IPCS INCHEM Home



    PESTICIDE RESIDUES IN FOOD - 1997


    Sponsored jointly by FAO and WHO
    with the support of the International Programme
    on Chemical Safety (IPCS)




    TOXICOLOGICAL AND ENVIRONMENTAL
    EVALUATIONS 1994




    Joint meeting of the
    FAO Panel of Experts on Pesticide Residues
    in Food and the Environment
    and the
    WHO Core Assessment Group 

    Lyon 22 September - 1 October 1997



    The summaries and evaluations contained in this book are, in most
    cases, based on unpublished proprietary data submitted for the purpose
    of the JMPR assessment. A registration authority should not grant a
    registration on the basis of an evaluation unless it has first
    received authorization for such use from the owner who submitted the
    data for JMPR review or has received the data on which the summaries
    are based, either from the owner of the data or from a second party
    that has obtained permission from the owner of the data for this
    purpose.



    FENAMIPHOS

    First draft prepared by
    S. Geertsen
    Health Evaluation Division, Pest Management Regulatory Agency
    Health Canada, Ottawa, Ontario, Canada

         Explanation 
         Evaluation for acceptable daily intake
              Biochemical aspects
                   Absorption, distribution, excretion
                   Biotransformation
                   Effects on enzymes and other biochemical parameters
              Toxicological studies
                   Acute toxicity
                   Short-term toxicity
                   Long-term toxicity and carcinogenicity
                   Genotoxicity
                   Reproductive toxicity
                        Multigeneration reproductive toxicity
                        Developmental toxicity
                   Special studies
                        Dermal and ocular irritation and dermal
                        sensitization
                        Delayed neuropathy
                        Neurotoxicity
                        Potentiation
              Comments
              Toxicological evaluation
              References

    Explanation

    Fenamiphos was first evaluated for toxicological effects by the JMPR
    in 1974 (Annex 1, reference 22), at which time an ADI of 0-0.0006
    mg/kg bw was established. In 1985, following a direct request for
    re-evaluation by a Member State, additional data were reviewed and the
    ADI was reduced to 0-0.0003 mg/kg bw and made temporary because of
    concern about fetotoxicity seen in a study in rabbits (Annex 1,
    reference 44). The 1985 JMPR requested submission of the results of an
    on-going study of oncogenicity in rats, a full, legible report and raw
    data from the study of developmental toxicity in rats, and a new study
    of developmental toxicity in rabbits to clarify the observation of
    fetotoxicity at low dietary levels. The results of these studies were
    considered by the 1987 JMPR (Annex 1, reference 50), which established
    an ADI of 0-0.0005 mg/kg bw. Fenamiphos was evaluated at the present
    Meeting within the CCPR periodic review programme.

    Evaluation for acceptable daily intake

    1.  Biochemical aspects

     (a)  Absorption, distribution, and excretion

    Wistar rats were given 3 mg/kg bw 14C-fenamiphos (purity, > 99%;
    specific activity, 62.6 µCi/mg; labelled in the benzene ring), and one
    rat was sacrificed 0.5, 2, 8, 24, and 48 h after treatment. Within 0.5
    h, radiolabel was distributed throughout the body, except for compact
    bone structures, the spinal marrow, and the brain, indicating minimal
    ability to cross the blood-brain barrier. The highest concentrations
    were seen in the stomach contents, some segments of the small
    intestine, the kidney, and the urinary bladder, indicating very rapid
    urinary excretion. High concentrations were also found in the liver;
    this, and its presence in the intestines, indicate a biliary-faecal
    route of elimination. Blood, lungs, salivary glands, parotids,
    hypophysis, and tissues with large amounts of connective tissue also
    had relatively high concentrations of radiolabel. Less was found in
    the lymph, adrenal gland, and spleen, and only low concentrations
    occurred in the musculature, fat, pancreas, and thymus. Over the
    course of the study, radiolabel was also found in the region of the
    hair follicles, suggesting slight elimination via this route.
    Fenamiphos was rapidly cleared from the body (little remained within 8
    h) with no evidence of tissue accumulation (Weber, 1988).

    In a study to assess the absorption, distribution, metabolism, and
    excretion of fenamiphos in Wistar rats, 14C-labelled compound
    (purity, > 99%; 62.6 µCi/mg; labelled in the benzene ring) was
    administered to groups of five rats of each sex in one of the
    following regimens: a single intravenous dose of 0.3 mg/kg bw labelled
    fenamiphos; a single oral dose of 0.3 mg/kg bw labelled fenamiphos; 14
    daily oral doses of 0.3 mg/kg bw unlabelled fenamiphos followed by a
    single oral dose of 0.3 mg/kg bw labelled fenamiphos; or a single oral
    dose of 3 mg/kg bw labelled fenamiphos. Urinary and faecal excretion
    were monitored for 48 h, when the animals were killed and tissue
    residue levels determined. Elimination of labelled carbon dioxide was
    monitored only in males given the high dose. The metabolites in the
    excreta were identified and quantified. Fenamiphos was rapidly
    excreted, 54-85% of the radiolabel being excreted renally within 4 h
    of treatment. After 48 h, >96% of the recovered radiolabel had been
    excreted by animals at all doses. Faecal elimination accounted for
    only 1.5-3.7% of the recovered radiolabel. Small amounts were found in
    the faeces after intravenous administration, indicating that a small
    amount of biliary excretion occurs. Negligible amounts (< 0.1%) of
    radiolabel were eliminated as carbon dioxide in the expired air. At 48
    h, most of the levels in tissues were below the limit of
    quantification, except in the animals given the high dose, in which
    the maximum tissue residue levels were seen in the liver (8.4 ppb in
    females, 3.5 ppb in males), kidney (2.1 ppb in males, 1.6 ppb in
    females), and skin (3.5 ppb in males, 1.6 ppb in females). Urinary
    excretion appeared to be slightly faster in males than females after
    oral dosing, faster after intravenous than oral dosing, and faster

    after the high than the low dose in females only. Prior treatment with
    unlabelled fenamiphos had no effect on the excretion characteristics
    (Ecker et al., 1989).

     (b)  Biotransformation

    The metabolic fate of labelled fenamiphos was studied in rats 
     in vivo and in rat liver microsomes  in vitro. The compounds were
    labelled with 14C in the ethyl or isopropyl position or with 3H in
    the thiomethyl position. Fenamiphos was excreted within 12-15 h after
    a single oral dose of 2 mg/kg bw.  In vitro a small quantity of an
    unknown metabolite, possibly resulting from the N-dealkylation of
    fenamiphos, was observed (Khasawinah & Flint, 1972). Apart from this
    minor component, metabolism in animals and plants followed the same
    pattern: oxidation of the thioether to the sulfoxide and sulfone,
    dearylation to yield the methyl thioether phenol (or its sulfoxide and
    sulfide), and potential dealkylation of the ethyl, isopropyl, or
    isopropylamino moiety of the phosphate ester. Treatment of rats with
    fenamiphos sulfoxide or sulfone produced the same excretion pattern
    and almost identical urinary metabolites (Gronberg, 1969; JMPR, 1974).

    Ecker et al. (1989; see above) also quantified the metabolites in the
    urine of all groups and in the faeces of animals at the high dose. The
    primary urinary metabolites (Figure 1) were found to be fenamiphos
    sulfoxide phenol sulfate (40-54%), fenamiphos sulfoxide phenol
    (4-22%), fenamiphos phenol sulfate (5-20%), and fenamiphos sulfone
    phenol sulfate (4-15%). Minor metabolites included fenamiphos sulfone
    phenol (2-10%), fenamiphos phenol (1-10%), fenamiphos sulfoxide
    (0-12%),3-hydroxymethyl fenamiphos sulfone phenol sulfate (0-11%), and
    desisopropyl fenamiphos sulfoxide (0-1.7%). About 60% of the faecal
    metabolites of animals at the high dose were identified and found to
    be restricted to fenamiphos sulfoxide, fenamiphos sulfoxide phenol,
    fenamiphos sulfone phenol, and fenamiphos phenol sulfate. No parent
    compound was identified in either urinary or faecal extracts. More
    than 93% of the radiolabelled metabolites were identified.
    Qualitatively, the metabolic profile was largely unaffected by sex,
    dose, route, or frequency of treatment. Quantitatively, there was
    greater production of fenamiphos phenol sulfate after intravenous
    administration (16-19%) than after oral administration (5-8%) (Ecker
    et al., 1989).

     (c)  Effects on enzymes and other biochemical parameters

    Fenamiphos, like other organophosphate esters, inhibits cholinesterase
    enzymes. The concentrations that inhibited activity by 50%  in vitro 
    were 5.1 × 10-5 mol/L in rat serum, 6.3 × 10-4 mol/L in rat
    erythrocytes, and 2.1 × 10-4 mol/L in rat brain. The maximum
    inhibition of whole-blood cholinesterase in rats  in vivo occurred
    3 h after treatment. The sensitivity of cholinesterase  in vivo 
    reflects the values for inhibition  in vitro, plasma cholinesterase
    being more sensitive than the erythrocyte enzyme (Löser & Kimmerle,
    1971; JMPR, 1974). The metabolites of fenamiphos are more active
    inhibitors than the parent molecule (Waggoner, 1972; JMPR, 1974). The

    FIGURE 1

    inhibitory activity of fenamiphos and its metabolites in horse serum
    cholinesterase  in vitro was in the order fenamiphos < sulfoxide =
    sulfone < unidentified metabolite.

    The concentrations that inhibited the activity of chicken and monkey
    liver aliesterase (triacetin hydrolysis) and monkey cholinesterase by
    50%  in vitro were 4.2 × 10-5 mol/L for chicken aliesterase, 1.0 ×
    10-6 mol/L for monkey aliesterase, and 4.6 × 10-6 mol/L for monkey
    cholinesterase (Coulston & Wills, 1974).

    Male albino rats were painted dermally on the clipped dorsal area with
    100-400 µg/cm2 fenamiphos, 25-800 µg/cm2 fenamiphos sulfoxide, or
    200-1600 µg/cm2 fenamiphos sulfone. The test materials were applied
    in acetone and allowed to dry. After 72 h, the animals were killed and
    erythrocyte acetylcholinesterase activity determined. Fifty percent
    inhibition was achieved with doses of 208 µg/cm2 fenamiphos, 262
    µg/cm2 fenamiphos sulfoxide, and 750 µg/cm2 fenamiphos sulfone
    (Knaak et al., 1981; JMPR, 1985).

    Blood samples were extracted from equal numbers of male and female
    Sprague-Dawley rats and then pooled for comparison of the
    cholinesterase inhibition induced by fenamiphos and five of its
    metabolites (fenamiphos sulfoxide, desisopropyl fenamiphos sulfoxide,
    fenamiphos sulfone, desisopropyl fenamiphos sulfone, and desisopropyl
    fenamiphos)  in vitro. Samples were incubated for 1 h before
    determination of the cholinesterase activity (Table 1). Erythrocyte
    acetylcholinesterase was less sensitive to fenamiphos and its
    metabolites than was plasma cholinesterase (Lamb & Landes, 1978; JMPR,
    1985).

    In order to assess the effects of fenamiphos on neurotoxic esterase
    activity, single doses of 0 or 25 mg/kg bw technical-grade fenamiphos
    (purity; 91.3%) were given by oral intubation to groups of nine
    atropinized Lohmann selected Leghorn hens. Groups of three surviving
    hens were killed one, two, and seven days after treatment. Even under
    atropine protection, one hen died within 24 h of treatment. Fenamiphos
    had no effect on neurotoxic esterase activity in the brains or spinal
    cords, whereas the positive control compound, tri- ortho-cresyl
    phosphate, had almost completely inhibited the activity 24 and 48 h
    after treatment (Flucke & Eben, 1988).

    2.  Toxicological studies

     (a)  Acute toxicity

    The results of studies on the acute toxicity of fenamiphos and its
    metabolites are summarized in Table 2. Impurities identified as
    components of the technical mixture (aryldiamide, diarylamide, diaryl
    ethyl ester, diethyl ester, diethylmonamide, di-SCH3 compound, ethyl
    aryl ester, ethyldiamide, and 4-methylthio- meta-cresol) were tested
    for their acute toxicity to rats. At doses 1.5 times the acute LD50
    of fenamiphos, none of the materials was toxic (Crawford & Anderson,
    1973; JMPR, 1974).


