. At the same time, significant amounts of DMAE, phosphatidyl-N-monomethylaminoethanol (PMME) and of phosphatidyl-N,N-dimethylaminoethanol (PDMAE) were present in the same brain areas. These results are evaluated and discussed in terms of providing a cause for the death of the MMAE- and DMAE-exposed neonatal rats.

It has been concluded that, in the presence of a dietary deficiency of Ch, MMAE increases the demand for methyl groups and is thus more toxic than DMAE which is already two-thirds methylated. The observations support this conclusion in as much as supplementation of the CD diet with 1 % MMAE, but not with 1 % DMAE, appears to compromise the growth of the dams as well, and indicates that availability of methyl groups may be the critical factor. Alterations in brain phospholipid metabolism could also be a likely cause of death of the MMAE- and DMAE-exposed pups.

The inclusion of 1 % MMAE or DMAE in the CD diet of dams limits the survival of pups after birth, most likely by interfering with the development or maturation of one or more vital systems of the fetuses.

Mode of Action Analysis / Human Relevance Framework

Additional Remark:

Reproduction and fertility have also been linked with kidney failure in male rats and kidney functional impairment (Nazian and Dietz, 1987, Menjívar et al., 2000). In these available studies male fertility was reported to be impaired by liver insufficiency as a secondary consequence (e.g. chronic nephrosis, uraemia in consequence of partial nephrectomy). Especially chronic nephrosis as an umbrella term for degenerative tubular kidney disease represents a comparable situation to what has been observed for MMEA. Ortiz et al. (1999) reported decreased male fertility in consequence of chronic nephrosis.

Considering the possible mechanism, effects on the choline-homeostasis could play a role. Various alkanolamines are known to produce choline-deficiency (e.g. diethanolamine DEA CAS 111-42-2).

References:

Menjíar M, Cárdenas M, Ortiz G, Pedraza-Chaverrí J(2000). Fertility Diminution in Female Rats with Experimental Chronic Nephrosis. Biol reproduction 63, 1549–1554

Nazian SJ, Dietz JR (1987). Reproductive Changes during the Early Stages of Chronic Renal Insufficiency in the Mal Rat. Biol of Reproduction 37: 105-111

Ortiz G, Vilchis F, Cárdenas M, Cruz C, Pedraza-Cahverris J, Menjívar M (1999). Reproduciton : Function in Male Rats with Chronic Nephrosis. Journal Reprod and Fertility 117: 223-228

Effects on Choline-homeostasis:

Choline is a vitamin-like compound with various physiological functions (i.e. building block of phospholipids and acetyl-choline, one-carbon-metabolism and DNA-methylation etc.). It could be demonstrated that certain alkanolamines exert an inhibitory effect on either choline-uptake and/or choline-metabolism. Thereby, alkanolamines cause a choline-depletion. A hallmark of choline-depletion is a fatty liver change (Zeisel, 1994). In line with this, liver enlargement concurrent with an increase in absolute absolute and relative liver weight has been reports in all dose groups after MMEA-treatment. Furthermore, minimal fatty changes and central hepatocellular hypertrophy have been observed in parallel. Similar effects have also been reported for DEA, where choline-deficiency caused liver and kidney effects (Melnick, 1992) in repeated dose toxicity tests. In long-term studies with DEA, liver and kidney tumours developed in mice but not in rats (NTP, 1992). In depth investigation on the possible mode-of-action revealed that the liver tumours formation could be attributed to an increase in hepatocellular proliferation probably due to a DEA-induced choline-depletion (Lehman-McKeeman and Gamsky, 2000; Lehman-McKeeman et al., 2002). However, an increase in hepatocellular proliferation has been reported for rodent hepatocytesin vitroonly whereas human primary hepatocytes did not respond. This indicates that rodent cell might be more sensitive and prone towards choline-depletion than human hepatocytes are. Thus, the human relevancy of the findings is questionable as no increase in proliferation was observed in the human hepatocytes (Stott, 2000; Kamendulis and Klaunig, 2005). Therefore, the mode-of-action of MMEA causing kidney lesions might rely on choline-depletion (as reported for various alkanoalmines as well). This mode-of-action has been demonstrated in the context of liver tumour formation to lack human relevance. It is thus concluded that MMEA-induced adverse kidney effects might arise in addition from a toxicokinetic difference with rodents being most sensitive species.This “choline” issue may also be causing some specific effect on membrane integrity that is resulting in a spectrum of toxic effects (anaemia, testes tubule degeneration, and possibly kidney tubule degeneration). This was considered to be part of the DEA toxicity spectrum as well, since some anemia was noted at the high dose levels of the DEA chronic studies.

