Pesticide residues in food -- 1999 Sponsored jointly by FAO and WHO with the support of the International Programme on Chemical Safety (IPCS) Toxicological evaluations Joint meeting of the FAO Panel of Experts on Pesticide Residues in Food and the Environment and the WHO Core Assessment Group Rome, 20-29 September 1999 CHLORPYRIFOS First draft prepared by D. Wagner Therapeutic Goods Administration,Department of Health and Aged Care, Canberra, ACT, Australia Explanation Evaluation for acceptable daily intake Biochemical aspects Absorption, distribution, and excretion Biotransformation Effects on enzymes and other biochemical parameters Toxicological studies Acute toxicity Short-term studies of toxicity Long-term studies of toxicity and carcinogenicity Genotoxicity Reproductive toxicity Multigeneration reproductive toxicity Developmental toxicity Special studies: Neurotoxicity Studies on metabolites Acute toxicity Short-term studies of toxicity Genotoxicity Developmental toxicity Observations in humans Experimental studies Case reports Monitored field trials Studies of morbidity Comments Toxicological evaluation References Explanation Chlorpyrifos [ O,O-diethyl O-(3,5,6-trichloro-2-pyridinyl) phosphorothioate] is a broad-spectrum organophosphorus pesticide. The toxicology of chlorpyrifos was first evaluated by the 1972 Joint Meeting (Annex 1, reference 18), when an ADI of 0-0.0015 mg/kg bw was established on the basis of a NOAEL of 0.014 mg/kg bw per day in a 1-month study in humans. Further biochemical and toxicological information was considered by the 1977 JMPR (Annex 1, reference 28), when the ADI was changed to 0-0.001 mg/kg bw. Additional reports on the toxicology of chlorpyrifos were reviewed by the 1982 Joint Meeting (Annex 1, reference 38), which increased the ADI to 0-0.01 mg/kg bw on the basis of a NOAEL of 0.1 mg/kg bw per day in humans exposed to chlorpyrifos for 9 days, with a 10-fold safety factor. This ADI was supported by findings in rats and dogs. Chlorpyrifos was reviewed at the present meeting within the periodic review pro-gramme of the Codex Committee on Pesticide Residues. Evaluation for Acceptable Daily Intake 1. Biochemical aspects (a) Absorption, distribution, and excretion Rats A dose of 50 mg/kg bw [36Cl]chlorpyrifos given orally to male Wistar rats by intubation was eliminated rapidly in the urine (about 90% of the dose) and faeces (about 10% of the dose). The compounds excreted in the urine were identified as 3,5,6-trichloro-2-pyridyl phosphated (5-80%), 3,5,6-trichloro-2-pyridinol (TCP; 15-20%), and traces of chlorpyrifos. The concentrations of residue were highest in liver and kidney 4 h after dosing, but the half-time in these tissues was < 20 h. The longest half-time, 62 h, was recorded in fat (Smith et al., 1967). By 72 h after a single oral dose of 19 mg/kg bw [14C]ring-labelled chlorpyrifos was given by intubation to male Sprague-Dawley rats, 83-87% had been eliminated, mainly in the urine (68-70%), faeces (14-15%), and expired air (0.15-0.39%). The residues found at this time represented about 1.7% of the total dose, and the concentration, while highest in fat, was < 1 ppm in any tissue (Branson & Litchfield, 1971a). In a study to investigate the breakdown of chlorpyrifos by microsomal enzymes in vitro (Branson & Wass, 1970), TCP was again identified as the major metabolite of chlorpyrifos. In cows fed chlorpyrifos at a dose of 5 ppm in the diet for four days, hydrolysis to the pyridinol was the major metabolic pathway (Gutenmann et al., 1968). Female Fischer 344 rats were given chlorpyrifos by oral gavage or by inhalation in nose-only or whole-body chambers, while another group was given [14C]chlorpyrifos by oral gavage to estimate the efficiency of recovery. Oral dosing led to 30-80% recovery of the administered dose in urine, while nose-only or whole-body inhalational exposure led to an average urinary excretion of 0.28-0.58 µg of TCP per ppb of chlorpyrifos in air, after adjustment for the route of exposure. Rats with whole-body exposure absorbed more chlorpyrifos than would have been expected to occur by inhalation alone, probably due to grooming activity. The total combined recovery of radiolabel from urine, faeces, and cage wash was 99.8% of the administered dose (Nolan et al., 1986). Single doses of [14C]ring-labelled chlorpyrifos (19 mg/kg bw) or TCP (7 mg/kg bw) were given by oral gavage to Sprague-Dawley rats, and the radiolabel was analysed in blood samples, expired air, and excreta. The main compound found in urine was TCP. The biological half-time of each chemical was estimated to be 8-17 h, and by 72 h > 98% of each compound had been eliminated, mainly in the urine (68-70% of chlorpyrifos, 73-76% of TCP) and faeces (14-15% of chlorpyrifos, 6-7% of TCP). Elimination in the form of radiolabelled carbon dioxide was a minor (< 1%) pathway. The residue concentrations were consistently low in the brain, and the highest were found in bone and intestine after administration of TCP and in fat and intestine after administration of chlorpyrifos. Administration of single oral doses of [14C]TCP or [14C]chlorpyrifos did not lead to accumulation of these compounds in rat tissues (Branson & Litchfield, 1970, 1971b). Groups of male and female Fischer 344 rats were given [14C]chlorpyrifos as a single oral dose of 0.5 or 25 mg/kg bw or unlabelled chlorpyrifos as 15 consecutive doses of 0.5 mg/kg bw per day followed on day 16 by 0.5 mg/kg bw of [14C]chlorpyrifos. By 3 days after exposure, essentially all of the radiolabel had been recovered (> 97%), mainly in the urine (84-92% of the administered dose), with 6-12% in the faeces. Repeated dosing induced a slight increase (6-7%) in urinary excretion as compared with the single dose of 0.5 mg/kg bw per day. At sacrifice, the residues in the carcass accounted for 0.2% of the administered dose of 25 mg/kg bw. The residues were found mainly in perirenal fat and livers of males and in the ovaries and fat of females. The half-time for excretion was 12 h for males and 23 h for females at 25 mg/kg bw. No unchanged chlorpyrifos was found in urine. The major metabolites were TCP (12%), its glucuronide conjugate (80%), and, tentatively, a sulfate conjugate (Nolan et al., 1987). Hens Groups of 36 laying hens were fed diets containing chlorpyrifos at 0 (two groups), 0.3, 1, 3, or 10 (three groups) ppm for 30-45 days before sacrifice. All control birds and 24 birds at each dose were killed after 30 days, and the remaining 12 hens at each dose were fed treated diet for a further 15 days; eggs were collected from these birds throughout treatment. The additional two groups of birds at 10 ppm were allowed to recover from treatment for 7 and 21 days, respectively, after the 30-day treatment period and were then killed. Chlorpyrifos residues were detected only in fat, whereas TCP was found in liver (0.15 ppm), kidney (0.33 ppm), and eggs (< 0.05 ppm) of hens given 10 ppm chlorpyrifos. No residues were detected after the 7-day withdrawal period (Dishburger et al., 1972a). Goats Two goats were fed [14C]ring labelled (positions 2 and 6) chlorpyrifos twice daily in capsules for 10 days at concentrations equivalent to 15-19 ppm in the feed. The majority (80%) of the radiolabel was recovered in urine, with smaller amounts in faeces (3.6%), gut (0.9%), tissues (0.8%), and milk (0.1%). The major urinary metabolite (> 75% of the residual radiolabel) was the ß-glucuronide conjugate of TCP, with smaller amounts of unconjugated TCP. The major residue in fat was chlorpyrifos (0.12 ppm), while TCP was the major residue in liver and kidney (Glas, 1981a). A similar pattern of elimination was seen in a study in which lactating goats were fed [14C]ring labelled chlorpyrifos twice daily by capsule; little radiolabel (0.05-0.14%), mainly associated with chlorpyrifos, was recovered in milk (Glas, 1981b). Pigs Groups of two boars and one sow were fed basal rations containing 0, 1, 3, or 10 ppm chlorpyrifos for 30 days, and tissue samples were collected after withdrawal periods of 0, 7, and 21 days. Residues were initially found predominantly in fat tissues, with lower concentrations in muscle, liver, and kidney, but no residues were detectable after 7 days of withdrawal (McKellar et al., 1972). Weanling piglets of each sex were given daily oral doses of[14C]TCP equivalent to 75 ppm in the diet for 7 days, and urine and tissue samples were collected after withdrawal periods of up to 7 days. The principal urinary excretion product was unchanged TCP. The liver and kidney initially contained the highest residues; while the concentrations decreased rapidly after withdrawal, the liver had the slowest rate of clearance (Bauriedel & Miller, 1981). Cattle Calves were fed TCP in the feed at concentrations < 100 ppm for 28 days, and tissue samples were taken on day 28 or after a 3-, 7-, or 21-day withdrawal period. The concentrations of residues were highest in liver and kidney, with lower concentrations in fat and muscle. These concentrations declined rapidly after the return to untreated feed (Glas, 1977a). One calf fed 2-methoxy-3,5,6-trichloropyridine for 28 days showed significant concentrations (> 2 ppm) of TCP but no 2-methoxy-3,5,6-trichloropyridine in liver, kidney, and fat (Glas,1977b). Female calves were fed one capsule of chlorpyrifos per day for 30 days at doses equivalent to daily concentrations of dry matter in the diet of < 100 ppm. Tissue samples were taken on day 30 or after a 1-5-week withdrawal period. A dose-related increase in the concentration of residues was seen in all tissues but especially in fat, whereas TCP was found predominantly in the liver and kidney. After the dose of 100 ppm, the residues of chlorpyrifos in fat were 0.93 ppm at 7 days and 0.02 ppm at 35 days after withdrawal of the test material (Dishburger et al., 1972b, 1977). Humans In persons poisoned with chlorpyrifos formulations, chlorpyrifos was detected in serum samples only and at lower concentration than the diethylphosphorus metabolites, which were excreted mainly in urine. The urinary diethylphosphorus metabolites were excreted by first-order kinetics, with an average elimination half-time of 6.1 ± 2.2 h in the fastest phase and 80 ± 26 h in the slowest (Drevenkar et al., 1993). Male volunteers received chlorpyrifos as an oral dose of 0.5 mg/kg bw and 1 month later a dermal dose of 5 mg/kg bw. The time to the maximal concentration of TCP in blood was 0.5 h after oral dosing and 22 h after dermal treatment. The elimination half-time, irrespective of the route of administration, was 27 h. The percentage of the administered dose recovered from the urine was 70% after oral dosing and 1.3% after dermal administration (Nolan et al., 1982, 1984). (b) Biotransformation A generalized metabolic pathway for chlorpyrifos is shown in Figure 1. Chlorpyrifos, like many of the most commonly used organophosphorus insecticides, is a phosphorothionate. Phosphorothionate insecticides are bioactivated by the microsomal cytochrome P450 system in the bodies of vertebrates and insects to their active oxon (phosphate ester) metabolites, which are about three orders of magnitude more potent as anticholinesterases than the parent compounds. Most bioactivation takes place in the liver, while detoxification takes place in the liver and plasma. Chlorpyrifos is rapidly metabolized by mixed-function oxidases to the highly reactive chlorpyrifos oxon by oxidative desulfuration (step 1). Oxidative metabolism involving concurrent activation and degradation via a common intermediate appears to be the general pattern for P=S esters (Yang et al., 1971; Wolcott et al., 1972; Nakatsugawa, 1992). The oxidative desulfuration is believed to proceed via an electrophilic phosphooxathiiran intermediate (Kamataki & Neil, 1976). The degradation step occurs by conversion directly to TCP and diethyl thiophosphate (step 2). The oxon can be deactivated by hydrolysis to diethylphosphate and TCP (step 4) (Ma & Chambers, 1994; Sultatos & Murphy, 1983). A minor reaction pathway is hydrolysis to monethyl 3,5,6-trichloro-2-pyridinyl phosphorothioate (step 3). The moderate toxicity of chlorpyrifos, when compared with some other phosphorothionates, may be due to hydrolytic detoxification of the oxon by A-esterases, such as paraoxonase, and high serum concentrations of paraoxonase may protect against poisoning by organophosphorus insecticides with paraoxonase substrates as active metabolites (Omenn, 1987; Geldmacher-von Mallinckrodt & Diepgen, 1988). This effect has been demonstrated in rats directly, in which injection of purified paraoxonase before dosing with chlorpyrifos oxon reduced the inhibition of brain acetylcholinesterase in these animals by 2.5 times when compared with controls (Costa et al., 1990). Twelve rats given chlorpyrifos at 5 mg/kg bw (3 mCi per rat) orally by gavage excreted 88% of the administered radiolabel in urine within 48 h. At least six metabolites were present, three of which accounted for 97% of the excreted radiolabel. These were identified as the glucuronide of TCP (80%), a glucoside of TCP (4%), and TCP itself (12%) (Bakke et al., 1976).(c) Effects on enzymes and other biochemical parameters Hydrolysis is the most important route of detoxification of organophosphorus esters. Hydrolytic esterases are distributed ubiquitously in the blood and tissues of virtually all animal organisms and catalyse the hydrolysis of a variety of esters, including organophosphorus esters, but have little activity on organophosphorus itself. Esterases that interact with organophosphoruses have been characterized as A- and B-esterases according to their sensitivity to inhibition by organophosphorus compounds. Plasma and tissues of mammals have significant concentrations of the calcium-activated A-esterases, arylesterase (EC 3.1.1.2), and the high-density lipoprotein-associated paroxonase (EC 3.1.8.1). The B-esterases, including aliesterases (EC 3.1.1.1; also called carboxylesterases) and cholinesterases (e.g. butyryl cholinesterase, EC 3.1.1.8), are inhibited by organophosphorus compounds such as chlorpyrifos oxon. While they bind them, they do not hydrolyse them (Derelanko & Hollinger, 1995). Butyrylcholinesterase is the predominant cholinesterase in human serum (> 99%), while in rats there is an approximately equal distribution of acetylcholinesterase and butyryl cholinesterase (Nolan, 1997). These enzymes detoxify organophosphorus compounds by sequestering them through binding to or phosphorylation by blood proteins, such as albumin, which stoichiometrically degrades them (Aldridge, 1953). These protein-bound organophosphorus esters are secreted into the bile. Erythrocyte acetylcholinesterase (EC 3.1.1.7) is similar to the acetylcholinesterase in nervous systems and can bind to and sequester or hydrolyse organophosphorus compounds. The enzyme activities attributable to A-esterases, B-esterases, and cholinesterases vary widely within populations and are influenced by genetic and environmental factors and disease states (Derelanko & Hollinger, 1995). The serum activity of paraoxonase in rabbits is some 40 times greater than that in rats. Most avians, including hens, have relatively low activity of A-esterases, and the mean value of serum chlorpyrifos oxonase activity in humans is 10 times that of rat serum (Furlong et al., 1989). In humans, a substrate-dependent polymorphism of serum paraoxonase is observed, in which one isoform of paraoxonase has a high turnover for paraoxon and the other a low turnover (Furlong et al., 1989; Smolen et al., 1991). The polymorphism is also observed with the oxons of methyl parathion, chlothion, and phenylphosphonothioic acid O-ethyl O-para-nitrophenyl ester. The two isoforms appear, however, to hydrolyse chlorpyrifos oxon and phenylacetate at the same rate. Cloning and sequencing of the human paraoxonase cDNA has revealed the molecular basis of the polymorphism: arginine at position 192 determines high paraoxonase activity, and glutamine at this position encodes low paraoxonase activity (Humbert et al., 1993). In addition to this polymorphism, a 13-fold variation in serum enzyme activity of a given genetic class is seen (Furlong et al., 1989). While chlorpyrifos oxon was hydrolysed by the same plasma fraction that hydrolysed paraoxon in a study of plasma samples from 320 white blood donors (Furlong et al., 1988), the population distribution of chlorpyrifos oxon hydrolysis was unimodal. This contrasted with a bimodal population distribution of the activity for paraoxon hydrolysis, and the variation in paraoxonase activity was about 11-fold, while that in chlorpyrifos oxonase activity showed a four- to fivefold variation. Correlation of the LD50 values for orally administered chlorpyrifos in rats with the activation rates measured in brain rather than liver (Chambers, 1992) suggested that local biotransformation in target tissues is an important determinant of its toxicity. In a more recent review, however, Chambers & Carr (1995) compared published LD50 or LC50 values for a variety of insecticides in several vertebrate species. Studies in rats indicated that the sensitivity of brain acetylcholinesterase activity to inhibition by various phosphorothionate oxons did not correlate with their acute toxicity. Chlorpyrifos oxon has a greater affinity for rat brain acetylcholinesterase than does paraoxon, with median inhibitory concentrations (IC50 values) of 4.0 and 22 nmol/L, respectively, but a lower LD50. This finding is consistent with the greater affinity (IC50, 0.75 nmol/L) of chlorpyrifos oxon for plasma aliesterases than for rat brain acetylcholinesterase, indicating that aliesterases provide protection against chlorpyrifos oxon. Further work by Chambers & Carr (1995) indicated longer inhibition of esterases after exposure chlorpyrifos than to parathion. This was attributed to the greater lipophilicity of chlorpyrifos than parathion (hexane:acetonitrile partition coefficients, 0.28 and 0.062, respectively) and the assumption that a substantial fraction of the dose of chlorpyrifos would have been sequestered by fat and released gradually for later bioactivation. Studies of hepatic microsomal metabolism of parathion and chlorpyrifos indicated that desulfuration of parathion was favoured over dearylation (activation vs deactivation), whereas the reverse was seen for chlorpyrifos (Ma & Chambers, 1994). In channel catfish, however, the sensitivity of acetylcholinesterase to inhibition by oxons reflects the acute toxicity of these insecticides, and may be largely responsible for their toxicity in this species. Thus, the metabolism of insecticides appears to be more influential in some species than in others in determining their toxicity. Chlorpyrifos oxonase is a calcium-dependent A-esterase that hydrolyses chlorpyrifos oxon, the active metabolite of chlorpyrifos. As this activity may be related to that of paraoxonase, it was determined spectrophotometrically by measuring the generation of TCP (Furlong et al., 1989). Mortensen et al. (1996) hypothesized that young rats have less chlorpyrifos oxonase activity than adults, and they measured the activity of this enzyme in the brain, plasma, and liver of male Long-Evans rats (CRL:(LE)BR) at postnatal day 4 and in adults. No activity was detected in brain at either age, but the activities in plasma and liver were markedly lower in younger than in adult animals, the activities in plasma and liver being 1/11 and 1/2 of those in adults, respectively. As the Michaelis-Menten constant for chlorpyrifos oxonase activity was high (210-380 µmol/L), experiments were performed to determine whether this enzyme could hydrolyse physiologically relevant concentrations (nanomolar to low micromolar) of chlorpyrifos oxon. On the assumption that tissue acetylcholinesterase inhibition provides a sensitive bioassay for the concentration of chlorpyrifos oxon, changes in the tissue acetylcholinesterase IC50 for chlorpyrifos oxon were measured in the presence and absence of chlorpyrifos oxonase. Brain acetylcholinesterase activity was determined spectrophotometrically, while plasma and liver cholinesterase activities were determined by a radiometric method with a [3H]acetylcholine iodide substrate. An increase in the 'apparent' IC50 would indicate that chlorpyrifos oxonase had hydrolysed substantial amounts of chlorpyrifos oxon during the 30-min preincubation with the chlorpyrifos oxon. In the adult rats, the apparent IC50 values for both plasma and liver cholinesterase were higher in the presence of chlorpyrifos oxonase, suggesting that the enzyme in those tissues could hydrolyse physiologically relevant concentrations of chlorpyrifos oxon within 30 min. Young animals, however, showed less of a shift in the IC50 curves than adults, confirming that they have a lower capacity to detoxify physiologically relevant concentrations of chlorpyrifos oxon with chlorpyrifos oxonase. 2. Toxicological studies (a) Acute toxicity (i) Lethal doses Numerous studies have been carried out with technical-grade chlorpyrifos to establish LD50 and LC50 values (Table 1). The lowest value after oral administration was 96 mg/kg bw (range, 96-480 mg/kg bw) in rats and 100 mg/kg bw (range, 100-150 mg/kg bw) in mice. The signs of acute intoxication were consistent with cholinesterase inhibition and included inactivity, salivation, dyspnoea, flaccid paralysis, vomiting, piloerection, exophthalmia, and diarrhoea; female animals were generally more sensitive to the acute effects of chlorpyrifos than males. The LD50 for dermally applied chlorpyrifos was consistently lower than that for oral or inhalational exposure and indicated the lower dermal absorption of chlorpyrifos (about 2% in humans) when compared with the 70-90% absorption after inhalational or oral exposure. (ii) Dermal and ocular irritation and dermal sensitization Ocular irritation: In a paper in which few procedural details were provided (Taylor & Olson, 1963), slight conjunctival redness was seen in rabbits treated with chlorpyrifos, regardless of whether the eyes were washed after administration of the test material. The redness persisted for more than 7 days in some animals. Transient conjunctival and iridial irritation, possibly due to a direct physical irritating effect of cholorpyrifos powder, was seen in three young adult female New Zealand white rabbits after each had received 100 mg of a finely ground commercial preparation in the left conjunctival sac. The eyes of all animals were normal after 24 h (Jones, 1985a). The ocular irritation potential of technical-grade chlorpyrifos (purity, 99%) was tested (in accordance with GLP requirements) in six young male New Zealand white rabbits, 100 mg of test material being placed into the right eye and the untreated left eye serving as a control. There were no signs of corneal or iridial irritation. Slight to moderate conjunctival redness (mean Draize score, 1 at 1 h, 0.83 at 24 h), slight chemosis (mean Draize score, 0.83 at 1 h, 0.16 at 24 h), and discharge (mean Draize score, 1.6 at 1 h, 0.83 at 24 h) were seen up to 24 h after instillation, but no signs of irritation were reported at 48 h (Jackson & Ogilvie, 1994b). The ocular irritation potential of 0.1 g of chlorpyrifos (purity, 99.3%) was assessed in three male and three female New Zealand white rabbits by instilling the test substance into the conjunctival sac of the right eye, the left eye remaining untreated and serving as control. This study was conducted according to GLP requirements. Diffuse corneal opacities (score 1 for degree and area of opacity) were observed in two animals at 24 h, but these effects had disappeared by 48 h and no other corneal effects were seen. Iridial inflammation (score 1) was noted in four animals at 1 h, and this effect persisted for 24 h in one of the animals and for 48 h in another. No iridial effects were observed after 72 h. Conjunctival irritation was observed in all animals, with redness (score 2: diffuse, deeper crimson red at 1-24 h; score 1: vessels definitely injected above normal at 48 h), chemosis (score 2: obvious swelling with partial eversion of lids up to 48 h), and discharge (scores 2-3: moistening of the lids and hairs just adjacent to lids or a considerable area around the eye only at 1 h). No signs of conjunctival irritation was observed at 72 h. The maximum individual irritation scores were seen at 1 h and ranged from 12 to 19, with a mean score of 16. The mean scores at 24, 48, and 72 h were 9.3, 2.5, and 0, respectively. The overall scores according to EEC Council Directive 67/548/EEC were 2 (mean, 0.11) for corneal opacity, 3 (0.17) for iridial inflammation, 11 (0.61) for conjunctival redness, and 8 (0.44) for conjunctival chemosis (Dreher, 1994d) Technical-grade chlorpyrifos (100 mg; purity, 96.2%) was instilled into the conjunctival sac of the left eye of Himalayan albino rabbits, and the right eyes of the animals served as controls. A group of control animals was similarly treated with distilled water only. All animals were observed for signs of ocular irritation at 1, 4, 24, 48, and 72 h, and then daily up to day 7. At the 4-h observation, very slight iridial irritation (score 1) and conjunctival irritation (score 1) were reported. It was not clear from the report whether the conjunctival irritation was redness, discharge, or chemosis. No signs of irritation were reported at 24 or 72 h. A 'quality assurance statement' was issued for this study (Frederick Institute of Plant Protection and Toxicology, 1995d). Table 1. Studies of the acute toxicity of technical-grade chlorpyrifos Species Strain Sex Vehicle LD50 (mg/kg bw; GLP Reference 95% CI or range) or QA Oral administration Mouse SmithWebster M 1% aqueous gum 102 (94-110) Coulston et al. (1971) tragacanth Mouse NAMRU F Soya bean oil 152 (143-162) Berteau & Deen (1978) Mouse Swiss albino M&F Vegetable oil 109 (93-127) QA Fredrick Institute (1995a) Rat NR M 5% corn oil 163 (97-276) Taylor & Olson (1963) F 135 (97-188) Rat Sprague-Dawley F Soya bean oil 169 (146-196) Berteau & Deen (1978) Rat Sprague-Dawley M Arachis oil 276 (167-455) GLP Dreher (1994a) F 350 (285-429) M&F 320 (260-393) Rat Wistar M&F Vegetable oil 134 (102-163) QA Frederick Institute (1995b) Rat Sprague-Dawley M Maize oil 264 GLP Wilson & MacBeth (1994) F 141 M&F 192 Rat Sprague-Dawley M Maize oil 475 (311-727) Buch & Gardner (1981) F 337 (220-515) Rat Sprague-Dawley M Corn oil 221 (181-69) Nissimov & Nyska (1984a) F 144 (105-200) Table 1. (continued) Species Strain Sex Vehicle LD50 (mg/kg bw; GLP Reference 95% CI or range) or QA Rat Sprague-Dawley M Not reported 205 (134-299) Henck & Kociba (1980) F 96 (72-140) M 248 (170-379) F 97 (80-112) M 270 (188-426) F 174 (130-244) Guinea-pig NR M Corn oil 504 (300-850) GLP Lackenby (1985a) Rabbit NR M Corn oil 1000-2000 GLP Lackenby (1985a) Chicken NR M Capsule 32 (14-72) GLP Lackenby (1985a) Chicken Leghorn M Diet 25 (21-31) Sherman et al. (1967) Chicken Leghorn M Capsule 32 Stevenson (1963) Chicken White Rock M&F Capsule 50-63 Stevenson (1966a) Chicken NR M&F Capsule 20-50 Ross & Roberts (1974) Chicken NR M&F Corn oil 102 (64-169) Ross & Roberts (1974) Chicken NR M Capsule, 32 (14-72) Taylor & Olson (1963) undiluted Turkey Beltsville NR NR 32-63 Stevenson (1967) small white Table 1. (continued) Species Strain Sex Vehicle LD50 (mg/kg bw; GLP Reference 95% CI or range) or QA Inhalation Mouse NAMRU F 65% xylene 94 (83-106) Berteau & Deen (1978) (27-50 min, whole body, aerosol) Rat Albino M&F Vapour (4 h, > 200a GLP Hardy & Jackson (1984) (HC/CFHB) whole body) Rat Sprague-Dawley F 65% xylene 78 (57-108) Berteau & Deen (1978) (60-180 min, whole body, aerosol) Rat Sprague-Dawley M&F Undiluted (4 h, > 230b (max. GLP Anderson et al. (1995) nose only, attainable powder) concentration) Rat Sprague-Dawley M 40% xylene > 4070a Buch (1980) F (4 h, nose only, 2890b (2010-4160) aerosol) Rat Wistar M&F Undiluted (4 h, > 1020a GLP Kenny et al. (1987) whole body, aerosol) Rat Fischer 344 M&F 1% aqueous > 3200a GLP Phillips & Lomax (1989) solution (4 h, whole body, aerosol) Table 1. (continued) Species Strain Sex Vehicle LD50 (mg/kg bw; GLP Reference 95% CI or range) or QA Rat Sprague-Dawley M&F Vapour (4 h, > 36a (max. GLP Blagden (1994) nose only) attainable concentration) Rat Wistar M&F Acetone (4 h, 560b (360-950) Fredrick Institute (1996a) whole body, nebulized particles < 5 µm) Dermal application Rat Sprague-Dawley M&F PEG > 2000 Lackenby (1985b) Rat Sprague-Dawley M&F Undiluted > 2000 GLP Jackson & Ogilvie (1994a) Rat NR M&F Undiluted > 2000 QA Nissimov & Nyska (1984b) Rat Fischer M&F Undiluted > 2000 GLP Jeffrey et al. (1986) Rat Sprague-Dawley M&F Arachis oil > 2000 GLP Dreher (1994b) Rat Sprague-Dawley M&F Saline > 5000 (intact Buch et al. (1980) and abraded) Rabbit Himalayan M&F Water 1233 (993-1531) QA Fredrick Institute (1995c) Rabbit New Zealand M&F Undiluted 1580 (828-2606) Henck & Kociba (1980) white M&F 1598 (1243-1919) M&F 1801 (1023-3152) Table 1. (continued) Species Strain Sex Vehicle LD50 (mg/kg bw; GLP Reference 95% CI or range) or QA Rat CFY M&F Tween 20/ 147 (120-179) Davies & Kynoch (1970) DMSO/water 2:3:5 (subcutaneous) M, male; F, female; NR, not reported; GLP, good laboratory practice; QA, quality assurance; PEG, polyethylene glycol; DMSO, dimethyl sulfoxide Technical-grade chlorpyrifos (100 mg; fine powder) was placed into the right eye of nine young adult albino rabbits. The eyes of three animals were irrigated with saline solution 30 s after treatment, while the eyes of the remaining animals were not irrigated. Ocular examinations were conducted after 24, 48, and 72 h and at 4 and 7 days. Most of the animals displayed slight initial pain after instillation of the test material. No signs of irritation were observed in the cornea or the iris. Slight conjunctival redness was observed in all animals with unwashed eyes, and in two animals whose eyes were irrigated after 30 s. The irritation disappeared in most animals by days 3-4 but persisted for 7 days in a single animal with unirrigated eyes. The mean irritation scores in rabbits with unwashed eyes were 2.3 at 24 h and 0.3 at 7 days, while in the rabbits with irrigated eyes the scores were 1.3 and 0, respectively (Buch & Gardner, 1980a). Dermal irritation: A finely ground commercial preparation of chlorpyrifos (500 mg) moistened with distilled water was applied under an occlusive dressing to about 6 cm2 of clipped skin on the back of three young adult female New Zealand white rabbits as a single 4-h application. No primary irritation was observed (Jones, 1985b). The dermal irritation potential of technical-grade chlorpyrifos (purity, 99%) was tested in a study conducted according to GLP in six young male New Zealand white rabbits, to which 500 mg of the material (moistened with water) were applied under a gauze patch on clipped intact skin on the trunk and then covered with an occlusive dressing. The dressings were removed after 4 h and the skin wiped to remove residual material. The skin was examined for signs of irritation 1, 24, 48, and 72 h after removal of the patch and scored for irritation on the basis of a system recommended by the US Environmental Protection Agency. Further assessments were conducted after 4, 5, and 6 days to determine the reversibility of the effects. Slight erythema (score 1) was noted on three of six test sites at 1 h, and this effect persisted until 72 h at one site and until day 5 in another animal. No skin irritation was seen on day 6 (Jackson & Ogilvie, 1994dc). A technical-grade, fine-powder commercial preparation (purity not stated) was impregnated at a dose of 0.5 g onto unmedicated lint patches and tested in six young adult male New Zealand white rabbits on shaved abraded and intact sites on the back. The patches were held in place under impermeable dressings, which were removed after 23 h. The reactions at the test sites were assessed by the method of Draize at 24 and 72 h and 7 days after administration of the test material. Well-defined or very slight erythema was seen in all animals at 24 h, on both intact and abraded sites. Very slight oedema was also seen at the intact and abraded test sites of one animal at 24 h, another at 72 h, and a third at 7 days. No skin reactions were seen in other animals at these intervals. The primary irritation index was calculated to be 0.67 (Buch & Gardner, 1980b). Technical-grade chlorpyrifos (purity, 96.2%) was applied at a dose of 500 g to clipped intact and abraded skin on the trunks of three male and three female Himalayan albino rabbits, and the trunks were wrapped with plastic sheets held in place with tapes for 24 h. A control group was treated with distilled water. After exposure, the coverings were removed, but the report did not indicate whether the test sites were washed or cleaned. The skin was observed 24 and 72 h after application for signs of irritation. No sign of erythema or oedema was reported. A 'quality assurance statement' was issued for this study (Frederick Institute of Plant Protection and Toxicology, 1995e). Technical-grade chlorpyrifos (purity, 99.3%), moistened with 0.5 ml distilled water, was applied at a dose of 0.5 g to clipped areas on the backs of New Zealand white rabbits in a study carried out in accordance with GLP. Surgical gauze was applied over the sites and held in place with a strip of surgical adhesive tape, and the trunk of each rabbit was wrapped in an elasticized corset. After the 4-h contact period, the corset and patches were removed, and the treated skin was wiped