        Table 1. Inhibition of cholinesterase (%) by fenamiphos and its metabolites in vitro

                                                                                                                                         

    Compound                             Plasma                                          Erythrocytes
                                         (Dose: ppm in whole blood)                      (Dose: ppm in whole blood)
                                                                                                                                         
                                         6.88     68.8      688      5440      6880      6.88      68.8      688       5440      6880
                                                                                                                                         

    Fenamiphos                           -        0         10       49        69        -         0         0         0         23
    Fenamiphos sulfoxide                 18       41        48       -         85        5         0         5         -         52
    Fenamiphos sulfone                   0        13        50       -         87        8         -         -         -         -
    Desisopropyl fenamiphos sulfoxide    6        40        90       -         93        4         0         47        -         50
    Desisopropyl fenamiphos sulfone      0        20        69       -         90        6         0         32        -         61
    Desisopropyl fenamiphos              -        2         13       71        91        -         8         0         32        53
                                                                                                                                         

    -, not determined

    Table 2. Acute toxicity of fenamiphos, its metabolites, and impurities

                                                                                                                             

    Species         Route               Sex     Purity       LD50/LC50       Reference
                                                (%)          (mg/kg bw
                                                             or µg/L)
                                                                                                                             

    Fenamiphos

    Mouse           Oral                M       NR           22.7            Loser & Kimmerle (1971)
                    Intraperitoneal     M/F     80           3.4             DuBois et al. (1967)
                    Inhalation          M       NR           approx. 60      Kimmerle & Solmecke (1971)
                    (1 h static)

    Rat             Oral (fasted)       M       88-99.7      2.4-6.0         Crawford & Anderson (1973, 1974a);
                                        F                    2.4-6.1         Lamb & Matzkanin (1975); Mihail (1980);
                                                                             Heimann (1981, 1984, Krotlinger (1988)
                    Oral                M       80-90.2      8.1-17.2        DuBois et al. (1967); Loser & Kimmerle (1971);
                    (not fasted)        F                    9.6-19.4        Kimmerle & Solmecke (1971); Kimmerle (1972a);
                                                                             Heimann (1981, 1984)
                    Dermal              M       80-92.2      72-73           Dubois et al. (1967); Flucke (1980)
                                                             84-92
                    Dermal              M       NR           approx. 500     Kimmerle & Solmecke (1971)
                    Inhalation          M       89.8         110-175         Kimmerle (1972b); Kimmerle & Solmecke
                                        (1 h)   F            130-150         Thyssen (1979a)
                    Inhalation          M       89.8         91-100          Kimmerle & Solmecke (1971); Thyssen (1979a)
                    (4 h)                                    100
                    Intraperitoneal     M       80           3.0-3.7         DuBois et al. (1967); Loser & Kimmerle (1971);
                                        F                    4.2-4.9         Kimmerle & Solmecke (1971)
    Guinea-pig      Oral                M       NR           56-100          DuBois et al. (1967); Loser & Kimmerle (1971);
                                                                             Kimmerle & Solmecke (1971)
                    Intraperitoneal     M       NR           17.3            DuBois et al. (1967)
    Rabbit          Oral                M       NR           10-17.5         Loser & Kimmerle (1971); Kimmerle &
                                                                             Solmecke (1971)
                    Dermal              M       NR           225             Crawford & Anderson (1972)
                                        F                    179
    Cat             Oral                M       NR           approx. 10      Loser & Kimmerle (1971); Kimmerle &
                                                                             Solmecke (1971)

    Table 2. (continued)

                                                                                                                             

    Species         Route               Sex     Purity       LD50/LC50       Reference
                                                (%)          (mg/kg bw
                                                             or µg/L)
                                                                                                                              

    Dog             Oral                M       NR           approx. 10      Loser & Kimmerle (1971); Kimmerle &
                                                                             Solmecke (1971)
    Chicken         Oral                F       NR           5.3-12          Loser & Kimmerle (1971); Kimmerle &
                                                                             Solmecke (1971); DuBois et al. (1967)

    Fenamiphos sulfone
    Rat             Oral (fasted)       M       NR           2.6             Crawford & Anderson (1974b)
                                        F                    2.4
                    Oral                F       NR           1-25            Thyssen (1974a)
                    (not fasted)

    Fenamiphos sulfoxide
    Rat             Oral (fasted)       M/F     NR           2.4             Crawford & Anderson (1974b)
                    Oral                F       NR           10-25           Thyssen (1974b)
                    (not fasted)

    Desisopropylfenamiphos
    Rat             Oral (fasted)       M       NR           1.4             Lamb & Matzkanin (1977)
                                        F                    2.1

    Desisopropylfenamiphos sulfone
    Rat             Oral (fasted)       M       95           4.1             Lamb & Matzkanin (1975)
                                        F                    3.7

    Fenamiphos sulfoxide phenol
    Rat             Oral (fasted)       M       99           1418            Crawford & Anderson (1974a)
                                        F                    1175
                    Oral                F       NR           500-1000        Thyssen (1974c)
                    (not fasted)

    Table 2. (continued)

                                                                                                                             

    Species         Route               Sex     Purity       LD50/LC50       Reference
                                                (%)          (mg/kg bw
                                                             or µg/L)
                                                                                                                             

    Fenamiphos sulfone phenol
    Rat             Oral (fasted)       M       95           1250            Crawford & Anderson (1974a)
                                        F                    1854
                    Oral                F       NR           > 1000          Thyssen (1974d)
                    (not fasted)

    4-Methylthio-meta-cresol
    Rat             Oral (fasted)       M       96.4         1418            Crawford & Anderson (I974a)
                                        F                    1333
                    Oral                F       NR           > 2500          Thyssen (1974e)
                    (not fasted)
                                                                                                                             

    NR, not reported
    

    Fenamiphos (purity, 97%) was given orally to fasted Wistar (Bor:WISW
    (SPF-Cpb) rats at doses of 1-8 (females) or 1-100 (males) mg/kg bw.
    All deaths among males at 100 mg/kg bw occurred within 10 min, and
    those among animals receiving 5 mg/kg bw or more within 1.5 h.
    Clinical signs of toxicity noted at doses >4 (males) or 5 (females)
    mg/kg bw included apathy, palmospasms, laboured breathing, diarrhoea,
    piloerection, clonic cramps, and dyspnoea. Females at does > 1
    mg/kg bw had only diarrhoea (Krötlinger, 1988).

    Studies in which rodents were given fenamiphos by inhalation as a
    static spray at doses up to 230 µg/L for 1-4 h showed that rabbits and
    guinea-pigs are more tolerant of the acute effects than rats and mice
    (Kimmerle & Solmecke, 1971).

    Male and female TNO/W74 albino rats were exposed to aerosolized
    fenamiphos (purity, 89.8%) for 4 h per day on five consecutive days at
    concentrations of 0, 0.3, 0.6, 3.3, 4, 9, or 28 µg/L. Plasma and
    erythrocyte cholinesterase activities were measured before exposure,
    after the first, third, and fifth exposures, and 72 h after the fifth
    exposure; brain acetylcholinesterase activity was not measured. The
    cholinesterase activities were depressed in a dose-related manner;
    that of plasma cholinesterase was the most significantly depressed,
    and females were more sensitive than males. Plasma cholinesterase
    activity was 32-90% lower than before treatment in males at > 3.3
    µg/L and 31-96% lower in females at > 0.3 µg/L. The activity
    remained depressed by 19-44% for 72 h after the fifth exposure in
    females at concentrations of > 9 µg/L. Erythrocyte
    acetylcholinesterase activity in males was inhibited by < 20% at
    > 9 µg/L; however, that in females was depressed by up to 28% at 9
    µg/L and 33% at 28 µg/L (JMPR, 1985, slightly modified by reference to
    the original report of Thyssen, 1979b).

    In male rats, administration of atropine and/or 2-pralidoxime or
    obidoxime after poisoning reduced the acute lethal dose by a factor of
    about two. As with other organophosphorus compounds, rapid
    administration of atropine and oxime reactivator after poisoning
    affords some protection and alleviates the cholinergic signs of
    poisoning (DuBois et al., 1967; Kimmerle, 1972c; JMPR, 1974).

     (b)  Short-term toxicity

     Rats

    Three groups of 10 male and 10 female albino Wistar TNO/W 74 rats were
    exposed to technical-grade fenamiphos (purity, 92.2%) diluted with a
    1:1 mixture of ethanol and polyethylene glycol 400 for aerosolization
    in a dynamic flow inhalation chamber at doses of 0, 0.03, 0.25, or 3.5
    µg/L for 6 h per day, five days per week for three weeks; 98% of the
    particles were 3 µm or less. No toxic signs or effects on mortality,
    body-weight gain, or the results of haematology, urinalysis, or
    clinical chemistry were seen. A significant decrease (48-79%) in
    plasma cholinesterase activity and a slight decrease (9-18%) in
    erythrocyte acetylcholinesterase activity were seen in animals of each

    sex at 3.5 µg/L; brain acetylcholinesterase activity was not affected.
    There were no gross or histopathological changes or effects on organ
    weights. The NOAEL was 3.5 µg/L (Thyssen, 1979b).

    Groups of 15 male Sprague-Dawley rats were given fenamiphos orally or
    by intraperitoneal injection on five days per week for 60 days. All of
    the animals survived doses ranging from 1.5 mg/kg bw per day
    intraperitoneally to 1.7 mg/kg bw per day orally with no cumulative
    toxicological effect (Kimmerle & Solmecke, 1971; JMPR, 1974). In
    another study, female rats survived daily intraperitoneal
    administration of 1 mg/kg bw per day for 60 days, while 40% of those
    given 2 mg/kg bw per day and all rats at 3 mg/kg bw per day died
    (DuBois & Flynn, 1968; JMPR, 1974). These studies indicate little if
    any cumulative toxicity.

    Groups of 20 Fischer 344 rats of each sex were given diets containing
    fenamiphos (purity, 89%) at 0, 0.37, 0.57, or 0.91 ppm (equal to 0,
    0.03, 0.045, or 0.072 mg/kg bw per day for males and 0, 0.035, 0.053,
    or 0.084 mg/kg bw per day for females) for three months. Animals were
    monitored for clinical signs of toxicity, mortality, abnormality,
    masses, food consumption, and body weight throughout the study.
    Cholinesterase activity in plasma and erythrocytes was determined in
    10 rats of each sex per group at 5, 9, 12, and 14 weeks and in brain
    at termination of the study. No effects attributable to treatment were
    seen on food consumption or body weight or during clinical
    observation. Statistically but not toxicologically significant
    decreases in both plasma and erythrocyte cholinesterase activity were
    observed in treated rats of each sex when compared with controls. The
    maximal inhibition of plasma cholinesterase was 17% at week 14 in
    males at 0.37 ppm, with lesser inhibition at higher doses, and 24% at
    week 9 in females at the high dose. Erythrocyte acetylcholinesterase
    activity was inhibited by < 10% in all cases. No toxicologically
    significant, treatment-related change was seen in brain
    acetylcholinesterase activity. It was concluded that no biologically
    significant cholinesterase activity inhibition had occurred in this
    study. The NOAEL was 0.9 ppm, equal to 0.07 mg/kg bw per day, the
    highest dose tested (JMPR, 1987, slightly modified by reference to the
    original report of Hayes, 1986a).

    Groups of Wistar SPF rats of each sex (15 per treated group, 30
    controls) were fed diets containing fenamiphos (purity, 82%) at 0, 4,
    8, 16, or 32 ppm (equivalent to 0.2, 0.4, 0.8, or 1.6 mg/kg bw per
    day) for three months. Male and female rats at the highest dose showed
    signs of cholinergic stimulation during the first two months; no
    behavioural changes were seen in the other animals. Average food
    consumption and growth were similar for treated and control animals.
    The only effects on the haematological parameters examined were
    decreased plasma cholinesterase activity (> 20%) in animals at 8 ppm
    and inhibition of erythrocyte acetylcholinesterase activity (< 12%)
    in those at 8 ppm throughout the study, with peak inhibition (56%) at
    16 ppm by week 8. Gross and microscopic examination of tissues and
    organs at the end of the experiment showed slightly increased liver
    weights in males at 16 or 32 ppm, which was not reflected in the

    calculated relative organ:body weight ratios or on histological
    examination. Brain acetylcholinesterase activity was not measured
    (JMPR, 1974, slightly modified by reference to the original reports of
    Löser, 1968a; Mawdesley-Thomas & Urwin, 1970a). Although the NOAEL was
    originally considered to be 4 ppm on the basis of inhibition of plasma
    cholinesterase activity (JMPR, 1974), the present Meeting decided that
    the NOAEL was 8 ppm, equivalent to 0.4 mg/kg bw per day, on the basis
    of inhibition of erythrocyte acetylcholinesterase activity.

     Rabbits

    An aqueous formulation of technical-grade fenamiphos (purity, 89.8%)
    was applied to a clipped dorsal area of groups of six male and six
    female New Zealand rabbits at doses of 0, 2.5, or 10 mg/kg bw per day
    for 6 h per day, five days per week for three weeks. Two additional
    groups were similarly treated at 0 and 0.5 mg/kg bw per day. The skin
    of half of the animals in each group was abraded. Haematology,
    clinical chemistry, and urinalysis were performed before treatment and
    at the end of the study. Plasma and erythrocyte cholinesterase
    acetylcholinesterase activities were measured before treatment and
    after the tenth and last exposures.

    No signs of toxicity or mortality were observed. Although the authors
    concluded that body-weight gains were decreased in animals of each sex
    at 10 mg/kg bw per day (JMPR, 1985), the differences were less than
    10% and were not statistically significant. Furthermore, the range of
    individual weight gains seen in animals at the high dose was well
    within that observed in the controls. Slight erythema was observed in
    all groups only at the abraded skin sites during the initial week,
    which cleared by day 7. There were no apparent differences in
    haematological, urinary, or clinical chemical parameters between test
    and control groups. Gross necropsy, histopathology, and organ weight
    measurements showed no remarkable changes in comparison with controls.
    Plasma and erythrocyte cholinesterase activity was significantly
    depressed (by up to 65 and 34%, respectively) in male and female
    rabbits at 10 mg/kg bw per day; however, the females were somewhat
    more sensitive, with depressed plasma cholinesterase activity (by
    22-31%) at 2.5 mg/kg bw per day as well. Brain acetylcholinesterase
    activity was statistically significantly depressed in females at 2.5
    mg/kg bw per day (by 19%) and 10 mg/kg bw per day (by 21%). The NOAEL
    was 0.5 mg/kg bw per day (JMPR, 1974, slightly modified by reference
    to the original report by Mihail & Schilde, 1980).

     Dogs

    Groups of two beagle dogs of each sex were fed fenamiphos (purity,
    82%) in the diet at 0, 2, 6, or 18 ppm (equivalent to 0.05, 0.15, or
    0.45 mg/kg bw per day) for three months. Behavioural abnormalities
    with signs of cholinergic stimulation were evident in animals at 18
    ppm, and the growth of females at this dose was reduced.
    Haematological and clinical chemical parameters, including the results
    of tests for liver and kidney function and urinalyses, were not
    affected by treatment. The averages values for plasma and erythrocyte

    cholinesterase activity were depressed in males and females at 6 ppm;
    in animals at 2 ppm, no effect was found on average values for
    erythrocyte acetylcholinesterase activity, while those for plasma
    cholinesterase were depressed (20-30%). Brain acetylcholinesterase
    activity was not measured. Gross morphological examination of tissues
    and organs at the end of the study showed no abnormal effects (JMPR,
    1974, slightly modified by reference to the original report by Löser,
    1968b).