The dramatic reduction in male and female fertility (10 and 11 %, respectively) occurred in the range of morbidity already in the high dose of 450 mg/kg bw/day. Twenty per cent of the males (2/10) were either found dead (1/10) or had to be sacrificed due a poor general status (1/10). In the mid dose (150 mg/kg bw/day) reduction in fertility was still evident in the presence of adverse and severe kidney effects and haemolytic anaemia. However, no animal died treatment related in the mid dose group.

References:

Kamendulis LM and Klaunig JE (2005).Species differences in the induction of hepatocellular DNA synthesis by Diethanolamnin. Toxicological Sciences 87(2),328-336.

Lehman-McKeeman LD, Gamsky EA (2000). Choline supplementation inhibits diethanolamine-induced morphological transformation in Syrian hamster embryo cells: evidence for a carcinogenic mechanism. Toxicol Sci, 55, 303-10

Lehman-McKeeman, L. D. et al.(2002). Diethanolamine Induces Hepatic Choline Deficiency in Mice. Toxicological sciences 67, 39-45

Melnick R (1992). NTP technical report on the toxicity studies of Diethanolamine (CAS No. 111-42-2) Administered Topically and in Drinking Water to F344/N Rats and B6C3F1 Mice. Toxic Rep Ser. 20:1-D10.

National Toxicology Program (1992). Toxicity Studies of Diethanolamine (CAS No. 111-42-2) Administered Topically and in Drinking Water to F344/N Rats and B6C3F1 Mice. Tech. Rep. Ser. No. 20; NIH Publication No. 92-3343), Department of Health and Human Services, Research Triangle Park, NC. Report no.: TR20.

Stott WT, Bartels MJ, Brzak KA, Mar M, Markham DA, Thornton CM, Zeisel SH (2000).Potential mechanisms of tumorigenic action of diethanolamine in mice. Toxicol. Lett., 114, 67-75.

Zeisel SH and Blasztajn JK (1994). Cholin and human nutrition. Ann. Rev. Nutr. 14: 269-296

Justification for classification or non-classification

N-methylethanolamine caused severely impaired fertility in rats treated at dose levels of 150 and 450 mg/kg bw/d in the Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test (OECD 422; BASF, 2010). Although mating (male and female mating indices) was not influenced no lifeborn pups were delivered for both test groups. Signs of general toxicity were observed in animals of only the highest dose group (450 mg/kg bw). Changed levels of clinical chemistry parameters together with histopathological findings in organs of treated animals points to the systemic toxicity hazard by prolonged exposure. This is a summary of the most relevant findings which are very likely accountable for these effects:

Test group 3 (450 mg/kg body weight/day):

Males (xout of 10 animals):

Target: gonades

·        Testes: tubular degeneration (8 out of 10 animals)

·        Epidymides: oligospermia (9)

 

Target: kidney

·        Kidneys: tubular degeneration (10)

·        Decreased urea clearance (mean of test group)

·        Blood in urine (mean of test group)

 

Target: liver

·        Liver: central fatty change (5)

·        Liver: peripheral fatty change (2)

·        Increased albumin level (mean of test group)

 

Target: blood

·        Spleen: extramedullar haematopoiesis (8)

·        Haemolytic anaemia (mean of test group)

 

Target: stomach

·        Forestomach: erosion/ulceration (3)

·        Glandular stomach: erosion/ulceration (2)

 

Dams (xout of 10 animals):

Target: gonades

·        Ovaries: vacuolization of sex cord stroma (10 out of 10 animals)

 

Target: kidney

·        Kidneys: tubular degeneration (9)

·        Decreased urea clearance (mean of test group)

·        Blood in urine (mean of test group)

 

Target: liver

·        Liver: central hypertrophy (9)