with cotton-wool soaked in distilled water. One hour and approximately 24, 48, and 72 h after removal of the patches, the test sites were examined for evidence of dermal irritation and scored according to the method of Draize. Very slight (score 1) to well-defined (score 2) erythema and very slight (score 1) to slight (score 2) oedema were observed in all animals 1-24 h after removal of the patches. At 48 h, very slight erythema was observed in three animals and very slight oedema in one. No sign of skin irritation was seen at 72 h. The sum of the readings at 24 and 72 h was 10, and the primary irritation index (the sum of the readings/12) was 0.8. This score indicates mild irritation according to Draize (Dreher, 1994d). Dermal sensitization: The skin sensitizing potential of technical-grade chlorpyrifos (purity, 99.3%) was assessed in female albino Dunkin-Hartley guinea-pigs in a study conducted according to GLP. Very slight (score 1; 10 animals) to well-defined (score 2; 2 animals) erythema and very slight (score 1; 1 animal) oedema were observed 1 h after topical induction, but no signs of irritation were observed at 24 h. No skin reaction was observed in control animals, and no skin reactions were observed at the test or vehicle control sites after topical challenge 21 days after induction. One animal was found dead at this stage of the study, but the cause of death was not determined. The test material was not a skin sensitizer in this study (Dreher, 1994e). Technical-grade chlorpyrifos (purity, 96.2%) was prepared as a 1% solution in 0.5% carboxymethyl cellulose and tested at a dose of 100 mg in male Hartley guinea-pigs. Test and positive control groups received induction by exposure of the shaven skin of the left flank, and challenge 14 days after the third induction dose. Control animals received only a challenge with 0.5 ml of a 0.1% solution. The test animals had barely perceptible (score 1; 4/10 animals) erythema or slight oedema (score 1; 2/10 animals) 24 h after the challenge, but no signs of irritation were observed at 48 h. A single control animal had barely perceptible erythema at 24 h, but no other signs of skin irritation were seen in this group. Positive control animals had barely perceptible to deep-red erythema (score 1-3) and slight to marked oedema (score 1-3) 24 h after the challenge and barely susceptible erythema and slight to marked oedema at 48 h. This study was issued a 'quality assurance statement' (Frederick Institute of Plant Protection and Toxicology, 1996b). After a study to determine a suitable dosing regimen, six male and six female young albino Dunkin-Hartley guinea-pigs were treated with a commercial preparation of chlorpyrifos in a standard Beuhler skin sensitization assay. The test substance (0.3 ml, 100% w/v in polyethylene glycol on a lint pad) was applied for 6 h under an occlusive bandage to the freshly clipped left flank of the animals on days 1, 8, and 15. Challenge doses of chlorpyrifos and solvent were applied for 6 h on day 29 to the right and left flanks, respectively. The negative controls were treated only on day 29, when they received the same treatment as the test animals. The challenge sites of both groups were evaluated 24 and 48 h after removal of the patch. No adverse skin reactions were seen after exposure to chlorpyrifos or vehicle alone (Jones, 1985c). Technical-grade chlorpyrifos (purity, 99%) was tested in young female Dunkin-Hartley guinea-pigs by the Magnusson-Kligman maximization test in a study conducted according to GLP. The test consists of an induction with intradermal injections of the test material followed after 1 week by topical application, and a challenge 3 weeks after induction. The test sites were assessed for irritation 24, 48, and 72 h after injection and 24 and 48 h after removal of the patch (48-h exposure). A concentration of 25% of the test material in maize oil was selected for injection in the induction phase and 75% in maize oil for the topical induction phase; a concentration of 75% in maize oil was also selected for the challenge. During the induction phase, slight or discrete erythema was observed in all test and control animals 1 and 24 h after injection and 1 h after topical application. A number of test and control animals also showed slight or discrete erythema 24 h after topical application. During application, two test animals became ataxic, thin, and subdued and were removed from the study, and one test animal was removed from the study before the challenge application because it had lesions at the test site. A single test animal showed a positive response to challenge, with slight to discrete erythema. None of the animals given the vehicle alone reacted to the challenge application (Jackson & Ogilvie, 1994a). Technical-grade chlorpyrifos (purity, 96.8%) dissolved in 100% dimethylsulfoxide and diluted in the same solvent to achieve the desired concentrations was tested in a study that conformed to GLP in albino Dunkin-Hartley guinea-pigs. In animals treated with a single dose, irritation ranging from scattered mild redness to moderate and diffuse redness was seen at 24 h in 5/10 animals; only mild redness was seen at 48 h in 4/10 animals. In 20 animals induced and then challenged with chlorpyrifos, only one had scattered mild redness after the challenge dose at 24 h, and no skin reaction was seen at 48 h. In a positive control group, 9/10 animals had mild redness at 24 h and 4/10 at 48 h (Berman, 1987). (b) Short-term studies of toxicity Mice In a 2-week study conducted according to GLP requirements, young adult CD-1 mice, were given a commercial preparation of technical-grade chrlopyrifos (purity, 96.3%) at dietary concentrations of 0 (control), 75, 150, 300, 600, or 1200 ppm, equaivalent to 0, 14-18, 30-32, 56-67, 89-100, and 130-180 mg/kg bw per day, respectively. Deaths and treatment-related clinical signs, including tremor, ocular opacity, ocular staining, hunching, and lachrymation, were seen mainly in animals at the highest dose. Marked reductions in body weight and feed consumption were observed in animals at doses > 600 ppm. In males, cholinesterase activity was reduced at all doses. At 75 ppm, plasma cholinesterase activity was reduced by > 95%, erythrocyte acetylcholinesterase activity by almost 40%, and brain acetylcholinesterase activity by about 50%. Inhibition of erythrocyte acetylcholinesterase activity was not dependent on dose, and approximately 35% inhibition being seen at all doses, but plasma and brain cholinesterase activities were reduced in a dose-related manner, with an inhibition of 99% in plasma and about 89% in brain at the high dose. Similar patterns of cholinesterase inhibition were seen in females. On the basis of the results of this study, doses of 5, 50, 200, 400, and 800 ppm were selected for a 13-week dietary study in mice (Crown et al., 1984a). Groups of 40 male and 40 female CD-1 mice were given chlorpyrifos (purity, 95.7%) in the diet at 0 or 15 ppm, equal to 2.7 mg/kg bw per day for males and 3.4 mg/kg bw per day for females; 20 animals of each sex per group were killed after 1 week, and 20 were treated for a total of 4 weeks. There were no significant clinical signs or unscheduled deaths. Food consumption was unaffected by treatment, but the body-weight gain of male mice was decreased by 25% at the end of the study. Organ weights were not affected by treatment, and there were no significant differences between groups. The findings at necropsy were within normal limits for all groups. After 1 week, plasma cholinesterase activity was depressed by 88-91% and erythrocyte acetylcholinesterase activity was depressed by 40-53%; after 4 weeks, plasma cholinesterase activity was depressed by 91% and erythrocyte acetylcholinesterase activity by 53%. Brain acetylcholinesterase activity was unaffected by treatment at either time. There was no difference between the sexes in the alterations in plasma and erythrocyte cholinesterase activity (Davies et al., 1985). Groups of 12 Crl CD-1 mice of each sex were given chlorpyrifos (purity, 93.5%) in the diet at concentrations of 0, 5, 50, 200, 400, or 800 ppm (equivalent to 0, 0.7-1.3, 7.1-14, 32-53, 41-140, and 110-300 mg/kg bw per day, respectively) for 13 weeks. This study was conducted to the requirements of GLP. Dose-related increases in the mortality rate and the frequency of ocular opacities were seen at 400 and 800 ppm. Body weights were reduced by 10-15% in mice at 800 ppm throughout the study. Plasma cholinesterase activity was markedly decreased at all doses, and decreased erythrocyte acetylcholinesterase activity was seen, but with no clear dose-dependency; brain acetylcholinesterase activity was inhibited in a dose-dependent manner in males at doses > 200 ppm and in females at doses > 50 ppm (Table 2). Table 2. Group mean values for inhibition of cholinesterase activity in Crl CD-1 mice given chlorpyrifos in the diet (expressed as a percentage of control values) Cholinesterase Sex Dose (ppm) 5 50 200 400 800 Plasma Male 45*** 95*** 98*** 99*** 99*** Female 36*** 97*** 98*** 99 *** 99 *** Erythrocyte Male 34a 21 16 12 26 Female 11 57*** 44** 48** 44** Brain Male 43a 14* 80*** 85*** 87** Female 3a 28*** 58*** 84*** 88*** Plasma, butyryl cholinesterase; erythrocyte and brain, acetylcholinesterase * p < 0.05; ** p < 0.01; *** p < 0.001 a Increased in comparison with controls A single malignant lymphoma was reported in a female at the high dose at terminal sacrifice, but no other neoplastic lesions were reported. Given the isolated nature of this finding, it was considered not to be related to treatment. The absolute and relative organ weights were generally unaffected by treatment, although the relative liver weights were increased in females at 200, 400, and 800 ppm. The clinical signs included urogenital staining in males at doses > 200 ppm and in females at 800 ppm. Ocular opacities occurred in some animals at 400 and 800 ppm. Dose-related histopathological findings were seen in the adrenal glands (including lipogenic pigmentation at doses > 200 ppm in females and at > 400 ppm in males) and in the eyes (acute or subchronic keratitis in two males and four females at 800 ppm and in a single female at 400 ppm). Erythrocyte acetylcholinesterase activity was variable, and the effect of treatment on this parameter was equivocal. The NOAEL was 5 ppm, equal to 0.7 mg/kg bw per day, on the basis of inhibition of brain acetylcholinesterase activity at higher doses (Crown et al., 1987). Rats In a 2-week study to establish the doses to be used in a 13-week study, technical-grade chlorpyrifos (purity, 95.5%) was administered to groups of five rats of each sex at dietary concentrations of 0 (controls), 10, 30, 84, 240, or 694 ppm for 14 days, equivalent to 0, 1.4-1.9, 4.1-5.2, 12-14, 34-39, and 66-95 mg/kg bw per day, respectively. At the high dose, clinical signs of intoxication were reported, consisting of irritability, hunching, tremor, ataxia, urogenital staining, pigmented orbital secretion, failure to groom, and proneness. No treatment-related clinical signs were observed at other doses, and treatment-related deaths were confined to animals at the high dose. Body weights were reduced in males and females at 694 ppm and in females at 240 ppm, and food consumption was also reduced in animals at 694 ppm. Dose-related decreases in blood cholinesterase activity were seen at all doses, the inhibition ranging from 42% at 10 ppm to 93% at 694 ppm. On the basis of these findings, the doses selected for the 13-week study were 0.5, 10, and 100 ppm (Crown et al., 1984b). Plasma, erythrocyte, and brain cholinesterase activities were markedly reduced in rats after dietary administration of chlorpyrifos for 14 days at doses of 5 or 10 mg/kg bw per day in a study carried out according to GLP. Inhibition of cholinesterase activity occurred in the absence of clinical signs of intoxication. Feed consumption, body weights, and gross pathological appearance were similar in control and treated groups. Statistically significant increases in absolute and relative adrenal weights (about 20% compared with controls) were observed in females only at 10 mg/kg bw per day (Liberacki et al., 1990). Fischer 344 rats received technical-grade chlorpyrifos (purity, 95%) by nose-only inhalation for 6 h/day for 5 days at a target concentration of 20 ppb in a study that conformed to GLP. No deaths or treatment-related clinical signs were reported. Exposure of female rats to a concentration of 0.34 mg/m3 (23 ppb) resulted in a significant decrease in plasma cholinesterase activity. No effect on erythrocyte or brain acetylcholinesterase activity was seen in females or on plasma, erythrocyte, or brain cholinesterase activity in males (Newton, 1988a). Chlorpyrifos was administered by nose-only inhalation to female Fischer 344 rats at a time-weighted average concentration of 12 ppb (equivalent to 172 µg/m3) for 6 h/day on 5 days per week for 2 weeks in a study that met GLP requirements. No clinical signs of intoxication or treatment-related changes in body weights were reported, and all rats survived until the scheduled termination. Clinical chemistry showed no treatment-related changes in haematological, clinical chemical, or urinary parameters, including plasma, brain, and erythrocyte cholinesterase activity (Landry et al., 1986). In a study to determine a dose range, conducted to GLP requirements, technical-grade chlorpyrifos was administered by whole-body exposure of groups of Wistar rats for 6 h/day, 5 days per week for 2 weeks at target concentrations of 0 (air control), 10, 100, and 400 mg/m3 (actual concentrations, 0, 10, 94, and 388 mg/m3). Exposure of animals to the high dose was terminated after five exposures because they became moribund, with rapid weight loss and deterioration of their general condition, and all surviving animals were killed on day 9. No deaths was observed at other doses. There was no NOAEL in this study, as significant inhibition of plasma, erythrocyte, and brain cholinesterase activity was seen at all doses in males and females. Plasma cholinesterase activity was inhibited by 78, 86, and 88% at the three doses, respectively, in males and by 91, 93, and 95%, respectively, in females when compared with controls. Erythrocyte cholinesterase activity was inhibited by 78, 58, and 75% in males and by 60, 80, and 73% in females, respectively. Brain cholinesterase activity was inhibited by 50% at 10 mg/m3, about 70% at 94 mg/m3, and about 72% at 388 mg/m3 in males and females. Clinical signs of toxicity (tremors, salivation, and lachrymation), decreased food consumption, and decreased body weights were observed at the two higher doses. Increased adrenal weights were seen at 94 and 388 mg/m3, and effects in the forestomach including thickening and congestion were observed at 388 mg/m3 (Kenny et al., 1988). Fischer 344 rats were exposed to time-weighted average concentrations (nominal) of 0, 1, or 5 ppb (0, 0.014, and 0.072 mg/m3) of chlorpyrifos vapour for 6 h/day, 5 days per week for 2 weeks in a study conducted according to GLP. The mean daily time-weighted average concentrations found by analysis were 0.7 and 5 ppb, respectively. Although two control groups were used, their cholinesterase activity did not differ statistically significantly, and the activity in test groups was compared with that of the combined control group. Statistically significant decreases in plasma, erythrocyte, and brain cholinesterase activity were observed at 5 ppb, but as the reductions in brain and erythrocyte acetylcholinesterase activity did not reach 20% when compared with the control group, the effect was considered not to be of toxicological significance. Plasma cholinesterase activity was inhibited by > 20% in females at 5 ppb. A statistically significant decrease in plasma cholinesterase activity in females at 0.7 ppb was considered to be incidental to treatment. There were no treatment-related deaths, clinical signs of toxicity, or changes in body weight during the study. No treatment-related effects were seen on body or organ weights, and no lesions were found at gross examination. No significant adverse treatment-related effects were observed at 5 ppb (approximately 0.07 mg/m3) (Landry et al., 1985; Streeter et al., 1987). Groups of 10 CDF Fischer 344 rats of each sex were fed diets containing chlorpyrifos (purity, 95.7%) at concentrations that provided doses of 0, 0.1, 1, 5, or 15 mg/kg bw per day for 13 weeks in a study that conformed to GLP. The treatment-related effects seen at the highest dose consisted of decreased body weight and body-weight gain in males, increased fatty vacuolation of the adrenal zone fasciculata in males, and changes in haematological and clinical chemical parameters consisting of decreased erythrocyte counts in both sexes; increased platelet counts, reduced serum total protein, albumin, and globulin concentrations, and decreased alanine aminotransferase and alkaline phosphatase activity in males; and decreased serum glucose concentration and increased urinary specific gravity in females. Plasma and erythrocyte cholinesterase activities were depressed at doses > 1 mg/kg bw per day, and the activity of brain acetylcholinesterase was depressed at 5 and 15 mg/kg bw per day. The NOAEL was 0.1 mg/kg bw per day on the basis of depression of erythrocyte and brain acetylcholinesterase activity (Szabo et al., 1988). Groups of 10 rats of each sex were given diets containing chlorpyrifos at 0, 10, 30, 300, or 1000 ppm, but treatment at the highest concentration was discontinued after 4 weeks because of severe clinical signs. Plasma cholinesterase activity was reduced in a dose-related manner and was significantly lower than that of controls at all doses. Erythrocyte cholinesterase activity was similarly significantly reduced at all doses, but no dose-dependency was demonstrated. Brain cholinesterase activity showed a clear dose-related depression in animals of each sex at doses > 30 ppm. At 30 ppm, the activity was inhibited by 22% in males and 24% in females 41 days after administration. No other significant effects were found on haematological or clinical chemical parameters, but few were examined. No histopathological lesions were found that could be linked to treatment. The NOAEL for inhibition of brain cholinesterase activity was 0.5 mg/kg bw per day, and the LOAEL was 1.5 mg/kg bw per day. There was no NOAEL for inhibition of erythrocyte or plasma cholinesterase activity, as effects were seen at concentrations > 10 ppm (0.5 mg/kg bw per day) (JMPR 1972; modified by reference to the original reports of Beatty & McCollister, 1964; Beatty & McCollister, 1971). Five groups of 10 rats (strain not specified) of each sex received chlorpyrifos (purity not specified) in the diet at concentrations that provided doses of 0.03-10 mg/kg bw per day. The doses of 0 and 0.3 mg/kg bw per day were fed for 90 days; the doses of 1, 3, and 10 mg/kg bw per day were fed for 28 days, when these animals were fed control diets for 3 weeks and then diets containing 0, 0.03, or 0.1 mg/kg bw per day. Behaviour and growth were affected at 3 and 10 mg/kg bw per day. Cholinesterase activity was depressed at 0.3 mg/kg bw per day. During the period of removal from treated diet, the cholinesterase activity returned to pre-test levels; on resumption of feeding at 0.1 mg/kg bw per day, only a slight depression was observed in plasma and erythrocyte cholinesterase activity. In view of of the unusual design of this study, the toxicological relevance of this effect is considerede equivocal. No depression of brain or blood enzymes was seen at 0.03 mg/kg bw per day (Blackmore, 1968). Groups of 10 Crl rats of each sex were fed diets containing chlorpyrifos (purity, 97.5%) at concentrations that provided a dose of 0.3 mg/kg bw per day for 13 weeks or 1, 3, or 10 mg/kg bw per day for 4 weeks. No deaths occurred during the study. After 4 weeks, there was a clear dose-response relationship for inhibition of erythrocyte and plasma cholinesterase activity at all doses, and animals at higher doses showed clinical signs of toxicity. These animals were therefore removed from exposure and allowed to recover for 3 weeks, during which time the plasma and erythrocyte cholinesterase activities returned to control values. These groups were then fed chlorpyrifos at doses of 0, 0.03, or 0.1 mg/kg bw per day for 13 weeks. The dose of 0.03 mg/kg bw per day had no unequivocal effect on cholinesterase activity in erythrocytes, plasma, or brain in either sex, but the dose of 0.1 mg/kg bw per day significantly reduced the erythrocyte cholinesterase activity. The NOAEL was 0.03 mg/kg bw per day on the basis of inhibition of erythrocyte cholinesterase activity after 13 weeks at 0.1 mg/kg bw per day (Hazleton Laboratories, 1968). Groups of 20 Sprague-Dawley rats of each sex were given chlorpyrifos (purity not stated) in the diet at 0, 0.03, 0.15, or 0.75 mg/kg bw per day for 6 months; an interim sacrifice of five animals of each sex per dose was conducted after 3 months. A total of 16 animals across dose groups died from chronic murine pneumonia. Cholinesterase activity was inhibited at the high dose in both erythrocytes (50%) and plasma (65%), but the activity in brain was not inhibited at any dose. Treatment did not change body-weight gain, food consumption, or haematological or other clinical chemical parameters. The NOAEL was 0.15 mg/kg bw per day, on the basis of inhibition of erythrocyte acetylcholinesterase activity at 0.75 mg/kg bw per day (JMPR, 1972; modified by reference to the original report of Coulston et al., 1971). Groups of 20 Crl CD Sprague-Dawley-derived rats of each sex were given chlorpyrifos (purity, 95.5%) at dietary concentrations of 0 (control), 0.5, 10, or 200 ppm for 13 weeks in a study conducted according to GLP. The minimum and maximum achieved doses of the test material were calculated to be 0, 0.03 and 0.078, 0.6 and 1.5, and 13 and 31 mg/kg bw per day, respectively. All animals were inspected once or twice daily for signs of ill-health and other treatment-related signs and all were examined closely each week. No deaths or significant clinical signs were observed at any dose. Clinical chemistry, urinary analysis, and ophthalmoscopy revealed no findings that were considered to be related to treatment. At 200 ppm (approximately 13 mg/kg bw per day), reduced body weights, erythrocyte count, erythrocyte volume fraction, and haemoglobin and increased food consumption were observed. Statistically significant reductions (> 20% inhibition compared with controls) in plasma cholinesterase activity were observed in males at doses > 0.5 ppm (approximately 0.03 mg/kg bw per day) at 12 weeks and in females at doses > 10 ppm (approximately 0.6 mg/kg bw per day). The inhibition was dose-related in males but not in females. Erythrocyte and brain cholinesterase activities were not measured in this study. The NOAEL was 10 ppm, equal to 0.6 mg/kg bw per day, on the basis of reduced body weights and statistically significant reductions in packed cell volume, haemoglobin, and erythrocyte volume fraction at 200 ppm (Crown et al., 1985). Groups of 10 Fischer 344 rats of each sex were exposed to chlorpyrifos (purity, 100%) by nose only at concentrations of 0, 5.2, 10.3, or 20.6 ppb (0, 75, 150, or 300 µg/m3) for 6 h/day, 5 days per week for 13 weeks in a study that conformed to GLP. No treatment-related deaths and only minimal clinical signs were observed during the study. Body weights and urinary, haematological, and clinical chemical parameters were unaffected by treatment. No effects on plasma, erythrocyte, or brain cholinesterase activity were seen at any dose. Gross and histopathological examination revealed no effects associated with administration of chlorpyrifos (Corley et al., 1986). Groups of 15 Fischer 344 rats of each sex were exposed to chlorpyrifos (purity, 95%) by nose only for 6 h/day, 5 days per week, for 13 weeks at target concentrations of 0 (control), 5, 10, and 20 ppb (0, 0.07, 0.14, and 0.28 mg/m3, respectively). The study was conducted according to GLP requirements. There were no treatment-related deaths or ophthalmic, haematological, or clinical chemical changes. Plasma cholinesterase activity was inhibited by 23% in males at the high dose ( p < 0.01), but other changes in plasma cholinesterase activity were considered not to be related to treatment. Erythrocyte and brain cholinesterase activities were unaffected. Pathological and histopathological examination showed no effect of treatment. The NOAEL was 20 ppb (0.28 mg/m3), on the basis of inhibition of plasma cholinesterase activity. The absence of frank toxicity at any dose in this study makes it difficult to draw conclusions about the toxic potential of inhaled chlorpyrifos at doses > 0.28 mg/m3 (Newton, 1988b). In a study in Fischer 344 rats treated dermally, conducted according to GLP requirements, the animals received chlorpyrifos (purity, 100%) at 0, 0.1, 0.5, or 5 mg/kg bw per day in corn oil for a total of 15 days over a 21-day period. No treatment-related effects were observed at any dose, and plasma, erythrocyte, and brain cholinesterase activities were not inhibited. Gross and microscopic examination did not reveal any treatment-related changes, and body weights and organ weights were unaffected by treatment. In the 4-day range-finding component of this study, decreased plasma cholinesterase activity (45%) and erythrocyte cholinesterase activity (16%) were observed at 10 mg/kg bw per day, in the absence of clinical signs of intoxication (Calhoun & Johnson, 1988, 1989). Rabbits No skin irritation was found when a formulation containing 61.5% chlorpyrifos and 35% xylene was applied to the skin of the back and abdomen of groups of two rabbits at doses of 5, 10, 25, and 50 mg/kg bw 20, 4, 3, and 1 times, respectively. Both plasma and erythrocyte cholinesterase activities were significantly inhibited in each dose regimen. The values for plasma recovered more quickly than those for erythrocytes, which were still significantly inhibited at day 40 after the 3-, 4-, and 20-day exposure patterns (Pennington & Edwards, 1971). Three rabbits were exposed for 5 min to a formulation containing 61.5% chlorpyrifos and 34.5% xylene from an ultra-light vapour cold aerosol fog generator delivering 3.8 L/h. Two rabbits were exposed at a distance of 8 m at a height of 1.3 m and one at a height of 0.8 m. Exposure was terminated after 5 min because of ocular and pulmonary irritation. The recorded concentration of chlorpyrifos in breathing-space air was about 108 mg/L (range, 83-133 mg/L). By 24 h after treatment, the rabbits had decreased activities of plasma cholinesterase (< 33%) and erythrocyte cholinesterase (< 12%), but these values had recovered almost to the control level by 72 h after treatment (Pennington & Edwards, 1971). Chickens White Leghorn chickens were given chlorpyrifos in their drinking-water at 1 ppb, 1 ppm, or 100 ppm for up to 84 days. Only plasma cholinesterase activity was reported because of technical difficulties with blood sampling. Significant inhibition (54%) of plasma cholinesterase activity was recorded on day 84 in birds at 100 ppm (Stevenson, 1965). Dogs Technical-grade chlorpyrifos (purity, 95.8%) was administered to groups of two pure-bred beagles of each sex orally in gelatine capsule at doses of 0 (control; lactose only), 0.01, 0.03, 0.5, or 5 mg/kg bw per day for 4 weeks, although only one animal of each sex was exposed at 0 and 0.5 mg/kg bw per day. This study met GLP requirements. No deaths occurred during the study, and no clinical signs associated with treatment were observed. All animals gained weight steadily during treatment, and food consumption was similar in all groups. Rapid inhibition of plasma cholinesterase activity was observed at 0.5 and 5 mg/kg bw per day at all intervals, and the inhibition was both dose- and time-dependent. Plasma cholinesterase activity was reduced at 0.03 mg/kg bw per day, but the inhibition was considered not to be significant owing to variation between groups and the small group sizes. In animals at 5 mg/kg bw per day, erythrocyte cholinesterase activity was 47% that of controls on day 7 and was still inhibited by > 70% at the end of the study; brain cholinesterase activity was inhibited by 32% in comparison with concurrent controls at the end of the study. In animals at 0.5 mg/kg bw per day, erythrocyte cholinesterase activity was inhibited by 30% on days 27-28 only, and the effect was considered not to be toxicologically significant owing to its transient nature. Macroscopic post-mortem examination showed no changes that were considered to be of toxicological significance. The NOAEL was 0.5 mg/kg bw per day, on the basis of inhibition of erythrocyte and brain cholinesterase activity at 5 mg/kg bw per day (Harling et al., 1989a). Five dogs received 10 applications of a 0.087% commercial aerosol formulation of chlorpyrifos in dichlorophene on their backs (with the fur brushed against the grain) for 2 weeks, each application lasting 30 s. No deaths occurred, and no abnormalities were seen in clinical signs, body-weight gain, or opthalmoscopic, haematological or clinical chemical parameters. Erythrocyte cholinesterase activity was not decreased, but plasma cholinesterase activity was inhibited from the beginning of treatment, returning to normal 72 days after treatment. The inhibition of plasma cholinesterase activity was not accompanied by clinical signs (Sharp & Warner, 1968). Groups of two young adult beagles of each sex were given daily doses of chlorpyrifos orally by capsule at doses of 0, 0.03, 0.1, 0.3, or 1 mg/kg bw per day for 90 days. Plasma cholinesterase activity was inhibited at all doses. The NOAEL for inhibition of erythrocyte cholinesterase activity was 0.03 mg/kg bw per day, and that for inhibition of brain cholinesterase activity was 1 mg/kg bw per day (Blackmore, 1968). Technical-grade chlorpyrifos (purity, 95.8%) was administered orally by capsule to groups of four pure-bred beagles of each sex at doses of 0 (control), 0.01, 0.22, or 5 mg/kg bw per day for 13 weeks. There were no unscheduled deaths or unequivocal clinical signs during the study. The group mean body weights of animals at the high dose were reduced. Haematology, clinical chemistry, urinary analysis, and ophthalmoscopy revealed no findings that were considered to be related to treatment. No treatment-related pathological macroscopic or microscopic change was found. Statistically significant, dose-related reductions in plasma cholinesterase activity were seen at 0.01 mg/kg bw per day (bitches only; week 6 only; < 25%) and above (dogs and bitches; all sample intervals; < 87%). Erythrocyte cholinesterase activity was significantly inhibited in a dose-dependent manner at 0.22 mg/kg bw per day (< 46%) and at 5 mg/kg bw per day (< 85%). At 0.01 mg/kg bw per day, erythrocyte cholinesterase was inhibited by 17-18% at week 12, but the reduction was not statistically significant at any sample interval. Brain cholinesterase activity was inhibited in animals of each sex (by 46%) at 5 mg/kg bw per day, and the NOAEL for this effect was 0.22 mg/kg bw per day. The NOAEL for the study, on the basis of inhibition of erythrocyte acetylcholinesterase activity, was 0.01 mg/kg bw per day (Harling et al., 1989b). Beagles aged 10 and 11 months were given diets containing chlorpyrifos (purity, 97.2%) at concentrations that provided doses of 0, 0.01, 0.03, 0.1, 1, or 3 mg/kg bw per day for 1 year (one animal of each sex per dose), 1 year with a 3-month recovery period (two animals of each sex per dose), or 2 years (four animals of each sex per dose). Clinical signs were recorded daily, and body weights were recorded weekly for the first 6 months and every 2 weeks thereafter. Food intake was recorded weekly during the first 3 months and monthly thereafter. The clinical investigations in the groups receiving 0, 1, or 3 mg/kg bw per day included haematology (packed cell volume, haemoglobin, erythrocyte and leukocyte counts, and prothrombin time) and urinary analyses (specific gravity, occult blood, ketones, solids, pH, albumin, and sugar). The serum concentration of blood urea nitrogen and the activities of alkaline phosphatase and alanine and aspartate aminotransferases were measured in all dogs before treatment and at various intervals thereafter. Bromsulphalein retention was measured before treatment and at the 1-year sacrifice of animals at 0, 1, or 3 mg/kg bw per day. Cholinesterase activity was determined in plasma and erythrocytes in most groups before treatment and at various intervals during the study, and the activity in brain was measured at necropsy. Fasted dogs were necropsied and the brain, heart, liver, kidney, spleen, and testes were weighed. Portions of these organs and a standard set of tissues were preserved from the controls and animals at 1 (after 1 year only) and 3 mg/kg bw per day for histopathological examination. The animals fed chlorpyrifos for 2 years were given complete physical examinations before the end of the study, including routine neurological and opthalmoscopic evaluations. The data were evaluated only with Student's t test. The body weights were not significantly affected by treatment, and food consumption was comparable in groups of the same sex. Consumption of the test compound was monitored regularly and reported to approximate the nominal concentrations closely. The organ weights were not