    Groups of two male and two female beagle dogs (three of each sex as
    controls) were fed fenamiphos (purity, 99.4%) in the diet at levels of
    0, 1, 2, or 5 ppm (equivalent to 0.025, 0.05, or 0.13 mg/kg bw per
    day) for three months. Treatment at < 5 ppm in the diet had no
    effect on the average values of haematological parameters, liver
    function tests, clinical chemistry, or kidney function tests or on the
    gross or microscopic appearance of tissues and organs. As in other
    studies, cholinesterase activity was the only parameter significantly
    affected, the females being more susceptible than the males and plasma
    cholinesterase activity being more sensitive to inhibition than that
    of erythrocytes. The latter was unchanged at 2 ppm while plasma
    cholinesterase activity was slightly, transiently depressed; 1 ppm
    fenamiphos in the diet had no effect on plasma cholinesterase
    activity. Brain acetylcholinesterase activity was not measured (Löser,
    1969; Mawdesley-Thomas & Urwin, 1970b; JMPR, 1974).

    As an extension of the previous study, an additional two dogs of each
    sex were fed fenamiphos(purity, 99.4%) in the diet at 0 or 10 ppm
    (equivalent to 0.25 mg/kg bw per day) for three months. No deaths
    occurred during the experiment, although a very slight deviation in
    the average body weight of treated animals was noted. Because of the
    small number of animals, the slight weight differences cannot be fully
    evaluated. There was no effect on haematological or urinary
    parameters, clearance, blood sugar, or cholesterol levels.
    Cholinesterase activity was significantly depressed in both plasma and
    erythrocytes in male and female dogs; brain acetylcholinesterase
    activity was not measured. Gross and microscopic examination of
    tissues and organs showed no significant differences between treated
    and control animals (Löser, 1970; Thomson et al., 1972a; JMPR, 1974).

    Groups of four male and four female, four-month-old beagle dogs were
    fed diets containing fenamiphos (purity, 89%) at doses of 0, 0.6, 1,
    or 1.7 ppm (equivalent to 0.015, 0.025, or 0.042 mg/kg bw per day) for
    three months. They were observed twice daily, and body weight and food
    consumption were recorded weekly. Plasma and erythrocyte
    cholinesterase activity was determined at 0, 4, 6, 8, 10, and 12
    weeks; brain acetylcholinesterase was determined at termination of the
    study. Haematology, clinical chemistry, and urinalysis were not
    performed, and tissues and organs were not examined grossly or
    histologically. No toxicological symptoms or effects on body weight
    and food consumption were seen; however, plasma cholinesterase
    activity was depressed by 28-35% in males given 1.7 ppm in the diet.
    The author concluded that females at this dose were not similarly
    sensitive (JMPR, 1985), on the basis of the < 20% inhibition relative

    to control levels; however, when inhibition was calculated over the
    course of the study, the reductions in plasma cholinesterase activity
    in females at all doses were 2-14% in controls, 13-23% in animals at
    0.6 ppm, 18-28% at 1 ppm, and 31-41% at 1.7 ppm, which were comparable
    to those in males: 10-15% in controls, 17-23% in dogs at 0.6 ppm,
    20-29% at 1 ppm, and 34-41% at 1.7 ppm. Erythrocyte and brain
    acetylcholinesterase activities were unaffected (JMPR, 1985, slightly
    modified by reference to the original report by Hayes, 1983).

    Technical-grade fenamiphos (purity, 88.9%) was administered in the
    diet to groups of four male and four female beagle dogs at
    concentrations of 0, 1, 3, or 12 ppm (equal to 0, 0.03, 0.089, or 0.31
    mg/kg bw per day in males and 0, 0.03, 0.083, and 0.35 mg/kg bw per
    day in females) for one year. The animals were observed for mortality,
    clinical signs, body weight, food consumption, ophthalmic alterations,
    and haematological, urinary, and clinical chemical parameters. Plasma
    and erythrocyte cholinesterase activity was measured before treatment
    and quarterly thereafter. One-half of each brain was taken at necropsy
    for measurement of acetylcholinesterase activity. At necropsy, 11
    organs were taken from each dog and weighed, and about 40 tissues from
    each dog were examined grossly and histopathologically.

    Treatment did not affect survival, clinical signs, growth, food
    consumption, ophthalmoscopic or urinary parameters, or organ weights.
    Them were no gross or histopathological findings that could be
    attributed to treatment. Plasma cholinesterase activity was
    statistically significantly inhibited relative to the levels before
    treatment in both males and females in a dose-related fashion at doses
    > 1 ppm: 1 ppm, 20-32%; 3 ppm, 41-53%; 12 ppm, 54-65%; the control
    values were variably reduced by up to 14% over the course of the
    study. Erythrocyte acetylcholinesterase activity was also
    statistically significantly decreased in both males and females at
    3 ppm (by 17-36%) and 12 ppm (by 58-68%); the activity in the controls
    and dogs at 1 ppm was variably, statistically nonsignificantly reduced
    by 5-24% and 10-23%, respectively, during the course of the study.
    Brain acetylcholinesterase activity was nonsignificantly decreased by
    12% in males and sigificantly reduced by 17% in females at 12 ppm.
    Males at this dose also had mild, transient anaemia characterized by
    significantly decreased erythrocyte counts, haemoglobin
    concentrations, and haematocrit, with a concomitant increase in mean
    corpuscular volume. The authors concluded that there was no NOAEL
    because of the effects on plasma cholinesterase activity at the lowest
    dose (Riethet al., 1991). Owing to the variability in the activity of
    plasma cholinesterase in controls seen in a six-month follow-up (Jones
    & Loney, 1993; see below), the authors reconsidered the results of the
    one-year study and decided that the changes seen at 1 ppm were within
    the normal biological range seen in historical controls (Jones &
    Greufe, 1993). The Meeting agreed that the NOAEL was 3 ppm, equal to
    0.083 mg/kg bw per day, on the basis of decreased brain
    acetylcholinesterase activity and anaemia at the highest dose.

    In the supplemental study (Jones & Loney, 1993), technical-grade
    fenamiphos (purity, 89%) was administered in the diet to groups of
    four male and four female beagle dogs at concentrations of 0 or 0.5
    ppm, equal to 0.011 mg/kg bw per day, for six months. The only
    parameters measured were mortality, clinical and ophthalmologic signs,
    body weight, food consumption, and plasma and erythrocyte
    cholinesterase activity. No signs of toxicity were observed.

    Groups of four male and four female pure-bred beagle dogs were fed
    fenamiphos in the diet at levels of 0, 0.5, 1, 2, 5, or 10 ppm (equal
    to 0.015, 0.029, 0.06, 0.15, or 0.31 mg/kg bw per day for males and
    0.014, 0.036, 0.063, 0.17, and 0.34 mg/kg bw per day for females) for
    two years. There were no significant effects on growth, food
    consumption, or the results of any of the standard clinical and
    physiological examinations made during the course of the study. Gross
    and histological examination of all tissues and organs at the
    conclusion of the study showed no abnormal developments considered to
    be related to treatment. The only significant physiological effect
    observed was inhibition of plasma cholinesterase activity (> 20%) at
    doses > 2 ppm and of erythrocyte acetylcholinesterase activity at
    > 5 ppm. Brain acetylcholinesterase activity was not measured
    (JMPR, 1974, slightly modified by reference to the original report by
    Löser, 1972a; Thomson et al., 1972b).

     Cattle

    Groups of three dairy cows were fed fenamiphos sulfoxide in their
    diets at levels of 2, 6, or 20 ppm for 29 days; one untreated cow was
    used as a control. There were no apparent effects on behaviour, feed
    consumption, milk production, or body-weight gain. No adverse effects
    on whole-blood cholinesterase activity were seen with 2 or 6 ppm, but
    a significant depression (51%) was seen at 20 ppm (Wargo, 1978; JMPR,
    1985).

     (c) Long-term toxicity and carcinogenicity

     Mice

    Groups of 50 male and 50 female six-week-old outbred CD1 albino mice
    were given fenamiphos (purity, 89.5%) in the diet at doses of 0, 2,
    10, or 50 ppm (equal to 0, 0.3, 1.4, or 7.4 mg/kg bw per day for males
    and 0, 0.3, 1.8, or 8.8 mg/kg bw per day for females) for 20 months.
    Toxic effects were monitored daily, and body weights and food
    consumption were determined weekly. Haematological parameters were
    analysed in 10 mice of each sex in each group at 6, 12, 18, and 20
    months of the study. All animals underwent complete necropsy, and the
    liver, kidney, heart, lungs, gonads, spleen, brain, and adrenals were
    weighed. A full complement of tissues and organs from all animals were
    examined histopathologically. Cholinesterase activity was not
    measured. Daily observations were not reported. Survival was
    comparable in all groups, with only a marginal decrease at the high
    dose. Survival at 20 months was 32-45%. Body weights were
    statistically significantly reduced in both males and females at 50

    ppm, but the differences were less than 10%. Food consumption and
    haematological parameters were unaffected by treatment.

    Contrary to the conclusions of the 1985 JMPR with regard to changes in
    organ weights, reexamination of the data indicates that the absolute
    weights of the brains of female mice at all doses were slightly (4-6%)
    but statistically significantly lower than those of controls. The
    relative weights of the brains of animals at 50 ppm were, however,
    increased in both males (7%, statistically nonsignificant) and females
    (9%, statistically significant). The absolute and relative ovarian
    weights were decreased by 16-18% in animals at 10 ppm and by 42-55% in
    those at 50 ppm; and the absolute and relative splenic weights were
    reduced in animals of each sex at 50 ppm. Other findings include
    reductions in absolute heart, lung, liver, and kidney weights
    (generally 10-15%, of variable significance) in males and females at
    50 ppm, although the relative weights were comparable to those in
    controls. No gross or microscopic parallel to these weight changes was
    found. The most frequent pathological findings reported were chronic
    multifocal interstitial nephritis, chronic peribronchiolitis,
    pulmonary congestion, and acute and chronic myocarditis in all treated
    animals, but no significant difference associated with dose was found.
    Cystic endometrial hyperplasia was also frequent in all treated
    females. A significant degree of fatty diffuse change was seen in the
    liver, again with no relation to dose. In all groups, uniformly spread
    amyloidosis was observed in many organ systems, including liver,
    spleen, adrenals, kidneys, small intestines, thyroids, ovaries, and
    submaxillary salivary glands. Fenamiphos had no oncogenic potential at
    any dose. The NOAEL was 2 ppm, equal to 0.3 mg/kg bw per day, on the
    basis of decreased splenic and ovarian weights at doses > 10 ppm
    (JMPR, 1985, slightly modified by reference to the original report by
    Hayes, 1982).

     Rats

    Groups of 40 male and 40 female SPF-derived Wistar rats were fed
    fenamiphos in the diet at concentrations of 0, 3, 10, or 30 ppm (equal
    to 0, 0.17, 0.56, or 1.7 mg/kg bw per day for males and 0, 0.23, 0.76,
    or 2.2 mg/kg bw per day for females) for two years. Behavioural
    abnormalities due to cholinergic stimulation were seen only during the
    first six weeks of treatment at 30 ppm. The obvious effect on
    cholinesterase activity disappeared after six weeks of feeding and was
    not seen for the remainder of the study. Average growth, mortality,
    food consumption, haematological and clinical chemical parameters, and
    the results of liver and kidney function tests were unchanged. Urinary
    parameters and blood sugar and cholesterol values were normal. The
    thyroid weights and the thyroid:body weight ratios of females at 30
    ppm were larger than those of controls but were not accompanied by
    abnormal tumour development, goitre, or unusual histological findings.
    All other major tissues and organs appeared normal at gross and
    microscopic examination.

    Feeding of 10 ppm fenamiphos in the diet inhibited plasma
    cholinesterase activity by up to 55% in females but by < 20% in
    males. In animals at 30 ppm, plasma cholinesterase activity was
    inhibited by up to 60% and that of erythrocyte acetylcholinesterase by
    up to 54% in males and 65% in females. No significant effects were
    observed at 3 ppm. Although the NOAEL was originally considered to be
    3 ppm, the present Meeting determined it to be 10 ppm (equal to 0.56
    mg/kg bw per day) on the basis of inhibition of erythrocyte
    acetylcholinestrase activity at the next highest dose (JMPR, 1974,
    slightly modified by reference to the original report by Löser, 1972b;
    Cherry & Newman, 1973).

    Groups of 50 male and 50 female Fischer 344 rats were fed diets
    containing technical-grade fenamiphos (purity, 89.3%) at a mean
    concentration of 0, 1.7, 7.8, or 37 ppm (equal to 0, 0.098, 0.46, or
    2.5 mg/kg bw per day for males and 0, 0.12, 0.6, or 3.4 mg/kg bw per
    day for females) for two years. Clinical signs, mortality, food
    consumption, haematological and blood chemical parameters (in 20 rats
    of each sex per group), urinary parameters (in 10 rats of each sex per
    group), and body weights were monitored throughout the study. Plasma
    and erythrocyte cholinesterase, activity was monitored in 10 rats of
    each sex per group at weeks 6, 10, and 15 and at 6, 12, 18, and 24
    months. Additional groups of 10 rats of each sex were given 0 or 37
    ppm fenamiphos for one year. Brain acetylcholinesterase activity was
    determined at the end of the study in all rats in these satellite
    groups and in 10 rats of each sex per group in the main study. All
    rats, including those found dead or killed  in extremis during the
    study and those killed at 12 or 24 months, were examined grossly;
    their organs were weighed, and more than 40 tissues were evaluated
    microscopically. Ophthalmological examinations were performed on 10
    rats of each sex per dose before and at the end of the study.