·        Increased albumin level (mean of test group)

 

Target: blood

·        Spleen: extramedullar haematopoiesis (8)

·        Haemolytic anaemia (mean of test group)

 

Target: stomach

·        Forestomach: erosion/ulceration (1)

 

Additional finding

·        Mesenteric lymph nodes: sinus erythrocytosis (5)

 

Test group 2 (150 mg/kg body weight/day):

Males (xanimals):

Target: kidney

·        Kidneys: tubular degeneration (10)

·        Decreased urea clearance (mean of test group)

·        Blood in urine (mean of test group)

 

Target: liver

·        Liver: peripheral fatty change (5 out of 7 animals)

 

Target: blood

·        Haemolytic anaemia (mean of test group)

Dams (xanimals):

Target: gonades

·        Ovaries: vacuolization of sex cord stroma (4 out of 7 animals)

 

Target: kidney

·        Kidneys: tubular degeneration (9)

·        Blood in urine (mean of test group)

 

Target: blood

·        Spleen: extramedullar haematopoiesis (1)

·        Haemolytic anaemia (mean of test group)

 

Additional finding

·        Mesenteric lymph nodes: sinus erythrocytosis (1 out of 2 animals)

 

Test group 1 (50 mg/kg body weight/day):

Males (xanimals):

Target: kidney

·        Kidneys: tubular degeneration (6)

 

The kidneys of males of all treatment groups as well as in females of test groups 2 (150 mg/kg bw/d) and 3 (450 mg/kg bw/d) revealed a minimal to severe tubular degeneration (see next table) which was regarded to be treatment-related. The severity increased dose-dependently:

Tubular degeneration in the kidney:

 

Male animals

Female animals

Test group
(mg / kg bw / day)

0

 

1

(50)

2

(150)

3

(450)

0

 

1

(50)

2

(150)

3

(450)

Number of animals

10

10

10

10

10

10

10

10

Degeneration, tubular

-

6

10

10

-

-

9

9

minimal

-

3

1

1

-

-

9

7

slight

-

3

4

1

-

-

-

2

moderate

-

-

5

6

-

-

-

-

severe

-

-

-

2

-

-

-

-

 

Tubular degeneration in the testes / epididymides:

 

Male animals

Test group

(mg / kg bw / day)

0

1 (50)

2 (150)

3 (450)

Number ofanimals

10

10

10

10

Diffuse tubular degeneration

-

 

7

10

minimal

-

-

7

1

slight

-

-

-

1

moderate

-

-

-

5

severe

-

-

-

3

 

Tubular degeneration in kidneys and testes was dose dependent and assessed as an adverse effect. This might influence also the fertility effects. Furthermore, the effects in kidneys are probably related to anaemia. Since serum bilirubin was high, some amount of haemoglobin in the urine can be present, especially if the kidneys are damaged. It was manifested in discoloured urine of the treated animals.

Vacuolization of sex cord stroma

 

Femaleanimals

Test group

(mg / kg bw / day)

0

1 (50)

2 (150)

3 (450)

Number ofanimals

10

10

10

10

Occurrence of cysts

-

-

-

10

Vacuolization of sex cord stroma

-

-

4

10

 

The occurrence of cysts and vacuolization of sex cord stroma in ovaries was related to treatment and was considered to be adverse. This is another key factor to fertility effects.

Conclusion on classification and labelling:

Based on the findings observed in the Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test (OECD 422; BASF, 2010) in conjunction with the outcome of the above presented Weight-of-Evidence approach classification as toxic for reproduction Cat 2. (H316 fd - suspected of damagingf fertility. Suspected damging the unborn child) is warranted for N-methylethanolamine in accordance with the criteria of EU Classification, Labelling and Packaging of Substances and Mixtures (CLP) Regulations No 1272/2008.

Furthermore, adverse effects on i.e. kidney and anaemia do occur, which trigger the classification with a “hazard category 2” for “Specific Target Organ Toxicity Repeated Exposure” (STOT RE Cat. 2 - H373: May cause damage to organs through prolonged or repeated exposure”. Target organs are the kidney, tests, epididymides, ovaries, liver, and spleen. This classification is warranted for Methyl-Monoethanolamine (MMEA; CAS 109-83-1) according to the criteria of EU Classification, Labelling and Packaging of Substances and Mixtures (CLP) Regulations No 1272/2008.