affected by treatment, except for an increase in the mean relative liver weight in males at 3 mg/kg bw per day (3.5 g/100 g vs 2.5 g/100 g in controls) in the 2-year study. Details of the recorded clinical signs were not presented, and the authors simply stated that "No clinical signs of toxicity were observed in any of the dogs in either phase of the experiment." The results of clinical pathology were presented as individual and summary data. There were no unequivocal treatment-related differences between control and treated groups in the limited haematological parameters measured nor in urinary analyses. No effect of treatment was found on blood urea nitrogen, alkaline phosphatase or alanine and aspartate aminotransferase activities or Bromsulphalein retention. The results of gross pathological examination were reported in the form of a general summary. None of the findings indicated an effect of treatment. The results of the limited histopathological examinations (control and high dose only) were reported in a summary table with no indication of the severity of lesions or abnormalities; individual data were not presented. The available data did not indicate any treatment-related effects. Cholinesterase activity was presented for individual animals and for groups (Table 3). Inhibition was evident within nine days (the first assay time) of the start of the study, and the values recorded at termination were similar to those recorded at interim assays. Plasma cholinesterase activity was decreased in a dose-related manner at both times, and erythrocyte cholinesterase activity was similarly depressed, females being slightly more severely affected than males. Brain cholinesterase activity was the least affected by treatment. The cholinesterase activity of animals that were allowed to recover, assayed after 2, 6, and 13 weeks on normal diet, showed a return to control levels after 2 weeks for plasma activity and after 13 weeks for erythrocyte activity. Table 3. Percentage inhibition of cholinesterase activity in beagles treated with chlorpyrifos in the diet in comparison with concurrent controls Dose Inhibition of cholinesterase activity (%) (mg/kg bw per day) Plasma Erythrocytes Brain Male Female Male Female One year (males and females) 3 72 70 79 82 8 1 62 62 63 61 3 0.1 39 36 18 30 0 0.03 18 17 2 33 0 0.01 1 1 16 13 0 Two years (males) 3 77 67 83 70 20 1 69 50 68 66 7 0.1 52 38 27 41 8 0.03 23 22 0 6 7 0.01 2 0 14 6 1 Average values at terminal sacrifice Inadequacies in data collection and recording were noted. Although the long-term effects of dietary intake of chlorpyrifos could not be evaluated in the absence of complete pathological and histopathological examination, the data were adequate for analysis of inhibition of cholinesterase. Plasma cholinesterase activity was inhibited in animals of each sex at 0.03 mg/kg bw per day. Erythrocyte cholinesterase activity was inhibited by > 20% at 0.1 mg/kg bw per day at both the 1- and 2-year intervals, but the inhibition did not consistently reach statistical significance when compared with the values in concurrent controls or with pre-treatment values. This was due in part to the small group sizes and the variation in this parameter between groups, including the control group, the activity differing by as much as twofold between individual animals. Under these conditions, the erythrocyte cholinesterase activity would have had to be inhibited by as much as 50% in comparison with controls before statistical significance was achieved. The NOAEL was 0.1 mg/kg bw per day on the basis of inhibition of erythrocyte cholinesterase activity at higher doses. Inhibition of brain cholinesterase activity was seen only at 3 mg/kg bw per day after 2 years (JMPR, 1972; modified by reference to the original report of McCollister et al., 1971a). A supplementary report provided some of the data that were lacking or inadequate in the original report, but the observations made in individual animal during life were limited and did not consistently include basic observations. Individual body weights, ophthalmological data, the results of pre-treatment and terminal physical examinations, and an inventory of histopathological findings were included. The conclusions of the original study were unchanged, with the additional observation that no effect of treatment was detected by ophthalmology (Kociba et al., 1985). Rhesus monkeys Groups of one or two rhesus monkeys of each sex were given technical-grade chlorpyrifos in 1% aqueous gum tragacanth by stomach tube at doses of 0, 0.08, 0.4, or 2 mg/kg bw per day for 6 months. There were no treatment-related changes in body-weight gain, food consumption, clinical findings, or haematological, clinical chemical, or histopathological parameters. Plasma cholinesterase activity was depressed at all doses at 16 weeks and at > 0.4 mg/kg bw per day at 24 weeks, and erythrocyte cholinesterase activity was depressed at 0.4 and 2 mg/kg bw per day (Table 4). The NOAEL for erythrocyte cholinesterase inhibition was 0.08 mg/kg bw per day and that for brain cholinesterase inhibition was 2 mg/kg bw per day. The usefulness of this study was limited by the small number of animals tested (JMPR 1972; modified by reference to the original paper of Coulston et al., 1971). Table 4. Mean cholinesterase activity (and percent inhibition in comparison with controls) in rhesus monkeys given chlorpyrifos by stomach tube Cholinesterase Time Dose (mg/kg bw per day) activity (weeks) 0 0.08 0.4 2 Plasma 16 6.0 4.4 (26%) 1.6 (72%) 1.8 (70%) 24 4.4 3.8 (14%) 1.7 (61%) 2.0 (55%) Erythrocytes 16 9.5 8.2 (13%) 6.5 (32%) 2.6 (72%) 24 8.6 7.6 (13%) 6.3 (27%) 5.4 (38%) (c) Long-term studies of toxicity and carcinogenicity Mice In a study of carcinogenicity conducted in accordance with test guidelines 83-2 of the US Environmental Protection Agency and OECD 451 and in compliance with GLP standards, technical-grade chlorpyrifos (stated purity, 95.9%) was mixed in maize oil and incorporated into a commercially available powdered animal diet to produce dietary concentrations of 0 (control), 5, 50, and 250 ppm, calculated to be equal to 0.7-1.1, 6.1-12, and 32-55 mg/kg bw per day in males and 0.7-1.2, 6.6-12, and 34-62 mg/kg bw per day in females. The test diets were administered to groups of 64 CD-1 of each sex, 3 weeks old, for at least 79 weeks, terminal sacrifice being undertaken during weeks 80-82. The animals continued to receive the treated diets until scheduled necropsy. An additional 12 males per group were treated identically to the mice in the main groups and were used to replace animals for reasons unrelated to treatment (such as excessive aggression). All of the unused spares were discarded after 10 weeks of treatment. Each animal was weighed weekly for the first 13 weeks of the study and every 2 weeks thereafter, while food consumption was measured weekly for 13 weeks and monthly thereafter. The animals were examined daily for clinical signs of intoxication, and a careful examination, including palpation, was conducted weekly. Blood smears were prepared from all mice after 12 and 18 months for differential leukocyte counts, but only those from controls and animals at the high dose were examined. Plasma, erythrocyte, and brain cholinesterase activity was determined in five animals of each sex at each dose at 9 and 18 months. All animals were examined grossly, the examination including all external surfaces, the cranial, thoracic, abdominal, and pelvic cavities, and the carcass. A large number of organs and tissues from all animals were fixed for histopathological examination; the eyes, kidneys, livers, lungs, and thyroids/parathyroids from animals at all doses were examined, as were other tissues from control and animals at the high dose and those from animals that died during the study. There was no adverse effect on survival, and no treatment-related increase in the incidence of neoplastic lesions. Treatment-related clinical signs (ocular opacities, excess lachrymation, and cranial hair loss) and reductions in body weights and food consumption were observed at 250 ppm (approximately 32 mg/kg bw per day). The effects on cholinestrase activity are summarized in Table 5. Plasma cholinesterase activity was inhibited at all doses. Determinations of erythrocyte cholinesterase activity revealed > 20% inhibition at 42 weeks and about 30% inhibition at week 78 in animals at 250 ppm but considerable intra-group variation at other doses. In addition, the erythrocytes were washed with saline before the cholinesterase determinations at 78 weeks but not at 42 weeks. The NOAEL for inhibition of erythrocyte cholinesterase activity was 5 ppm, with a statistically significant inhibition of 29% in males at 78 weeks and 41% in females at 42 weeks in animals at 50 ppm. Significant inhibition of brain cholinesterase activity ( p < 0.001 or p < 0.01) was observed in males and females at 250 ppm at 42 and 78 weeks, the activity being reduced by 80-86% when compared with controls. At 50 ppm, decreases in activity of 43-47% were seen in males at 42 and 78 weeks and in females at 42 weeks, but this finding reached statistical significance ( p < 0.05) in females only at 42 weeks and in males only at 78 weeks. At 5 ppm, brain cholinesterase activity was reduced in males at 78 weeks (27% reduction), but this Table 5. Percent inhibition of cholinesterase activity in groups of five mice fed diets containing chlorpyrifos Cholinesterase Interval Dose (ppm) (weeks) 5 50 250 Male Female Male Female Male Female Plasma 42 4*** 45*** 95*** 97*** 98*** 99*** 78 49* 50*** 95*** 96*** 98*** 98*** Erythrocyte 42 1 15 11 41** 22 23 78 - +10a 29* 4 31* 29** Brain 42 8 1 43 46* 80** 85*** 78 27 +13a 47 9 86** 84*** * p < 0.05; ** p < 0.01; *** p < 0.001 a Increased activity in comparison with controls finding was not statistically significant and was considered to be incidental to treatment. The NOAEL for inhibition of brain cholinesterase activity was 5 ppm. Non-neoplastic effects were observed at the high dose, in the livers (slight subchronic pericholangitis, histiocytic proliferation, and centrilobular hepatocytic fatty vacuolation) of males and in the eyes of males and females. No treatment-related lesions were seen at lower doses. In males at the high dose, an increased incidence of lung nodules was seen macroscopically, with rates of 5/31 in controls, 4/26 at 5 ppm, 2/37 at 50 ppm, and 14/49 at 250 ppm. The nodules were diagnosed histologically as bronchio-alveolar adenomas or carcinomas, but the incidences were stated to be within the range in historical controls. Nevertheless, two sets of data were reported for the incidence of neoplasms in historical controls: values obtained from 24-month studies and a published report on spontaneous neoplasms in CD-1 mice in a number of testing laboratories. Those used in the report were less than ideal. The use of background incidence from 24-month studies would be expected to increase the range of spontaneous tumours over that in the 78-week protocol used in this study. In addition, the tumour incidence was reported in a published paper (Maita et al., 1988) as a percentage of the animals examined, while the historical data from the 24-month study and the tumour incidence in this study were given as percentages of the number of organs examined. Histopathological examination did not reveal any statistically significant increase in the incidence of neoplasms in treated animals when compared with controls. The NOAEL was 5 ppm (0.7 mg/kg bw per day) on the basis of inhibition of erythrocyte and brain acetylcholinesterase activity (Gur et al., 1991). Groups of 56 CD-1 mice of each sex were maintained on diets containing 0, 0.5, 5, or 15 ppm of chlorpyrifos (purity, 99.6%) for 105 weeks. They were observed daily for clinical signs, body weights were recorded monthly, and daily food consumption was calculated from a 3- or 4-day measurement made once a month. Gross pathological examination was performed on all animals that died, when the weights of the brain, testes, heart, kidneys, and liver were recorded and a range of other tissues were collected. Sections were prepared of all the these tissues for histopathology. Haematology was limited to examination of smears of blood collected from the tail vein at terminal sacrifice. The body weights and absolute and relative organ weights were not significantly affected by treatment over the course of the study, and the mean body weight of all treated female mice was greater (< 13%) than that of controls from day 84 onwards, although food consumption was comparable in all groups. The mortality rate was also generally unaffected by treatment, and treated males had slightly higher survival rates than controls: 39% in controls, 46% at 0.5 ppm, 50% at 5 ppm, and 48% at 15 ppm; the survival rates of females were lower than those of controls only at the highest dose: 46% in controls, 59% at 0.5 ppm, 46% at 5 ppm, and 38% at 15 ppm. There were no clinical signs or pathological findings that indicated an effect related to treatment. A significant increase was observed in the incidence of spindle-cell hyperplasia of the adrenal gland in animals of each sex given 0.5 ppm chlorpyrifos and in males given 5 ppm. This finding was not considered to be related to treatment as there was a high background incidence in ageing mice and no dose-response relationship. Vacuolation was found in sciatic nerve preparations from 2/56 male and 3/54 female controls, 5/54 males and 8/55 females at 0.5 ppm, 2/56 males and 2/52 females at 5 ppm, and 6/52 males and 8/53 females at 15 ppm. The incidences of alveologenic adenomas and hyperplastic nodules in the liver were significantly increased only in males at 5 ppm. These findings were considered to be incidental. This study was barely adequate for assessing the carcinogenicity and long-term toxicity of chlorpyrifos, as it had serious shortcomings in design, method, and data collection. No clinical pathology was performed, and in particular cholinesterase activity was not measured; no clinical signs were reported; individual data were not presented for several parameters, including histopathology; and intestinal parasites (nematodes) were present in all groups (JMPR, 1982; modified by reference to the original report of Warner et al., 1980). Rats In a 2-year study, groups of 25 male and 25 female Sherman rats received chlorpyrifos (purity, 97.2%) in the diet at doses of 0, 0.01, 0.03, 0.1, 1, or 3 mg/kg bw per day (dietary concentrations not provided). Supplementary animals were included at each dose to allow interim sacrifices. The design of the study is shown in Table 6. Clinical signs were recorded 'frequently', body weights were recorded twice weekly for the first 4 weeks, weekly for 2-6 months, and every 2 weeks for the remainder of the study. The food intake of the main group was recorded continuously during the first 3 months and once a month thereafter. The clinical investigations included haematology (packed cell volume, haemoglobin, and erythrocyte and leukocyte counts) and urinary analysis (solids, pH, albumin, sugar, occult blood, ketones) for five rats of each sex at 0, 1, and 3 mg/kg bw per day; the serum concentration of urea nitrogen and the activities of alkaline phosphatase and alanine aminotransferase were measured in all rats at sacrifice at 12 and 18 months, in all male survivors and in four to five females at 24 months. Cholinesterase activity was determined in plasma, erythrocytes, and brain samples, as shown in Table 6. Rats in groups A, H, I, and K were necropsied in a standard manner, and the weights of the brain, heart, liver, kidney, spleen, and testes were recorded. Portions of these organs and a standard set of tissues were preserved for histopathology, but only the slides for the controls, animals at the high dose, and animals at 1 mg/kg bw per day at the 12-month necropsy were examined. Some tissues from animals that died during the study and from those animals killed when moribund were also examined histologically. Statistical evaluation of the data was limited to Student's t test. Body weights (data presented only as graphs) were generally not significantly affected by treatment. No details of the recorded clinical signs were presented; rather, the authors stated that "No changes in appearance or demeanour or signs of toxicity were observed grossly in any of the rats. No evidence of a cholinergic response was noted at any time." The results of clinical pathology were presented as individual and summary data, and no unequivocal treatment-related differences between control and treated groups were found for the limited haematological parameters measured or for the results of urinary analyses. Treatment had no effect on serum concentrations of urea nitrogen or on alkaline phosphatase and alanine aminotransferase activity. Plasma cholinesterase activity was significantly decreased at 3 mg/kg bw per day at all assay times, the inhibition ranging from 20 to 40% in males and 55 to 74% in females; the activity was less severely inhibited at 1 mg/kg bw per day, but from 6 months onwards the inhibition was 18-38% in males and 50-69% in females when compared with controls, and these differences were statistically significant at most assay times. Erythrocyte cholinesterase activity was severely inhibited in animals of each sex at the higher doses at all assay times, with rates of 60-100% at 3 mg/kg bw per day, 13-90% at 1 mg/kg bw per day, and little effect at the other doses. Animals that had Table 6. Design of the long-term study of toxicity and carcinogenicity in groups of 56 mice fed diets containing chlorpyrifos Group No. of rats of Sacrifice interval End-point examined each sex per dose A 25 2 years Necropsy, blood and brain cholinesterase B 5 1 week Blood cholinesterase C 5 1 month Blood cholinesterase D 5 3 months Blood cholinesterase E 5 6 months Blood and brain cholinesterase F 5 9 months Blood cholinesterase G 6 12 months Blood and brain cholinesterase H 5 12 months Necropsy I 7 12 months, Necropsy, blood and brain 7-8-week recovery cholinesterase J 7 18 months Blood and brain cholinesterase K 7 18 months Necropsy been allowed to recover showed full restoration of cholinesterase activity. Brain cholinesterase activity was significantly inhibited by treatment at 3 mg/kg bw per day (30-53% inhibition) at all assay times, to a lesser degree at 1 mg/kg bw per day (3-16%), and was comparable to that of controls at other doses. Animals that were allowed to recover showed restoration of brain cholinesterase activity. This study was considered to be inadequate for assessing the carcinogenicity and long-term effects of chlorpyrifos, other than clinical chemical findings including cholinesterase inhibition, as there were serious shortcomings in data collection, and the reports of gross and histopathology were inadequate and no clinical findings were reported. The NOAEL for inhibition of plasma and erythrocyte cholinesterase activity was 0.1 mg/kg bw per day on the basis of significant inhibition at 1 mg/kg bw per day. The NOAEL for inhibition of brain acetylcholinesterase activity was 1 mg/kg bw per day on the basis of significant inhibition at 3 mg/kg bw per day (JMPR, 1972; modified by reference to the original report of McCollister et al., 1971b). A supplementary report provided some of the data that were lacking or inadequate in the original report, but the observations made in individual animal during life were limited and did not include basic observations such as abnormal behaviour or gait. Individual body weights and gross and histopathological findings were included in the supplement, but the histopathology was limited and variable, only a small number of tissues from each rat being examined consistently. The results were not presented as summary tables of incidence and severity. The conclusions of the original study remain unchanged by this supplement (McCollister et al., 1985). In a 2-year study conducted according to accepted test guidelines (OECD 451, USEPA 83-5) and GLP, groups of 60 Fischer 344 rats of each sex were fed diets containing technical-grade chlorpyrifos (purity, 96.1%) at concentrations of 0, 0 (vehicle control), 0.2, 5, or 100 ppm. The vehicle used was 0.04% w/w maize oil. The diets were prepared weekly and assayed regularly for stability and accuracy of dosing. Body weights were generally recorded weekly; food consumption was recorded weekly for 13 weeks and monthly thereafter. Clinical signs were recorded daily and palpation performed weekly. Cholinesterase activity was measured before treatment and at weeks 14, 32, 45, 50 (interim sacrifice of five animals of each sex per group for measurement of brain acetylcholinesterase activity), 78, and 104 weeks (terminal sacrifice with measurement of brain acetylcholinesterase activity in 10 animals of each sex per group), and generally on 10 rats of each sex per dose. Haematological parameters were measured in blood smears from 10 rats of each sex in the vehicle control group and at 100 ppm at months 12 and 18. Gross pathological findings were recorded for all animals at necropsy, including those that died during the study or were killed at early sacrifices. Necropsy included removal of the eyes with the optic nerve and adnexa. The weights of the brain, testes, thyroids, adrenals, kidneys, and liver were recorded, and a range of tissues was collected. All gross lesions were sectioned, and sections from all control animal and those at 100 ppm were examined. Appropriate statistical tests were used to analyse the data. The mean consumption of chlorpyrifos was estimated to be 0.012, 0.3, and 6 mg/kg bw per day for animals of each sex. Mortality rates and the incidences of clinical signs or palpable masses were not affected by treatment. A 5% reduction in the mean body weights of males at 100 ppm was significant in comparison with controls at most times during weeks 3-94, and the weights of females at this dose were significantly lower (about 4%) than those of controls at most times during weeks 2-64. A variety of non-neoplastic and neoplastic lesions were recorded, and the incidence of these lesions occasionally showed a positive trend with dose; however, the incidence was generally within the range of historical controls in the laboratory or within the range of published values from the National Toxicology Program in the USA. The incidences of neoplastic lesions did not show unequivocal or statistically significant dose-response relationships. The only observation that was considered to be related to treatment was an increased incidence of cataracts and diffuse retinal atrophy in females (Table 7). Table 7. Incidences of ocular abnormalities found microscopically in rats given diets containing chlorpyrifos Abnormality Sex Dose (ppm) 0 0 (vehicle) 0.2 5 100 Diffuse retinal atrophy Male 4/60 0/60 3/23 1/28 3/60 Female 5/60 15/59* 9/60 5/58 24/60* Cataract Male 31/60 31/60 7/23 6/28 30/60 Female 38/60 38/59 33/60 32/58 51/60* * Significantly different from controls at p < 0.01 or p < 0.001 Significant (> 20%) inhibition of plasma cholinesterase activity relative to the control values was observed at doses > 0.3 mg/kg bw per day (Table 8). While erythrocyte cholinesterase activity was inhibited in animals at 100 ppm, the variation in the data and the lack of statistical significance and/or dose-response relationships mitigated against an unequivocal finding. The NOAEL for inhibition of brain cholinesterase activity was 0.3 mg/kg bw per day (Crown et al., 1988). In a study conducted in compliance with GLP standards, groups of 60 Fischer 344 rats of each sex received diets containing chlorpyrifos (purity, 98.5%) at concentrations that provided doses of 0 (control), 0.05, 0.1, 1, or 10 mg/kg bw per day for 24 months. Ten rats of each sex at each dose were randomly designated at the start of the study for interim sacrifice at 12 months. Blood was collected for haematology, clinical chemistry, and measurement of plasma and erythrocyte cholinesterase activity at 6, 12, and 18 months as well as at the 24-month terminal sacrifice. The groups were observed daily for deaths and signs of toxicity. All rats were examined clinically at least once a week from after the sixth month. All animals were palpated for externally detectable masses before treatment, before the 12-month kill, and monthly thereafter. Body weights and feed consumption were determined weekly for the first 3 months and monthly thereafter. All rats were weighed, but feed consumption was determined for only 20 rats of each sex per group. All clinical laboratory procedures scheduled for 6 and 12 months were performed on rats designated for the 12-month interim kill, whereas tests scheduled for 18 and 24 months were performed on rats designated for terminal sacrifice. Blood samples were obtained for haematology and clinical chemistry by orbital sinus puncture under Table 8. Group mean percent inhibition of cholinesterase activity in comparison with vehicle controls in rats given diets containing chlorpyrifos Assay time Dose Cholinesterase inhibition (%) (weeks) (ppm) Plasma Erythrocytes Brain Male Female Male Female Male Female 50 0 0 7 0* 31 0 35 0.2 1 4 0 42 0 14 5 15 51 0* 39 9 10 100 93 98 13 45 57* 80* 78 0 0 3 27 0 0.2 4 3 11 0 5 28* 47* 0 0 100 93* 97* 10 0 104 0 8 9 0 0 18 4 0.2 13 0 0 0 0 0 5 36 37* 17 11 0 0 100 95* 96* 34 18 58* 61* Values the same as or higher than those of the vehicle control are recorded as 0 inhibition. * Significantly different from control at p < 0.01 or p < 0.001 light anaesthesia. The haematological determinations consisted of packed cell volume, haemoglobin concentration, erythrocyte count, total and differential leukocyte counts, and platelet count. The clinical biochemical determinations included blood urea nitrogen, alkaline phosphatase and alanine and aspartate aminotransferase activities, glucose, total protein, albumin, globulin (calculated), creatine phosphokinase, total bilirubin, cholesterol, calcium, phosphorus, sodium, potassium, and chloride. Urine samples were obtained 1-2 weeks before the scheduled sacrifices at 12 and 24 months and at 6 and 18 months from 10 rats of each sex per group. The urinary parameters measured included specific gravity and semi-quantitative estimates of bilirubin, glucose, ketones, occult blood, pH, protein, and urobilinogen. Microscopy of a pooled sample was also conducted. Plasma and erythrocyte cholinesterase activities were assayed in 10 rats of each sex per group at 6, 12, 18, and 24 months, and acetylcholinesterase activity was measured in half-brain samples obtained at the 12-month and 24-month scheduled necropsies. All animals killed at the interim and terminal sacrifices or which died or were killed when moribund were necropsied and subjected to a complete gross examination. The brain, liver, kidneys, testes, ovaries, and adrenal glands were weighed, and many organs and tissues were removed and preserved in neutral, phosphate-buffered 10% formalin for subsequent histopathological evaluation. Histological sections of the formalin-fixed tissues from all controls and those at the highest dose were prepared, stained with haematoxylin and eosin and examined microscopically. Histopathological examination of tissues from animals at the three lower doses was limited to the liver, kidneys, adrenals, and tissues with gross lesions at both sacrifices. At terminal sacrifice, the lungs, spleen, testes, pituitary, and thyroid/parathyroid were also examined microscopically. Appropriate statistical tests were applied to the data. Males at the high dose showed a consistent decrease in body-weight gain relative to controls in the absence of reduced food consumption, depression of plasma (56-87%), erythrocyte (20-40%) and brain (56-58%) cholinesterase activities, and an increase in the weight of their adrenal glands which was characterized microscopically by exacerbated fatty vacuolation of the zona fasciculata. Similar effects were observed in females at this dose, but were generally less pronounced than in males: for example, a transient decrease in body-weight gain relative to controls with no reduction in food consumption, depression of plasma (82-95%), erythrocyte (generally > 20%), and brain (57-61%) cholinesterase activities, and an increase in adrenal weight at the terminal sacrifice with no associated histopathological lesions. At 1 mg/kg bw per day, the only effects attributable to treatment were inhibition of plasma cholinesterase activity (39-71% in males and 60-86% in females) and erythrocyte cholinesterase activity (20-40% in males and < 22% in females); brain cholinesterase activity was not affected. These results are summarized in Table 9. No treatment-related effects were observed at the two lower doses. There was no increase in the incidence of any type of tumour. The NOAEL for inhibition of erythrocyte cholinesterase activity was 0.1 mg/kg bw per day, on the basis of toxicologically (> 20%) or statistically significant inhibition at 1 mg/kg bw per day, and the NOAEL for inhibition of brain cholinesterase activity was 1 mg/kg bw per day on the basis of toxicologically and statistically significant inhibition at 10 mg/kg bw per day (Young & Grandjean, 1988). Chickens Hens were fed diets containing chlorpyrifos at concentrations of 0, 25, 50, or 200 ppm (approximately 0, 2.5, 5, and 20 mg/kg bw per day) for 52 weeks. Mortality was unaffected by treatment, with rates of 13% in controls, 3% at 25 ppm, 7% at 50 ppm, and 10% at 200 ppm. Plasma cholinesterase activity was assayed in three hens at each dose 1, 3, 7, 14, 22, and 29 days after the start of treatment, monthly thereafter, and 1, 2, and 3 weeks after the end of treatment. The onset of inhibition of cholinesterase activity was rapid and dose-related, with 22% inhibition in week 1 at 25 ppm, 45% inhibition at 50 ppm, and 76% inhibition at 200 ppm. It persisted at similar levels throughout the study but returned to control levels during the Table 9. Group mean cholinesterase activities in rats given diets containing chlorpyrifos, expressed as percent of the respective control values for each sampling period Cholinesterase Sex Sampling period Dose (mg/kg bw per day) (months) 0 0.05 0.1 1 10 Plasma Male 6 100 96.5 95.1 60.9* 44.2* 12 100 94.5 97.8 28.6* 13.3* 18 100 93.4 80.1 36.6* 22.5* 24 100 92.4 85.5 40.0* 19.7* Female 6 100 97.9 91.0 34.6* 16.9* 12 100 104 87.0* 13.8* 4.8* 18 100 99.1 85.8* 30.4* 12.4* 24 100 103 94.4 39.7* 17.9* Erythrocytes Male 6 100 107 88.8 76.1* 75.6* 12 100 107 93.2 67.4 63.1 18 100 109 95.3 66.2* 71.0* 24 100 105 108 86.5 73.5* Female 6 100 93.3 106 104 87.5 12 100 88.2 109 82.2 59.5* 18 100 114 99.7 78.0 81.9 24 100 107 87.2 83.6 79.9 Brain Male 12 100 93.8* 93.1* 91.0* 42.1* 24 100 102 100 103 44.3* Female 12 100 97.9 102 95.3* 38.9* 24 100 101 100 96.2 42.9* * Statistically significantly different from control by Dunnett's or Wilcoxon's test, alpha = 0.05, one-sided recovery period. Overall feed consumption, body weight, egg production, feed efficiency, egg weight, and shell thickness were not affected by treatment. There was no NOEL for inhibition of plasma cholinesterase activity in this study, as significant inhibition was seen at the lowest dose tested (Sherman & Herrick, 1973). (d) Genotoxicity Chlorpyrifos was not genotoxic in vitro or in vivo in a range of studies (Table 10). Table 10. Results of studies for the genotoxicity of chlorpyrifos End-point Test object Concentration Purity Results GLP Reference (%) or QA In vitro Gene mutation S. cerevisiae D3 NR NR Negative Poole et al. (1977; rec locus abstract only) Gene mutation B. subtilus H17, 20 (100, 200, 500, 1000 95.83 Negative Shirasu et al. (1980) M45 2000 µg/plate in DMSO Reverse mutation S. typhimurium TA98, 10, 50, 100, 500, 1000, 95.83 Negativea Shirasu et al. (1980) TA100, TA1535, 5000 µg/plate in DMSO TA1537, TA1538 Reverse mutation S. typhimurium TA98, 1, 3.162, 10, 31.62, 95.7 Negativea,b QA Bruce & Zempel (1986) TA100, TA1535, 100 µg/plate in DMSO TA1537, TA1538 Reverse mutation S. typhimurium TA98, 0.01, 0.1, 1, 5, 10, 95 Negativea,c Jai Research Foundation TA100, TA1535, 100 µg/plate in DMSO (1996a) TA1537 Reverse mutation S. typhimurium TA98, 30, 100, 300, 3000, 96.8 Negativea GLP Loveday et al. (1987) TA100, TA1535, 10 000 µg/plate in DMSO TA1537, TA1538 Reverse mutation S. typhimurium TA98, NR NR Negative Poole et al. (1977; TA100, TA1535, abstract only) TA1537 Reverse mutation E. coli WP2 NR NR Negative Poole et al. (1977; abstract only) Table 10. (continued) End-point Test object Concentration Purity Results GLP Reference (%) or QA Relative toxicity E. coli and NR NR Negative Poole et al. (1977; B. subtilis abstract only) Reverse mutation S. typhimurium TA98, 5, 500, 5000 µg/plate 96.2 Negativea,d QA Fredrick Institute TA100, TA1535, in DMSO (1996c) TA1537, TA1538 Forward mutation Chinese hamster ovary 10, 20, 25, 30, 40, 95.7 Negativea Mendrala (1985) cells, hprt locus 50 µmol/L Forward mutation Chinese hamster ovary 5, 10, 25, 50, 75 µg/ml, 96.8 Negativea,e GLP Tu (1987) cells, hprt locus 16 h, - S9 5, 10, 20, 30, 40, 50 µg/ml, 16 h, - S9 30, 50, 100, 300, 1000 µg/ml, +S9, in DMSO Sister chromatid Human lymphocytes 0.02, 0.2, 2 or 20 µg/ml NR Negativea,f Sobti et al. (1982) exchange (Laz-007) in ethanol Chromosomal Chinese hamster ovary 0.975, 1.47, 2.93, 4.89, 96.8 Negativea,g GLP Loveday (1987) aberration cells 9.75, 14.7, 29.3, 48.9, 97.5, 147 µg/ml, 19 