    Survival at termination of the study was not related to treatment and
    was 58-88% in males and 62-84% in females. Clinical observation
    indicated a higher incidence of rough coat and alopecia in females at
    the high dose than in controls. A statistically significant decrease
    in body-weight gain was seen in male and female rats at the high dose
    throughout the study, although no treatment-related effect on feed
    consumption was seen. There was a dose-related, statistically
    significant decrease in plasma cholinesterase activity in all treated
    rats throughout the study: in mate rats, 7-38% at 1.7 ppm, 19-68% at
    7.8 ppm, and 56-88% at 37 ppm; in females, 22-96% throughout the
    study. Erythrocyte acetylcholinesterase activity was also
    statistically significantly inhibited in treated animals: by 0-7% at
    1.7 ppm, 12-25% at 7.8 ppm, and 56-81% at 37 ppm in males and 0-11% at
    1.7 ppm, 21-43% at 7.8 ppm, and 62-81% at 37 ppm in females. A
    statistically significant decrease in brain acetylcholinesterase
    activity was observed in male rats at the high dose at termination
    (-14%) and in animals of each sex (-25% in males and -24% in females)
    at interim sacrifice after one year of treatment. There were
    statistically significant increases in the relative weights of the
    brain, heart, and lung in both males and females at the high dose at
    the end of the study; females at the high dose also had significantly

    increased relative kidney weights. At interim sacrifice, females had
    statistically significantly increased relative weights of adrenals,
    brain, heart, kidneys, and ovaries. These changes were considered to
    be related to the decrease in body weight observed in rats of each sex
    at 37 ppm. The only statistically significant change in absolute organ
    weight at termination was a decrease in liver weight and an increase
    in lung weight in animals of each sex at 37 ppm. No treatment-related
    change was seen during ophthalmological examination, in food
    consumption, in haematological, clinical chemical, or urinary
    parameters, or on gross pathology. No treatment-related neoplastic
    lesions were observed during histopathological examination, but
    statistically significantly higher incidences of non-neoplastic
    inflammatory lesions were observed in the nasal, laryngeal, and lung
    tissues of rats receiving 37 ppm fenamiphos in the diet when compared
    with controls. These changes were attributed by the author of the
    study to the marked inhibition of cholinesterase activity in these
    animals. No treatment-related non-neoplastic changes were noted at 1.7
    or 7.8 ppm. The author concluded that fenamiphos was not oncogenic.
    Contrary to the conclusions of the 1987 JMPR, the present Meeting
    determined that the NOAEL was 7.8 ppm, equal to 0.46 mg/kg bw per day,
    on the basis of inhibition of brain acetylcholinestrase activity and
    changes in body-weight gain, organ weights, and histoptahological
    appearance at the next highest dose (JMPR, 1987, slightly modified by
    reference to the original report by Hayes, 1986a).

     (d)  Genotoxicity

    Fenamiphos has been tested adequately in a battery of tests for
    mutagenicity (Table 4). It was weakly clastogenic only  in vitro; no
    similar response was seen  in vivo in tests for micronucleus
    formation and dominant lethal mutation. The Meeting concluded that
    fenamiphos is not genotoxic.

     (e)  Reproductive toxicity

     (i)  Multigeneration reproductive toxicity

     Rats

    A standard three-generation (two litters per generation) study of
    reproductive toxicity was performed in which groups of 10 male and 20
    female FB30 rats were given fenamiphos in the diet at 0, 3, 10, or 30
    ppm throughout mating, gestation, and suckling. Immediately after
    birth, pups were examined for malformations and were then prepared for
    another generation or killed. Five weanling rats per group from the
    F3b. generation were killed, and macroscopic and microscopic
    examinations were performed on the major tissues and organs. There
    were no apparent differences in the limited indices of reproduction
    investigated, including fertility, litter size, lactation index, or
    growth of young, or in the incidence of malformations (Löser, 1972c;
    Cherry et al., 1972; JMPR, 1974).

    In a two-generation study of reproductive toxicity, groups of 30 male
    and 30 female albino CD Sprague Dawley rats received technical-grade
    fenamiphos (purity, 89%) in the diet at concentrations of 0, 2.5, 10,
    or 40 ppm (equal to 0.17, 0.64, or 2.8 mg/kg bw per day for males and
    0.2, 0.73, or 3.2 mg.kg bw per day for females) for 70 days before
    mating. Oestrous cycles were characterized over two weeks in 10 F0
    and F1 females per dose before mating After weaning, 30 F1 animals
    of each sex per dose were treated for 70 days and then bred to produce
    the second generation (F2); treatment was continued throughout
    mating, gestation, and lactation. Groups of 10 F0 and F1 adults of
    each sex per dose were used to assess cholinesterase activity in
    plasma and erythrocytes in week 8 before mating and just before
    sacrifice and in brain at the time of sacrifice. Plasma, erythrocyte,
    and brain cholinesterase activities were measured in one pup of each
    sex from each of 10 litters at the time of culling and on day 21 of
    lactation. F0 and F1 females were killed after their pups had been
    weaned or on day 24 of gestation. The males were killed after the last
    litters were delivered. The histologic al examinations concentrated on
    reproductive tissues.

    There were no treatment-related deaths or clinical signs of toxicity
    in the parental animals. In F0 and F1 dams at 40 ppm, statistically
    significant reductions were seen in body-weight gain during lactation
    (by 72 and 65%) and food consumption (by up to 11 and 19%). F1 and
    F2 pups also had significant reductions in body-weight gain beginning
    on day 7 of lactation. The body weights of F1 adults at 40 ppm were
    significantly reduced throughout the premating period (by about 10% in
    males and 7% in females), which the author attributed to their reduced
    body weights at the start of the F1 premating period. Although not
    reported by the author, the overall weight gain of F1 males before
    mating was also reduced during the first four weeks, by 12% in gain
    those at 10 ppm and by 15% in those at 40 ppm. The relative ovarian
    weights of F0 females were significantly reduced, by 13% at 2.5 ppm,
    11% at 10 ppm, and 20% at 40 ppm but were unaffected in F1 females.
    Although the author concluded that this effect was related to
    treatment at 40 ppm, no histopathological lesions were observed in the
    ovaries, reproductive parameters were unchanged, and there was no
    comparable effect in the F1 generation.

    Plasma cholinesterase activity was significantly inhibited by > 20%
    in all treated adult females of both generations, in F1 males at 10
    ppm at the time of sacrifice, and in both F0 and F1 males at 40 ppm
    both before mating and at sacrifice. Erythrocyte acetylcholinesterase
    activity was consistently significantly inhibited in females at doses
    > 10 ppm but only at 40 ppm in F0 and F1 males. At 40 ppm, brain
    acetylcholinesterase activity was significantly inhibited by 21% in
    F0 females, by 29% in F1 females, and by only 6% in F0 males. In
    pups, plasma cholinesterase activity was significantly inhibited on
    day 21 of lactation in both males and females of both generations at
    doses > 10 ppm. Erythrocyte acetylcholinesterase activity was
    inhibited by > 20% in males and females of both generations at 40 ppm
    only. Brain acetylcholinesterase was not affected.


        Table 4. Results of tests for genotoxicity with fenamiphos

                                                                                                                     

    End-point             Test system        Concentration              Purity    Results       Reference
                                                                        (%)
                                                                                                                     

    In vitro
    Reverse mutation      S. typhimurium     4, 20, 100, 500, 2500      NR        (Negative)a,b Herbold (1979)
                          TA1535, TA1537     µg/plate (DMSO)
                          TA98, TA100

    Reverse mutation      S. typhimurium     20, 100, 125, 250, 500,    92.4      Negativea     Herbold (1985a,b)
                          TA98, TA100        1000, 2000, 2500
                          TA1535, TA1537     µg/plate (DMSO)

    Forward mutation      Chinese hamster    Unactivated: 100, 110,     85        Negativea,c   Yang et al. (1984)
                          ovary cells        120, 130 µg/ml (DMSO)
                          (CHO-K1-BH4)       Activated: 170, 190,
                                             210, 230 µg/ml (DMSO)

    Unscheduled           Rat hepatocytes    1.5, 5, 15, 50, 100 µg/ml  89.5      Negative      Curren (1988)
    DNA synthesis                            (DMSO)

    Chromosomal           Human              25, 100, 400 µg/ml         91.3      Positivea,d   Herbold (1987)
    aberrations           lymphocytes        (DMSO)

    Chromosomal           Human              Unactivated:               91.9      Negative      Herbold (1988)
    aberrations           lymphocytes        25, 50, 75, 100 µg/ml
                                             (DMSO)

                                             Activated: 100, 150,                 Weakly
                                             225, 350 µg/ml (DMSO)                positivee

    Sister chromatid      Chinese hamster    Activated: 10, 20, 40,     NR        Negativef     Chen et al. (1982)
    exchange              cell line (V79)    80 µg/ml

    Table 4. (continued)

                                                                                                                     

    End-point             Test system        Concentration              Purity    Results       Reference
                                                                        (%)
                                                                                                                     

    In vivo
    Micronucleus          Mouse (NMRI)       0.625, 1.25, 2.5 mg/kg     92.5      (Negative)g   Herbold (1980a)
    formation             bone-marrow cells  bw

    Dominant lethal       Mouse (male        5 mg/kg bw                 92.5      Negative      Herbold (1980b)
    mutation              NMRI) germ cells
                                                                                                                     

    NR, not reported; DMSO, dimethyl sulfoxide
    a   With and without metabolic activation
    b   The 1985 JMPR concluded that the test protocol was unacceptable. Another test was conducted in only 
        one strain (TA 1537), without activation, at doses of 125, 250, 500, and 1000 µg/plate.
    c   Positive controls (0.2 µl/ml ethylmethylsulfonate; 2 µg/ml benzo[a]pyrene) yielded the expected positive responses.
    d   Statistically significant increase (52% mitotic index) seen at 100 µg/ml without activation and at 400 µg/ml with 
        activation (< 0.1% mitotic index). Haemolysis was seen at 400 µg/ml. The author concluded that the increase in 
        aberrations was due exclusively to cytotoxicity.
    e   Statistically significant response seen only at 350 µg/ml with activation (35% mitotic index). The author concluded 
        that the increase in aberrations was due to cytotoxicity.
    f   Positive control (5 µg/ml cyclophosphamide) gave expected positive response.
    g   The mice were dosed twice, 24 h apart, and the bone marrow was sampled once, 6 h after the second dose. The 1985 
        JMPR concluded that the test protocol was unacceptable. The protocol was also considered unacceptable in the report 
        of the Gene-Tox Program (Mavournin et al., 1990).
    

    Oestrous cycles, mating, fertility, and gestation indices, sex ratio,
    and pup viability indices were unaffected, and no treatment-related
    gross or histological lesions seen in any tissue from either parental
    animals or pups. The pups showed no clinical signs of toxicity. The
    NOAEL for systemic toxicity was 2.5 ppm, equal to 0.17 mg/kg bw per
    day, on the basis of decreased body-weight gain. The NOAEL for
    reproductive toxicity was 10 ppm, equal to 0.64 mg/kg bw per day, on
    the basis of decreased pup body weights during lactation (Eigenberg,
    1991).

     (ii)  Developmental toxicity

     Rats

    Four groups of 25 female FB30 rats mated overnight with untreated
    males (in a ratio of one male to two females) were given fenamiphos
    (purity, 92.5%) in a 0.5% aqueous Cremophor emulsion at daily doses of
    0, 0.3, 1, or 3 mg/kg bw per day by gavage on days 6-15 of gestation;
    the first day of gestation was that on which sperm was found in a
    smear obtained the morning after mating. Control females received the
    same volume (10 ml/kg bw) of the aqueous emulsion. On day 20 of
    gestation, the dams were narcotized with carbon dioxide and the
    fetuses were removed. Litter size, average fetus weight per litter,
    sex, external and visceral abnormalities, and skeletal malformations
    and development were noted. Cholinesterase activity was not measured.

    Eighteen dams receiving 3 mg/kg bw per day showed signs of toxicity
    (trembling and recumbency), and two died; however, the time of death
    was not given and the cause of death could not be established. No
    treatment-related signs of toxicity or deaths occurred in rats
    receiving 0, 0.3, or 1 mg/kg bw per day. A 15% reduction in average
    weight gain was seen during treatment among dams receiving 3 mg/kg bw
    per day in comparison with controls, but the author reported that the
    difference was not statistically significant. A total of six females
    -- one control, two at 0.3 mg/kg bw per day, two at 1 mg/kg bw per
    day, and one at 3 mg/kg bw per day -- were not fertilized. The average
    placental weight of animals at the high dose was significantly lower
    than that in controls. Treatment did not affect the number of
    fertilized or pregnant females, litter size, number of resorptions,
    number of fetuses, average fetal weight, sex ratio, incidence of
    alterations in development, or the type or number of malformations.
    The most frequent manifestations were nodulations on ribs, which were
    found in two fetuses from one litter of a dam at 0.3 mg/kg bw per day
    and in four fetuses of three litters of dams at 1 mg/kg bw per day.
    The other malformations observed were general oedema, abdominal
    fissure, and anophthalmia in one fetus at 0.3 mg/kg bw per day. No
    malformations were observed in fetuses at the high dose. The lower
    placental weight observed at the high dose was considered to be not
    toxicologically significant because the average weight was within the
    normal range and no effects were seen on embryonic or fetal
    development. Thus, fenamiphos at doses < 3 mg/kg bw per day was not
    embryotoxic or teratogenic; it was, however, toxic to dams at 3 mg/kg
    bw per day (Schlüter, 1981; JMPR, 1987).

    Groups of 33 mated female Crl:CD-BR rats were given fenamiphos
    (purity, 88.7%) at doses of 0, 0.25, 0.85, or 3 mg/kg bw per day by
    gavage on days 6-15 of gestation. Five dams from each group were
    killed on day 16 in order to measure plasma, erythrocyte, and brain
    cholinesterase activities; the remaining dams were killed on day 20 of
    gestation and necropsied grossly. All dams were examined for number of
    corpora lutea and implantation sites, and their uteri and placentas
    were weighed. The fetuses were weighed and sexed; about half were
    examined externally and in the viscera, and the other half were
    processed for skeletal examination. Brain acetylcholinesterase
    activity was measured in 20 fetuses per group.

    Six dams at 3 mg/kg bw per day died between days 7 and 14 of
    gestation, one female with convulsions on the day of its death.
    Clinical signs of toxicity, such as tremors, salivation, lachrymation,
    urine staining, and hypoactivity, were seen to varying extents in the
    survivors. Body-weight gain and food consumption were significantly
    reduced throughout treatment at this dose. Gross necropsy revealed no
    treatment-related abnormalities, and gestational parameters were
    unaffected. Fetal body weights were unchanged, and they had no
    treatment-related variations or malformations. Plasma and erythrocyte
    cholinesterase activities were statistically significantly reduced (by
    50 and 42%, respectively) in dams at 3 mg/kg bw per day on day 16 of
    gestation. By day 20, the plasma activity had returned to normal,
    while that of erythrocyte cholinesterase was still significantly
    reduced by 30%. Brain acetylcholinesterase activity was reduced by 28%
    in adults at 0.85 mg/kg bw per day and by 12% in those at 3 mg/kg bw
    per day; as the differences were not significant or dose-related, they
    were not considered to be related to treatment. Fetal brain
    acetylcholinesterase activity was unaffected. The NOAEL for maternal
    toxicity was 0.85 mg/kg bw per day. Fenamiphos was not teratogenic or
    fetotoxic under the conditions of this study (Clemens et al., 1989).