 

The rationale is as follows:

 

1.Substance and treatment-related adverse effects on kidneys (i.e. tubular degeneration) after repeated dose exposure occur in the range of the low dose (i.e. 50 mg/kg bw/day).

2.Substance and treatment-related adverse effects on blood (i.e. haemolytic anaemia) occurred in the range of the mid and high doses (150 and 450 mg/kg bw/day).

3.The observed effects on fertility trigger a classification and labelling as toxic to male and female fertility and as no offsprings were generated also as toxic to the unborn child. The observed effects on kidney and blood trigger a classification and labelling as a specific target organ toxicant after repeated exposure.

 

Details on the rationale:

Ad 1) MMEA exerted treatment-related and adverse effects on the kidneys in males and females (i.e. tubular degeneration was observed at all doses tested. Additionally, changes in urine parameters were reported that corroborate the impaired functionality of the renal system at 150 and 450 mg/kg bw/day (e.g. occurrence of blood in the urine, reduction in urea clearance). The effects may be secondary to anaemia.

Ad 2) Haemolytic anaemia has been reported to be caused substance- and treatment-related by MMEA. A significant decrease in red blood cell count and in total haemoglobin content could be observed in the mid and high doses. In consequence, a decrease in haematocrit was observed in line with the before mentioned findings representing anaemia. Further indications for a haemolytic anaemia can be derived from the fact that an increase in hemosiderin storage in the spleens has been reported. Additionally, an increase in bilirubin levels was reported to occur in the urine indicative for the haemolytic character of the anaemia.

Ad 3) MMEA is subject for classification for adverse effects on fertility. In the available OECD 422 the adverse effects on fertility were reported to occur in the range of other significant toxic effects (i.e. kidney toxicity and haemolytic anaemia as described above). After evaluation of the data sets of the 4 analogues substances monoethanolamine (MEA; CAS 141-43-5), diethanolamine (DEA; CAS 111-42-2; also known as 2,2’iminodiethanol), methyldiethanolamine (MDEA; CAS 105-59-9) and 2-dimethylaminoethanol (DMAE; CAS 108-01-0) within a weight-of-evidence approach, it became evident that DEA is the most similar substance to MMEA with regard to the toxicological effects and their dose-response relationship. The observed effects for both substances do point to a similar mode-of-action of MMEA and DEA – a choline depletion-mediated toxicity. In the recent Extended One-Generation Reproductive Toxicity Study (EOGRTS) according to OECD TG 443 available for DEA (Extended One-Generation Reproductive Toxicity Study with the developmental neurotoxicity and immunotoxicity (DNT/DIT) cohorts but without the extension of Cohort 1B to mate the F1 animals to produce an F2 generation), the same general toxicty effects were observed. In detail, DEA also induced anaemia in the highest dose group of 128 mg/kg bw/d in males and females of the F0 and F1 generation. Furthermore, signs of degeneration and regeneration in the proximal tubules of the kidneys and a fatty change in the liver were observed in the mid- and high-dose-group). Thus, the classification of DEA regarding reproductive toxicity (cat. 2; H361fd) was adopted accordingly for MMEA.

Furthermore, MMEA should be classified for specific target organs toxicity (STOT RE Cat. 2; H373); affected organs are kidney, testes, epididymides, ovaries, liver and spleen.

 

Besides the substance is classified as:

GHS:

- Acute toxicity - oral: Cat 4, H302:harmful if swallowed

- Acute toxicity -dermal: Cat 4, H312:harmful in contact with skin.

-Skin Corr.1B, H314:causes severe skin burns and eye damage

- Eye Damage 1:H318:causes serious eye damage.

- STOT SE Cat.3, H335:may cause respiratory irritation, C>=5%

and

- STOT RE Cat 2,H373: May cause damage to organs <or state all organs affected, if known> through prolonged or repeated exposure <state route of exposure if it is conclusively proven that no other routes of exposure cause the hazard>; Affected organs: other: the kidney, testes, epidymides, ovaries, liver, and spleen).

Additional information