h, - S9 1.56, 3.12, 5.2, 10.4, 15.6, 31.2, 52, 104, 156 µg/ml, 10 h, - S9 9.75, 14.7, 29.3, 48.9, 97.5, 147, 293 µg/ml, 19 h, +S9 1, 1.5, 3, 5, 10, 15, 30, 50, 100 µg/ml, 10 h, + S9 2.95, 4.95, 9.85, 14.8, 29.6, 49.4, 98.5, 296 µg/ml, 10 h, +S9 Table 10. (continued) End-point Test object Concentration Purity Results GLP Reference (%) or QA Chromosomal Rat hepatocytes 16.7, 50, 167, 500, 1667, 98.6 Negativea,h GLP Linscombe et al. (1992) aberration 5000 µg/ml harvested after 24 h and 5, 16.7, 50, 167 µg/ml harvested after 24 and 48 h, in DMSO Sister chromatid Chinese hamster ovary 1, 10, 100 µg/ml in acetone NR Negative Muscarella et al. (1984) exchange cells Unscheduled DNA Rat hepatocytes 1, 3.16, 10, 31.6, 95.7 Negative Mendrala & Dryzaga synthesis 100 µmol/L in DMSO (1986) Reverse mutation S. typhimurium TA98, 0, 1, 3.16, 10, 31.6, 95.7- Negativea Gollapudi et al. (1995) TA100, TA1535, 100 µg/plate in DMSO 98.6 TA1537, TA1538 Forward mutation Chinese hamster ovary 0, 3.5, 7, 8.8, 10.5, 14, 95.7-98.6 Negativea,i Gollapudi et al. (1995) cells, hprt locus 17.5 µg/ml in DMSO Chromosomal Rat lymphocytes 16.7-5000 µg/ml in 95.7-95.6 Negativea,j Gollapudi et al. (1995) aberration DMSO Unscheduled Rat hepatocytes 1, 3.16, 10, 31.6, 95.7-95.6 Negative Gollapudi et al. (1995) DNA synthesis 100 µmol/L in DMSO In vivo Micronucleus Mouse (CD-ICR BR) 0, 7, 22, 70 mg/kg bw 95.7 Negative QA Gollapudi et al. (1985) formation marrow cells orally in corn oil Micronucleus Mouse (Swiss albino) 0, 15, 30, 60 mg/kg bw 96.2 Negative QA Fredrick Institute formation marrow cells orally in vegetable oil (1996d) Table 10. (continued) End-point Test object Concentration Purity Results GLP Reference (%) or QA Chromosomal Mouse (Swiss albino) 0.6, 3, 15 mg/kg bw per 96.2 Negative QA Fredrick Institute aberration marrow cells day orally in vegetable oil (1996e) Chromosomal Chick embryo (Cornell 1.11, 11.1, 111, 1110, NR Negativek Muscarella et al. (1984) aberration K strain eggs) 2220 µg/embryo Chromosomal Bovine blastocysts NR NR Negative Muscarella et al. (1984) aberration Micronucleus Mouse (CD-1 (ICR) BR) 90 mg/kg bw orally in 97.9 Negative GLP McClintock & Gollapudi formation marrow cells corn oil (1989a) QA, quality assurance; GLP, good laboratory practice; NR, not reported; S9, exogenous metabolic activation system from 9000 × g fraction of rat liver; DMSO, dimethyl sulfoxide Positive control substances were used in all assays and gave the expected results. a With and without metabolic activation b Cytotoxicity observed at 100 µg/plate in TA100, 1535, 1537, 1538 with precipitation of test material; in TA98, precipitation in the absence of toxicity c Cytotoxicity at > 10 µg/plate; dose-related increase in revertant frequency (> twofold than controls) in all strains ± S9, but increases lower than in positive controls by 2-50-fold and not statistically significant d Increase in revertants at all dose, - S9 in TA1537 only; no dose-response relationship e Cytotoxicity at 50 µg/ml in one assay - S9 f At 2 and 20 µg/ml, frequency was statistically significantly different from controls but not double the control frequency g - S9: cytotoxicity at highest doses; increase in gaps only at 52 µg/ml and 10-h exposure; no increases in other aberrations +S9: cytotoxicity at 15 µg/ml and 10-h incubation; in one 10-h assay, a significant increase in number of cells with aberrations (including gaps) at 3 and 10 µg/ml; incidence of aberrations (excluding gaps) not statistically significantly increased and not dose-dependent In repeat 10-h assay, no increase in incidence of aberrations. h Cytotoxicity at > 500 µg/ml -S9 and > 167 µg/ml +S9 in first assay; no mitotic index measurable at 50 µg/ml - S9 or 167 µg/ml ±S9 i Test material precipitated at 10.5, 14, and 17.5 µg/ml ± S9 j Test material cytotoxic at > 50 µg/ml ± S9 k Increased mortality at 1110 and 2220 µg/embryo (e) Reproductive toxicity (i) Multigeneration reproductive toxicity Rats In a three-generation study, Sprague-Dawley rats (two litters per generation) were given diets containing chlorpyrifos (purity not specified) at concentrations that provided doses of 0, 0.03, 0.1, or 0.3 mg/kg bw per day for the first generation and 0, 0.1, 0.3, or 1 mg/kg bw per day for the second and third generations. The sizes of the groups were consistent in all generations, the controls comprising 20 males and 40 females and the treated groups comprising 10 males and 20 females. Treatment was started at 58 days of age, and F0 parents were mated at 118 days of age at a 1:2 male:female ratio. Evidence of conception (vaginal plug or sperm) was considered to be day 0 of gestation. Pairing for the second litter in each generation was started approximately 10 days after the first litter was weaned at 21 days, and the treated diets were fed to the litters from weaning. When the progeny of the F1b or F2b numbered more than 10 per litter, each litter was reduced to 10 after 5 days of age. The F3b fetuses were examined for teratogenic changes. F2b parent animals were continued on the test diet throughout the breeding period, except that females were placed on normal diet during organogenesis and the test substance was administered by gavage in acetone or corn oil. Dams were killed on day 20 of gestation and the fetuses removed surgically and examined for external abnormalities. Onethird were fixed for examination of soft tissues, and two-thirds were stained with alizarin red for skeletal examination; however, only the controls and rats at 1 mg/kg bw per day were examined. Clinical observations were stated to have been performed 'frequently'. Body weights and food consumption were recorded weekly until breeding commenced. During gestation and lactation, the diets were adjusted on the basis of an average feed consumption of 25 and 40 g/day; rats were weighed before breeding to allow dietary adjustment. In the investigation of teratogenic effects, the body weights of the dams were recorded on days 0, 6, 15, and 20 of gestation, while food consumption was measured during the gestational intervals 0-6, 6-16, and 16-20 days. Various indices of reproductive performance were calculated: fertility index as pregnancies per mating; gestation index as live litters born per number of pregnancies; viability as rats alive at day 5 per rats born alive; and lactation index as rats alive at day 21 per rats alive at day 5. All animals that died and five pups of each sex per dose from the F1a, F2a, and F3a litters were examined grossly. An extensive set of tissues was prepared and fixed from these pups, but in the absence of gross abnormalities only tissues from the F3a pups were examined histopathologically. Clinical chemistry was limited to the F2 animals, and consisted of measurements of plasma and erythrocyte cholinesterase activity in 11 control and five or six treated dams at the time of removal of their litters and in five males per group. Statistical analysis was limited. No clinical signs of toxicity were seen in the parents or offspring. The parental body weights were not significantly affected by treatment, and food consumption was variable but unaffected. The fertility, gestation, and lactation indices were comparable between groups and generations. The viability index was decreased at 1 mg/kg bw per day, but this effect was considered not to be clearly related to treatment in view of the isolated nature of the finding and the study design. The mean body-weight gain of the dams that produced the F3b pups showed a dose-related increase, with gains of 114 g in controls, 120 g at 0.1 mg/kg bw per day, 123 g at 0.3 mg/kg bw per day, and 126 g at 1 mg/kg bw per day. This generation showed no treatment-related effects on fertility (per cent pregnant), the mean number of corpora lutea or implantations, viable litter size, or pup weight after surgical removal. The skeletons of the pups showed common minor variants such as incomplete ossification of sternebrae and the occurrence of extra ribs. Visceral examination of the pups showed that minor variants of the urogenital system (hydronephrosis or left, right, or bilateral hydroureter) were more common in the treated group. A single fetus with multiple abnormalities was found at 1 mg/kg bw per day, but this isolated finding was considered not to be related to treatment. The group average activity of plasma and erythrocyte cholinesterase was decreased by > 20% at 1 mg/kg bw per day in males and females of the F2 generation. The NOAEL was 0.1 mg/kg bw per day on the basis of decreased plasma and erythrocyte cholinesterase activity at 1 mg/kg bw per day and decreased erythrocyte cholinesterase activity at 0.3 mg/kg bw per day (JMPR, 1972; modified by reference to the original report of Thompson et al., 1971). To supplement the finding of an equivocal decrease in neonatal survival at 1 mg/kg bw per day, groups of 30 Sprague-Dawley rats of each sex were fed diets containing chlorpyrifos (purity, 96.6-99%) at concentrations providing doses of 0, 0.5, 0.8, or 1.2 mg/kg bw per day for 135 days and then bred to produce F1 litters. On day 21 of lactation, 30 pups of each sex were selected randomly, dosed in the same manner as the F0 animals for 120 days, and then bred to produce the F2 litters. The F2 pups were weaned on day 21 of lactation. The F0 and F1 matings were in a 1:1 ratio, with pairing for 5 days, a 7-day rest, and a pairing with a different male for a further 5 days. The litters were culled on day 4 post partum, and the litter weights and pup survival were recorded on days 1, 4, 7, 14, and 21; individual pups were weighed on day 21. The parental animals were examined daily for signs of toxicity, and body weight and food intake were recorded weekly for animals of each sex until pairing; thereafter, males were weighed after pairing until sacrifice, and females were weighed on days 1, 4, 7, 14, and 21 of lactation. The date of parturition, litter size at birth, and the numbers of live and dead pups at birth were recorded. The dietary concentrations were adjusted weekly during the pre-mating period according to the food consumption and body weights of the rats; the concentrations were then maintained at the last adjusted concentration from commencement of cohabitation until sacrifice. Appropriate statistical tests were applied. The parental animals showed no significant clinical signs in either generation. Body weight and food intake were unaffected by treatment. The mean fertility index (number of females delivering a litter expressed as the percentage of the total number of females placed with a male) was reduced at 0.5 and 0.8 mg/kg bw per day but not at the highest dose, and a similar inverted dose-response relationship was seen for length of gestation; data on individual animals were not presented. The litter sizes (10-11) were comparable at all doses, as were the survival indices (> 91% in all groups on days 1, 4, 7, 14, and 21) during lactation. The pup weights (presumably litter weight per number of pups) were also comparable. In F1 adults, the body weight of males was reduced at 1.2 mg/kg bw per day on days 160-182 after cohabitation, and those of females showed slight, sporadic increases when compared with controls. Food intake was sporadically decreased in males but not in females. No effects were seen on female body weight during lactation. The fertility index in treated groups exceeded the control values. The litter sizes were comparable in all groups, as were the pup weights and pup survival during lactation. The adequacy of this study for regulatory purposes was limited by the doses selected. As no adverse effects were found at the highest dose, it was unclear whether the study clearly demonstrated the reproductive toxicity of the test material in rats. The lack of data for individual animals and the inverted dose-response relationships for some parameters rendered the findings equivocal. In the absence of clear adverse effects on reproductive parameters at any dose, the NOAEL was 1.2 mg/kg bw per day (Dietz et al., 1983). In a study conducted in compliance with GLP standards and the requirements of the US Environmental Protection Agency 83-4, technical-grade chlorpyrifos (stated purity, 95.8%) was given in the diet to groups of male and female CrL: COBS CD (SD) BR rats at concentrations of 0, 2, 10, or 50 ppm over two generations. These concentrations corresponded to achieved intakes of 0.1-0.2, 0.5-0.9, and 2.5-4.5 mg/kg bw per day, respectively, in F0 males and 0.1-0.2, 0.6-0.9, and 2.9-4.6 mg/kg bw per day, respectively, in F0 females. The corresponding intakes in the F1 generation were 0.1-0.3, 0.7-1.6, and 3.3-8.1 mg/kg bw per day in males and 0.2-0.3, 0.8-1.6, and 4-8.1 mg/kg bw per day in females. The animals of the F0 generation (28 of each sex per dose) were approximately 7 weeks of age at the beginning of treatment and were maintained on their respective diets for at least 56 days before mating. They were mated at a 1:1 ratio for 20 days, and then the dams were allowed to rear their young to day 21 post partum. Before weaning, all F1 offspring in all litters were examined for developmental indices (surface righting reflex, startle reflex, air righting reflex, and pupil reflex). At day 21 post partum, 24 pups from each group were selected for mating; excess pups were sacrificed on day 22 post partum and examined macroscopically, and specified organs were weighed and tissues preserved for histopathological examination from one animal of each sex per litter. Shortly after the F1 pups were weaned, the F0 adults were killed and examined macroscopically, and a wide range of organs and tissues were preserved, the ovaries, pituitary, prostate, testes with epididymides, and uterus with cervix and vagina being examined histopathologically.The animals in the F1 generation that were selected for mating were reared on their respective diets for at least 56 days (i.e. until they were approximately 12 weeks of age) and were then mated 1:1 for 20 days; the dams were allowed to rear their young until day 21 post partum. Pathological examination was conducted as for the F0 generation. All animals were handled regularly and examined for signs associated with treatment. All animals that died or were killed were examined. The weight of each animal was determined at the start of each generation and subsequently at weekly intervals. All pups were weighed at birth and at 4, 8, 12, and 21 days post partum. Cholinesterase activity was not determined in the study. The doses were selected on the basis of those that decreased cholinesterase activity in a preliminary study in which chlorpyrifos was given in the diet for a single generation at doses up to 250 ppm. In this preliminary study, plasma cholinesterase activity was inhibited when compared with that in controls by 86% in adult females and by about 60% in weanling males and females at 50 ppm. At 50 ppm, erythrocyte cholinesterase activity was also inhibited in adult females (70% inhibition) and weanlings (about 50% inhibition), and brain cholinesterase was inhibited by 38% in adult females Food consumption was similar in control and treated groups and no adverse, treatment-related effects on group mean body weights were observed at any dose in either generation. Mating performance and the duration of gestation were similar in control and treated groups. Slight inter-group variation was observed in the incidence of total pre-birth loss in both the F0 and F1 generations, but these findings were considered not to be related to treatment in the absence of a consistent relationship between dose and effect. The F0 generation showed statistically significant increases in litter size and litter weight and decreased mean pup weight at a number of doses at most sample intervals (Table 11), but these values were generally only slightly different from those in controls and showed no consistent relationship with dose. The decrease in mean pup weights was probably related to the increased litter size. The data for the litters of the F1 generation were similar for control and treated groups, with the exception of a single, statistically significant decrease in litter weight at 50 ppm on day 21. This isolated finding was considered to be incidental to treatment. No consistent, dose-related effect on organ weights was reported during the study. Pathological examination revealed no macro- or microscopic effects that were related to treatment. Under the conditions of this study, no adverse treatment-related effects were observed at any dose. The slight increase in mean body weights of F0 males at the high dose was the only effect attributed to treatment. The NOAEL was 50 ppm, equal to 2.5 mg/kg bw per day (James et al., 1988). Four groups of Sprague-Dawley rats were used in a two-generation (one litter per generation) study of reproductive toxicity in which diets containing chlorpyrifos (purity, 97.8-98.5%) at doses of 0, 0.1, 1, or 5 mg/kg bw per day were administered. The study was conducted in accordance with FIFRA guideline 83-4, OECD guideline 416, and GLP principles. The F0 parental animals (30 of each sex per dose) were Table 11. Statistically significant differences in group data for litters of the F0 generation of rats given diets containing chlorpyrifos Time Litter parameter Dose (ppm) 0 2 10 50 At birth Litter size, total 12 13 14** 13 Litter size, live 12 13 14** 13 Litter weight (g) 70 72 79* 71 Pup weight (g) 6.1 5.6*** 5.7** 5.7* Day 4 Litter size, live 11 13* 14*** 12 Litter weight (g) 100 110 120* 110 Mean pup weight (g) 9.3 8.7 8.6* 9.2 Day 8 Litter size, live 11 13** 13*** 12 Mean pup weight (g) 15 14* 14* 15 Day 12 Litter size, live 11 13** 13*** 12 Litter weight (g) 240 270 280** 250 Mean pup weight (g) 22 21 21* 22 Day 21 Litter size, live 11 13* 13*** 12 Litter weight (g) 460 520 530* 480 Mean pup weight (g) 44 42 40* 42 * p < 0.05; ** p < 0.01; *** p < 0.001 6 weeks of age at the beginning of the study and were mated after 10 weeks of exposure to produce the F1 litters. Groups of 30 rats of each sex per dose were selected from the F1 weanlings and were treated for 12 weeks before breeding to produce the F2 litters. Pairing in both generations allowed for three periods of 7 days' cohabitation in a 1:1 ratio, the males being changed weekly. Care was taken to avoid sibling pairings. Females were removed from pairing when vaginal lavage showed sperm, and this was considered to be day 0 of gestation. The F1 and F2 litters were culled if appropriate to a total of eight pups on day 4. Clinical observations were performed daily on all animals; litter size at birth, the numbers of live and dead pups on days 0, 1, 4, 7, 14, and 21 post partum, and the sex and weight of each pup were recorded on days 1, 4, 7, 14, and 21 of lactation. Body weights and food consumption were recorded weekly before and after breeding. The body weights of dams were recorded on days 0, 7, 14, and 21 of gestation and on days 1, 4, 7, 14, and 21 of lactation, whereas food consumption was recorded once during the first week of lactation, twice during the second week, and every 2-3 days during the third week. Inhibition of cholinesterase activity in plasma, erythrocytes, and brain was measured in 10 F0 and F1 parental animals of each sex at necropsy after 19 and 21 weeks of exposure. Complete necropsies were conducted on all F0 and F1 adults, and included ocular examinations and collection ofmany tissues. Histopathological examinations were made of the adrenals, brain, gross lesions, and reproductive tissues (cervix, coagulating glands, epididymides, ovaries, oviducts, pituitary, prostate, seminal vesicles, testes, uterus, and vagina) of controls and animals at the high dose. The livers of 10 F1 parental males in the control and high-dose groups were also examined microscopically. Only the adrenals from animals at the intermediate and low doses were examined. Ten F1 and F2 pups of each sex per dose were autopsied grossly. Appropriate statistical tests were applied to all data. No significant effects of treatment were seen on clinical signs, food intake, or body weight. A slight but not statistically significant decrease in body weight was seen in F1 males at 5 mg/kg bw per day, and F1 females at this dose showed reduced body-weight gain during lactation. Plasma cholinesterase activity was inhibited in parental F0 and F1 rats of each sex at 1 mg/kg bw per day (40-57%) and at 5 mg/kg bw per day (< 72%). Plasma cholinesterase activity was also inhibited in animals of both generations at 0.1 mg/kg bw per day as part of a dose-related trend, although the inhibition was generally not statistically significant at this dose. Erythrocyte cholinesterase activity was strongly inhibited at 1 and 5 mg/kg bw per day, but that of brain was inhibited only at 5 mg/kg bw per day (Table 12). Gross observation of F0 and F1 parents revealed no significant alterations, and the only significant histopathological change in the parental animals was vacuolation consistent with fatty changes in the adrenal zone fasciculata. Treatment had no effect on fertility, length of gestation, survival dring gestation, time to mating, sex ratio, or litter size in either generation. F1 pups at 1 mg/kg bw per day showed slightly decreased body-weight gain during lactation and statistically significantly decreased body-weight gain and survival at 5 mg/kg bw per day. No effect of treatment was seen during gross or daily observation of the F1 weanlings. F2 pups did not show dose-related decreases in body-weight gain during lactation, but the survival of pups at 0 and 5 mg/kg bw per day was decreased, with total loss of three and five litters, respectively; this effect was stated to be due to maternal neglect, since the stomachs of pups in these litters contained no milk. No effect of treatment was seen during gross or daily observation of the F2 weanlings. The NOAEL for inhibition of erythrocyte cholinesterase in adults was 0.1 mg/kg bw per day and that for inhibition of brain cholinesterase activity and materal toxicity was 1 mg/kg bw per day. The NOAEL for developmental effects was 1 mg/kg bw per day, and the NOAEL for effects on fertility and reproductive effects was 5 mg/kg bw per day (Breslin et al., 1991). Table 12. Mean cholinesterase activity in F0 and F1 parent rats at necropsy (percentage inhibition compared with controls) Generation Dose (mg/kg Cholinesterase activity (% inhibition) bw per day) Plasma (IU/ml) Erythrocytes (IU/ml) Brain (IU/g) Male Female Male Female Male Female F0 0 0.54 2.0 1.1 1.0 9.3 9.0 0.1 0.46 (15%) 1.6 (20%) 1.0 (5%) 1.0 9.2 (1%) 9.1 1 0.30a (44%) 0.8a (59%) 0.3b (92%) 0.36b (65%) 8.8 (6%) 8.7 (3%) 5 0.21a (61%) 0.6a (67%) 0.3b (92%) 0.31b (70%) 4.9b (48%) 4.6a (49%) F1 0 0.53 1.8 1.1 0.96 9.7 9.4 0.1 0.43 (19%) 1.6 (15%) 0.98a (13%) 0.97 9.7 9.2 (2.5%) 1 0.30a (43%) 0.93a (52%) 0.37a (67%) 0.32b (67%) 9.4 (3%) 9.0 (4.5%) 5 0.19a (64%) 0.52a (72%) 0.33a (70%) 0.24b (75%) 4.6a (52%) 4.0b (58%) a Statistically significantly different from control mean by Wilcoxon's test b Statistically significantly different from control mean by Dunnett's test (ii) Developmental toxicity Mice In the range-finding portion of a study of developmental toxicity, pregnant mice were given chlorpyrifos at doses of up to 60 mg/kg bw per day by gavage. No toxic effects were seen in the dams or offspring at 3, 10, and 20 mg/kg bw per day. In the main study, groups of 40-47 pregnant CF-1 mice were initially given chlorpyrifos (purity, 96.8%) at 0, 1, 10, or 25 mg/kg bw per day by gavage in cottonseed oil on days 6-15 of gestation. The day on which a vaginal plug was observed was considered to be day 0 of gestation. Owing to severe maternal toxicity at 25 mg/kg bw per day, additional groups of 35-41 mice were given 0, 0.1, 1, or 10 mg/kg bw per day by gavage on days 6-15 of gestation. The mice were observed for signs of toxicity and were weighed daily on day 6-16 of gestation and then sacrificed on day 18. At the time of surgical removal of their fetuses, the weights of the liver and gravid uterus with ovaries were recorded. The uteri of non-pregnant mice were stained with sodium sulfide to ensure that no implantations had occurred. The fetuses were weighed, measured (crown-rump length), sexed, and examined externally. One-third of the fetuses were dissected to examine the soft tissues. All of the heads were preserved in Bouin's solution and subsequently sectioned. All fetuses were stained with Alizarin red for examination of the skeletons. Additional mice were added to the study to assess clinical changes. Cholinesterase (erythrocyte and plasma) activity was measured in groups of 4-10 mice given 1, 10, or 25 mg/kg bw per day on day 6, days 6-10, or days 6-15 of gestation, and these measurements were also made in a group treated with 0, 0.1, 1, or 10 mg/kg bw per day. Blood was collected for assay of cholinesterase 5 h after dosing on days 6, 10, and 15 of gestation, respectively. A homogenate of fetal tissue was prepared from litters of mice killed on day 15 of gestation. Data were not provided on the body weights or food and water consumption of the animals in these groups. Appropriate statistical analysis was applied to the experimental data: fetal body weights and body measurements, maternal body weights, weights of maternal livers and uteri, and food and water consumption were analysed by one-way analysis of variance and Dunnett's test. The incidences of fetal resorptions and alterations were analysed by a modified Wilcoxon test. The Fisher exact test was applied to other data. In the dose-ranging study, severe cholinergic signs and consequent reproductive failure were found at doses > 30 mg/kg bw per day. In the first main study, dose-related increases in the incidence of clinical signs including mortality, excessive salivation, tremors, urine-soaked coat, ataxia and lethargy were found at 10 and 25 mg/kg bw per day. A single death and isolated clinical signs at 1 mg/kg bw per day were considered not to be related to treatment. Body-weight gain was reduced at 25 mg/kg bw per day, but no effects were observed on litter size, resorption incidence, or sex ratio. Fetal body weight and crown-rump length were significantly reduced at 25 mg/kg bw per day. Plasma cholinesterase activity was depressed in a dose-related manner, while the activities in erythrocytes and fetal tissue homogenate were inhibited at 10 and 25 mg/kg bw per day (Table 13). Exencephaly was reported in all groups, but with no dose-response relationship, and a statistically significant incidence of delayed ossification was seen at 25 mg/kg bw per day (Table 14). The second main study showed no cholinergic signs in dams at any dose up to 10 mg/kg bw per day. Body weight, food intake, maternal liver weights, gravid uterine weight, and adjusted body weights were also unaffected. Cholinesterase activity was rapidly and significantly inhibited in plasma and erythrocytes at 1 and 10 mg/kg bw per day but was depressed in fetal tissue homogenates only at the high dose. No effects were observed on materna1 parameters or on litter size, the incidence of resorptions, the incidence of dead fetuses, the sex ratio, fetal body weight, or crown-rump length (Table 15). Significant fetal alterations are shown in Table 16. Neither study provided evidence of teratogenic activity at any dose, but fetal toxicity was seen at 25 mg/kg bw per day. The NOAEL for developmental toxicity was therefore 10 mg/kg bw per day. The NOAEL for inhibition of erythrocyte cholinesterase activity was 0.1 mg/kg bw per day, and the NOAEL for maternal toxicity was 1 mg/kg bw per day (JMPR, 1982; modified by reference to the original reports of Deacon et al., 1979, 1980). Rats In a range-finding study of developmental toxicity, which was given a statement of quality assurance, Fischer 344 rats were fed chlorpyrifos (purity, 96.6%) by gavage at doses of 0, 3, 10, 15, or 30 mg/kg bw per day during organogenesis. Slight maternal toxicity was observed at 15 mg/kg bw per day, and severe toxicity was observed at 30 mg/kg bw per day, with typical cholinergic signs of excessive salivation, lachrymation, urination, defaecation, and body tremors and general observations of unkempt appearance, decreased body size, and matting of perineal and facial hair. Deaths were also seen at 30 mg/kg bw per day, and animals in this group had decreased body-weight gain, decreased food consumption, enlarged adrenal glands, decreased liver weight, and shrunken thymus glands. Both plasma and erythrocyte cholinesterase activities were depressed at all doses. Maternal toxicity was thus observed at all doses, but fetal toxicity was seen only at 30 mg/kg bw per day (Ouelette et al., 1983a). In the main study, which also had a statement of quality assurance, groups of 31-33 pregnant Fischer 344 rats were given chlorpyrifos (purity, 96.6%) by gavage in corn oil at doses of 0, 0.1, 3, or 15 mg/kg bw per day on days 6-15 of gestation. They were mated 1:1, and the day sperm were observed in a vaginal smear was considered to be day 0 of gestation. The rats were observed daily for signs of toxicity, were weighed daily on days 6-16 and day 21 of gestation, and Table 13. Significant findings in mice and offspring given chlorpyrifos by gavage on days 6-15 of gestation Finding Dose (mg/kg bw per day) 0 1 10 25 No. of females 51 40 44 47 No. of animals with cholinergic signs 0 2 9 32 Deaths 0 1 1 4 Total pregnanta 40 (4) 31 (1) 32 (1) 34 (1) No. with litters 36 29 30 29 Maternal body weight gain (g)b 21 ± 5 22 ± 3 22 ± 4 18 ± 4* No. of implantation sites per dam 13 ± 3 13 ± 2 13 ± 1 12 ± 3 No. of fetuses per litter 11 ± 3 12 ± 2 12 ± 2 11 ± 3 Fetal body weight (g) 1.14 ± 0.10 1.13 ± 0.07 1.15 ± 0.08 1.03 ± 0.17* Fetal crown-rump length (mm) 25.0 ± 1.2 25.2 ± 0.9 25.3 ± 0.9 24.4 ± 1.6* Plasma cholinesterase activityc 100 (10) 15 (7) 4 (8) 2 (9) Erythrocyte cholinesterase activityc 100 (10) 86 (6) 57 (9)* 43 (9)* Fetal homogenate cholinesterase activity 100 81 65 35 * Significantly different from control at p < 0.05 a Numbers in parentheses are pregnancies detected by sodium sulfide staining (not resulting in litters). b Days 6-17 of gestation c Percent of control; numbers in parentheses are numbers of animals Table 14. Incidences (no. of fetuses/no. of litters) of significant alterations in fetuses of mice given chlorpyrifos by gavage on days 6-15 of gestation Alteration Dose (mg/kg bw per day) 0 1 10 25 No. of fetuses examined 408 347 359 326 Exencephalya 1/1 5/5* 1/1 4/3 Ablephariaa 1/1 1/1 1/1 2/2 Cleft palatea 0 2/2 1/1 2/1 Small cerebral hemispherea 1/1 1/1 1/1 0 Multiple skeletal malformationsa 0 0 1/1 0 Fused ribsa 1/1 0 1/1 0 Skull: delayed ossification 5/5 9/5 10/6 34/10* Sternebrae: delayed ossification 37/17 24/11 19/11 78/22* Sternebrae: unfused 2/2 7/6* 3/3 8/7* Sternebrae: fused 24/16 9/16* 23/12 6/5* * Significantly different from control group a Considered to be a major malformations were sacrificed on day 21. At the time their fetuses were surgically removed, the body and liver weights were recorded. The number of corpora lutea and the number and position of live and resorbed fetuses were recorded at necropsy. Fetuses were sexed, examined, and measured. The uteri of non-pregnant rats were stained with sodium sulfide to determine whether implantations had occurred in the absence of litters. Separate groups of 10 rats were dosed on days 6-15 of gestation and killed, when samples of maternal blood were collected by heart puncture to measure erythrocyte and plasma cholinesterase activity. Appropriate statistical analysis of the data was reported. There were no unscheduled deaths. Clinical signs of maternal toxicity were found, including excessive salivation, urine staining in the perineal region, porphyrin deposits around the eyes, and vaginal bleeding. Tremors were observed only at 15 mg/kg bw per day, and animals at this dose also had decreased body-weight gain; no other maternal findings were reported at necropsy. Both plasma and erythrocyte cholinesterase activities were depressed at doses of 3 and 15 mg/kg bw per day (Table 17). No adverse effects were observed on reproductive parameters, and no teratogenicity was observed. Variations and malformations were found which were distributed randomly by dose, and no effect of treatment on skeletal development was seen. The NOAEL for maternal toxicity was 0.1 mg/kg bw per day, on the basis of inhibition of erythrocyte cholinesterase activity in all adult animals at higher doses. No fetal toxicity was observed (Ouelette et al., 1983b). Table 15. Significant findings in mice and offspring given chlorpyrifos by gavage on days 6-15 of gestation Finding Dose (mg/kg bw per day) 0 0.1 1 10 No. of females bred 41 35 36 35 Total no. pregnanta 30 (0) 25 (1) 23 (0) 28 (3) No. with litters 30 24 23 25 Maternal body weight gain (g)b 12 ± 6 14 ± 5 14 ± 5 14 ± 5 No. of implantation sites per dam 10 ± 3 10 ± 3 10 ± 3 9 ± 4 No. of fetuses per litter 8 ± 3 7 ± 3 7 ± 3 7 ± 3 Fetal body weight (g) 1.04 ± 1.11 1.07 ± 0.15 1.08 ± 0.18 1.09 ± 0.12* Fetal crown-rump length (mm) 24.4 ± 1.1 24.8 ± 1.6 24.9 ± 2.1 24.8 ± 1.2* Plasma cholinesterase activityc 100 (8) 86 (6) 25 (7)* 3 (6)* Erythrocyte cholinesterase activityc 100 (8) 88 (6) 75 (7)* 50 (6)* Fetal homogenate cholinesterase activityc 100 84 95 77 * Significantly different from control at p < 0.05 a Numbers in parentheses are pregnancies detected by sodium sulfide staining. b Days 6-17 of gestation c Percent of control; numbers in parentheses are