     Rabbits

    Groups of 20 double-mated female New Zealand rabbits were given
    fenamiphos (purity, 88.8%) orally in corn oil at doses of 0, 0.1, 0.3,
    or 1 mg/kg bw per day on days 6-18 of gestation. They were observed
    daily for clinical signs of toxicity and were weighed initially,
    periodically during the test, and at termination of the study. Once
    the pups had been removed, the ovaries and uteri of the dams were
    examined, and the fetuses were examined grossly and prepared for
    evaluations of soft tissues and the skeleton. The numbers of corpora
    lutea, implantations, resorptions, live and dead fetuses, and
    anomalies were also determined. Cholinesterase activity was not
    assessed. Dams given doses > 0.3 mg/kg bw per day showed signs of
    toxicity, with decreased body-weight gain, bloody nasal discharge, and
    white, mucoid ocular discharge. Treatment did not affect the number of
    litters, number of pups per litter, pregnancy rate, the number of
    corpora lutea, implantations, or gross abnormalities. Mean fetal
    weight was slightly depressed at 1 mg/kg bw per day. One dam at 0.3
    mg/kg bw per day aborted one dead pup, and two at 1 mg/kg bw per day

    aborted eight dead pups and had seven late resorptions. In addition,
    one dead fetus was found in each of two litters at the high dose.

    The commonest developmental variation observed was the left carotid
    arising from the innominate, which occurred in six to eight fetuses
    (7-9%) in each of three litters (25%) at doses > 0.1 mg/kg bw per
    day. This anomaly was not seen in the controls and in only one of 31
    litters (3.2%) of historical controls at the laboratory where the
    study was performed. More recent data (through July 1985) for
    historical controls revealed incidences of 11/336 (3.3%) in fetuses
    and 9/53 (17%) litters. The newer historical control data provide some
    evidence that the incidence of left carotid anomalies was increased in
    superovulated or artificially inseminated rabbits. Furthermore, data
    for rabbits of the same strain in different labs indicate that the
    anomaly is a frequent finding, occurring in about 8% of fetuses and
    25% of litters; the historical data are for artificially inseminated
    rabbits, while the animals used in this study were naturally bred. The
    biological significance of this finding in relation to treatment is
    dubious.

    An increased incidence of accessory skull bones was also seen in all
    treated groups, but it did not occur in a dose-related manner and was
    thus not considered to be related to treatment. A significant increase
    in the incidence of chain-fused sternebrae was seen in five fetuses in
    three litters at 1 mg/kg bw per day, and this anomaly was also seen in
    one fetus in one litter at 0.3 mg/kg bw per day. This anomaly is of
    questionable biological significance. Two fetuses in one litter at the
    high dose had aortic arches with a common truncus, which was
    considered to be a major malformation. Other skeletal malformations
    which occurred only at doses > 0.3 mg/kg bw per day included fused
    ribs, scoliosis, absent vertebrae (thoracic, lumbar, sacral, and
    caudal), and bipartite or malformed centra. The 1985 Meeting concluded
    that these findings were related to treatment; however, these
    anomalies occurred in only one or two fetuses in single litters, often
    with no relation to dose. Furthermore, several of the anomalies were
    clustered within a single fetus, indicating that they were not likely
    to be related to treatment. The NOAEL for maternal toxicity was 0.1
    mg/kg bw per day, and that for developmental toxicity was 0.3 mg/kg bw
    per day. Fenamiphos had no teratogenic effects at any dose (JMPR,
    1985, modified by reference to the original report by MacKenzie,
    1982).

    In a study to determine the doses for a study of embryotoxicity
    (including teratogenicity), groups of three mated (1:1) female
    Chinchilla rabbits were given single daily doses of fenamiphos
    (purity, 91%) in distilled water with 0.5% Cremophor EL0 at doses of
    0.1, 0.8, or 3 mg/kg bw per day by gavage on days 6-18  post coitum. 
    Cholinesterase activity was measured in plasma and erythrocytes before
    the first dose and just after the last dose; brain
    acetylcholinesterase activity was not assessed. All of the animals
    were killed on day 28  post coitum, and the fetuses were removed,
    sexed, weighed, and examined for gross external and internal
    abnormalities.

    One female at the high dose lost weight from day 10 and died on day 12
     post coitum; body-weight loss and reduced food consumption were seen
    throughout treatment in all animals at this dose when compared with
    controls. Statistically significant reductions in both plasma (89%)
    and erythrocyte cholinesterase activity (90%) relative to controls
    were noted in rabbits at the high dose on day 18  post coitum. Five
    preimplantation losses and one fetal resorption were observed in does
    at the high dose but in no other group. Marginal effects on body
    weight and food consumption were seen at 0.8 mg/kg bw per day. No
    treatment-related effects were seen on the numbers of corpora lutea,
    implantations, live or dead fetuses, or on fetal weight (Becker et
    al., 1986; JMPR, 1987).

    In the main study, four groups of 16 single-mated female Chinchilla
    rabbits were given fenamiphos (purity, 91%) in distilled water
    containing 0.5% Cremophor as single daily doses of 0, 0.1,0.5, or 2.5
    mg/kg bw per day by gavage on days 6-18  post coitum. The does were
    killed on day 28  post coitum, and the fetuses were removed. Only
    does with at least one living fetus were used in calculating
    body-weight gain, food consumption, and reproductive parameters. They
    were examined for the position of fetuses in the uterus and numbers of
    corpora lutea, implantations, resorptions, and live and dead fetuses.
    The fetuses were weighed, sexed, and examined for external and
    internal malformations, skeletal abnormalities, and development.
    Cholinesterase activity was not assessed. Dosing solutions were
    prepared daily. The concentration of fenamiphos in triplicate samples
    taken for determination of homogeneity before the study was found to
    vary considerably (14-140% of the nominal concentration), with mean
    concentrations of 71 ± 8.3, 84 ± 64, and 70 ± 16% of the nominal
    concentration at doses of 0.1, 0.5, and 2.5 mg/kg bw per day,
    respectively. At the next sampling 10 days later, the mean
    concentrations were all within 90% of the nominal concentration,
    although the variability was still high (92 ± 37, 97 ± 45, and 99 ±
    41% of the nominal concentration at the three doses, respectively).
    Although it is stated that homogeneity was maintained during dosing by
    use of a magnetic stirrer, it is not clear if the samples taken for
    testing were subjected to the same treatment before analysis.

    Four females at 2.5 mg/kg bw per day died as a result of treatment
    after 3, 5, and 10 days and on day 21  post coitum. Treatment-related
    signs of toxicity (salivation and dyspnoea) were observed in these
    females and in five other females at the high dose between days 7 and
    18  post coitum. Ataxia was seen in two females that died, and
    diarrhoea was observed in another. No signs of toxicity were found in
    animals at 0, 0.1, or 0.5 mg/kg bw per day. A treatment-related,
    statistically significant decrease in mean food consumption (29%) and
    a treatment-related reduction in body-weight gain (56%) were observed
    during treatment in does at the high dose when compared with controls.
    Food consumption was significantly increased in this group on days
    24-28.

    No treatment-related or significant difference was observed between
    treated and control animals with regard to the mean numbers of
    implantations, corpora lutea, live or dead fetuses, or resorptions. In
    fetuses, no effect that could be attributed to treatment was seen in
    sex ratio, body weight, external or internal malformations, skeletal
    abnormalities, or development. One fetus at the high dose had
    encephalocele with reduced brain size, but this finding was not
    considered to be related to treatment. A number of skeletal changes
    unrelated to treatment were seen in fetuses at all doses. The NOAEL
    for maternal toxicity was 0.5 mg/kg bw per day. Although questions
    remain about the doses actually administered, because of the problems
    of homogeneity, the study showed no embryotoxicity or teratogenicity
    at maternally toxic doses (JMPR, 1987, modified by reference to the
    original report by Becker, 1986).

     (f)  Special studies

     (i)  Dermal and ocular irritation and dermal sensitization

    Technical-grade fenamiphos painted on the skin of New Zealand white
    rabbits in an acetone solution at 50 mg/kg bw resulted in slight
    erythema but was not considered to be a primary irritant. Application
    of technical-grade fenamiphos to the conjunctival sac of New Zealand
    white rabbits as 100 mg of a crystalline material resulted in
    irritation considered to be mechanical rather than physiological
    (Crawford & Anderson, 1971; JMPR, 1974).

    Technical-grade fenamiphos (purity, 90.7%) was melted at 35°C, and
    0.5 ml was painted onto the intact or abraded skin of six
    Japanese-derived albino rabbits for 24 h under occlusion. Irritation
    was scored according to Draize 24, 48, 72, and 168 h after
    application. Minimal irritation was observed, characterized by slight
    erythema and oedema during the first 48 h. The mean primary irritation
    index, based on readings at 24 and 72 h, was 0.42 (Kato 1984a).

    Technical-grade fenamiphos (purity, 90.7%) was heated to liquidity
    (temperature not specified) and applied at 0.1 ml to the conjunctival
    sacs of nine Japanese-derived albino rabbits. The eyes of three of the
    rabbits were washed with water. Irritation was scored according to
    Draize 24, 48, 72, 96, 168, and 240 h after administration. In the
    absence of washing, fenamiphos was a moderate irritant, with a maximum
    average score of 31.3 at 24 h. The irritation scores slowly declined,
    and all of the eyes were clear by 240 h. Fenamiphos was not irritating
    when the eyes were washed with water after application. Midriasis,
    persisting until day 2 or 3, was seen in all animals when the eyes
    were not washed within 10 min of application. The translation of the
    report leaves some uncertainty about other ocular effects, but signs
    of systemic toxicity peaked within 3-4 h of application; these
    included salivation, increased respiration, cyanosis, and slight
    convulsions (number affected not specified). All animals were
    reportedly normal within 6 h. No systemic toxicity was seen in the
    animals when the eyes were washed (Kato, 1984b).

    The potential of fenamiphos to sensitize skin was examined in a
    maximization test in which 20 male Hsd Win:DH guinea-pigs were induced
    intradermally with 1% fenamiphos in saline containing 2% Cremophor EL.
    One week later, they were induced topically with 25% fenamiphos in
    saline (with 2% Cremophor EL); they were challenged three weeks later
    with 12 and 25% solutions of fenamiphos in the saline solution. Patchy
    erythema was seen 24 h after removal of the patch in 22% of the
    animals challenged with the 25% solution, whereas none of the 10
    induced control animals showed irritation. The author considered a
    response rate of > 30% to be indicative of sensitization, and
    concluded that fenamiphos produced no relevant sensitization; however,
    according to the original protocol of Magnusson and Kligman, these
    results would indicate that fenamiphos is a mild sensitizer (Stropp,
    1995).

     (ii)  Delayed neuropathy

    Groups of eight hens were fed fenamiphos in the diet at levels of 0,
    1,3, 10, or 30 ppm, equal to 0, 2, 5, 16, or 26 mg/kg bw per day, for
    30 days. At the end of treatment, some birds were killed and the
    remainder were observed for four weeks for neurological signs of
    poisoning. Food consumption was depressed in birds at 30 ppm, and the
    average body weight and growth of hens at this dose was reduced.
    Whole-blood cholinesterase activity was decreased after 30 days at
    doses > 1 ppm, although no signs of cholinergic poisoning were
    observed. There were no indications of delayed neurotoxicity, and
    microscopic examination of brain, spinal cord, and sciatic nerve
    (stained with haematoxylin and eosin) did not indicate delayed
    neuropathy (Kimmerle, 1970; Spicer, 1970; JMPR, 1974).

    Groups of 10 hens given an LD50 a dose of fenamiphos (5.0 mg/kg bw)
    orally were observed for three weeks and then killed. No evidence of
    delayed neurotoxicity was observed either clinically or
    histologically, whereas signs were observed with tri- ortho-cresyl
    phosphate (Kimmerle, 1971; Spicer, 1971; JMPR, 1974).

    Fenamiphos (purity, 91.3%) in a 2% Cremophor solution was administered
    twice by intubation to 30 Lohmann selected Leghorn hens at 25 mg/kg bw
    per day at a 21-day interval. Five hens serving as positive controls
    received tri- ortho-cresyl phosphate at 375 mg/kg bw per day. The
    hens treated with fenamiphos received atropine intramuscularly at 100
    mg/kg bw before treatment and subcutaneously at 30-50 mg/kg bw 7, 24,
    or 30 or 48 h after treatment. The birds were observed for body
    weight, clinical signs, forced motor coordination, and gross and
    histopathological changes. Although statistically significant,
    treatment-related weight loss (during week 1) and signs of severe
    poisoning were seen, the birds showed no impairment of motor
    coordination indicative of delayed neurotoxicity. Histological
    examination of tissues from the peripheral and central nervous systems
    showed no changes indicative of delayed neuropathy. Birds given
    tri- ortho-cresyl phosphate, however, showed clinical signs of
    delayed neurotoxicity (ataxia and paresis) two weeks after treatment
    and were killed in moribund condition on day 17. The histopathological

    changes in these birds were typical of neuropathy (Flucke & Kaliner,
    1987).

     (iii)  Neurotoxicity

     Rats

    Groups of 12 Wistar (Hsd Win:WU) rats of each sex were given single
    doses of technical-grade fenamiphos (purity, 95.2%) at 0.37, 1.52, or
    2.31 mg/kg bw by garage. They were observed for mortality, clinical
    signs, and body weight. Functional observational and motor or
    locomotor activity tests were conducted on all animals before
    treatment, within 30 min of treatment, and 7 and 14 days later.
    Plasma, erythrocyte, and brain cholinesterase activity was measured in
    additional satellite groups of six animals of each sex at each dose,
    which were killed within 1 h of treatment. One-half of the animals in
    the main group were perfused, and various neural and skeletal muscle
    tissues, including the brain, spinal cord and spinal ganglia, eyes and
    optic nerves, peripheral nerves, gastrocnemic muscle, and trigeminal
    ganglia, were processed for histological examination.

    In animals at the lowest dose, plasma cholinesterase activity was
    statistically significantly inhibited in females (by 55%) and
    nonsignificantly decreased in males (by 23%). Erythrocyte
    acetylcholinesterase activity was significantly inhibited only in
    males (by 24%), but this was not considered to be an adverse effect
    because no corroborating clinical signs were seen in the functional
    and motor activity tests. At the next highest dose, both plasma and
    erythrocyte cholinesterase activity was significantly inhibited in
    both males (by 64 and 70%, respectively) and females (by 77 and 51%,
    respectively). Uncoordinated gait and muscle fasciculations were also
    seen in males during the functional observational battery of tests. At
    the highest dose, similar signs were seen, with decreased grip
    strength and deaths among both males and females and decreased motor
    activity in malesś Other behavioural or physiological changes seen in
    animals of each sex in the functional observational battery of tests
    included piloerection, nasal, oral, and lachrymal staining,
    salivation, and decreased activity and rearing in the open field.
    Brain acetylcholinesterase activity was unaffected at all doses. No
    treatment-related histopathological changes were seen. The NOAEL was
    0.37 mg/kg bw, the lowest dose tested, on the basis of uncoordinated
    gait and muscle fasciculations in males at the next highest dose
    (Dreist, 1995).

    In a study of similar design, technical-grade fenamiphos (purity,
    95.6%) was administered in the diet to groups of 12 Wistar (Hsd
    Cpb:WU) rats of each sex for 13 weeks at dietary concentrations of 0,
    1, 10, or 50 ppm, equal to 0, 0.06, 0.61, or 3.1 mg/kg bw per day in
    males and 0.08, 0.8, or 4 mg/kg bw per day in females. The animals
    were observed for deaths, clinical signs, body weight, and food and
    water consumption. Functional observational and motor or locomotor
    activity tests were conducted before treatment and in weeks 4, 8, and
    13. Plasma and erythrocyte cholinestemse activity was measured in six

    rats of each sex at each dose on week 4 and before terminal sacrifice
    at week 15, and brain acetylcholinesterase activity only at terminal
    sacrifice.

    All females at the highest dose had muscle fasciculations during the
    first three weeks of the study. Plasma and erythrocyte cholinesterase
    activities were statistically significantly reduced in both males
    (> 68%) and females (> 86%). Brain acetylcholinesterase activity
    was also significantly reduced (by 12%) in females, but the author
    considered that this was not biologically significant. At 10 ppm,
    plasma cholinesterase activity was significantly reduced in both males
    (by 30-39%) and females (by 71-77%; rho < 0.01) in weeks 4 and 15.
    Erythrocyte acetylcholinesterase activity was inhibited by about 25%
    in week 15 in males (rho < 0.05) and in weeks 4 and 15 in females
    at 10 ppm. The lowest dose resulted in nonsignificant decreases (about
    30%) in plasma cholinesterase activity in females in weeks 4 and 15.
    Body weights, food and water consumption, brain weights, and gross and
    histopathological appearance were all unaffected by treatment. The
    author extrapolated the results back to a 20% level of inhibition of
    plasma cholinesterase activity and estimated that the NOAEL in females
    was 0.4 ppm. The overall NOAEL was 10 ppm, equal to 0.8 mg/kg bw per
    day, on the basis of inhibition of brain acetylcholinesterase activity
    in females at the highest dose (Dreist & Popp, 1995).

     (iv)  Potentiation

    In male rats given fenamiphos orally in combination with disulfoton or
    E 154, no potentiation of the acute toxicity was seen (Kimmerle,
    1972c; JMPR, 1974).

    The LD50 for fenamiphos (purity, 91.8%) administered orally to male
    Wistar rats was 4.6 mg/kg bw, while that of carbofuran was 8.1 mg/kg
    bw. When the two compounds were given concomitantly, essentially as a
    2:1 ratio of carbofuran:fenamiphos, the LD50 was 6 mg/kg bw,
    indicating that the toxicity of the combination was additive but not
    synergistic (Mihail, 1980 JMPR, 1985).

    Comments

    In rats, fenamiphos was rapidly excreted, with over 96% of the
    administered dose eliminated within 48 h. Excretion was primarily in
    the urine, with less than 4% of the dose eliminated in the faeces. At
    48 h, the levels of tissue residues were below the limit of
    quantification, except following a high dose (3 mg/kg bw), when the
    maximal tissue levels observed were 3.5-8.4 µg/kg in the liver,
    1.6-2.1 µg/kg in the kidney, and 1.6-3.5 µg/kg in the skin. Fenamiphos
    was completely metabolized in rats. Metabolites retaining
    anticholinesterase activity, such as fenamiphos sulfoxide and
    desisopropyl fenamiphos sulfoxide, were seen in variable but generally
    low proportions (rarely greater than 3%). Most of the products were
    dephosphorylated phenol, sulfoxide phenol, or sulfone phenol
    metabolites and their corresponding sulfates.

    Fenamiphos is extremely hazardous after single oral doses to rats,
    mice, rabbits, cats, dogs, and chickens (LD50 values = 2 4-23 mg/kg
    bw) and highly hazardous after dermal administration to rats and
    rabbits (LD50 values, 75-230 mg/kg bw). It is moderately hazardous
    after inhalation in rats and mice (LC50 values < 100 µg/L, 4 h).
    WHO has classified fenamiphos as 'extremely hazardous' (WHO, 1996).
    The sulfoxide, sulfone, and desisopropylated sulfone metabolites of
    fenamiphos are similarly toxic to rats after oral administration
    (LD50 values = 1.4-4.1 mg/kg bw). The sulfoxide and sulfone phenol
    metabolites are only slightly toxic to rats after oral administration,
    with LD50 values ranging from 1200 to 1900 mg/kg bw.

    Fenamiphos inhibited plasma cholinesterase more effectively than
    erythrocyte acetylcholinesterase, both  in vitro and  in vivo. 
    Fenamiphos sulfoxide, fenamiphos sulfone, desisopropyl fenamiphos,
    desisopropyl fenamiphos sulfoxide, and desisopropyl fenamiphos sulfone
    inhibited plasma and erythrocyte cholinesterase  in vitro more
    effectively than fenamiphos itself.

    In evaluating the following studies, inhibition of erythrocyte
    acetylcholinesterase activity was not used as an indicator of adverse
    effects in the nervous system when information on brain
    acetylcholinesterase activity was also available. In the absence of
    this information, NOAELs were determined on the basis of inhibition of
    erythrocyte acetylcholinesterase (of < 20%). Statistical
    significance was used as a criterion for considering depression of
    brain acetylcholinesterase activity to be adverse.

    In a three-week study in which rats were exposed by inhalation to
    atmospheres containing fenamiphos at 0, 0.03, 0.25, or 3.5 µg/L for 6
    h per day, five days per week, the only finding was inhibition of
    plasma cholinesterase at the highest dose. Erythrocyte and brain
    acetylcholinesterase were unaffected. The no-observed-adverse-effect
    concentration (NOAEC) was 3.5 µg/L.

    In a three-month study in rats, fenamiphos given at dietary
    concentrations of 0, 0.37, 0.57, or 0.91 ppm inhibited plasma
    cholinesterase activity only at the highest dose. No treatment-related
    changes in erythrocyte or brain acetylcholinesterase activity were
    seen at any dose. In a second study of short-term toxicity, rats were
    fed diets containing 0, 4, 8, 16, or 32 ppm fenamiphos for three
    months. Erythrocyte acetylcholinesterase activity was inhibited at
    doses of 16 ppm (equivalent to 0.8 mg/kg bw per day) and above. This
    was considered an adverse effect as brain acetylcholinesterase was not
    measured in this study. The overall NOAEL was 8 ppm, equivalent to 0.4
    mg/kg bw per day.

    Rabbits received fenamiphos by dermal application at doses of 0, 0.5,
    2.5, or 10 mg/kg bw per day for three weeks (6 h per day, five days
    per week). Body-weight gain was slightly reduced in animals of each
    sex at 10 mg/kg bw per day. In females, reductions in cholinesterase
    activity in the brain (by 20%) and plasma were noted at 2.5 mg/kg bw
    per day and above. Erythrocyte acetylcholinesterase activity, however,

    was affected only at 10 mg/kg bw per day. In males, the only findings
    were decreased plasma and erythrocyte cholinesterase activity at 10
    mg/kg bw per day. The NOAEL was 0.5 mg/kg bw per day.

    In a series of studies, dogs were fed diets containing 0, 0.5, 0.6, 1,
    1.7, 2, 3, 5, 6, 10, 12, or 18 ppm fenamiphos for periods ranging from
    three months to two years. In dogs treated at 18 ppm (equivalent to
    0.45 mg/kg bw per day) for three months, muscle tremors were seen. In
    dogs treated at 12 ppm (equal to 0.31 mg/kg bw per day) for one year,
    brain acetylcholinesterase activity was inhibited in females (by 17%),
    and males showed slight anaemia. Erythrocyte acetylcholinesterase
    activity was inhibited at doses of 3 ppm (equal to 0.083 mg/kg bw per
    day) and above for one year. Plasma cholinesterase activity was
    inhibited at doses of 1.7 ppm (equivalent to 0.042 mg/kg bw per day)
    and above in a three-month study, No other parameters were affected.
    Since no information on brain acetylcholinesterase activity was
    available at doses between 3 and 12 ppm, the Meeting considered 3 ppm
    (equal to 0.083 mg/kg bw per day) to be the overall NOAEL in dogs.

    In mice fed diets containing 0, 2, 10, or 50 ppm fenamiphos for 20
    months, there were marginal decreases in survival and body-weight gain
    at 50 ppm (equal to 7.4 mg/kg bw per day). The relative ovarian and
    spleen weights were reduced at 10 ppm (equal to 1.4 mg/kg bw per day)
    and above. There were no non-neoplastic changes that could be
    attributed to treatment, and fenamiphos was not carcinogenic at any
    dose. Cholinesterase activity was not measured. The NOAEL was 2 ppm,
    equal to 0.3 mg/kg bw per day.

    In rats fed diets containing 0, 3, 10, or 30 ppm fenamiphos for two
    years, the only treatment-related effects were inhibition of
    erythrocyte acetylcholinesterase activity throughout the study and
    behavioural changes during the first six weeks of the study in animals
    at 30 ppm. Brain acetylcholinesterase activity was not measured. The
    NOAEL was 10 ppm, equal to 0.56 mg/kg bw per day.

    Rats were fed diets containing 0, 1.7, 7.8, or 37 ppm fenamiphos for
    two years. At 37 ppm, equal to 2.5 mg/kg bw per day, body-weight gain
    in both males and females was decreased. Erythrocyte
    acetylcholinesterase activity was inhibited at 7.8 ppm (equal to 0.46
    mg/kg bw per day) and above. Brain acetylcholinesterase activity was
    inhibited only at 37 ppm; inhibition was 25% in animals of each sex
    killed after one year, and 14% in males at termination of the study.
    Animals of each sex at 37 ppm also had an increased frequency of
    non-neoplastic inflammatory lesions of the nasal, laryngeal, and lung
    tissues and increased relative weights of the brain, heart, and lungs.
    Fenamiphos was not carcinogenic at any dose. The NOAEL was 7.8 ppm,
    equal to 0.46 mg/kg bw per day.

    Fenamiphos was adequately tested in a battery of tests for
    genotoxicity. It was found to be mildly clastogenic at cytotoxic doses
     in vitro but not  in vivo. It did not cause reverse or forward
    mutation, unscheduled DNA synthesis, or sister chromatid exchange 
     in vitro. The Meeting concluded that fenamiphos is not genotoxic.

    In a two-generation study of reproductive toxicity, rats were treated
    with 0, 2.5, 10, or 40 ppm fenamiphos. Parental toxicity was
    characterized by reduced weight gain in F0 and F1 dams at 40 ppm
    (equal to 2.8 mg/kg bw per day) during lactation, and in F1 males at
    10 ppm (equal to 0.64 mg/kg bw per day) and above before mating.
    Pathological changes in the salivary gland were seen in F0 males and
    females at 40 ppm. Erythrocyte acetylcholinesterase activity was
    consistently inhibited at 10 ppm and above in females but only at 40
    ppm in males. Brain acetylcholinesterase activity was inhibited at the
    highest dose in adult F0 and F1 females (by 21-29%) and in F1 males
    (by 6%) but not in pups of either sex. In pups at 40 ppm, erythrocyte
    acetylcholinesterase activity was inhibited only on day 21 of
    lactation. The only reproductive effect was decreased weight gain of
    F1 and F2 pups at 40 ppm, beginning on day 7 of lactation. The NOAEL
    for systemic toxicity was 2.5 ppm, equal to 0.17 mg/kg bw per day. The
    NOAEL for reproductive toxicity was 10 ppm, equal to 0.64 mg/kg bw per
    day.

    In a study of developmental toxicity, mated rats were treated with 0,
    0.3, 1, or 3 mg/kg bw per day on days 6-15 of gestation. Maternal
    toxicity was seen at the highest dose, characterized by mortality,
    tremors, and reduced weight gain. The fetuses were not affected at any
    dose. Cholinesterase activity was not measured in this study. The
    NOAELs were 1 mg/kg bw per day for maternal toxicity and 3 mg/kg bw
    per day for developmental toxicity.

    Fenamiphos was also administered to mated rats at doses of 0, 0.25,
    0.85, or 3 mg/kg bw per day on days 6-15 of gestation. The highest
    dose resulted in maternal deaths, tremors, salivation, lachrymation,
    urine staining, and hypoactivity. Body-weight gain and food
    consumption were also significantly reduced. Erythrocyte
    acetylcholinesterase activity was reduced at this dose, but the
    changes in brain acetylcholinesterase activity were not statistically
    significant or dose-related. The fetuses were unaffected at 3 mg/kg bw
    per day. The NOAELs were 0.85 mg/kg bw per day for maternal toxicity
    and 3 mg/kg bw per day for developmental toxicity.