numbers of animals. Table 16. Incidences (no. of fetuses/no. of litters) of significant alterations in fetuses of mice given chlorpyrifos by gavage on days 6-15 of gestation Alteration Dose (mg/kg bw per day) 0 0.1 1 10 Exencephalya 1/1 1/1 0 1/1 Ablephariaa 1/1 0 0 0 Cleft palatea 0 0 1/1 0 Omphalocelea 0 0 0 1/1 Skull: delayed ossification 18/10 11/6 3/2 3/3 Sternebrae: delayed ossification 50/20 25/13 25/13 17/11 Sternebrae: unfused 3/3 6/5 0 1/1 Sternebrae: fused 13/7 9/7 3/3 12/8 a Considered to be major malformations Female CD Sprague-Dawley rats were mated 1:1 with stock males from the same source and strain. Technical-grade chlorpyrifos (stated purity, 96.1%) was formulated daily as solutions in maize oil and given to groups of 32 mated females in a volume of 5 ml/kg bw by oral gavage at doses of 0.5, 2.5, or 15 mg/kg bw per day on days 6-15 post coitum. A further group received maize oil only. This study was conducted in accordance with US Food and Drug Administration and Environmental Protection Agency guidelines for GLP and testing guidelines of the OECD (No. 414) and US Environmental Protection Agency (section 83-3). All females were examined daily for signs associated with treatment. Animals were weighed on days 0, 3, 6-15, 17, and 20 of gestation, and food consumption was measured twice weekly. Ten mated females were bled on day 15 for measurement of plasma cholinesterase activity, blood samples being obtained from the retro-orbital sinus under ether anaesthesia. The same procedure was repeated for an additional 10 animals per dose immediately before necropsy on day 20. The animals used for cholinesterase determinations on day 15 were discarded without necropsy after being bled. All remaining animals were killed on day 20, and the following were recorded for each animal: any macroscopic abnormality of the reproductive tract; number of corpora lutea in each ovary; weight of gravid uterus; distribution of live and dead fetuses and distribution of resorption sites in each uterine horn; individual placental weights; individual fetal weight, length, and sex; and external anomalies of individual fetuses. The thoracic and abdominal contents of approximately one-half of each litter were dissected and examined, and these fetuses were used for skeletal staining and evaluation; the remaining fetuses were used for preparation of free-hand sections and examination of visceral organs. Table 17. Adverse effects in rats given chlorpyrifos by gavage Finding Dose (mg/kg bw per day) 0 0.1 3 15 No. of females 31 32 33 31 No. of animals with cholinergic signs 0 0 0 12 Total no. pregnanta 30 (0) 28 (0) 26 (0)b 29 (1) No. of litters 29 26 24 26 Maternal body weight gain (g)c 65 ± 11 64 ± 14 71 ± 12 58 ± 11 No. of implantation sites per dam 11 ± 2 10 ± 2 10 ± 2 10 ± 3 No. of fetuses per litter 10 ± 3 9 ± 2 10 ± 3 9 ± 3 Mean fetal body weight (g) 4.28 4.37 4.52* 4.46* Mean fetal crown-rump length (mm) 43.91 43.93 43.96 43.64 Plasma cholinesterase activity (IU/ml)d 46.0 42.8 (93%) 4.9 (11%) 1.5 (3%) Erythrocyte cholinesterase activity (IU/ml)d 12.1 11.9 (98%) 3.1 (26%) 2.5 (21%) * Significantly different from control a Numbers in parentheses are pregnancies detected by sodium sulfide staining. b Two dams removed during dosing for reasons not related to treatment c Days 6-20 d Mean of 6-8 dams on day 15, IU/ml (percentage of control) No treatment-related deaths occurred during the study, and clinical signs of intoxication were confined to tremors late in the study in three animals at 15 mg/kg bw per day. Slight but statistically significant decreases in mean food consumption (8% reduction during day 79 only) and body-weight gain were seen at 15 mg/kg bw per day. Statistically significant, dose-related decreases in plasma cholinesterase activity were observed at all doses; erythrocyte and brain cholinesterase activities were not determined. The effects on litters are summarized in Table 18. The total live litter sizes were unaffected by treatment, but at 15 mg/kg bw per day the mean number of live male fetuses was statistically significantly decreased and post-implantation loss was slightly elevated. Statistically significant increases in pre-implantation loss were seen at all doses, but there was no consistent dose-response relationship and the incidence was within historical control values for this laboratory; the finding was considered not to be related to treatment. At 15 mg/kg bw per day, slight but statistically significant increases were seen in mean fetal weight and mean crown-rump length, but as the increase in fetal weight was not accompanied by a decrease in litter size, this finding may have been due to advanced fetal development and was considered to be of no toxicological significance. Similarly, the statistically significant decrease in the incidence of small fetuses observed in all test groups was considered not to be toxicologically significant. A number of minor structural variations were reported, including a single incidence of anophthalmia at 0.5 mg/kg bw per day and instances of microphthalmia at 0.5 and 2.5 mg/kg bw per day, but the incidence of such findings was low and/or were not related to dose. These findings were therefore considered to be incidental to treatment. Skeletal examination showed no adverse treatment-related findings. The NOAEL for frank maternal toxicity was 2.5 mg/kg bw per day, on the basis of reduced body weight, tremors, and transient reductions in food consumption at 15 mg/kg bw per day. The NOAEL for fetal toxicity was 2.5 mg/kg bw per day, on the basis of a slight increase in post-implantation loss at 15 mg/kg bw per day, probably associated with maternal toxicity (Rubin et al., 1987a). Rabbits Groups of 14 female New Zealand white rabbits were mated 1:1 with stock males from the same source and strain and given technical-grade chlorpyrifos (stated purity, 96.1%), formulated daily as solutions in maize oil, by oral gavage at doses of 1, 9, 81, or 140 mg/kg bw per day (in a volume of 2 ml/kg bw) on days 7-19 post coitum. The doses for animals at 81 and 140 mg/kg bw per day were prepared directly, and those for the animals at 1 and 9 mg/kg bw per day were prepared by diluting the dose of 81 mg/kg bw per day with maize oil. A further group received maize oil only. This study was conducted in accordance with US Food and Drug Administration and Environmental Protection Agency guidelines for GLP and testing guidelines of the OECD (No. 414) and US Environmental Protection Agency (section 83-3). The doses were selected after a preliminary range-finding study. On day 15, three does at 81 mg/kg bw per day were accidentally given a dose of 140 mg/kg bw, and the group was expanded to 21 animals so that the overdosed animals could be excluded from the study if that was found to be warranted. Two of the three animals were pregnant, but no apparent difference was found from the females treated at 81 mg/kg bw per day, and the maternal and fetal data for these animals were included in the report. All animals were examined daily for clinical signs associated with treatment. Any animals found dead or killed in extremis were autopsied to determine the cause of death. Animals were weighed on days 0, 3, 7-19, 22, 25, and 29 of gestation, and food consumption was determined twice weekly. Plasma cholinesterase activity was measured before mating and again after at least 10 days of dosing. The does were killed on day 29 and the following were recorded: any macroscopic abnormality of the reproductive tract; the number of corpora lutea in each ovary; the weight of gravid uterus; the distribution of live and dead fetuses and of resorption sites in each uterine horn; individual placental weights; individual fetal weight and crown-rump length; sex; and external anomalies of individual fetuses. The thoracic and abdominal contents were dissected and examined, and the skull of each fetus was sectioned transversely through the frontal-parietal suture, and the brain examined. Each fetus was used for skeletal staining and evaluation. Table 18. Effects on fetuses of rats given chlorpyrifos by gavage Effect Dose (mg/kg bw per day) 0 0.5 2.5 15 Pre-implantation loss (%) 8.6 12.2*** 11.5*** 11.4*** Post-implantation loss (%) 7.0 6.0 5.8 9.0** Mean fetal weight (g) 3.33 3.40 3.44 3.51** Mean crown-rump length (mm) 36.0 36.2 36.3 36.7** Live fetuses: Male 7.6 8.0 6.8 6.0* Female 6.8 7.0 7.3 7.5 Total 14.4 15.0 14.1 13.4 Small fetuses (no. observed) 48 (317) 25 (315)** 20 (311)*** 13 (282)*** Ablepheron (no. observed) 0 (317) 0 (315) 1 (311) 0 (282) Major cranio-facial malformations: 0 (317) 1 (315) 0 (311) 0 (282) proboscis, anophthalmia (no. observed) Hydronephrosis (no. observed) 0 (161) 0 (158) 1 (158) 0 (146) Hydroureter 0 (161) 0 (158) 1 (158) 0 (146) Anophthalmia: free-hand section 0 (156) 1 (157)a 0 (153) 0 (136) Microthalmia: free-hand section 0 (156) 1 (157)a 0 (153) 0 (136) *, p < 0.05; **, p < 0.01; ***, p < 0.001 a Same fetus No treatment-related deaths were observed during the study, and no treatment-related clinical signs were reported. Maternal group mean food consumption was unaffected by treatment, and group mean body weights and body-weight gains were not affected by treatment at doses up to 81 mg/kg bw per day. At 140 mg/kg bw per day, body-weight gain was inhibited during most of the treatment period, although it was greater than normal after cessation of treatment, and by day 29 the animals in this group had attained body weights similar to those in the control group. Statistically significant, dose-related decreases in plasma cholinesterase activity were seen at all doses after 10 days of treatment, with inhibition of 56% at 1 mg/kg bw per day, 68% at 9 mg/kg bw per day, 70% at 81 mg/kg bw per day, and 72% at 140 mg/kg bw per day compared with controls. Erythrocyte and brain cholinesterase activities were not determined. Statistically significant decreases in pre-implantation loss were observed at most doses, but this finding was considered not to be toxicologically significant. Statistically significant increases in post-implantation loss were observed at 9 and 140 mg/kg bw per day but not at 81 mg/kg bw per day. In the absence of a consistent dose-response relationship, the toxicological significance of this finding was unclear; no historical control data were supplied for this effect from the laboratory. A slight but statistically significant decrease in mean fetal crown-rump length and a decrease in mean fetal weight were observed in animals at 140 mg/kg bw per day, and these effects were considered to be related to treatment. A number of fetal anomalies were observed but were considered not to be treatment-related as the incidence was generally low and unrelated to dose. An increased incidence of fetuses with fifth sternebra and/or unossified xiphisternum seen at 140 mg/kg bw per day was considered to be related to slightly delayed development at this dose, as indicated by reduced fetal weight and length in this group. Given the variation in the incidence of other skeletal observations in treated groups, however, this finding may have been due to chance. The NOAEL for frank maternal toxicity was 81 mg/kg bw per day, on the bais of decreased body-weight gain at the higher dose. The NOAEL for fetal toxicity was 81 mg/kg bw per day, on the basis of a slight decrease in mean fetal crown-rump length, decreased mean fetal weight, and an increased incidence of fetuses with fifth sternebra and/or unossified xiphisternum at the higher dose (see Table 19) (Rubin et al., 1987b). (f) Special studies: Neurotoxicity Chickens No signs of delayed ataxia or paralysis were reported in groups of three hens treated with single oral doses of chlorpyrifos at 40, 75, 100, or 150 mg/kg bw. The two higher doses resulted in deaths and other clinical signs of toxicity (Stevenson, 1966a). Similarly, groups of 10 hens given single oral doses of chlorpyrifos at up to 100 mg/kg bw did not display delayed neurotoxicity, and no neuropathological findings associated with treatment were detected (Rowe et al., 1978). In another study, single oral doses of chlorpyrifos at up to five Table 19. Effects on litters and fetuses of rabbits given chlorpyrifos by gavage Effect Dose (mg/kg bw per day) 0 1 9 81 140 Number of fetuses examined 108 117 126 142 99 Pre-implantation loss (%) 15.2 13.1 8.9*** 11.9* 11.5* Post-implantation loss (%) 6.5 8.6 13.8*** 8.7 13.9*** Mean fetal weight (g) 45.4 44.3 43.3 41.8 40.7 Mean crown-rump length (mm) 97.4 95.9 95.0 94.2 93.2* Teeth absent 0 0 1 0 0 Arthrogryposis, forelimb 0 0 2 0 0 Irregularly shaped thorax 0 0 0 0 1 Hydrocephalus 0 0 1 0 0 Enlarged aortic arch 0 0 1a 0 0 Rudimentary pulmonary artery 0 0 1a 0 0 Agenesis of interventricular cardiac septum 0 0 1a 0 0 Hydronephrosis (bilateral) 2 0 0 4 1 Hydronephrosis (unilateral) 2 2 1 2 1 Diffuse opacity of eye 0 0 0 2 0 No. of fetuses examined for skeletal defects 108 117 126 142 99 Reduced or incomplete ossification of interparietal bone 3 4 3 5 2 Unossified interparietal bone 0 1 0 0 3 Reduced or incomplete ossification of hyoid bone 2 10* 17*** 16** 8* Irregular ossification of one or more of sternebrae 1-4 1 2 7* 8* 1 Bony plaque adjacent to sternebra 10 6 2** 3* 1** Unossified fifth sternebra and/or xiphisternum 1 1 2 3 6* Multiple malformations of ribs and lumbar spine 0 0 0 0 1 *, p < 0.05; **, p < 0.01; ***, p < 0.001 a Same fetus times the oral LD50 in hens (> 150 mg/kg) resulted in significant inhibition of neuropathy target esterase and cholinesterase activity (Capodicasa et al., 1991), with signs consistent with organophosphate-induced delayed neuropathy. Extensive, aggressive antidotal treatment, both before and throughout the treatment and recovery periods, was required for the birds' survival. Technical-grade chlorpyrifos (purity, 96.8%) was tested for acute toxicity and delayed neurotoxicity in adult domestic hens in a study carried out according to GLP. In the assessment of neurotoxicity, groups of 10 birds were given corn oil, tri- ortho-cresyl phosphate (positive control; 20% in corn oil; 500 mg/kg), chlorpyrifos (110 mg/kg bw) plus atropine sulfate (intramuscular injection; 10 mg/kg bw), chlorpyrifos (110 mg/kg bw; 5.5% in corn oil) plus pyridine 2-aldoxime methane sulfonate (intramuscular injection; 50 mg/kg bw; 5% aqueous solution), or were not treated and were maintained as a source of additional birds if necessary. The hens were observed for 21 days, at which time all those given the corn oil were given a second dose, and all treated birds were given chlorpyrifos, followed by another 21-day observation period. No clinical signs of toxicity were observed in those given the vehicle. In birds given tri- ortho-cresyl phosphate, no signs of toxicity were observed immediately after dosing, but all birds developed signs of delayed locomotor ataxia as early as day 11 after treatment. The birds given chlorpyrifos showed signs of toxicity after either dose, including subdued appearance, unsteadiness, inability to stand, and weakness. Deaths were observed in both chlorpyrifos-treated groups, four birds that died after the first dose with atropine sulfate (no deaths after the second dose) and three after the first dose of chlorpyrifos with pyridine 2-aldoxime methane sulfonate (one death after the second dose). Surviving birds recovered completely within 5-8 days of treatment. Additional doses of atropine sulfate were given to several birds in both groups treated with chlorpyrifos owing to the severity of the clinical signs observed. Histopathological examination revealed a low incidence of minor neurological changes (grade II on a scale of I-V of increasing severity) in several birds given the vehicle, which was considered to show a normal background incidence of axonal degeneration. Most of the birds given tri- ortho-cresyl phosphate had grade III changes at at least one level of spinal cord and grade III or IV changes in at least one level of peripheral nerve, indicating significant treatment-related axonal degeneration. Three birds treated with chlorpyrifos showed grade II changes at one level of peripheral nerve only, and these findings were considered to be similar to the incidence and severity of changes seen in the vehicle controls. Thus, oral administration of a single dose of chlorpyrifos at 110 mg/kg bw to hens and another dose after 21 days did not induce clinical signs of delayed neurotoxicity, as confirmed by histopathological examination of nervous tissues (Roberts et al., 1987) When chlorpyrifos was given to chickens for 91 days in the diet at doses of < 10 mg/kg bw per day in a study conducted according to GLP, the only compound-related clinical signs were transient, slight gait alterations in a number of birds at the high dose. No histopathological lesions of nerve tissues characteristic of organophosphate-induced delayed neuropathy were seen. Concurrent positive controls showed both toxic signs and histopathological lesions of nerve tissues typical of those induced by a delayed neurotoxicant (Barna-Lloyd et al., 1986). To determine whether repeated doses of chlorpyrifos at the maximum tolerated dose caused cumulative inhibition of neuropathy target esterase and organophosphate-induced delayed neuropathy, chlorpyrifos (purity, 100%) was given orally to 15 white Leghorn hens, 18 months of age, for 20 days at a dose of 10 mg/kg bw per day. Corn oil was given to a control group of 18 birds. Physical observations (including posture and gait) were made daily. Three hens per group were killed on days 0 (controls only), 4, 10, 15, 20, and 48, and brain acetylcholinesterase, plasma butyryl cholinesterase, and lymphocyte and brain neuropathy target esterase were assayed. Body weights were decreased during treatment, by 25% when compared with controls on day 20, but had recovered to about 87% of that of controls by the end of the study. Some birds showed acute effects (staggering gait and diarrhoea) during week 1 of treatment. Diarrhoea was also seen in controls and may have been associated with the corn oil. No behavioural or perching defects were seen during the observation period. During days 4-20, brain cholinesterase activity was depressed by 58-70% and plasma cholinesterase activity by 49-80% when compared with control values, but after the 4-week observation period brain cholinesterase activity had recovered to 86% and plasma cholinesterase to 134% of the controls values; neuropathy target esterase activity in brain and lymphocytes throughout the study was 82-99% and 85-128% of that in controls, respectively. This finding was not statistically significant for lymphocytes, but the 18% inhibition of brain neuropathy target esterase activity on days 10 and 20 was statistically significant. There were no indications of delayed neurotoxicity after repeated administration of a dose that caused significant inhibition of brain cholinesterase activity and loss of body weight (Richardson et al., 1993a). In order to clarify the magnitude of the neuropathic risk associated with exposure to chlorpyrifos, groups of three 18-month-old white Leghorn hens were given single oral doses of chlorpyrifos (purity, 100%) at 0, 75, 150, or 300 mg/kg bw with atropine (20 mg/kg bw subcutaneously at the time of the first dose of chlorpyrifos and additional doses as needed to maintain the hens for 4 days). Four days after treament, when the hens were killed, brain acetylcholinesterase activity was depressed by 0, 58, 75, and 86% and neuropathy target esterase activity by 0, 21, 40, and 77% at the three doses, respectively. In a further study, 18 hens were given corn oil alone and 15 atropine-protected hens were given chlorpyrifos at a single oral dose of 150 mg/kg bw. Three controls were killed on day 0, and three treated and three control hens were killed on days 1, 2, 4, 8, and 16. Brain acetylcholinesterase activity was inhibited by 86, 82, 72, 43, and 29% on days 1, 2, 4, 8, and 16 after treatment, and brain neuropathy target esterase activity was inhibited by 30, 28, 38, 29, and 6%. No signs of organophosphate-induced delayed neuropathy were observed. In studies in vitro in which chlorpyrifos-oxon was added to whole brain homogenate, the rate constant for the enzyme-inhibitor complex was 15.5 per mm per min for acetylcholinesterase and 0.145 per mm per min for neuropathy target esterase. The calculated 20-min fixed-time IC50 values were 2.2 nmol/L for acetylcholinesterase and 240 nmol/L for neuropathy target esterase. The measured IC50 values based on fixed-time (20 min) preincubation were 2.2 nmol/L for acetylcholinesterase and 210 nmol/L for neuropathy target esterase. The ratio of the calculated values for neuropathy target esterase:acetylcholinesterase is 110. As a ratio of > 1 generally indicates that the dose that would be required to cause organophosphate-induced delayed neuropathy would be in excess of the LD50, acute exposure to chlorpyrifos, except at doses well in excess of the LD50, would not be anticipated to induce this condition (Richardson et al., 1993b). The potential of chlorpyrifos to cause acute delayed neurotoxicity was tested in white Leghorn hens given doses < 12 mg/kg bw per day for 21 days by oral gavage in peanut oil. Dose- and time-related increases in the incidence and severity of ataxia were observed. Plasma and whole blood cholinesterase activities were reduced at 5 and 12 mg/kg bw per day. Ataxia was observed at 2, 5, and 12 mg/kg bw per day. Necropsy did not reveal any findings associated with treatment. Histopathological examination of the brain and spinal cord revealed a low incidence of several lesions, with no relation to dose, and these findings were considered to be incidental to treatment. Since continued administration of chlorpyrifos resulted in ataxia, it would have been difficult to observe any delayed neurological effects had they occurred (Jai Research Foundation, 1996b). Rats In rats, single oral doses of chlorpyrifos at 50-100 mg/kg bw resulted in treatment-related effects and clinical signs for several days after dosing. No deaths occurred at any dose. At 10 mg/kg bw, effects were confined to isolated observations of perineal staining and/or decreased activity. No neuropathological lesions were reported (Wilmer, 1992; Mattsson et al., 1996). In a study in adult Long-Evans rats carried out in accordance with GLP, oral administration of chlorpyrifos at doses < 10 mg/kg bw per day for 4 weeks resulted in dose-related inhibition of plasma and erythrocyte cholinesterase activity at > 1 mg/kg bw per day and of brain cholinesterase activity at > 3 mg/kg bw per day. These effects were accompanied by clinical signs of intoxication at 10 mg/kg bw per day. Cognitive behavioural tests indicated some non-cognitive changes associated with impaired motor activity at 10 mg/kg bw per day, but no clear treatment-related effects on cognitive function were observed (Maurissen et al., 1996). After oral administration of chlorpyrifos to rats at single doses < 100 mg/kg bw in a study that conformed to GLP, no treatment-related effects on neuropathy target esterase activity were observed at any dose. Plasma, erythrocyte, and heart cholinesterase activity was statistically significantly lower than that in controls at doses > 5 mg/kg bw, and brain cholinesterase activity was decreased at > 50 mg/kg bw. No significant inhibition was observed at 0.5 or 1 mg/kg bw (Dittenber, 1997). The neurotoxic potential of chlorpyrifos (purity, 99.5%) was assessed in groups of 20 male Long-Evans hooded rats, approximately 70 days old with a mean body weight of 350 g. The test material was dissolved in corn oil and given to the rats by oral gavage in a volume of 1 ml/kg bw at doses of 0, 10, 30, 60, or 100 mg/kg bw. All rats were evaluated for behavioural signs 3.5 h after dosing with a functional observational battery of tests consisting of evaluations of home cage, open-field, and manipulative neurobehaviour. The measures were autonomic activity (lachrymation, salivation, miosis, defaecation), motor activity (rearing, home cage posture), convulsive activity (tremors), neuromuscular activity (gait score, landing foot splay, righting reaction), general reactivity (arousal, removal reactivity), sensorimotor activity (responses to tail pinch, click, approach), and physiological reactions (body temperature, body weight). Data on motor activity were recorded during a 1-h session in a maze shortly after testing. Tissue samples were obtained after the motor activity tests, 4.5 and 25 h after dosing. Half of the animals in each group were killed at 3.5 h, and the remaining 10 animals were examined again at 24 h and then killed. Immediately after sacrifice, the brain, retina, liver, heart, diaphragm, and quadriceps femoris muscle were collected from all animals. The brain was sectioned mid-sagitally, and the remaining half-brain was further sectioned to obtain the frontal cortex, hippocampus, striatum, hypothalamus, cerebellum, and pons/medulla. Blood was collected from the trunk. Muscarinic receptor density was determined in all brain regions and in the heart and retina. Cholinesterase activity was determined in brain, muscle, retina, plasma, erythrocytes, whole blood, and liver. Cholinesterase activity was significantly inhibited in a range of tissues, including brain, plasma, and erythrocytes, at all doses at both 4.5 and 25 h (Table 20). As the data were represented only as graphs, the percentages are approximations. Erythrocyte cholinesterase activity was slightly more sensitive than that of plasma at the doses used in this study, and plasma cholinesterase activity recovered more quickly than that of erythrocytes. The degree of inhibition of whole-blood cholinesterase activity lay between that of plasma and erythrocyte activity at both 4.5 and 25 h. Whole-brain cholinesterase activity can be considered representative of that in various regions of the brain, although a number of regions showed less inhibition than was measured in the half-brain in this study. In peripheral tissues, Table 20. Percentage inhibition of cholinesterase activity compared with controls in rats given chlorpyrifos by gavage Time (h) Dose Inhibition of cholinesterase activity (%) (mg/kg bw) Plasma Erythrocytes Blood Half-brain Retina Heart Diaphragm Quadriceps Liver 4.5 10 80 90 85 40 40 55 40 5 70 30 85 95 90 70 65 65 55 20 70 60 85 100 90 85 80 70 70 70 80 100 90 100 95 90 90 75 85 85 85 2.5 10 35 75 55 30 30 40 5 + 10 25 30 75 90 85 60 60 75 35 20 70 60 95 100 95 80 75 90 65 35 85 100 95 100 95 85 85 90 75 55 85 When inhibition was > 20% in comparison with corresponding controls, the change in cholinesterase activity was statistically significant ( p < 0.05). the inhibition was much lower in the quadriceps than in other tissues at 4.5 h, and inhibition of heart, liver, and retina cholinesterase activity was similar at 25 h. Clinical and behavioural signs were observed at doses > 30 mg/kg bw at 4.5 h. The doses selected for this study resulted in a skew of some correlations, especially for cholinesterase inhibition in blood and blood components, with behavioural and clinical findings. In order to examine the relationship between clinical and behavioural findings and cholinesterase inhibition, the authors grouped rats according to the extent of cholinesterase inhibition (instead of by dose) and indicated the incidence of cholinergic signs related to individual cholinesterase inhibition. The 10 'cardinal' cholinergic signs that were used for this analysis were temperature, gait, motor activity, smacking, foot splay, tail pinch, tremors, lachrymation, pupil response, and salivation (Table 21). None of these signs was observed in the 18 animals with < 60% inhibition of brain cholinesterase activity. The incidence of these findings increased in relation to brain cholinesterase inhibition. In animals with < 80% inhibition of whole-blood cholinesterase activity, no signs were observed, while all of the signs were seen at varying incidences in animals with 90-100% inhibition. The correlation between these end-points and inhibition of cholinesterase in other tissues or in erythrocytes or plasma was not provided. These results were interpreted by the authors as indicating a threshold effect, since no clinical signs were observed until a certain percentage inhibition of cholinesterase activity was reached. Because of the doses selected for this study, marked inhibition was observed at the lowest dose in the absence of clinical findings. The use of a single dose also limited the usefulness of the findings. No clinical signs were seen when brain cholinesterase activity was inhibited by < 60% or when whole blood cholinesterase activity was inhibited by < 80% after a single dose of chlorpyrifos. The data do not suggest that these values could be extrapolated to repeated doses (Nostrandt et al., 1997). Technical-grade chlorpyrifos (purity, 98.1%) was given to groups of 10 male and 10 female Fischer 344 rats in the diet for 13 weeks at doses of 0, 0.1, 1, 5, or 15 mg/kg bw per day. The dietary concentrations were adjusted on the basis of measured weight and food consumption, but the range of dietary concentrations used to achieve these target doses was not provided. The study was conducted in accordance with GLP requirements and to meet US Environmental Protection Agency FIFRA guideline No. 82-7. Body weights and feed consumption were measured before treatment and weekly during treatment. Ophthalmic examinations were conducted before treatment and at necropsy. Cage-side observations were made twice daily and clinical examinations weekly. A functional observational battery of tests was applied to all rats before treatment and during weeks 4, 8, and 13 of treatment. Five rats of each sex per dose were examined for gross pathological alterations. Tissues for histopathological evaluation were prepared only from rats in the control and high-dose groups. Nine transverse sections of the brain included the olfactory bulb, cerebral cortex (frontal, parietal, temporal, and occipital lobes), Table 21. Incidence (%) of cardinal cholinergic signs in rats given chlorpyrifos by gavage and grouped according to extent of inhibition of cholinesterase activity (numbers of rats in parentheses) End-point Inhibition of brain cholinesterase activity (%) Inhibition of blood cholinesterase activity (%) 90-100 (10) 80-90 (8) 70-80 (7) 60-70 (7) < 60 (18) 90-100 (25) 80-90 (14) < 80 (11) Temperature 100 100 86 57 0 92 36 0 Gait 100 100 57 71 0 84 43 0 Motor activity 90 88 86 57 0 84 36 0 Smacking 90 63 57 0 0 64 14 0 Foot splay 40 38 29 0 0 32 14 0 Tail pinch 70 38 0 0 0 40 0 0 Tremors 90 25 0 0 0 44 0 0 Lachrymation 80 13 0 0 0 32 7 0 Pupil response 60 0 0 0 0 24 0 0 Salivation 30 0 0 0 0 12 0 0 thalamus/hypothalamus, midbrain, pons, cerebellum, medulla oblongata, and nucleus gracilis/cuneatus. Other tissues examined were the trigeminal ganglion and nerve, pituitary, eyes (retina and optic nerve), spinal cord (cervical and lumbar), olfactory epithelium, and skeletal muscles (gastrocnemius and anterior tibial). A low incidence of perineal staining was seen in females at 5 and 15 mg/kg bw per day. No decrease in body weight was recorded at any dose. Gross pathological examination did not reveal any treatment-related effects. Histopathological examination revealed a small number of isolated findings that were similar in control and treated groups and which were not considered to be related to treatment. These included slight degeneration of individual nerve fibres in the trapezoid body of the medulla oblongata in four control males and three control females and in one male and two females at the high dose; slight degeneration of individual nerve fibres in the cervical spinal cord of a single male at the high dose; and myelin ellipsoids in the lumbar spinal cord of one male at the high dose and one female control. A slight reduction in motor activity was observed at 15 mg/kg bw per day during week 4 only, but the toxicological significance of this finding was unclear. Cholinesterase activity was not measured in this study. No neuropathological findings related to treatment were observed at any dose. The NOAEL for cholinergic effects was 1 mg/kg bw per day, and that for neuropathy was 15 mg/kg bw per day (Shankar et al., 1993). In a study designed to investigate the effects of age on the toxicity of chlorpyrifos, rats received corn oil or chlorpyrifos orally at doses of 80 mg/kg bw for adults and 15 mg/kg bw for young rats (at postnatal day 17). These doses were equally effective in inhibiting cholinesterase activity. In adult rats, peak behavioural changes and cholinesterase inhibition occurred in males 3.5 h after dosing, while in females the onset of functional changes occurred sooner, the time course was more protracted, and recovery was slower. In young rats, the maximal behavioural effects and cholinesterase inhibition occurred 6.5 h after dosing, with no sex-related difference. Partial to full recovery from the behavioural changes was seen at 24-72 h, whereas the cholinesterase activity recovered more slowly. The blood and brain cholinesterase activity in young rats had almost recovered by 1 week after dosing, whereas the brain cholinesterase activity in adults had not recovered by 2 weeks. Assays for binding to muscarinic receptors showed apparent down-regulation in some areas of the brain, mostly at 24 and 72 h. Rats at postnatal day 17 generally showed more receptor down-regulation than adults, whereas only adult