    In a study of developmental toxicity in rabbits, animals received 0,
    0.1, 0.3, or 1 mg/kg bw per day on days 6-18 of gestation. At 0.3
    mg/kg bw per day and above, fenamiphos was maternally toxic, resulting
    in decreased body-weight gain, bloody nasal discharge, and white
    ocular discharge. Fetotoxicity, characterized by chain fusion of the
    sternebrae, was seen only at 1 mg/kg bw per day. The NOAEL for
    maternal toxicity was 0.1 mg/kg bw per day, and that for developmental
    toxicity was 0.3 mg/kg bw per day. Cholinesterase activity was not
    measured in this study.

    In a second study, mated rabbits were treated with 0, 0.1, 0.5, or 2.5
    mg/kg bw per day on days 6-18 of gestation. Clear maternal toxicity
    was seen at the highest dose, which included mortality, salivation,
    dyspnoea, ataxia, diarrhoea, and decreased weight gain and food
    consumption during treatment. Although some questions remain about the
    doses that were actually administered (because of uncertain

    homogeneity), no embryotoxic or teratogenic effects were seen at the
    maternally toxic dose of 2.5 mg/kg bw per day.

    Fenamiphos was minimally irritating to rabbit skin and moderately
    irritating to rabbit eyes and was a mild skin sensitizer in the
    guinea-pig.

    A single dose of 25 mg/kg bw fenamiphos had no effect on neuropathy
    target esterase activity in the brains or spinal cords of hens, under
    atropine protection. Fenamiphos did not induce delayed neuropathy in
    three studies in hens when tested at doses of 0, 2, 5, 16, or 26 mg/kg
    bw per day for 30 days or when given once at doses of 0 or 25 mg/kg
    bw.

    In a study of acute neurotoxicity, rats were given single doses of 0,
    0.37, 1.5, or 2.3 mg/kg bw fenamiphos by gavage. Erythrocyte
    acetylcholinesterase activity was inhibited at the lowest dose tested
    in males only, but in both males and females at higher doses. At 1.5
    mg/kg bw, males showed uncoordinated gait and muscle fasciculation. At
    the highest dose, decreased motor activity was also seen in males, and
    clinical signs, decreased grip strength, and deaths occurred in rats
    of each sex. There was no effect on brain acetylcholinesterase
    activity, at any dose. The NOAEL was 0.37 mg/kg bw per day. In a
    further study, rats were fed diets containing 0, 1, 10, or 50 ppm
    fenamiphos for 13 weeks. At 10 ppm and above, erythrocyte
    acetylcholinesterase activity was inhibited in animals of each sex. At
    the highest dose, brain acetylcholinesterase activity was inhibited
    (by 12%) in females only. A battery of functional observational and
    motor activity tests revealed no treatment-related effects. The NOAEL
    was 10 ppm, equal to 0.61 mg/kg bw per day.

    An ADI of 0-0.0008 mg/kg bw was established on the basis of an overall
    NOAEL of 0.083 mg/kg bw per day in the dog, and a safety factor of
    100.

    Toxicological evaluation

     Levels that cause no toxic effect

         Mouse:    2 ppm in the diet, equal to 0.3 mg/kg bw per day
                   (20-month study of toxicity and carcinogenicity)

         Rat:      0.37 mg/kg (single doses, study of neurotoxicity) 10
                   ppm, equal to 0.61 mg/kg bw per day (three-month study
                   of neurotoxicity)
                   2.5 ppm, equal to 0.17 mg/kg bw per day (parental
                   toxicity in a study of reproductive toxicity)
                   10 ppm, equal to 0.64 mg/kg bw per day (study of
                   reproductive toxicity)
                   0.85 mg/kg bw per day (maternal toxicity in a study of
                   developmental toxicity)
                   3 mg/kg bw per day (developmental toxicity in a study
                   of developmental toxicity)

        Toxicological criteria for estimating guidance values for dietary and non-dietary exposure to fenamiphos

                                                                                                                            

    Human exposure     Relevant route, study type, species              Results, remarks
                                                                                                                            

    Short-term         Oral neurotoxicity, rat                          NOAEL = 0.37 mg/kg bw per day: effects observed
    (1-7 days)                                                          during battery of functional observational tests
                       Oral toxicity, rat (fasted)                      LD50 = 2.4-6 mg/kg bw
                       Inhalation toxicity, 4 h, rat                    LC50 = 91-100 µg/L
                       Inhalation toxicity, 5 days, rat                 NOAEL = 4 µg/L
                       Dermal toxicity, rat                             LD50 = 72-92 mg/kg bw
                       Dermal irritation, rabbit                        Minimally irritating
                       Ocular irritation, rabbit                        Moderately irritating
                       Dermal sensitization, guinea-pig                 Mildly sensitizing

    Medium-term        Repeated inhalation toxicity, 3 weeks, rat       NOAEL = 3.5 µg/L (highest dose tested)
    (1-26 weeks)       Repeated dermal toxicity, 3 weeks, rabbit        NOAEL = 0.5 mg/kg bw per day: inhibition of brain
                                                                        acetylcholinesterase activity
                       Repeated oral, reproductive toxicity, rat        NOAEL = 0.17 mg/kg bw per day: parental toxicity
                                                                        NOAEL = 0.64 mg/kg bw per day: reproductive
                                                                        toxicity
                       Repeated oral, developmental toxicity, rabbit    NOAEL = 0.1 mg/kg bw per day: maternal toxicity
                                                                        NOAEL = 0.3 mg/kg bw per day: developmental
                                                                        toxicity

    Long-term          Repeated oral, 1 - 2 years, dog                  NOAEL = 0.083 mg/kg bw per day: inhibition of
    (> 1 year)                                                          acetylcholinesterase activity, anaemia
                                                                                                                            
    

                   7.8 ppm, equal to 0.46 mg/kg bw per day (two-year study
                   of toxicity and carcinogenicity)

         Rabbit:   0.1 mg/kg bw per day (maternal toxicity in a study of
                   developmental toxicity)
                   0.3 mg/kg bw per day (fetotoxicity in a study of
                   developmental toxicity)

         Dog:      3 ppm in the diet, equal to 0.083 mg/kg bw per day
                   (overall assessment)

     Estimate of acceptable daily intake for humans

         0-0.0008 mg/kg bw

     Estimate of acute reference dose

    The available data did not permit the Meeting to establish an acute
    reference dose different from the ADI (0-0.0008 mg/kg bw). Although
    the results of a study of neurotoxicity in rats given single doses was
    available, the dog was found to be the more sensitive species.
    Information on acute effects in dogs may allow the establishment of an
    acute reference dose in the future.

     Studies that would provide information useful for continued
     evaluation of the compound

    1.   Effects of single doses in dogs (with appropriate evaluation of
         functional changes in the cholinergic nervous system, including
         brain acetylcholinesterase activity).
    2.   Observations in humans

    References

    Becker, H. (1986) Embryotoxicity (including teratogenicity) study with
    SRA 3886 in the rabbit. Unpublished report No. R 3917 (project No.
    065261) from Research & Consulting Co., Itingen, Switzerland.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Becker, H., Mueller, E., Luetkemeir, H. & Sachsse, K. (1986)
    Dose-finding embryotoxicity (including teratogenicity) study with SRA
    3886 in the rabbit. Unpublished report No. R3693 (project No. 065250)
    from Research & Consulting Co., Itingen, Switzerland. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Chen, H.H., Sirianni, S.R. & Huang, C.C. (1982) Sister chromatid
    exchange in Chinese hamster cells treated with seventeen
    organophosphorus compounds in the presence of a metabolic activation
    system.  Environ. Mutag., 4, 621624. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Cherry, C. & Newman, A. (1973) Pathology report of Bayer 68138.
    Chronic toxicological studies in rats. Unpublished report (addendum to
    report No. 3539) from Huntingdon Research Centre, Huntingdon, United
    Kingdom. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Cherry, C., Urwin, C. & Newman, A. (1972) Pathology report of BAY
    68138. Rat breeding study. Unpublished report (addendum to report No.
    3424) from Huntingdon Research Centre, Huntingdon, United Kingdom.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Clemens, G.R., Troup, C.M. & Hartnagel, R.E., Jr (1989) Teratology
    study in the rat with Nemacur technical. Unpublished report No. 1145
    (study no. MTD0108) from Toxicology Department, Miles Inc., Elkhart,
    IN, USA. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Coulston, F. & Wills, J.H. (1974) Summary of research by the Institute
    of Comparative and Human Toxicology, Albany Medical College, on
    phenamiphos. Report dated October 1974. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Crawford, C. & Anderson, R. (1971) The skin and eye irritating
    properties of BAY 68138 technical to rabbits. Unpublished report No.
    29988 from Chemagro Division of Baychem Corp. Submitted to WHO by
    Bayer AG, Leverkusen, Germany.

    Crawford, C. & Anderson, R. (1972) The acute dermal toxicity of
    Nemacur technical to rabbits. Unpublished report No. 34216 from
    Chemagro Division of Baychem Corp. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Crawford, C.R. & Anderson, R.H. (1973) Comparative oral toxicity in
    rats of several impurities and a technical compound of Nemacur(R)
    with analytical grade Nemacur(R). Unpublished report No. 34446 from
    Chemagro Division of Baychem Corp. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Crawford, C.R. & Anderson, R.H. (1974a) The acute oral toxicity of two
    Nemacur(R) phenolic metabolites and MTMC to male and female rats.
    Unpublished report No. 39700 from Chemagro Division of Baychem Corp.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Crawford, C.R. & Anderson, R.H. (1974b) The acute oral toxicity of
    Nemacur(R)-sulfone and Nemacur(R)-sulfoxide to rats. Unpublished
    report No. 40215 from Chemagro Division of Baychem Corp. Submitted to
    WHO by Bayer AG, Leverkusen, Germany.

    Curren, D. (1988) Unscheduled DNA synthesis in rat primary
    hepatocytes. Unpublished report No. 991 (study no. T5724.380) from
    Microbiological Associates Inc., Rockville, MD, USA. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Dreist, M. (1995) SPA 3886 (common name:Fenamiphos) acute oral
    neurotoxicity screening study in Wistar rats. Unpublished report No.
    24408 (study No. T6058166) from the Institute of Toxicology
    Agrochemicals, Bayer AG, Wuppertal, Germany. Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Dreist, M. & Popp, A. (1995) SRA 3886 (common name: Fenamiphos)
    subchronic neurotoxicity screening study in Wistar rats (thirteen-week
    administration in the diet). Unpublished report No. 24948 (study no.
    T9058277) from Institute of Toxicology Agrochemicals, Bayer AG,
    Wuppertal, Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    DuBois, K. & Flynn, M. (1968) The subacute parenteral toxicity of BAY
    68138 to rats. Unpublished report No. 22178 from Toxicity Laboratory,
    University of Chicago, Chicago, IL, USA. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    DuBois, K., Flynn, M. & Kinoshita, F. (1967) The acute toxicity and
    anticholinesterase action of Bayer 68138. Unpublished report No. 20810
    from Toxicity Laboratory, University of Chicago, Chicago, IL, USA.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Ecker, W., Weber, H. & Brauner, A. (1989) [Phenyl-1-14C]Nemacur:
    General metabolism study in the rat. Unpublished report No. PF3175
    (study no. M182 0189-9) from Bayer AG, Pflanzenschutz-Zentrum Monheim,
    Leverkusen, Germany. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Eigenberg, D.A. (1991) A two generation reproduction study in rats
    using Fenamiphos (Nemacur(R)). Unpublished report No. 5762 (study
    no. 88-671-BC) from Mobay Corp, Stilwell, KS, USA. Submitted to WHO by
    Bayer AG, Leverkusen, Germany.

    Flucke, W. (1980) Determination of acute toxicity. Unpublished report
    dated 18 July 1980 from Bayer AG, Institute of Toxicology, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Flucke, W. & Eben, A. (1988) SPA 3886 Technical (common name:
    Fenamiphos): Study of the effect on the neurotoxic esterase (NTE)
    following oral administration to hens. Unpublished report No. 17388
    from Fachbereich Toxikologie, Bayer AG, Wuppertal-Elberfeld, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Flucke, W. & Kaliner, G. (1987) Delayed neurotoxicity studies on hens
    following acute oral administration. Unpublished report No. 16187
    (study no. T 1020919) from Fachbereich Toxikologie, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Gronberg, R.R. (1969) The metabolic fate of ethyl-4-(methylthio)-m-
    tolyl isopropylphosphoramidate (BAY 68138), ethyl-4-(methylsulfinyl)-
    m-tolyl isopropylphosphoramidate (BAY 68138 sulfoxide) and ethyl-4-
    (methylsulfonyl)-m-tolyl-isopropylphosphoramidate (BAY 68138 sulfone)
    in white rats. Unpublished report No. 26759 from Chemagro Division of
    Baychem Corp. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Hayes, R.H. (1982) Technical fenamiphos (Nemacur(R)) oncogenicity
    study in mice. Unpublished report No. 241 (study No. 78CCM02) from
    Stanley Research Center, Mobay Chemical Corp, Stilwell, KS, USA.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Hayes, R.H. ( 1983 ) Ninety-day cholinesterase study on dogs with
    fenamiphos in diet. Unpublished report No. 444 (study No. 83-174-01)
    from Environmental Health Research, Mobay Chemical Corp, Stilwell, KS,
    USA. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Hayes, R.H. (1986a) Ninety-day cholinesterase study on rats with
    technical fenamiphos (Nemacur) in diet. Unpublished report No. 717
    (study No. 83-171-01) from Corporate Toxicology Dept, Mobay Chemical
    Co., Stilwell, KS, USA. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Hayes, R.H. (1986b) Combined chronic toxicity/oncogenicity study of
    technical fenamiphos (Nemacur) with rats. Unpublished report No. 721
    (study No. 83-271-01) from Corporate Toxicology Dept, Mobay Chemical
    Co., Stilwell, KS, USA. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Heimann, K G (1981) Determination of acute toxicity (LD50 Unpublished
    report dated 18 August 1981 from Bayer AG/Institute of Toxicology,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Heimann, K G (1984) Determination of acute toxicity (LD) Unpublished
    report dated 14 February 1984 from Bayer AG/Institute of Toxicology,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Herbold, B. (1979) Salmonella/microsome test for detection of
    point-mutagenic effects. Unpublished report No. 8730 (study no.
    Nemacur/003) from Bayer AG, Institut für Toxikologie, Wuppertal,
    Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Herbold, B. (1980a) Micronucleus test on mouse to evaluate Nemacur
    technical for potential mutagenic effects. Unpublished report No. 8805
    (study no. SRA 3886/002) from Bayer AG Institute of Toxicology,
    Wuppertal, Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Herbold, B. (1980b) SRA 3886/dominant lethal study on male mouse to
    test for mutagenic effects. Unpublished report No. 8838 (study no. SRA
    3886/001) from Institute of Toxicology, Bayer AG, Wuppertal, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Herbold, B. (1985a) SRA 3886: Salmonella/microsome test to evaluate
    for potential point mutation. Unpublished report No. 13365 (study no.
    T2017617) from Bayer AG, Institute of Toxicology, Wuppertal-Elberfeld,
    Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Herbold, B. (1985b) Addendum to report SRA 3886: Salmonella/microsome
    test to evaluate for potential point mutation. Unpublished report No.
    13365A (study no. T2017617) from Bayer AG, Institute of Toxicology,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Herbold, B. (1987) SRA 3886: Cytogenetic study of human lymphocyte
    cultures  in vitro to test for chromosome damage. Unpublished report
    No. 15406, (study No. T3022874) from Bayer AG, Fachbereich Toxicology,
    Wuppertal, Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Herbold, B. (1988) SRA 3886:  In vitro cytogenetic study with human
    lymphocytes for the detection of induced clastogenic effects.
    Unpublished report No. 16690, (study No. T1025572) from Bayer AG,
    Fachbereich Toxicology, Wuppertal, Germany. Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Jones, R.D. & Greufe (1993) Response to US-EPA review: Chronic feeding
    toxicity study with technical grade Fenamiphos (Nemacur(R)) with
    dogs. Unpublished report dated 12 July 1993 (supplement to Mobay Corp.
    study No. 88-274-BB) from Miles Inc., Stilwell, KS, USA. Submitted to
    WHO by Bayer AG. Leverkusen, Germany.

    Jones, R.D. & Loney, M.L. (1993) A subchronic feeding toxicity study
    with technical grade Fenamiphos (Nemacur(R)) in dogs. Unpublished
    report No. 91 - 176-KP (supplement to Mobay Corp. study No. 88-274-BB)
    from Miles Inc., Stilwell, KS, USA. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Kato, M.C. (1984a) Primary dermal irritation study of fenamiphos
    technical in rabbitś Unpublished report No. B-561 from Bozo Research
    Center Inc., Tokyo, Japan (study No. B-561). Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Kato, M.C. (1984b) Primary eye irritation study of fenamiphos
    technical in rabbitś Unpublished report No.B-562 from Bozo Research
    Center Inc., Tokyo, Japan (study No. B-562). Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Khasawinah, A.M. & Flint, D.R. (1972) Metabolism of Nemacur(R)
    [ethyl-4-(methylthio)-m-tolyl isopropylphosphoramidate] by rat liver
    microsomes  in vitro. Unpublished report No. 34217 from Chemagro
    Division of Baychem Corp. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Kimmerle, G. (1970) Subchronic neurotoxicity studies on hens.
    Unpublished report No. 1831 from the Institute for Toxicology, Bayer
    AG, Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Kimmerle, G. (1971) Acute neurotoxicity studies on hens. Unpublished
    report No. 2829 from Institute for Toxicology, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Kimmerle, G. (1972a) Acute toxicity of SRA-3886 in combination with
    S-276 and with E-154 to rats. Unpublished report No. 3438 from
    Institute for Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Kimmerle, G. (1972b) Acute inhalation toxicity study with Nemacur(R)
    active ingredient on rats. Unpublished report No. 3559 from the
    Institute for Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Kimmerle, G. (1972c) Antidotal experiments on rats. Unpublished report
    No. 3560 from the Institute for Toxicology, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Kimmerle, G. & Solmecke, B. ( 1971) BAY 68138 toxicological studies.
    Unpublished report No. 2767 from Institute for Toxicology, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Knaak, J.B., Ackerman, K.Y., Fredrickson, A.S. & Lee, P. (1981)
    Reentry research: Dermal dose red cell cholinesterase-response curves
    for fenamiphos, fenamiphos sulfoxide and sulfone. Unpublished report
    No. 69689 from California Department of Food and Agriculture.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Krötlinger, F. ( 1988 ) Study for acute oral toxicity in rats.
    Unpublished report No. 16744 (study No. T0027669) from Bayer AG,
    Fachbereich Toxicologies, Wuppertal, Germany. Submitted to WHO by
    Bayer AG, Leverkusen, Germany.

    Lamb, D.W. & Landes, A.M. (1978) In vitro inhibition of cholinesterase
    by (R)Nemacur and Nemacur metabolites. Unpublished report No. 66068
    (reference 77-060) from Mobay Chemical Corp., Chemagro Agricultural
    Division, Kansas City, MO, US A. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Lamb, D.W. & Matzkanin, C.S. (1975) The acute oral toxicity of Nemacur
    technical, desisopropyl Nemacur sulfoxide and desethyl Nemacur.
    Unpublished report No. 44531 (reference 74-175) from Chemagro
    Agricultural Division, Mobay Chemical Corp. Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Lamb, D.W. & Matzkanin, C.S. (1977) The acute oral toxicity of Nemacur
    desisopropyl to rats. Unpublished report No. 51597 (reference 75-144)
    from Chemagro Agricultural Division, Mobay Chemical Corp. Submitted to
    WHO by Bayer AG, Leverkusen, Germany.

    Löser, E. (1968a) Subchronic toxicological studies on rats.
    Unpublished report No. 745 from the Institute for Toxicology, Bayer
    AG, Wuppertal-Elberfeld. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Löser, E. (1968b) Subchronic toxicological studies on dogs.
    Unpublished report No. 837 from the Institute for Toxicology, Bayer
    AG, Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Löser E (1969) Subchronic toxicological studies on dogs (3 month
    feeding test). Unpublished report No. 1655 from the Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Löser, E. (1970) Subchronic toxicological studies on dogs. Unpublished
    report No. 2008 from the Institute for Toxicology, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Löser, E. (1972b) Chronic toxicological studies on dogs (2 year
    feeding experiment). Unpublished report from the Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Löser, E. (1972b) Chronic toxicological studies on rats (2 year
    feeding experiment). Unpublished report No. 3539 from the Institute
    for Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to
    WHO by Bayer AG, Leverkusen, Germany.

    Löser, E. (1972c) Generation studies on rats. Unpublished report No.
    3424 from the Institute for Toxicology, Bayer AG, Wuppertal-Elberfeld,
    Germany. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Löser, E. & Kimmerle, G. (1971) Acute and subchronic toxicity of
    Nemacur(R), active ingredient. Pflanzenschutz-Nachrichten Bayer,
    24(1), 69-113. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    MacKenzie, K.M. (1982) Teratology study with Nemacur in rabbits.
    Unpublished report No. 308 (study No. 81165) from Hazelton Raltech,
    Inc., Madison, WI, USA. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Mavournin, K.H., Blakey, D.H., Cimino, M.C., Salamone, M.F. & Heddle,
    J.A. (1990) The in vivo micronucleus assay in mammalian bone marrow
    and peripheral blood. A report of the US Environmental Protection
    Agency Gene-Tox Program.  Mutat. Res., 239, 29-80.

    Mawdesley-Thomas, L. & Urwin, C. (1970a) Pathology report of BAY
    68138. Subchronic toxicological studies in rats. Unpublished report
    (addendum to report No, 745) from Huntingdon Research Centre,
    Huntingdon, United Kingdom. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    Mawdesley-Thomas, L. & Urwin, C. (1970b) Pathology report of BAY
    68138. Subchronic toxicity tests in dogs. Unpublished report (addendum
    to report No. 1655) from the Huntingdon Research Centre, Huntingdon,
    United Kingdom. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Mihail F. (1980) SRA 3885 and Curaterr(R): Evaluation for acute
    combination toxicity. Unpublished report No. 9294 from Institute of
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Mihail, F. & Schilde, B. (1980) SPA 3886 (active ingredient of
    Nemacur(R)) subacute dermal toxicity study on rabbits. Unpublished
    report No, 9297 from Institute of Toxicology, Bayer AG,
    Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Rieth, J.P., Moore, K.D. & Elcock, L.E. (1991 ) Chronic feeding
    toxicity study of technical grade Fenamiphos (Nemacur(R)) with dogs.
    Unpublished report No. 6243 (study No. 88-274-BB) from Mobay Corp.,
    Stilwell, KS, USA. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Schlüter, G. (1981) SRA 3886 (Nemacur): Evaluation for embryotoxicity
    and teratogenic effects in orally dosed rats. Unpublished report No.
    9785 from Institute for Toxicology, Bayer AG, Wuppertal, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Spicer, J. (1970) Pathology report of BAY 68138. Subchronic
    neurotoxicity tests on hens. Unpublished report (addendum to report
    No. 1831 ) from the Huntingdon Research Centre, Huntingdon, United
    Kingdom. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Spicer, J. (1971) Pathology report of BAY 68138. Hen study.
    Unpublished report (addendum to report No. 2829) from Huntingdon
    Research Centre, Huntingdon, United Kingdom. Submitted to WHO by Bayer
    AG, Leverkusen, Germany.

    Stropp, G. (1995) Study for the skin sensitization effect in guinea
    pigs (guinea pig maximization test according to Magnusson and
    Kligman). Unpublished report No. 24341 (study No. T9058358) from Bayer
    AG, Fachbereich Toxikologie, Wuppertal, Germany. Submitted to WHO by
    Bayer AG, Leverkusen, Germany.

    Thomson, C., Newman, A. & Urwin, C. (1972a) Pathology report of BAY
    68138. Subchronic toxicity study in dogs. Unpublished report (addendum
    to report No. 2008) from Huntingdon Research Centre, Huntingdon,
    United Kingdom. Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Thomson, C., Newman, A. & Urwin, C. (1972b) Pathology report of BAY
    68138. Chronic toxicity study in dogs (administration in diet for 2
    years). Unpublished report (addendum to report No. 3561) from
    Huntingdon Research Centre, Huntingdon, United Kingdom. Submitted m
    WHO by Bayer AG, Leverkusen, Germany.

    Thyssen, J. (1974a) Nemacur(R) sulfone, Acute toxicity in rats.
    Unpublished report dated 11 February 1974 from Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Thyssen, J. (1974b) Nemacur(R) sulfoxide, Acute toxicity in rats.
    Unpublished report dated 11 February 1974 from Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Thyssen, J. (1974c) 3-Methyl-4-methylmercaptophenol, Acute toxicity in
    rats. Unpublished report dated 25 February 1974 from Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Thyssen, J. (1974d) 3-Methyl-4-methane-sulfonylphenol, acute toxicity
    in rats. Unpublished report dated 25 February 1974 from Institute for
    Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.

    Thyssen, J. (1974e) 4-Methylmercapto-m-cresol, acute toxicity in rats.
    Unpublished report dated 29 April 1974 from Institute for Toxicology,
    Bayer AG, Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Thyssen, J. (1979a) SPA 3885 (Nemacur(R) active ingredient)/acute
    inhalation toxicity studies. Unpublished report No. 8210 from Bayer
    AG, Wuppertal-Elberfeld, Germany. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Thyssen, J. (1979b) Nemacur(R) active ingredient (SRA 3886)/subacute
    inhalational toxicity studies. Unpublished report No. 8669 from
    Institute of Toxicology, Bayer AG, Wuppertal-Elberfeld, Germany.
    Submitted to WHO by Bayer AG, Leverkusen, Germany.

    Waggoner, T.B. (1972) Metabolism of Nemacur [ethyl 4-(methylthio)-N-
    tolyl isopropylphosphoramidate] and identification of two metabolites
    in plants.  J. Agric. Food Chem., 20, 157-160. Submitted to WHO by
    Bayer AG, Leverkusen, Germany.

    Wargo, J.P. (1978) The effect of feeding Nemacur(R) sulfoxide to
    dairy cattle. Unpublished report No. 66212 from Chemical Agricultural
    Division, Mobay Chemical Corp. Submitted to WHO by Bayer AG,
    Leverkusen, Germany.

    Weber, H. (1988) [Phenyl-1-14C] Nemacur: Whole-body autoradiographic
    distribution of the radioactivity in the rat. Unpublished report No.
    PF3055 (study No. M181 0188-7) from Bayer AG Agrochemicals,
    Leverkusen, Germany. Submitted to WHO by Bayer AG, Leverkusen,
    Germany.

    WHO (1996)  The WHO Recommended Classification of Pesticides by 
     Hazard and Guidelines to Classification 1996-1997 (WHO/PCS/96.3),
    Geneva, International Programme on Chemical Safety.

    Yang, L.L., Putnam, D.L. & Conklin, P.M. (1984) CHO/HGPRT mutation
    assay in the presence and absence of exogenous metabolic activation --
    Nemacur(R). Unpublished report No. 620 (study No. T2600.332) from
    Microbiological Associates Inc., Bethesda, MD, USA. Submitted to WHO
    by Bayer AG, Leverkusen, Germany.
    


    See Also:
       Toxicological Abbreviations
       Fenamiphos (ICSC)
       Fenamiphos (WHO Pesticide Residues Series 4)
       Fenamiphos (Pesticide residues in food: 1977 evaluations)
       Fenamiphos (Pesticide residues in food: 1978 evaluations)
       Fenamiphos (Pesticide residues in food: 1980 evaluations)
       Fenamiphos (Pesticide residues in food: 1985 evaluations Part II Toxicology)
       Fenamiphos (Pesticide residues in food: 1987 evaluations Part II Toxicology)
       Fenamiphos (JMPR Evaluations 2002 Part II Toxicological)