females showed receptor changes in striatal tissue that persisted for 2 weeks. Thus, young rats showed similar behavioural changes and cholinesterase inhibition to adults, although at a fivefold lower dose; the onset of maximal effects was somewhat delayed in the young rats; cholinesterase activity tended to recover more quickly in the young rats; young rats appeared to have more extensive muscarinic receptor down-regulation; and young rats showed no sex-related difference in functional changes (Moser & Padilla, 1998). Chlorpyrifos was administered subcutaneously to neonatal rats at 1 or 5 mg/kg bw per day on postnatal days 1-4 or daily at 5 or 25 mg/kg bw per day on days 11-14, and the two groups of pups were examined at postnatal days 5 and 10 or 15 and 20, respectively. Pups treated at postnatal days 1-4 had a significant increased mortality rate (> 50%) at 5 but not at 1 mg/kg bw per day; the body weights of the affected pups were decreased by 10-15% and the brain was significantly smaller than that of controls. The survivors showed severe cell loss in the brainstem; brainstem growth was maintained by enlargement of the remaining cells. The lower dose did not compromise survival or growth. Neither dose had an adverse effect on the forebrain, despite the fact that the brainstem and forebrain have comparable cholinergic projections. Pups treated on postnatal days 11-14 showed no increase in mortality rate at 5 mg/kg bw per day but a significant increase (> 80%) at 25 mg/kg bw per day at day 15. The body weights of the affected pups were significantly decreased (> 40%), and the major target for cell loss shifted from the brainstem to the forebrain. The pups at 5 mg/kg bw per day had a small, transient loss in body weight; there was no significant effect on whole brain weight, but these pups showed loss of forebrain cells between 15 and 20 days of age, and the total amount of DNA in the forebrain declined whereas an increase was seen in the forebrains of control pups. The brainstem showed a smaller but not significant cell loss during this period. The cerebellum differed from the other regions in that it showed short-term increases in DNA after exposure to chlorpyrifos in either the early or the late postnatal period; nevertheless, the values then regressed to subnormal, in parallel with loss of cells in other regions. Although regions rich in cholinergic projections, such as the brainstem and forebrain, may be more severely affected than non-cholinergic regions such as the cerebellum, the time of maturation of each region (brainstem earliest, forebrain intermediate, cerebellum last) appears to be more important in determining vulnerability. The weights of the various regions remained within normal limits despite severe reductions in cell numbers, probably because reactive hypertrophy of the remaining cells masked the cell loss. These results suggest that chlorpyrifos induces cellular deficits in the developing brain even when growth or survival is unaffected; however, the route of administration and the doses used in this study limit the relevance of these findings to humans (Campbell et al., 1997). Chlorpyrifos was administered subcutaneously to neonatal rats at doses of 1 or 5 mg/kg bw per day on postnatal days 1-4, or at 5 mg/kg bw per day on days 11-14, and the two groups of pups were examined on postnatal days 5 and 10 or 15 and 20, respectively. Signs of interference with the adenyl cyclase signalling cascade were determined in the heart and developing brainstem and cerebellum. Pups treated on postnatal days 1-4 at 1 mg/kg bw per day showed no signs of toxicity, while there was significant mortality (> 50%) at 5 mg/kg bw per day. Pups treated on postnatal days 11-14 at 5 mg/kg bw per day showed no deaths or clinical signs. No significant changes in the weights of the brain region were found at these apparently non-toxic doses. Cholinesterase activity in the brainstem of the pups given 1 mg/kg bw per day was inhibited by 25% when measured 24 h after the last dose, but recovery was substantial (< 10% inhibition) by day 10. Cholinesterase activity in the brainstem of the pups given 5 mg/kg bw per day on days 11-14 was inhibited by > 60% when measured 24 h after the last dos, but recovery was again substantial (< 30% inhibition) by postnatal day 20. Separate examinations of the forebrain, cerebellum, and heart showed that chlorpyrifos had evoked deficits in multiple components of the adenyl cyclase cascade: the expression and activity of adenyl cyclase itself, functioning of the G-proteins that link neurotransmitter and hormone receptors to cyclase activity, and expression of neurotransmitter receptors that act through this cascade. Disruption of signalling function was not restricted to transduction of cholinergic signals but extended to adrenergic signals as well. In most cases, the adverse effects were not evident during administration of chlorpyrifos but appeared after several days. Most of the results of this study were presented in summary form only (Song et al., 1997). The acute neurobehavioural effects (as determined by observational tests of function and motor activity) of two carbamates, carbaryl and aldicarb, and five organophosphates, chlorpyrifos, diazinon, parathion, fenthion, and diisopropyl fluorophosphate, were evaluated in 10-week-old male Long-Evans rats on the day of dosing at the time of the peak effect, at 1 and 3 days, and 1 week after dosing. All doses were administered by oral gavage in corn oil in a volume of 1 ml/kg bw, except carbaryl, which was given in 2 ml/kg bw. A weight loss of about 10% was recorded with each substance only at the high dose. Generally, all of the cholinesterase inhibitors induced autonomic signs of cholinergic over-stimulation (salivation, lachrymation, and miosis), hypothermia, mild tremors, mouth-smacking (chewing motions), decreased motor activity, decreased tail-pinch response, and altered neuromuscular function (gait changes and increased foot splay). The measures found to be most sensitive on the day of dosing were body temperature, motor activity, gait, and the presence of mouth-smacking and fine tremors, although no single measure was the most sensitive for all compounds. Differences in the slopes of the dose-response curves were evident for some measures. Many effects were still present at 24 h, but the rats recovered from the effects of all compounds. Interestingly, residual effects were found at 72 h with the carbamates and with the organophosphate fenthion, but not with the other compounds; this may be due, at least in the case of the carbamates, to slow absorption from the gut. Thus, the overall clinical toxicity profile of these cholinesterase inhibitors was similar, but compound-specific differences emerged in terms of individual measures, dose-response relationships, and time course; the behavioural effects induced by these compounds may be due to both cholinergic and non-cholinergic mechanisms. Only summary data were available for evaluation (Moser, 1995). Chlorpyrifos was administered subcutaneously to neonatal rats at 2 mg/kg bw on postnatal day 1 or at 11 mg/kg bw per day on postnatal days 6-9. The developing brain regions (brainstem, forebrain, and cerebellum) were examined for signs of interference with cell development by using markers of cell division. The 1-day-old rats showed significant inhibition (about 15%) of DNA synthesis in all brain regions within 4 h of treatment. Equivalent results were obtained when a small dose (0.6 or 2 µg/g brain) was introduced directly into the brain by intracisternal injection, indicating that the effect was not secondary to systemic toxicity. Comparable inhibition (> 10%) of DNA synthesis was seen in these animals at 8 days of age, but at this point there was regional selectivity, with sparing of the cerebellum (about 1% inhibition). In another phase of this study, 6-8-day-old animals were pretreated with mecamylamine, a nicotinic receptor antagonist, and then with chlorpyrifos at 11 mg/kg bw. The pretreatment caused a decrease in DNA synthesis and also prevented any further decrement in DNA synthesis due to chlorpyrifos. Administration of chlorpyrifos at 1 day of age caused severe (> 30%) inhibition of protein synthesis throughout the brain; the effect was distinct from that on DNA synthesis, as it had diminished substantially by 8 days of age (< 10%) and did not show regional selectivity. Assays of ornithine decarboxylase activity 4 and 48 h after treatment with chlorpyrifos in 1- and 8-9-day-old rat pups showed no significant alteration of activity in any brain region in comparison with appropriate controls. The authors concluded that the effects of chlorpyrifos on DNA and protein synthesis are not secondary to generalized cell damage or suppression of cell metabolism, as evidenced by the maintenance of normal ornithine decarboxylase activity. The results of this study were presented only in summary form (Whitney et al., 1995) Pregnant Sprague-Dawley rats were given chlorpyrifos at doses of 0, 0.3, 1, or 5 mg/kg bw per day by gavage in corn oil from day 6 of gestation through day 11 of lactation, and their neurobehavioural performance was evaluated. A satellite group of mated females was dosed similarly every day on days 6-20 of gestation and used to obtain blood and brain samples for analysis of cholinesterase. This study was conducted in accordance with GLP standards. There were no deaths and only a few clinical signs (fasciculations, hyperpnoea, hyper-reactivity, and decreased body-weight gain) in the dams at the high dose. Between days 1 and 5 of lactation, three dams at 5 mg/kg bw per day had total litter loss and an additional 57 pups died or were cannibalized; these losses led to a decreased viability index. Necropsy of many of the dead pups indicated treatment-related lack of maternal care (no milk in the stomach). No similar effects of treatment were seen at the other doses. In dams, cholinesterase activity in plasma and erythrocytes was inhibited at all three doses, with > 40% inhibition at 0.3 mg/kg bw per day. Brain cholinesterase activity was inhibited at the two higher doses, but the inhibition at 1 mg/kg bw per day was not > 20% that of controls. Pups that were exposed to chlorpyrifos in utero and for a period post partum showed signs of toxicity at the high dose: the weights of male and female pups were significantly lower than those at the other doses, consistent with the decreased food consumption and lack of body-weight gain by dams at this dose. Morphometric measurements of the brain and brain sections of six pups of eachsex per dose showed a pattern of an increase at the low dose and a decrease at the high dose, which was more pronounced in male pups, paralleling the differences in brain weight between the groups. Hence, the NOAEL for maternal toxicity was 1 mg/kg bw per day. The LOAEL for inhibition of plasma and erythrocyte cholinesterase activity was 0.3 mg/kg bw per day, and the NOAEL for inhibition of brain acetylcholinesterase activity was 1 mg/kg bw per day. The NOAEL for toxic effects in the pups was 1 mg/kg bw per day on the basis of the decreased viability index, relative brain weight, and delayed sexual maturity, possibly associated with maternal toxicity and subsequent diminished maternal care at the high dose. Cognitive function (learning, memory, and habituation) in the pups were not affected by treatment (Hoberman, 1998). Adult male Long-Evans rats were trained to perform a test of memory and motor function and were then injected subcutaneously with single doses of 0, 60, 125, or 250 mg/kg bw of chlorpyrifos in peanut oil (2 ml/kg bw) and tested on 5 days/week for 7 weeks. Unconditioned behaviour was also rated for signs of cholinergic toxicity. Cholinesterase activity was measured in whole blood and in several brain regions, and muscarinic receptor density and the hypothermic effect of oxotremorine challenge were determined. Clinical signs of toxicity were seen in one rat at 250 mg/kg bw. Whole-blood cholinesterase activity was inhibited by 60-75% in a dose-dependent fashion on day 4 but had recovered to near control values by day 53 in animals given 60 mg/kg bw and by day 74 in those given 125 mg/kg bw. Brain cholinesterase activity was strongly inhibited in a dose-dependent manner and was inhibited by < 95% on day 7 in animals given 250 mg/kg bw. By day 21, partial recovery of cholinesterase activity was seen in some brain regions after the doses of 60 and 125 mg/kg bw, but no significant recovery was seen in animals given 250 mg/kg bw. Muscarinic receptor density decreased with dose and time. At 250 mg/kg bw, the receptor density in the hippocampus, frontal cortex, and striatum had fallen to 70-75% of the control values after 7 days and to 60% 21 days after dosing; the hypothalamic receptors were less severely affected. In comparison with controls, the hypothermia induced by oxotremorine challenge was significantly less pronounced in each group receiving chlorpyrifos on days 8 and 32, but was comparable to that of controls by day 52. Unconditioned behaviour was relatively unaffected by treatment, with fine tremor in the head and limbs, the only sign of cholinergic overstimulation, peaking on day 9 and no longer observed by 14 days after dosing. Functional deficits in working memory and motor function appeared within 2 days of injection of a dose of chlorpyrifos that caused severe cholinergic effects, but the animals recovered within 3 weeks. The definition of working memory in this paper was non-specific, and the short-term memory of the animals was unaffected by treatment (Bushnell et al., 1993). The authors contrasted these findings to those seen in rats dosed with the organophosphate diisopropyl fluorophosphate (Bushnell et al., 1991). These rats showed progressive, persistent impairment of cognitive and motor function over a 3-week period of daily exposure, despite neurochemical and pharmacological evidence of tolerance to diisopropyl fluorophosphate-induced inhibition of cholinesterase. This difference suggests that the diisopropyl fluorophosphate-induced behavioural changes cannot be attributed entirely to its effects on cholinesterase activity and changes in muscarinic receptor binding. Pregnant Sprague-Dawley rats were injected subcutaneously with either peanut oil or chlorpyrifos (purity, 98%) at 200 mg/kg bw as a single dose on day 12 of gestation and then killed on day 16 or 20 of gestation or on postnatal day 3 for measurement of maternal and developmental indicators of toxicity. While most treated dams showed no overt cholinergic signs, 4/28 showed moderate-to-severe signs 2-3 days after treatment, and these rats were omitted from further studies. Maternal body weight was decreased by 15% 3 days after treatment, after which the body-weight increase was comparable to that of controls. Chlorpyrifos did not significantly affect fetal body weights or brain weights when assayed on day 16 or 20 of gestation. Inhibition of acetylcholinesterase activity (by 82-88%) was seen in maternal brain at all three times after exposures. At days 16 and 20 of gestation, fetal brain acetylcholinesterase activity was inhibited by 42-44%. While some degree of recovery in acetylcholinesterase activity was seen in pup brain by postnatal day 3, it was still inhibited (by 30%) in treated pups cross-fostered by control dams. Maternal brain muscarinic receptor binding at day 20 of gestation and postnatal day 3 was more extensively reduced (30-32%) than in the developing brain (16 and 11%, respectively). The response to a simple postnatal reflex test of righting was transiently altered (present at on day 1 but not on day 3) by exposure to chlorpyrifos. The results suggested that acute exposure of dams during gestation induced more extensive neurotoxicological effects in the dams than in their developing fetuses, but the dose and the route of administration make the relevance of this finding unclear (Chanda et al., 1995). The maximum tolerated dose of chlorpyrifos in Sprague-Dawley rats injected subcutaneously in peanut oil was 45 mg/kg bw in 7-day-old pups and 280 mg/kg bw in adults (80-100 days old) (Pope et al., 1991). At this dose, the neonates showed an initial transient decrease in body weight when compared with controls. Adult rats also had an initial decrease in body weight, but this had recovered by the end of the 7-day observation period. Brain cholinesterase activity increased by about 42% in neonates on days 7-14, whereas it remained constant in adults. Neonatal brain cholinesterase activity was inhibited by 80% on day 1 and by 45% on day 7, whereas 90% inhibition was seen in adults on day 4, with little or no recovery. In plasma, inhibition of cholinesterase activity was rapid and marked (95%) in pups on day 1 but had recovered to 50% of control by day 7. In adults, such inhibition persisted through day 7, with no recovery of activity. In a study to determine whether a measure of inhibitory potency in vivo, the ED50, could be correlated with a measure of acute toxicity, the maximum tolerated dose, in neonatal and adult rats, chlorpyrifos was given by subcutaneous injection in peanut oil. Cholinesterase inhibition was measured at the time of peak inhibition (24 h in neonates and 4 days in adults). Depression of cholinesterase activity versus log dose gave a straight line, and the slope of the curve of inhibition of plasma cholinesterase was slightly steeper than that of brain cholinesterase. The ED50 values for brain cholinesterase activity were 20 (range, 17-23) in neonates and 44 (31-63) in adults, and those for plasma cholinesterase activity were 20 (13-31) in neonates and 70 (40-120) in adults. The correlation between the two values was good for neonates but not for adult rats (Pope & Chakraborti, 1992). Groups of five 3-month old Sprague-Dawley rats were given chlorpyrifos (purity, 98%) in peanut oil by subcutaneous injection at a dose of 280 mg/kg bw, and trunk blood, cerebral cortex, and corpus striatum were collected 2, 4, 6, and 12 weeks after dosing. Acetylcholinesterase activity was assayed, and the binding of [3H]quinuclidinyl benzilate to muscarinic receptors was measured in membranes of the cortex and striatum. Locomotor activity was measured for 30 min on 5 days/week. A challenge of scopolamine (1 mg/kg bw intraperitoneally) was given 2 weeks after chlorpyrifos treatment and then every two weeks for 12 weeks. Extensive inhibition of cortical and striatal cholinesterase activity was seen 2 (94-96%), 4 (about 80%), 6 (about 60%), and 12 (15-20%) weeks after treatment. Plasma cholinesterase activity was inhibited to a lesser extent, and recovery was more rapid, with 85% inhibition at 2 weeks, 55% at 4 weeks, and 30% at 6 weeks; the activity was normal at 12 weeks. The changes in cholinesterase activity were accompanied by reductions in muscarinic receptor binding sites in the cortex (34, 33, and 18% reductions in maximal binding) and striatum (48, 40, and 23% reductions) 2, 4, and 6 weeks after exposure. Tests of locomotor activity showed hypoactivity in treated rats for 2-3 days after treatment. After the scopolamine challenge, chlorpyrifos-treated rats showed increased activity at all times except week 10 (Pope et al., 1992). Chlorpyrifos was given to groups of six adult male Sprague-Dawley rats by subcutaneous injection at a dose of 280 mg/kg bw, and the animals were killed 2, 7, and 14 days after treatment. A significant ( p < 0.05), approximately 6% decrease in body weight was seen after 2-3 days, but the body weights recovered during the remainder of the study. Chlorpyrifos caused marked inhibition of cholinesterase activity in brain regions and plasma at all times, with about 90% inhibition in the cortex, striatum, and plasma at day 2, and there was virtually no recovery of this activity within the 14-day recovery period. Reductions of < 55% in atropine-sensitive quinuclidinyl benzilate binding (total muscarinic receptors) and binding in the cortex and striatum were also observed (Chaudhuri et al., 1993). Pregnant Sprague-Dawley rats were injected subcutaneously with either peanut oil or chlorpyrifos (purity, 98%) at 25 mg/kg bw per day on days 12-19 of gestation and killed on day 20 of gestation or postnatal day 3. In a separate study to determine the dose-response relationship, rats were similarly exposed to chlorpyrifos at 6.25 or 12.5 mg/kg bw per day on days 12-19 of gestation and killed on day 20 for analysis of various neurochemical markers. No maternal toxicity was seen at any dose. Treatment with 25 mg/kg bw per day induced only an initial, slight, transient decrease in maternal body weight. Treatment from day 12 of gestation did not affect fetal weight at day 16 or 20, but pup weight on postnatal day 1 was significantly reduced by treatment at this dose. Maternal brain cholinesterase activity measured on day 20 of gestation was significantly inhibited in a dose-related manner, with 75 and 90% inhibition at 6.25 and 25 mg/kg bw per day, respectively. Fetal brain cholinesterase activity was less severely inhibited, with 40 and 60% inhibition at the two doses, respectively. Extensive inhibition of acetylcholinesterase activity, to 10-17% of the control value, was seen in dams treated with 25 mg/kg bw per day. Significant, dose-related downregulation of muscarinic receptors in maternal and fetal brain was noted on day 20 of gestation after exposure at all doses. The responses to righting reflex and cliff avoidance tests were markedly altered after repeated exposures. Generally, the neurochemical effects were more severe in dams than in the developing fetuses, despite the greater sensitivity of fetal brain cholinesterase to inhibition by chlorpyrifos. Comparison of these results with those of an earlier study of acute exposure to chlorpyrifos (Chanda et al., 1995) indicated that repeated exposure causes greater down-regulation of muscarinic receptors than the equivalent acute dose (Chanda & Pope, 1996). Cats Technical-grade chlorpyrifos dissolved in methylene chloride and mixed in olive oil was given to adult male, colony-raised, short-hair cats at doses of 0, 0.1, 0.5, 1, 5, 10, or 50 mg/kg bw to establish a dose that would induce clinical signs of toxicosis but not death. Two cats were used in this segment of the study, and each received increasing doses of chlorpyrifos. On the basis of deaths at 50 mg/kg bw, a dose of 40 mg/kg bw was selected for the next phase of the study, in which three groups of six cats received corn oil, chlorpyrifos, or chlorpyrifos followed by injections of atropine sulfate every 12 h for 2 days. Two cats receiving the antidote also received pyridine 2-aldoxime methane sulfonate every 12 h until the clinical signs had abated. All cats given chlorpyrifos showed clinical signs of toxicity, and one cat died 4 days later. Whole-blood and plasma cholinesterase activity was reduced in all treated animals, but the levels had returned to pre-test levels after 7-28 days. Brain cholinesterase activity was normal in all animals that survived until the end of the study, 28 days after treatment. Gross pathology revealed no treatment-related lesions, and haematological parameters were unaffected (Hooser et al., 1988). Groups of five adult male cats received a single intramuscular injection of corn oil (vehicle), diisopropyl fluorophosphate (positive control; 5 mg/kg bw), or chlorpyrifos (purity, 99%; 300 mg/kg bw) and were observed for 60 days. Atropine and pyridine 2-aldoxime methane sulfonate were given to chlorpyrifos-treated animals once or twice daily for 14-24 days. Ataxia was observed in positive controls (mean onset, 16 days; range, 14-19 days) and in those given chlorpyrifos (mean onset, 19 days; range, 17-21 days) . Functional deficits were seen, characterized by a crouched-waddling gait, hypermetria, and proprioceptive deficits, and these effects were still present in both groups 56 days after exposure; the effects in chlorpyrifos-treated animals began to subside after day 35. Lymphocyte neuropathy target esterase activity was inhibited by < 96% in positive control animals (24 h after dosing) and by 46% in chlorpyrifos-treated cats (7 days after dosing) but had reached control levels by day 28 in the latter group. Whole-blood cholinesterase activity was reduced by < 86% by day 4 and slowly recovered to control levels by day 56. Diisopropyl fluorophosphate had inhibited cholinesterase activity by < 63% 24 h after dosing, but the recovery of activity was similar to that in chlorpyrifos-treated animals. Histological examination (Table 22) revealed degeneration of axons in animals treated with chlorpyrifos and with diisopropyl fluorophosphate, and, while the lesions were similar, they were more severe in chlorpyrifos-treated animals. The effects were characterized by axonal degeneration with vacuolar (spongy) changes in the white matter tracts of the cerebrum, cerebellum, and spinal cord and occasionally in the peripheral nerves. Vacuolization was seen, resulting from distension of axonal spaces associated with focal axonal swelling and degeneration of myelin and myelin-forming cells, with infiltration of snall numbers of macrophages (Fikes et al., 1992). The route of administration and magnitude of the dose in this study limit its usefulness. (g) Studies on metabolites (i) Acute toxicity The main metabolite of chlorpyrifos, TCP, was generally of lower acute toxicity than chlorpyrifos (Table 23). (ii) Short-term studies of toxicity Groups of 10 rats of each sex were fed diets containing 0, 0.01, 0.03, 0.1, 0.3, or 1% TCP for 90 days. The three lower doses had no treatment-related effects on body-weight gain, mortality, food consumption, haematological values, blood urea nitrogen, alkaline phosphatase, or average body and organ weights. The NOAEL was 0.1% in the diet, approximately 50 mg/kg bw per day, on the basis of reduced body-weight gain at 1% (Beatty, 1964). Groups of 15 rats of each sex were fed diets containing sodium TCP at concentrations providing doses of 0, 10, 30, or 100 mg/kg bw per day for 91 days. The dietary concentration was varied to allow for increasing body weight and ranged from about 85, 250, and 825 ppm at day 0 to about 160, 460, and 1540 ppm at day 84 at the three doses, respectively. Significantly increased body-weight gain was observed in males at 30 mg/kg bw per day from day 48 onwards. There were no treatment-related changes in haematology, urinary analysis, or blood chemistry values. The absolute weight of the heart was increased in males at 100 mg/kg bw per day, and increased absolute and relative liver and kidney weights were seen in animals of each sex at this dose. There were no gross or histopathological alterations due to treatment. The NOAEL was 30 mg/kg bw per day on the basis of increased relative liver and kidney weights (Barna-Lloyd & Szabo, 1985). Table 22. Occurrence and severity (score ± SD of histological changes in the nervous system of cats given a single intramuscular injection of chlorpyrifos, diisopropyl fluorophosphate (positive control), or corn oil Location Treatment Corn oil Diisopropyl fluorophosphate Chlorpyrifos No. affected Score No. affected Score No. affected Score Brain Cerebruma 2/5 0.40 ± 0.49 4/5 0.80 ± 0.40 4/5 2.00 ± 0.63 Cerebellumb 1/5 0.20 ± 0.40 2/5 0.40 ± 0.49 3/5 1.00 ± 0.89 Spinal cord C2 2/5 0.20 ± 0.49 4/5 0.80 ± 0.40 5/5 1.40 ± 0.49 C7 1/5 0.20 ± 0.40 3/5 0.60 ± 0.49 5/5 1.40 ± 0.49 T-L 4/5 0.80 ± 0.40 5/5 1.00 ± 0.0 4/5 0.80 ± 0.40 L4 0/5 0.00 ± 0.0 5/5 1.60 ± 0.49 5/5 1.80 ± 0.40 Sciatic nerve 0/5 0.00 ± 0.0 3/5 0.60 ± 0.49 1/5 0.20 ± 0.40 Severity of lesions (axonal swelling, vacuolar change, myelin degeneration) in each section was given the score of the most severely affected area. The lesions were scored as 0 = normal or equivocal, 1 = mild (1-5 nerve fibres affected/high power field), 2 = moderate (6-10 nerve fibres affected), 3 = marked (11-15 nerve fibres affected), and 4 = severe change (> 20 nerve fibres affected). a Anterior parietal cerebrum sectioned transversely immediately posterior to the optic chiasm and anterior to the infundibulum b Transverse section through the cerebellum and brain stem at the level of the trigeminal nerves Table 23. Acute toxicity of 3,5,6-trichloro-2-pyridinol (TCP) administered orally Species Strain Sex Vehicle LD50 (mg/kg bw Reference and 95% CI or range) Mouse Swiss M 0.5% hydroxypropylmethyl 380 (333-433) Gerbig & Emerson (1970a) F cellulose (Methocel) 414 (367-469) Rat Sprague-Dawley M 0.5% hydroxypropylmethyl 794 (709-889) Gerbig & Emerson (1970b) F 870 (758-1009) Doga Beagle NR Gelatine capsule > 1000 Gerbig & Emerson (1970c) NR, not reported a One dog received two doses of 1000 mg/kg bw TCP, a second received four doses of 500 mg/kg bw, and a third received eight doses of 50 mg/kg bw. Emesis was seen in all animals within 1-3 h of dosing. There were no deaths. Groups of two beagles of each sex were given diets containing TCP at concentrations providing doses of 0, 26, or 80 mg/kg bw per day for 93 days. At termination, haematological parameters were normal. There was no NOAEL as elevated serum alkaline phosphatase activity and histopathological evidence of liver damage and increased testicular weights were seen in the treated animals (JMPR, 1972; modified by reference to the original report of Copeland, 1964). Groups of three dogs of each sex were given diets containing TCP at concentrations providing doses of 1, 3, 10, or 30 mg/kg bw per day for 91 days. No toxic effects were seen at the three lower doses, but at 30 mg/kg tbw per day evidence of liver toxicity was seen, with increased activity of alkaline phosphatase and alanine and aspartate aminotransferases although there was no unequivocal sign of liver damage on histopathological examination (JMPR, 1972; modified by reference to the original report of Emerson & Gerbig, 1970). Groups of four beagles of each sex were fed diets containing TCP at concentrations providing doses of 0, 3, 12, or 48 mg/kg bw per day for 1 year in a study conducted according to GLP. No details of the clinical observations were reported. The body weights of males were not statistically significantly different between groups, but females at the high dose had consistently lower body weights than controls and statistically significantly lower body-weight gains. No changes in food consumption were found in treated groups, and no alterations in haematological or urinary parameters. The dose-related changes in clinical chemistry values that were attributed to treatment were increased serum alkaline phosphatase and alanine aminotransferase activities in males and females at 3, 6, and 12 months. The values were comparable to those of controls at 3 mg/kg bw per day, consistently elevated but generally not to a statistically significant degree at 12 mg/kg bw per day, and were statistically significantly increased at 48 mg/kg bw per day at most times. There were no gross or histopathological lesions that were associated with treatment and no changes in absolute or relative organ weights. The NOAEL for systemic effects was 12 mg/kg bw per day, on the basis of a lack of a consistent, statistically significant increase in enzyme activities at that dose (Zempel et al., 1987). (iii) Genotoxicity TCP was not genotoxic in a range of assays in vitro and in vivo (Table 24). (iv) Developmental toxicity In a range-finding study, Fischer 344 rats were given TCP (purity, 99.7%) at doses of 0, 43, 75, or 127.5 mg/kg bw per day on days 6-15 of gestation by oral gavage. Clinical signs (perineal soiling) were seen sporadically in all dams at the highest dose and in some at 75 mg/kg bw per day. Food and water consumption were unaffected by treatment. The signs of maternal toxicity were limited to reduced body-weight gain in dams at the high dose. The relative and Table 24. Results of tests for the genotoxicity of 3,5,6-trichloro-2-pyridinol End-point Test object Concentration Purity Results Reference (%) In vitro Reverse mutation S. typhimurium 3.16, 10, 31.6, 99.7 Negativea,b Zempel & Bruce (1986) TA98, TA100, 100, 316 µg/plate TA1535, TA1537, in DMSO TA1538 Unscheduled Rat hepatocytes 1-100 µg/ml in 99.7 Negativec Gollapudi (1987) DNA synthesis (Fischer CDF 344) DMSO Forward mutation Chinese hamster 62.5, 125, 250, 99.7 Negativea Linscombe & Gollapudi ovary cells, hprt locus 500, 750 µg/ml (1986) In vivo Micronucleus Mouse (CD-1) 250, 500, 750, 99.9 Negative McClintock & Gollapudi formation marrow cells 1000, 1500, 2000, (1989b; GLP) 3000 mg/kg bw orally in corn oil Micronucleus Mouse (CD-1) 0, 24, 76, 240 mg/kg 99.7 Negative Bruce et al. (1985) formation marrow cells bw orally in corn oil GLP, good laboratory practice; DMSO, dimethyl sulfoxide a With and without an exogenous metabolic system b Toxic at 100 (some strains) and 316 (all strains) µg/plate c Toxic at 50 and 100 µg/ml absolute weights of the liver and kidney were unaffected by treatment, and there was no observable effect on reproductive parameters (Scortichini et al., 1986a). Groups of Fischer 344 rats were given 0, 50, 100, or 150 mg/kg bw per day of TCP (purity, 99.7%) on days 6-15 of gestation by oral gavage in a study that conformed to GLP. No clinical signs of toxicity were seen in any group. Slight vaginal bleeding occurred in one animal at 50 mg/kg bw per day and two at 150 mg/kg bw per day on day 13. Food consumption was slightly decreased in animals at 100 mg/kg bw per day and decreased by about 10% in those at the high dose. Significant, dose-related decreases in body-weight gain were recorded throughout treatment at 100 and 150 mg/kg bw per day. Changes in the relative and absolute liver and kidney weights were restricted to a significant increase (4%) in the relative liver weight at 150 mg/kg bw per day, consequent to a significant decrease (5%) in body weight at this dose. There was a dose-related trend in the number of implantation sites per dam, which was reflected in the number of fetuses per litter and probably in gravid uterine weight; the latter trend may also reflect the trend to lower body weight with higher dose. Fetal examination revealed a small number of findings in each group, including controls. Overall, treatment had no significant effect on reproductive parameters or fetuses. The NOAEL for maternal effects was 50 mg/kg bw per day on the basis of decreased body-weight gain, and that for fetal toxicity and teratogenic effects was 150 mg/kg bw per day, the highest dose tested (Hanley et al., 1987a). In the first phase of a study in inseminated New Zealand white rabbits, the animals were given TCP (purity, 99.7%) on days 6-18 of gestation by oral gavage at doses of 0, 48.5, 80, or 130 mg/kg bw per day. As there was uncertainty that the maximum tolerated dose had been reached in this phase, additional groups of animals were given 130 or 210 mg/kg bw per day (phase 2). Maternal body-weight gain during gestation was depressed but not statistically significantly and was highly variable at the highest dose in both phases. Gross examination of all animals showed no significant findings or any differences between groups. At 210 mg/kg bw per day, resorptions were seen in all litters, and the percent implantations resorbed and the percent litters with resorptions were statistically significantly different from those in the comparable control group in phase 2. There was no observable effect of treatment on reproductive parameters at doses up to 130 mg/kg bw per day (Scortichini et al., 1986b). Groups of New Zealand white rabbits were given TCP (purity, 99.7%) by oral gavage on days 7-19 of gestation at doses of 0, 25, 100, or 250 mg/kg bw per day in a study that met GLP requirements. Female animals were given injections of human chorionic gonadotropin to synchronize estrus and were inseminated 3 weeks later, which was presumed to be day 0 of gestation. No clinical signs of toxicity were seen in any group. Water and food consumption were not reported. A dose-related decrease in body-weight gain was seen during treatment, which reached statistical significance at the high dose. There were no treatment-related changes in the relative or absolute weights of the liver or kidney. Fetal examination revealed a small number of malformations in each group, including controls. Overall, there was no statistically significant effect of treatment on reproductive parameters or fetuses, but an increased incidence of central nervous system malformations at the intermediate and high doses suggested a possible teratogenic effect, and the authors concluded that these findings provided evidence of teratogenic potential, despite a lack of statistical significance. The malformations included severe dilatation of the cerebral ventricles and hydrocephaly. The incidences of minor external, visceral, or skeletal alterations were unchanged. The NOAEL for maternal effects was 100 mg/kg bw per day on the basis of decreased body-weight gain during treatment. The NOAEL for fetal toxicity and teratogenic effects was 25 mg/kg bw per day, on the basis of central nervous system malformations at higher doses (Hanley et al., 1987b). 3. Observations in humans 3.1 Experimental studies A commercial preparation of chlorpyrifos containing 35% xylene was applied to the skin of the back and abdomen of four volunteers (skin area not stated) at a dose of 5, 10, 25, or 50 mg/kg bw 20 (5 days/week), four, three, and two times, respectively. The material was covered with an occlusive patch for 12 h, and after removal of the patch each site was scored for irritation before washing. Each site was used only once. Plasma and erythrocyte cholinesterase activity was determined before and several times after treatment (exact times not stated; for results, see Table 25). In another study, five volunteers received dermal doses of 94-620 mg/kg bw for 12 h and showed similar inhibition of erythrocyte and plasma cholinesterase activity 24, 48, and 72 h after exposure, even at the highest dose. No skin irritation was recorded. Plasma cholinesterase activity was significantly inhibited with two of the dose regimens, the activity reaching a minimum around 3 days after the last dose and then recovering to control values within 4-10 days; erythrocyte cholinesterase activity were not inhibited with any regimen (Kilian et al., 1970). Four volunteers were exposed for 5 min to a formulation containing 61.5% chlorpyrifos and 34.5% xylene from an ultra-low volume cold aerosol fog generator delivering 3.8 L/h. Air sampling showed a concentration of chlorpyrifos in the breathing space of about 108 mg/L (range, 83-133 mg/L). The subjects were exposed at a distance of 8 m and wore plastic coveralls allowing exposure of the heads and hands of two subjects and the heads, hands, and arms of the other two. Exposure was terminated after 5 min because of the ocular and pulmonary irritation induced. Plasma and erythrocyte cholinesterase activity was not depressed 24 h after exposure (Pennington & Edwards, 1971). Table 25. Cholinesterase activity after dermal dosing of humans (% of pre-treatment value) Dose regimen End of dosing Day 1 Day 3 Plasma Erythrocytes Plasma Erythrocytes Plasma Erythrocytes 10 mg/kg bw for 4 × 12 h 99 102 97 103 97 100 50 mg/kg bw for 2 × 12 h 100 100 100 100 102 100 25 mg/kg bw for 3 × 12 h 67 98 48 98 5 mg/kg bw for 20 × 12 h 67 103 64 104 74 104 One subject per dose regimen Groups of four healthy, adult, male volunteers received chlorpyrifos in tablet form at doses of 0, 0.014, 0.03, or 0.1 mg/kg bw per day for 27, 20, and 9 days, respectively, with breakfast. Control subjects received a placebo for 48 days. Although not specifically stated, the high dose appears to have been withdrawn because of the severity of plasma cholinesterase inhibition on day 9. No rationale was given for termination of treatment at the lower doses. Heparinized blood samples were obtained from the subjects twice weekly for determination of plasma and erythrocyte cholinesterase activity until all values had returned to pre-treatment levels. As it was not stated whether the blood samples were taken at the same time on each sampling day, the interval between administration and sampling is unknown. No indication was given of the frequency of observation for clinical signs after treatment or how such observations were made. A single subject at 0.1 mg/kg bw per day reported having a runny nose, blurred vision, and a feeling of faintness on the final day of dosing, and his plasma cholinesterase activity was found to be inhibited by 70% when compared with pre-treatment levels. No decrease in erythrocyte cholinesterase activity was seen in any subject during the study, and no other treatment-related effects were noted. Rapid, marked depression of plasma cholinesterase activity was observed in subjects at the high dose, and the activity did not return to control levels until about day 34. Similar results were obtained when the plasma cholinesterase activity in these subjects was compared with the baseline activity of the group before treatment. The mean plasma cholinesterase activity of men at 0.03 mg/kg bw per day was inhibited by > 20% on days 16-20 of treatment when compared with those given placebo, and the activity did not return to control levels for several weeks after cessation of treatment. The plasma cholinesterase activity of men at 0.014 mg/kg bw per day was inhibited by 20% only on day 13 and was similar to the baseline activity from day 20 to day 27, even with continued administration of chlorpyrifos. Two-way analysis of variance indicated that the inhibition of mean plasma cholinesterase activity was significant only at 0.1 mg/kg bw per day ( p < 0.05) when compared with controls. The NOAEL for inhibition of erythrocyte cholinesterase activity was 0.1 mg/kg bw per day, the highest dose tested (JMPR, 1972; modified by reference to the original report of Coulston et al., 1972). Six white male volunteers aged 27-50 years were used in another study. One volunteer was given a single oral dose of 0.5 mg/kg bw chlorpyrifos in a lactose capsule, about 30 min after food, and about 1 month later the other volunteers received the same treatment and the first volunteer was given a dermal dose of 0.5 mg/kg bw chlorpyrifos dissolved in methylene chloride. Two weeks after the first dermal dose, the first volunteer was given a further dermal dose of 0.5 mg/kg bw in dipropylene glycol methyl ether. The other volunteers were given a dermal dose of 5 mg/kg bw of chlorpyrifos in dipropylene glycol methyl ether 4 weeks after their oral dose. All urine was collected from volunteers from 24-48 h before dosing until 120 h after dosing. No signs or symptoms of toxicity were reported in any of the men during the study. After oral administration of 0.5 mg/kg bw, the plasma cholinesterase activity of the first volunteer was inhibited by 71% from that before treatment and that of the other men by a mean of 85% within 12-24 h of treatment. The range of individual values was not provided. Plasma cholinesterase activity had returned to > 80% of the mean baseline value by 30 days after oral treatment. After dermal application on day 30, the plasma cholinesterase activity had decreased to approximately 70% of baseline, and returned to about 80-90% of those levels by day 40 of the study. The intra-group variation was stated to be considerable, but the figures were not provided. Erythrocyte cholinesterase activity was not significantly inhibited by either oral or dermal doses of chlorpyrifos (Nolan et al., 1982, 1984). In a double-blind, randomized, placebo controlled study of the effects of single oral doses of chlorpyrifos (purity, 99.8%), groups of six fasted men and women aged 18-55 received doses of 0, 0.5, 1, or 2 mg/kg bw in lactose powder. The study was conducted in two phases separated by 14 days. The volunteers were dosed with 0, 0.5, or 1 mg/kg bw in the first phase and the results were assessed before administration of 0 or 2 mg/kg bw in the second phase. Blood samples were collected 10 and 0 h before treatment and 2, 4, 8, 12, 24, 36, 48, 72, 96, 120, 144, and 168 h after treatment and analysed for erythrocyte cholinesterase activity and chlorpyrifos and its metabolites. In addition, all urine voided from 48 h before dosing to 168 h after dosing was collected at 12- or 6-h intervals and analysed for chlorpyifos and metabolites. Haematology, clinical chemistry, urinary analysis and a brief physical examination were performed at completion of the study. Blood and urine were collected to determine each volunteer's paraoxonase status and the concentrations of chlorpyrifos and metabolites, but these data have not yet been reported. The volunteers were screened for general health according to set criteria and instructed to refrain from alcohol, strenuous exercise, and prescription medications before and during the study. The doses were taken by capsule after an overnight fast. The health status of subjects was monitored closely; vital signs (blood pressure, pulse, respiration, and temperature) were assessed before dosing and 1, 2, 4, 8, 12, 24, 48, and 168 h after treatment. The subjects were questioned about their well-being at each sampling time, and the symptoms were evaluated clinically. The subjects were aware of the signs and symptoms of cholinergic toxicity and were instructed to inform the study physician of any adverse effects. The subjects were unaware of their treatment, and the signs and symptoms were assessed and treated by a physician who was also unaware of the treatment status of the subject. There were no significant deviations from the study protocol. One male control in phase 1 and one woman receiving 2 mg/kg bw did not provide a complete series of blood and urine samples. Treatment had no effect on general health or on clinical chemical parameters measured 7 days after dosing. The only treatment-related effect was found in the woman who withdrew from the study, who had decreased erythrocyte cholinesterase activity when compared with her pre-treatment values at most sampling times, with 98.4% of the pretreatment value at 4 h after dosing, 77% at 8 h, 72% at 12 h, 74% at 24 h, 81% at 36 h, and 80% at 48 h (Table 26). When the data for this subject are removed from the analysis, the mean for women receiving 2 mg/kg bw is indistinguishable from the value for concurrent controls (Table 26). The NOAEL for clinical signs or symptoms was thus the highest dose tested, and the NOAEL for inhibition of erythrocyte cholinesterase activity was 1 mg/kg bw on the basis of significant inhibition in one of 12 subjects (Kisicki et al., 1999). 3.2 Case reports Blondell & Dobozy (1997) summarized the case reports, case series, statistical surveys, and epidemiological studies of acute and chronic health effects reported to be related to exposure to chlorpyrifos. The authors noted the limitations of their review, including inadequate documentation of exposure and effects, reporting biases, and absence of information on the denominator population at risk. They concluded, largely on the basis of an examination of reports from poison control centres, that chlorpyrifos is one of the leading causes of acute insecticide poisoning in the USA. Certain uses were considered to pose greater risks than others, the main concern being use of liquid formulations in households or by pest control operators indoors or outdoors, termite treatment, and application of liquid sprays and dips to domestic animals. Most of the more serious cases were associated with misuse or inappropriate use such as spills and inadvertent contamination by a pest control operator. Shurdut et al. (1997) commented that deficiencies in the report of Blondell & Dobozy (1997) invalidated its use for reaching a conclusion about the safety of chlorpyrifos. They concluded that the data from poison control centres had been misinterpreted and in fact indicated the relative safety of products containing chlorpyrifos. They also claimed that the report failed to respect the limitations of anecdotal information, failed to consider the extensive testing with regard to neurological damage and the long history of use of products containing chlorpyrifos, made inappropriate use of anecdotes and studies of organophosphates to characterize the safety of chlorpyrifos, failed to consider the extensive database on exposure after use of chlorpyrifos products, and presented the data selectively. In a case report, a previously well 3-year-old boy was found playing near an open, spilled bottle of insecticide containing chlorpyrifos (concentration not stated). The authors noted that clinical manifestations of organophosphate-induced polyneuropathy usually begin 1-3 weeks after an acute cholinergic crisis and that the results of examination of the child were consistent with distal, symmetric, predominantly motor polyneuropathy. In this patient, the symptoms of cholinesterase inhibition and manifestations of weakness and areflexia 11 days after ingestion and electromyographic findings confirmed the presence of acute transient polyneuropathy with a more Table 26. Mean erythrocyte cholinesterase activity as percent of mean baseline values in volunteers given single oral doses of chlorpyrifos Dose Time in relation to dosing (h) (mg/kg bw) - 10 0 4 8 12 24 36 48 Women 0.0a 99.5 (2.1) 100.5 (2.1) 98.3 (2.0) 102.1 (2.4) 100.7 (1.8) 99.7 (3.9) 98.4 (1.7) 100.2 (2.3) 0.0b 99.2 (4.4) 100.8 (4.4) 102.5 (5.9) 103.1 (5.4) 98.4 (3.2) 99.4 (3.7) 94.6 (5.3) 97.7 (5.1) 0.5 101.2 (6.7) 98.8 (6.7) 96.3 (6.5) 98.1 (2.1) 95.8 (3.9) 97.0 (3.6) 94.7 (4.7) 97.1 (4.1) 1 98.2 (0.9) 101.8 (0.9) 104.6 (4.2) 100.8 (3.5) 98.9 (3.7) 100.9 (5.6) 96.0 (4.1) 98.9 (2.1) 2 99.8 (1.8) 100.2 (1.8) 100.3 (3.2) 96.6 (6.81) 91.1 (9.7) 95.2 (10.6) 95.9 (7.4) 94.5 (7.7) Men 0.0a 100.6 (1.8) 99.4 (1.8) 99.1 (2.2) 99.6 (4.6) 99.8 (4.3) 98.4 (4.3) 97.6 (2.8) 98.2 (3.1) 0.0b 96.1 (5.1) 103.9 (5.1) 101.6 (4.0) 102.5 (5.2) 102.9 (5.8) 102.6 (2.3) 98.2 (3.8) 95.9 (6.8) 0.5 98.7 (1.1) 101.3 (1.1) 102.0 (3.0) 103.4 (4.9) 99.7 (3.0) 101.9 (3.3) 98.1 (3.0) 92.8 (4.7) 1 98.6 (1.9) 101.4 (1.9) 104.5 (7.8) 101.1 (4.2) 101.1 (4.7) 101.7 (4.7) 98.5 (4.6) 92.1 (2.0) 2 101.2 (2.3) 98.8 (2.3) 99.2 (3.1) 98.9 (3.5) 98.7 (4.0) 99.2 (2.6) 99.5 (5.1) 98.4 (3.1) a Phase 2 control b Phase 1 control proximal distribution than is usually seen in adults. The acute, reversible bilateral vocal cord paralysis reported in this patient had not been seen previously in organophosphate-induced delayed neuropathy, but the authors noted that the onset of stridor and vocal cord paralysis coincided with the onset of muscle weakness and resolved at the same time as normalization of the peripheral neuropathy (Aiuto et al., 1993a). Gutmann & Bodensteiner (1993), responding to the report of Aiuto et al. (1993), suggested that this case may not be a clear example of organophosphate-induced delayed neuropathy. They noted that chlorpyrifos has been associated with such symptoms only after severe intoxication characterized by extensive cholinesterase inhibition. The inhibition of plasma cholinesterase activity in the reported case was instead suggestive of mild chlorpyrifos intoxication. They also noted that the clinical findings associated with organophosphate-induced delayed neuropathy are predominantly motor polyneuropathy, flaccid weakness and atrophy primarily in the distal limb muscles, and often incomplete recovery usually after months or years. The case report implied that the evoked compound muscle action potentials and sensory nerve action potentials were of normal amplitude, which would run counter to a diagnosis of organophosphate-induced delayed neuropathy. In response to these comments, Aiuto et al. (1993b) noted that the patient's symptoms were consistent with > 90% inhibition of cholinesterase activity and that the plasma cholinesterase activity did not necessarily correlate with the severity of poisoning. The authors concluded that the patient had developed delayed, life-threatening neuropathy, although they were unable to determine the mechanism of the condition. In another case report, a 38-year-old man drank an undefined quantity of a solution of 25% chlorpyrifos. He was treated with 3 mg of atropine intravenously every 2 h for 6 days (total, 400 mg), but any attempt to stop the atropine treatment resulted in reappearance of respiratory distress. After 6 days, the patient was stuporous, catatonic, and suffering from respiratory distress with white mucous secretions, and no cholinesterase activity could be detected in his serum. When ipratropium was administered endotracheally at a dose of 0.5 mg as an aerosol mist over 10 min, the patient's clinical condition improved, although the salivary secretion continued. The patient continued to receive ipratropium at 1-2 mg/day, and respiratory support was discontinued on the fifth day. Severe catatonia and coarse tremor were relieved by administration of dantrolene at 10 mg intravenously, followed by 25 mg orally, three times daily. Cholinesterase activity was still undetectable in serum 1 month after the man was discharged (Shemesh et al., 1988). In a case in which a 26-year-old man intentionally ingested approximately 360 ml of a 6.7% chlorpyrifos formulation containing 2,4-D (10.8%) and mecoprop (11.6%) and a few granules of a warfarin (0.025%) concentrate, the main clinical findings on admission were coma, myoclonus, miosis, cardiac arrythmia with progression to hypotension, oliguria, and death after several episodes of asystole. The authors stated that such symptoms are consistent with those seen in patients who have ingested chlorophenoxy acetic acids such as 2,4-D and mecoprop. The patient did not show many of the usual signs of organophosphate poisoning, such as lachrymation, salivation, respiratory paralysis, or muscle fasciculation, but his erythrocyte cholinesterase activity was inhibited by about 70% at 13 h and by < 90% by 26 h. Plasma cholinesterase activity was inhibited completely at all times. Lymphocyte neurotoxic esterase activity was inhibited by about 50% after 13 h but was within normal limits after 19 h. The activity of neurotoxic esterase in the cerebral cortex was within normal limits but that in peripheral nerves was inhibited by about 70% when compared with normal levels. The inhibition of neurotoxic esterase activity was found in the absence of significant signs of organophosphate intoxication (Osterloh et al., 1983). A 21-year-old man who ingested an unknown quantity of a chlorpyrifos formulation required large amounts of atropine and pyridine 2-aldoxime methane sulfonate, endotracheal intubation, and ventilation to overcome the cholinergic effects, which consisted of pupillary constriction, excess secretions, tachycardia, and impaired consciousness. Bilateral vocal cord paralysis developed 16 days after extubation (28 days after exposure), and this finding was considered to be indicative of organophosphate-induced delayed neuropathy. The patient was asymptomatic within 4 weeks of the onset of symptoms (de Silva et al., 1994). A 42-year-old man who attempted suicide by drinking a commercial formulation of 41% chlorpyrifos (estimated dose, 300 mg/kg bw) went into coma and showed respiratory insufficiency, lachrymation, salivation, sweating, miosis, fasciculations, and bronchorrhoea on admission to hospital 18 h after exposure. A chest X-ray showed right basal bronchopneumonitis, electroencephalographic signs of diffuse irritation were recorded, and chlorpyrifos was detected in gastric lavage fluid. Plasma cholinesterase activity was inhibited by almost 100% by 36 h. The coma and cholinergic signs persisted for 17 days, during which time atropine, pyridine 2-aldoxime methane sulfonate, mechanical respiration, and antibiotics were used. The patient was asymptomatic on day 24 after exposure, and drug administration was stopped; no signs of peripheral or central nervous system involvement were observed. On day 30, lymphocytic neurotoxic esterase, blood acetylcholinesterase, and plasma butyryl cholinesterase activities were markedly below the normal range but recovered slowly thereafter. On day 43, weakness and paraesthesia in the legs with reduced tendon reflexes and sensory conduction velocity were noted. By day 62, the leg weakness was more severe, gait was impaired, and tendon reflexes were absent. Electromyography of the leg muscles showed symmetrical signs of denervation with spontaneous activity, and motor conduction velocity was reduced. Electrophysiological studies of the arms confirmed a symmetrical reduction of sensory conduction velocity of the ulnar nerves. Sural nerve biopsy on day 63 revealed a few altered myelinated fibres. Aspects of axon and myelin degeneration were also found occasionally. The changes were consistent with mild distal axonopathy. Chlorpyrifos was still detectable 10 days after exposure, and neurotoxic esterase activity was still inhibited by 60% 4 weeks after exposure (Lotti et al., 1986). In a study in which toxicological and/or electrophysiological examinations were conducted in 11 patients who had acute organophosphate poisoning, three of the patients, one of whom had been poisoned with chlorpyrifos, developed organophosphate-induced delayed neuropathy (Moretto & Lotti, 1998). The findings in this individual were reported by Lotti et al. (1986). The electro-physiological data indicated organophosphate-induced delayed neuropathy with sensory and motor components which persisted for 3 months, at which time the patient was lost to follow-up. Four incidents of birth defects allegedly associated with exposure to chlorpyrifos were reported. The children were found to have a range of birth defects, including ventricular, ocular, and palate defects and growth retardation (all children), hydrocephaly, microcephaly, mental retardation, blindness, hypotonia, widely spread nipples, and deformities of the teeth, ears, and external genitalia. The mothers of the affected children had reportedly been exposed to chlorpyrifos either at the workplace or at home during the pregnancy. Two of the children were born to the same mother. The exposure of the mothers to chlorpyrifos was poorly characterized, and it was not indicated whether it was severe or sustained during organogenesis. The range of birth defects reported was not consistent with the results of studies in laboratory animals (Sherman, 1996). In a comment on this paper, Gibson (1996) noted that the medical records of the cases indicated that the same effects were not seen in all children and that the effects in some of the children were consistent with a diagnosis of an autosomal recessive birth defect syndrome of the brain and eye known as cerebro-oculo-facio-skeletal syndrome or a closely related one known as the 'MICRO' syndrome. Eight cases were reported in which exposure to chlorpyrifos was claimed to have caused sensory neuropathy. The patients each presented with a range of symptoms, and characterization of exposure was poor. One patient who reported signs of cholinesterase inhibition, including lachrymation, muscle twitching, and diarrhoea, was a pesticide applicator who had reportedly been exposed to chlorpyrifos in a closed environment for 6 months. The patient's erythrocyte cholinesterase activity was reportedly to be 'low' initially but returned to normal levels within 2 months. Neurological evaluations were conducted only 6 weeks after the other symptoms were reported; at this time, sensory loss of all modalities was found in a 'stocking-glove distribution' test, with mild distal weakness and areflexia in the lower extremities. Studies of nerve conduction and quantitative sensory threshold revealed changes consistent with peripheral neuropathy of the distal anonopathy type. The decreased reflexes and mild distal weakness were consistent with polyneuropathy. Follow-up examination at 1 year revealed remission of all symptoms. In the other seven patients (four from the same family), the symptoms were nonspecific, and the absence of symptoms of cholinesterase inhibition suggested that these patients had not been as exposed to chlorpyrifos as extensively as the first patient. The lack of immediate electrodiagnostic testing, the poor exposure characterization, and the variation in the reported clinical findings make interpretation of the case studies difficult. Additionally, subjective questioning was used to obtain information from the patients. Follow-up examinations for a range of parameters were not conducted on several patients, and the patients did not undergo the same initial testing regimen. Thus, one patient was exposed to chlorpyrifos occupationally at a level sufficient to decrease cholinesterase activity and cause cholinergic symptoms; he also showed evidence of mild, reversible polyneuropathy, possibly in the presence of decreased erythrocyte cholinesterase activity. For two other patients, there was limited evidence of mild polyneuropathy (Kaplan et al., 1993). 3.3 Monitored field trials A monitored field test was used to determine the effect of chlorpyrifos on spraymen applying treatment at the rates used in programmes for the eradication of mosquitoes. Three studies were conducted in 1966, 1967, and 1968. Cholinesterase activity was measured, and exposure was terminated if the inhibition reached 50% of the baseline values. In the first study, three spraymen showed marked depression of plasma cholinesterase activity, with reductions of 68-82% from baseline values. The two remaining spraymen showed reductions of 52-56% over 9 days, but the baseline values were not available. The study was discontinued after 2 weeks because of the significant decrease in cholinesterase activity. In the second study, no treatment-related effect on cholinesterase activity was seen during a short period. In the third study, erythrocyte cholinesterase activity was not affected by treatment, but plasma cholinesterase activity was reduced by 50-80% in spraymen using a suspension formulation. The activity had generally returned to pre-exposure levels within 2 months of cessation of use. Four spraymen using an emulsion formulation did not show reduced plasma cholinesterase activity during the exposure period (Kenaga, 1967; Eliason et al., 1969). 3.4 Studies of morbidity In a study in which the sample size was small and the statistical power was limited, comparisons were made between 175 employees potentially exposed to chlorpyrifos at a manufacturing plant where they had held jobs between 1 January 1977 and 31 July 1985 and 335 unexposed controls matched on age (within 5 years), date of hire (within 5 years), sex, race, and pay status. The source of information for the observations (self, nurse, company physician, private physician, laboratory) was recorded, as were use of tobacco, alcohol, and cholinergic drugs. Exposure was grouped into high, moderate, and low on the basis of industrial hygiene surveys and consultation with veteran manufacturing personnel. The plasma cholinesterase activities of the potentially exposed employees before exposure were available, and follow-up was conducted at roughly monthly intervals. The mean inhibition of plasma cholinesterase activity was 19 ± 2.9% in the group with low exposure, 32 ± 2.8% in that with moderate exposure, and 32 ± 5.3% in that with high exposure. No cases of peripheral neuropathy were recorded in the study group. Inhibition of plasma cholinesterase activity was observed but was not associated with illness. The exposed groups reported symptoms of dizziness, malaise, and fatigue more frequently than the unexposed group, but the analysis did not indicate any correlation with increasing exposure (Brenner et al., 1989). In a follow-up to this study (Burns et al., 1998), the data were updated to include the period 1987-94, and additional medical disorders were considered. Data on self-reported paraesthesia were collected from 1982 to 1994. The prevalence of peripheral neuropathy was not significantly increased in the group of workers exposed to chlorpyrifos. Comments After oral administration to rats, radiolabelled chlorpyrifos was rapidly and extensively absorbed (up to about 90% of the dose) and eliminated, predominantly in the urine (68-93%) and faeces (6-15% of the dose), within about 72 h of administration. The urinary metabolites included the glucuronide (about 80%) and sulfate (about 5%) conjugates of chlorpyrifos, and 3,5,6-trichloro-2-pyridyl phosphate (TCP; about 12%). The tissue concentrations of residues of [14C]chlorpyrifos were very low (generally < 1 ppm) within 72 h of dosing. The longest half-time of residues in rats was 62 h in fat, and low levels were also detected in the fat of several other species and in the milk of goats. In humans who were poisoned with chlorpyrifos formulations, diethylphosphorus metabolites were excreted in the urine by first-order kinetics, with an average elimination half-time of 6.1 ± 2.2 h in the fast phase and of 80 ± 26 h in the slow phase. In volunteers, the time to maximal concentration of 3,5,6-TCP in the blood was 0.5 h after oral dosing and 22 h after dermal treatment, but the elimination half-time by both routes was 27 h, and the percentage of the administered dose recovered from the urine was 70% after oral dosing and 1.3% after dermal administration. Chlorpyrifos is rapidly metabolized by mixed-function oxidases to the highly reactive chlorpyrifos oxon by oxidative desulfuration. The oxon can be deactivated by hydrolysis to diethylphosphate and 3,5,6-trichloropyridinol, while a minor reaction pathway is hydrolysis to monoethyl 3,5,6-trichloro-2-pyridinyl phosphorothioate. The lowest oral LD50 value was 96 mg/kg bw (range, 96-475 mg/kg bw) in rats and 100 mg/kg bw (range, 100-150 mg/kg bw) in mice. Female rats were generally more sensitive to the acute effects of chlorpyrifos than males. The signs of acute intoxication with chlorpyrifos were consistent with cholinesterase inhibition. The acute dermal LD50 of chlorpyrifos was > 2000 mg/kg bw in rats and > 1200 mg/kg bw in rabbits. WHO (1999) has classified chlorpyrifos as 'moderately hazardous'. Chlorpyrifos was irritating to the eye and skin of rabbits, but it did not sensitize the skin of guinea-pigs in Magnusson-Kligman maximization or Buehler tests. In short-term studies, the NOAEL for inhibition of erythrocyte cholinesterase activity was 0.03 mg/kg bw per day in dogs and 0.1 mg/kg bw per day in rats. The NOAEL for inhibition of brain cholinesterase activity was 1 mg/kg bw per day in dogs and rats. The signs of toxicity were largely limited to cholinergic signs and decreased body weights and/or food consumption. The NOAEL for these effects in short-term studies was 1 mg/kg bw per day in rats, and the NOAEL for clinical signs was 3 mg/kg bw per day in dogs. In mice, ocular effects and histopathological alterations (including adrenal lipogenic pigmentation and ocular keratitis) were observed (NOAEL, 50 ppm; equal to 7 mg/kg bw per day). In rats, the NOAEL for increased fatty vacuolation of the adrenal zonal fasciculata and changes in haematological and clinical chemical parameters was 5 mg/kg bw per day. When rats received chlorpyrifos dermally for 21 days, the NOAEL for inhibition of cholinesterase activity in erythrocytes and brain was 5 mg/kg bw per day. In long-term studies, inhibition of cholinesterase activity was again the main toxicological finding in all species. In rats, the NOAEL was 0.1 mg/kg bw per day for inhibition of erythrocyte acetylcholinesterase activity and 1 mg/kg bw per day for inhibition of brain acetylcholinesterase activity, but clinical signs were not seen at doses up to 10 mg/kg bw per day, and the NOAEL for reduction in body weight was 1 mg/kg bw per day. In mice, erythrocyte and brain acetylcholinesterase activities were inhibited at 50 ppm, equal to 6.1 mg/kg bw per day, and the NOAEL was 5 ppm, equal to 0.7 mg/kg bw per day. Cholinergic signs and reductions in body weight were reported only at the highest dietary concentration of 250 ppm (equal to 32 mg/kg bw per day). Other treatment-related findings included effects on the liver in mice, with a NOAEL of 50 ppm (equal to 6.6 mg/kg bw per day), and increased adrenal weight in rats with a NOAEL of 1 mg/kg bw per day. There was no treatment-related increase in the incidence of neoplastic lesions in any of the long-term studies. The Meeting concluded that chlorpyrifos is unlikely to pose a carcinogenic risk to humans. Chlorpyrifos was not genotoxic in an adequate range of studies in vitro and in vivo. The Meeting concluded that chlorpyrifos is not genotoxic. In multigeneration studies of reproductive toxicity in rats, the treatment-related effects of chlorpyrifos were limited to inhibition of cholinesterase activity, consistent with that seen in other short- and long-term studies, and fetotoxicity characterized by reduced pup viability, body weights and survival. No significant, treatment-related clinical signs were reported. The NOAEL for inhibition of maternal acetylcholinesterase activity was 0.1 mg/kg bw per day for erythrocytes and 1 mg/kg bw per day for brain. The NOAEL for developmental toxicity was 1 mg/kg bw per day. No effects on reproductive parameters were observed at the highest dose tested, 5 mg/kg bw per day. In studies of developmental toxicity in mice, rats, and rabbits, the maternal effects included inhibition of erythrocyte and/or brain acetylcholinesterase activity and cholinergic signs (lowest NOAEL, 1 mg/kg bw per day in rats and mice) and reductions in body weight and food consumption (lowest NOAEL, 2.5 mg/kg bw per day in rats). The observed fetal toxicity (lowest NOAEL, 2.5 mg/kg bw per day in rats) and developmental toxicity (NOAEL, 1 mg/kg bw per day in rats) were consistent with treatment-related maternal toxicity; there was no evidence of treatment-related malformations in any of the studies. There was no effect on cognitive function (learning, memory, and habituation) in pups exposed to chlorpyrifos in utero and for a period post partum at doses up to and including the highest dose of 5 mg/kg bw per day, while inhibition of cholinesterase activity, decreased brain weight, and delayed development were seen at lower doses, consistent with findings in other studies. In studies of delayed neurotoxicity, chlorpyrifos was given to chickens as either single or repeated doses. Significant inhibition of both cholinesterase and neuropathy target esterase activity was observed, and mild delayed neuropathy was seen in a number of studies; aggressive antidotal therapy was always necessary to allow at least some of the treated birds to survive. Despite the marked cholinergic toxicity of chlorpyrifos, there was no evidence that it caused delayed neurotoxicity, and there was no increase in the incidence of histopathological lesions in the nerve tissues of birds treated at doses up to 10 mg/kg bw per day for up to 91 days. In a number of studies in rats given single doses of up to 100 mg/kg bw, repeated doses of up to 10 mg/kg bw per day for 4 weeks, or repeated doses of up to 15 mg/kg bw per day for 13 weeks, there were no treatment-related neurological lesions or effects on cognition and no inhibition of neuropathy target esterase activity, although significant inhibition of erythrocyte, brain, and peripheral tissue cholinesterase activity was seen at some doses. In a study that included a functional observational battery of tests, clinical signs of intoxication were observed after a single dose only when brain acetylcholinesterase activity was inhibited by more than 60% or when whole-blood cholinesterase activity was inhibited by more than 80%. When chlorpyrifos was applied as a single dose of up to 5 mg/kg bw to the skin of volunteers for 12 h, erythrocyte cholinesterase activity was not significantly inhibited. Plasma cholinesterase activity was inhibited after 20 12-h dermal exposures to 5 mg/kg bw per day over 4 weeks or after three daily 12-h exposures to 25 mg/kg bw per day on consecutive days, but erythrocyte cholinesterase activity was not inhibited under any treatment regimen. A single oral dose of up to 1 mg/kg bw or repeated doses of up to 0.1 mg/kg bw per day for 9 days did not significantly inhibit erythrocyte acetylcholinesterase activity in volunteers. No clinical signs were observed in these studies. Inhibition of erythrocyte acetylcholinesterase activity was observed in a single female volunteer (of a group of six men and six women) given a single oral dose of 2 mg/kg bw. In a case of human poisoning with chlorpyrifos at an estimated dose of 300-400 mg/kg bw, significant inhibition of neuropathy target esterase in lymphocytes and of plasma and erythrocyte acetylcholinesterase activity was reported, with severe cholinergic signs which required aggressive, extensive antidotal therapy and artificial ventilation. Mild distal axonopathy consistent with organophosphate-induced delayed polyneuropathy was reported some weeks after the poisoning incident. The ADI of 0-0.01 mg/kg bw established by the 1982 Meeting was based on a NOAEL of 0.1 mg/kg bw per day for inhibition of erythrocyte acetylcholinesterase activity in humans. The present Meeting affirmed this ADI on the basis of the NOAEL of 1 mg/kg bw per day for inhibition of brain acetylcholinesterase activity in studies in mice, rats, and dogs, using a 100-fold safety factor, and on the basis of the NOAEL of 0.1 mg/kg bw per day for inhibition of erythrocyte acetylcholinesterase activity in the study of human subjects exposed for 9 days, using a 10-fold safety factor. The Meeting allocated an acute reference dose of 0.1 mg/kg bw on the basis of the NOAEL of 1 mg/kg bw for inhibition of erythrocyte acetylcholinesterase activity in a study in which volunteers received a single oral dose of chlorpyrifos, with a safety factor of 10. Toxicological evaluation Levels that cause no toxic effect Mouse: 5 ppm, equal to 0.7 mg/kg bw per day (toxicity in a 79-week study of toxicity and carcinogenicity) 1 mg/kg bw per day (maternal toxicity in a study of developmental toxicity) 10 mg/kg bw/day (fetal toxicity in a study of developmental toxicity) Rat: 1 mg/kg bw per day (toxicity in 2-year studies of toxicity and carcinogenicity) 1 mg/kg bw per day (developmental and parental toxicity in a two-generation study of reproductive toxicity) 1 mg/kg bw per day (maternal and developmental toxicity in a study of developmental toxicity) 2.5 mg/kg bw per day (fetal toxicity in a study of developmental toxicity) Rabbit: 81 mg/kg bw per day (maternal and fetal toxicity in a study of developmental toxicity) Dog: 1 mg/kg bw per day (toxicity in a 2-year study of toxicity) Human: 0.1 mg/kg bw per day (no inhibition of erythrocyte cholinesterase activity at highest dose tested in men dosed orally for 9 days) 1 mg/kg bw (inhibition of erythrocyte cholinesterase activity in adult volunteers after a single oral dose) Estimate of acceptable daily intake for humans 0-0.01 mg/kg bw Estimate of acute reference dose 0.1 mg/kg bw Studies that would provide information useful for continued evaluation of the compound Further observations in humans Toxicological end-points relevant for setting guidance values for dietary and non-dietary exposure to chlorpyrifos Absorption, distribution, excretion, and metabolism in mammals Rate and extent of oral absorption Up to 90% in rats within 72 h; about 70% in humans within 96 h Dermal absorption Less than 2% in humans within 180 h; not determined in animals Distribution Initially widely distributed; highest residues in liver, kidneys and fat at 72 h in rats Potential for accumulation Elimination half-times of < 24 h and low tissue residues after 72 h in rats. No evidence of potential for accumulation Rate and extent of excretion > 95% within 72 h in rats, mainly in urine (68-93%) and faeces (6-15%) Metabolism in animals Rapidly metabolized by mixed-function oxidases to chlorpyrifos oxon via oxidative desulfuration and an electrophilic phosphooxathiiran intermediate. Degradation by conversion directly to 3,5,6-trichloro-2-pyridyl phosphate and diethyl thiophosphate. The oxon is hydrolysed to diethyl phosphate and 3,5,6-trichloropyridinol, while a minor reaction pathway is by hydrolysis to monoethyl-3,5,6-trichloro-2-pyridinol phosphorothioate. Toxicologically significant compounds Parent compound and oxon (animals, plants and environment) Acute toxicity Rat,, LD50, oral 96 mg/kg bw Rat LD50, dermal > 2000 mg/kg bw Rat, LC50, inhalation > 36 mg/m3 (4 h, vapour, nose-only exposure) 560 mg/m3 (4 h, nebulized particles < 5 mm, whole-body exposure) Rabbit, dermal irritation Slightly irritating Rabbit, ocular irritation Slightly irritating Guinea-pig, dermal sensitization Not sensitizing Short-term toxicity Target/critical effect Inhibition of brain cholinesterase activity Lowest critical oral NOAEL 1 mg/kg bw per day, dog, 2 years 1 mg/kg bw per day, rat, 13 weeks Lowest relevant dermal NOAEL 5 mg/kg bw per day, rat; 21 days Lowest relevant inhalation NOAEL 20.6 ppb (296 mg/m3), rat; 13 weeks Long-term toxicity and carcinogenicity Target/critical effect Inhibition of brain cholinesterase activity Lowest relevant NOAEL 2 years, rat; 1 mg/kg bw per day 78 weeks, mouse; 0.7 mg/kg bw per day Carcinogenicity Not carcinogenic in rats or mice Genotoxicity Not genotoxic Reproductive toxicity Reproductive target/critical effect Neonatal toxicity (reduced pup body weight and survival) Lowest relevant reproductive NOAEL Two-generation, rat; 1 mg/kg bw per day Developmental target/critical effect Fetal and perinatal toxicity at maternally toxic doses (including an increase in delayed ossification, reduced crown-rump length, reduced pup weight, increased postimplantation loss, delayed sexual maturity) Lowest relevant developmental NOAEL Rats; 1 mg/kg bw per day Neurotoxicity/Delayed neurotoxicity Reversible neurotoxicity consistent with cholinesterase inhibition. No evidence of delayed neurotoxicity or histopathological changes in nerves of hens (10 mg/kg bw per day) and rats (15 mg/kg bw per day) for up to 13 weeks. At high acute doses (up to 150 mg/kg bw), significant inhibition of neuropathy target esterase and mild delayed neuropathy in hens, but at this dose, extensive and aggressive antidote treatment were required for birds' survival. Other toxicological studies No effect on cognitive function in rat pups in a study of developmental toxicity at doses up to 5 mg/kg bw per day Medical data No inhibition of erythrocyte acetylcholinesterase activity in volunteers after repeated oral doses of up to 0.1 mg/kg bw per day (for 9 days), single oral doses of up to 1 mg/kg bw, or a single dermal doses of 5 mg/kg bw. Poisoning case presented with severe cholinergic effects, with evidence of delayed polyneuropathy and/or distal axonopathy at a dose that required antidotal treatment and artificial ventilation. Summary Value Study Safety factor ADI 0-0.01 mg/kg bw Rat, 2-year dietary 100 Rat, reproduction Mouse, developmental toxicity Dog, 2-year dietary Human, 9-day oral 10 Acute reference dose 0.1 mg/kg bw Human, single dose 10 References Aiuto, L.A., Pavlakis, S.G. & Boxer, R.A. (1993a) Life-threatening organophosphate-induced delayed polyneuropathy in a child after accidental chlorpyrifos ingestion. J. Pediatr., 122, 658-660. Aiuto, L.A., Pavlakis, S.G. & Boxer, R.A. (1993b) Organophosphate-induced delayed polyneuropathy (comment). J. Pediatr., 123, 837. Aldridge, W.N. (1953) Serum esterases I. J. Biochem., 53, 110-117. Anderson, B.T., Walker, S.A. & Punler, M.J. (1995) Chlorpyrifos tech: Acute inhalational toxicity study in rats. Unpublished report No. 10425 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Bakke, J.E., Feil, V.J. & Price, C.E. (1976). Rat urinary metabolites from O,O-diethyl-O(3,5,6-trichloro-2-pyridyl) phosphorthioate. J. Environ. Sci. Health Bull., 3, 225-230. Barna-Lloyd, T. & Szabo, J.R. (1985) Sodium 3,5,6-trichloro-2-pyridinol: 3-month rat dietary toxicity study. Unpublished report No. TexasT:K-065999-099 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Barna-Lloyd, T., Szabo, J.R. & Young, J.T. (1986) Chlorpyrifos: Subchronic organophosphate-induced delayed-neurotoxicity (OPIDN) study in laying chickens hens. Unpublished report No. TexasT:K-044793-064 from Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Bauriedel, W.R. & Miller, J.H. (1981) The metabolic fate of the sodium salt of C14-labelled 3,5,6-trichloro-2-pyridinol orally administered to swine. Unpublished report No. GH-C 1427 from Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Beatty, S.C. (1964) Results of 90-day dietary feeding studies of 3,5,6-trichloro-2-pyridinol in rats. Unpublished report from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Beatty, S.C. & McCollister, D.D. (1964) Results of 90-day dietary feeding studies of O,Odiethyl O-3,5,6-trichloro-2-pyridyl phosphorothioate in rats. Unpublished report from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Beatty, S.C. & McCollister, D.D. (1971) Results of 90-day dietary feeding studies of O,Odiethyl-O-3,5,6-trichloro-2-pyridyl phosphorothioate in rats. Unpublished report from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Berman, C.L. (1987) Evaluation of chlorpyrifos (Pyrinex) for dermal sensitization of guinea pig. Unpublished report No. 59487-01 from Arthur D. Little Inc., Cambridge, Massachusetts, USA. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Berteau, P.E. & Deen, W.A. (1978) A comparison of oral and inhalation toxicities of four insecticides to mice and rats. Bull. Environ. Contam. Toxicol., 19, 113-120. Blackmore, R.H. (1968) Oral administration -- dogs Dursban. Final report. Unpublished report No. 174-115 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Blagden, S.M. (1994) Chlorpyrifos technical: Acute inhalation toxicity study four-hour exposure (nose only) in the rat. Unpublished report No. CHA 10-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to WHO by Cheminova Agro A/S, Lemvig, Denmark. Blondell, J. & Dobozy, V.A. (1997) Review of chlorpyrifos poisoning data. Washington DC, Environmental Protection Agency. Branson, D.R. & Litchfield, N.H. (1970) Comparative absorption, elimination and distribution of DOWCO 179, its methyl analog DOWCO 214 and their major metabolite, 3,5,6-trichloro-2-pyridinol. Unpublished report No. 70 4479 from Dow Chemical Co., USA. Submitted to WHO by Dow Agrosciences, Indianapolis, IN, USA. Branson, D.R. & Litchfield, N.H. (1971a) Absorption, excretion and distribution of O,Odiethyl O-3,5,6-trichloro-2, 6-C14-2-pyridyl phosphorothioate in rats. Unpublished report No. NBA-9 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, IN, USA. Branson, D.R. & Litchfield, N.H. (1971b) Absorption, excretion and distribution of 3,5,6trichloro-2, 6-C14-2-pyridinol in rats. Unpublished report No. NBA-10 from Dow Chemical Co., USA Submitted to WHO by Dow Agrosciences, Indianapolis, IN, USA. Brenner, F.E., Bond, G.G., McLaren, E.A., Greene, S. & Cook, R.R. (1989) Morbidity among employees engaged in the manufacture or formulation of chlorpyrifos. Br. J. Ind. Med., 46, 133-137. Breslin, W.J., Liberacki, A.B., Dittenber, D.A., Brzak, K.A. & Quast, J.F. (1991) Chlorpyrifos: Two-generation dietary reproduction study in Sprague-Dawley rats. Unpublished report No. K-044793-088 from the Toxicology Research Laboratory, Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Bruce, R.J. & Zempel, J.A. (1986) Chlorpyrifos: Evaluation in the Ames Salmonella/mammalian-microsome mutagenicity assay. Unpublished report No. TexasT:K044793-075 from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Bruce, R.J., Gollapudi, B.B. & Hinze, C.A. (1985) Evaluation of 3,5,6-trichloro-2-pyridinol in the mouse bone marrow micronucleus test. Unpublished report No. K-038278-008 from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Buch SA (1980) Pyrinex tech: Acute inhalation toxicity in rats. Unpublished report No. 80/MA/025/362 from Life Science Research, Stock, Essex, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Buch, S.A. & Gardner, J.R. (1980a) Pyrinex tech: Irritance to rabbit eye. Unpublished report No. 80/MA/023/143 from Life Science Research, Stock, Essex, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Buch, S.A. & Gardner, J.R. (1980b) Pyrinex tech: Irritance to rabbit skin. Unpublished report No. 80/MAK/024/144 from Life Science Research, Stock, Essex, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Buch, S.A. & Gardner, J.R. (1981) Pyrinex technical: Acute oral toxicity in the rat. Unpublished report No. 81/MAK035/104 from Life Science Research, Stock, Essex, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Buch, S.A., Gardner, J.R. & Birnie, J.V. (1980) Pyrinex: Acute percutaneous toxicity study in rats. Unpublished report No. 80/MAK022/300 from Life Science Research, Stock, Essex, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Burns, C.J., Cartmill, J.B., Powers, B.S. & Lee, M.K. (1998) Update of the morbidity experienced of employees potentially exposed to chlorpyrifos. Occup. Environ. Med., 55, 65-70. Bushnell, P.J., Padilla, S.S., Ward, T., Pope, C.N. & Olszyk, V.B. (1991) Behavioral and neurochemical changes in rats dosed repeatedly with diisopropylfluorophosphate. J. Pharmacol. Exp. Ther., 256, 741-750. Bushnell, P.J., Pope, C.N. & Padilla, S. (1993) Behavioral and neurochemical effects of acute chlorpyrifos in rats: Tolerance to prolonged inhibition of cholinesterase. J. Pharmacol. Exp. Ther., 266, 1007-1017. Calhoun, L.L. & Johnson, K.A. (1988) Chlorpyrifos: 4-day dermal probe and 21-day dermal toxicity studies in Fischer 344 rats. Unpublished report No. HET K 044793-085,086 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Calhoun, L.L. & Johnson, K.A. (1989) Supplemental information to the report entitled: Chlorpyrifos: 4-day dermal probe and 21 day dermal toxicity studies in Fischer 344 rats. Unpublished report No. K-044793-085(S) from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Campbell, C.G., Seidler, F.J. & Slotkin, T.A. (1997) Chlorpyrifos interferes with cell development in rat brain regions. Brain Res. Bull., 43, 179-189. Capodicasa, E., Scapellato, M.L., Moretto, A., Caroldi, S. & Lotti, M. (1991) Chlorpyrifosinduced delayed polyneuropathy. Arch. Toxicol., 65, 150-155. Chambers, J.E. (1992) The role of target-site activation of phosphorothionates in acute toxicity. In: Chambers, J.E., Levi, P.E. & Levi, A.P., eds, Organophosphates, San Diego, Academic Press. Chambers, J.E. & Carr, R.L. (1995) Biochemical mechanisms contributing to species differences in insecticidal toxicity. Toxicology, 105, 291-304. Chanda, S.M. & Pope, C.N. (1996) Neurochemical and neurobehavioural effects of repeated gestational exposure to chlorpyrifos in maternal and developing rats. Pharmacol. Biochem. Behav., 53, 771-776. Chanda, S.M., Harp, P., Liu, J. & Pope, C.N. (1995) Comparative developmental and maternal neurotoxicity following acute gestational exposure to chlorpyrifos in rats. J. Toxicol. Environ. Health, 44, 189-202. Chaudhuri, J., Chakraborti, T.K., Chanda, S. & Pope, C.N. (1993) Differential modulation of organophosphate-sensitivity muscarinic receptors in rat brain by parathion and chlorpyrifos. J. Biochem. Toxicol., 8, 207-216. Copeland, J.R. (1964) Results of 93-day dietary feeding studies of 3,5,6-trichloro-2-pyridinol in beagle hounds. Unpublished report No. T35.12-38278-3 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Corley, R.A., Landry, T.D., Calhoun, L.L., Dittenber, D.A. & Lomax, L.G. (1986) Chlorpyrifos: 13-week nose-only vapor inhalation exposure study in Fischer 344 rats. Unpublished report No. HET K 044793-077 from Dow Chemical Company Laboratory, Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Costa, L.G., McDonald, B.E., Murphy, S.D., Omenn, G.S., Richter, R.J., Motulsky, A.G. & Furlong, C.E. (1990) Serum paraoxonase and its influence on paraoxon and chlorpyrifosoxon toxicity in rats. Toxicol. Appl. Pharmacol., 103, 66-76. Coulston, F., Golberg, L., Abraham, R. & Benitz, F.K. (1971) Final report on safety evaluations and metabolic studies on Dowco 179 (Indiana 151). Unpublished report No. K44793-(48)A from Institute of Experimental Pathology and Toxicology, Albany Medical College, Albany, New York, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Coulston, F., Golberg, L. & Griffin, T. (1972). Safety evaluation of Dowco 179 in human volunteers. Unpublished report from Institute of Experimental Pathology and Toxicology, Albany Medical College, Albany, New York, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Crown, S., Weiss, A., Nyska, A. & Waner, T. (1984a) Pyrinex. Toxicity in dietary administration to mice for two weeks: A range-finding study. Unpublished report No. MAK/109/PYR from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Crown, S., Nyska, A. & Waner, T. (1984b) Pyrinex technical. Two-week range-finding study in dietary administration to rats. Unpublished report No. MAK/057/a/PYR from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Crown, S., Gur, E., Nyska, A. & Waner, T. (1985) Pyrinex technical toxicity in dietary administration to rats for 13 weeks. Unpublished report No. MAK/058/PYRA from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Crown, S., Weiss, A., Nyska, A., Waner, T. & Pirak, M. (1987) Pyrinex technical. Toxicity in dietary administration to mice for 13 weeks. A preliminary study. Unpublished report No. MAK/105/PYR from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Crown, S., Nyska, A., Pirak, M. & Waner, T. (1988) Pyrinex technical. Oncogenicity study in the rat. Unpublished report No. MAK/095/PYR from Life Science Research Israel Ltd, Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Davies, R.E. & Kynoch, S.R. (1970) Acute subcutaneous toxicity of Dursban to rats and the effect of treatment with 1. Atropine sulphate 2. Atropine sulphate and PAM. Unpublished report No. A1A-275 or 3641/70/453 from Huntingdon Research Centre, Huntingdon, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Davies, D.B., Tollett, J.T. & Lomax, L.G. (1985) Chlorpyrifos: A four-week dietary study in the CD-1 mice. Unpublished report No. HET-K-044793-068 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Deacon, M.M., Murray, J.S., Pilny, M.K., Dittenber, D.A., Hanley, T.R., Jr & John, J.A. (1979) The effect of orally administered chlorpyrifos on embryonal and foetal development in mice. Unpublished report No. HET-K-44793-32 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Deacon, M.M., Murray, J.S., Pilny, M.K., Rao, K.S., Dittenber, D.A., Hanley, T.R., Jr & John, J.A. (1980) Embryotoxicity and fetotoxicity of orally administered chlorpyrifos in mice. Toxicol. Appl. Pharmacol., 54, 31-40. Derelanko, M.J. & Hollinger, M.A., eds (1995) CRC Handbook of Toxicology, Boca Raton, FL, CRC Press Inc. Dietz, F.K., Mensik, D.C., Hinze, C.A., Rachunek, B.K. & Taylor, H.W. (1983) Dursban insecticide: Assessment of neonatal survival in a two-generation reproduction study in rats. Unpublished report No. TexasT-K-44793-(45) from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Dishburger, H.J., McKellar, R.L. & Wetters, J.H. (1972a) Residues of chlorpyrifos and 3,5,6trichloro-2-pyridinol in tissues and eggs from chickens fed chlorpyrifos. Unpublished report No. GH-C 555 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Dishburger, H.J., Rice, J.R., McKellar, R.L. & Pennington, J.Y. (1972b) Determination of residues of chlorpyrifos, its oxygen analogue, and 3,5,6-trichloro-2-pyridinol in tissues of cattle fed chlorpyrifos. Unpublished report No. GH-C 566 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Dishburger, H.J., McKellar, R.L., Pennington, J.Y. & Rice, J.R. (1977) Determination of residues of chlorpyrifos, its oxygen analogue, and 3,5,6-trichloro-2-pyridinol in tissues of cattle fed chlorpyrifos. J. Agric. Food Chem., 25, 1325-1329. Dittenber, D.A. (1997) Chlorpyrifos: Evaluation of single oral doses on cholinesterase and neurotoxic esterase inhibition in F344 rats. Unpublished report No. 960036 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Dreher DM (1994a) Chlorpyrifos technical: Acute oral toxicity test in the rat. Unpublished report No. CHA 8-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to WHO by Cheminova Agro A/S, Lemvig, Denmark. Dreher DM (1994b) Chlorpyrifos technical: Acute dermal toxicity (limit test) in the rat. Unpublished report No. CHA 9-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to WHO by Cheminova Agro A/S, Lemvig, Denmark. Dreher DM (1994c) Chlorpyrifos technical: Acute eye irritation test in the rabbit. Unpublished report No. CHA 12-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to WHO by Cheminova Agro A/S, Lemvig, Denmark. Dreher DM (1994d) Chlorpyrifos technical: Acute dermal irritation test in the rabbit. Unpublished report No. CHA 11-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to WHO by Cheminova Agro A/S, Lemvig, Denmark. Dreher DM (1994e) Chlorpyrifos technical: Magnusson & Kligman maximisation study in the guinea pig. Unpublished report No. CHA 12-CYF from Safepharm Laboratories Ltd, Derby, United Kingdom. Submitted to Who by Cheminova Agro A/S, Lemvig, Denmark. Drevenkar, V., Vasilic, Z., Stengl, B., Frobe, Z. & Rumenjak, V. (1993) Chlorpyrifos metabolites in serum and urine of poisoned persons. Chem.-Biol. Interactions, 87, 315-322. Eliason, D.A., Crammer, M.F., von Windeguth, D.L., Kilpatrick, J.W., Suggs, J.E. & Schoof, H.F. (1969) Dursban premise applications and their effect on the cholinesterase levels of spraymen. Mosquito News, 29, 591-595. Emerson, J.L. & Gerbig, C.L. (1970) 91 day toxicology study in beagle dogs treated with 3,5,6-trichloro-2-pyridinol. Unpublished report from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Fikes, J.D., Zacchary, J.F., Parker, A.J. & Beasley, V.R. (1992) Clinical, biochemical, electrophysiologic and histologic assessment of chlorpyrifos induced delayed neuropathy in the cat. Neurotoxicology, 13, 663-678. Fredrick Institute of Plant Protection and Toxicology (1995a) Acute oral toxicity study of chlorpyrifos technical in Swiss albino mice. Unpublished report No. 2739 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1995b) Acute oral toxicity study of chlorpyrifos technical in Wistar rats. Unpublished report No. 2738 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1995c) Acute dermal toxicity study of chlorpyrifos technical in rabbits. Unpublished report No. 2748 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1995d) Irritation of chlorpyrifos technical to mucous membrane in rabbits. Unpublished report No. 2746 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1995e) Primary skin irritation of chlorpyrifos technical in rabbits. Unpublished report No. 2747 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1996a) Acute inhalation study with chlorpyrifos technical in Wistar rats. Unpublished report No. 2968 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1996b) Allergy and skin sensitization potential of chlorpyrifos technical in guinea pigs. Unpublished report No. 2928 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1996c) Mutagenicity evaluation of chlorpyrifos technical in the point-mutation Ames Salmonella typhimurium test system. Unpublished report No. 3049 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1996d) Mutagenicity evaluation of chlorpyrifos technical in the mouse micronucleus assay. Unpublished report No. 3054 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Fredrick Institute of Plant Protection and Toxicology (1996e) Mutagenicity evaluation of chlorpyrifos technical in the mouse bone marrow cytogenic assay. Unpublished report No. 3076 from Fredrick Institute of Plant Protection and Toxicology, Tamil Nadu, India. Submitted to WHO by Ficom Organics, Bombay, India. Furlong, C.E., Richter, R.J., Seidel, S.L. & Motulski, A.G. (1988) Role of genetic polymorphism of human plasma paraoxonase/arylesterase in hydrolysis of the insecticide metabolites chlorpyrifos oxon and paraoxon. Am. J. Hum. Genet., 43, 230-238. Furlong, C.E., Richter, R.J., Seidel, S.L., Costa, L.G. & Motulsky, A.G. (1989) Spectrophotometric assays for the enzymatic hydrolysis of the active metabolites of chlorpyrifos and parathion by plasma paraoxonase/arylesterase. Anal. Biochem., 180, 242-247. Geldmacher-von Mallinckrodt, M. & Diepgen, T.L. (1988) The human serum paraoxonase polymorphism and specificity. Toxicol. Environ. Chem., 18, 79-196. Gerbig,, C.G. & Emerson, J.L. (1970a) Oral median lethal dose (LD50) determination of 3,5,6-trichloro-2-pyridinol in mice. Unpublished report No. HH-240 from Dow Chemical Co., Zionsville, Indianna, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gerbig,, C.G. & Emerson, J.L. (1970b) Oral median lethal dose (LD50) determination of 3,5,6-trichloro-2-pyridinol in the rat. Unpublished report No. HH-239 from Dow Chemical Co., Zionsville, Indianna, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gerbig,, C.G. & Emerson, J.L. (1970c) Determination of oral minimum lethal dose of 3,5,6trichloro-2-pyridinol in beagle dogs. Unpublished report No. HH-253 from Dow Chemical Co., Zionsville, Indianna, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gibson, J.E. (1996) Critical review of allegations associating Dursban with human teratogenicity. Unpublished report No. JEG122396, from Dow Elanco, USA. Submitted by Dow Agrosciences, Indianapolis, Indiana, USA. Glas, R.D. (1977a) Residues of 3,5,6-trichloro-2-pyridinol and 2-methoxy-3,5,6trichloropyridine in tissues from calves fed 3,5,6-trichloro-2-pyridinol. Unpublished report No. GH-C 1053 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Glas, R.D. (1977b) Residues of 3,5,6-trichloro-2-pyridinol and 2-methoxy-3,5,6trichloropyridine in tissues from a calf fed 2-methoxy-3,5,6-trichloro-2-pyridine. Unpublished report No. GH-C 1052 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Glas, R.D. (1977c) Residues of triclopyr, 3,5,6-trichloro-2-pyridinol and 2-methoxy-3,5,6trichloropyridine in bovine tissues from calves fed trichlopyr. Unpublished report No. GH-C 1047 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Glas, R.D. (1981a) The metabolic fate of 14C-chlorpyrifos fed to lactating goats. Unpublished report No. GH-C 1408 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Glas, R.D. (1981b) Identification of 14C-labelled residues in milk from goats fed 14Cchlorpyrifos. Unpublished report No. GH-C 1470 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gollapudi, B.B. (1987) Evaluation of 3,5,6-trichloro-2-pyridinol (TCP) in the rat hepatocyte unscheduled DNA synthesis (UDS) assay. Unpublished report No. TexasT-K-38278-012 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gollapudi, B.B., Linscombe, V.A. & Wilkerson, J.E. (1985) Evaluation of chlorpyrifos in the mouse bone marrow micronucleus test. Unpublished report No. TexasT-K-044793-067 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gollapudi, B.B., Mendrala, A.L. & Linscombe, V.A. (1995) Evaluation of the genetic toxicity of the organophosphate insecticide chlorpyrifos. Mutat. Res., 342, 25-36. Gur, E., Nyska, A. & Waner, T. (1991) Pyrinex technical oncogenicity study in the mouse. Unpublished report from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Isreal. Gutenmann, W.H., St John, L.E., Jr & Lisk, D.J. (1968) Metabolic studies with O,O-diethyl O-(3,5,6-trichloro-2-pyridyl) phosphorothioate (Dursban) insecticide in a lactating cow. Unpublished report from Cornell University, Ithaca, New York, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Gutmann, L. & Bodensteiner, J.B. (1993) Organophosphate-induced delayed polyneuropathy (comment). J. Pediatr., 123, 837. Hanley, T.R., Jr, Zielke, G.J. & Lomax, L.G. (1987a) 3,5,6-Trichloro-2-pyridinol: Oral teratology study in Fischer 344 rats. Unpublished report No. K-038278-011 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Hanley, T.R., Jr, Zielke, G.J. & Lomax, L.G. (1987b) 3,5,6-Trichloro-2-pyridinol: Oral teratology study in New Zealand white Rrabbits. Unpublished report No. K-038278-015 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Hardy, C.J. & Jackson, G.C. (1984) Dursban technical: Acute inhalation toxicity in rats. Unpublished report No. DET 368 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Harling, R.J., Barker, M.H., Buist, D.P., Crook, D., Dawe, I.S. & Anderson, A. (1989a) Chlorpyrifos oral dose range finding toxicity study in beagle dogs (final report -- repeated daily dosage for 4 weeks). Unpublished report No. MBS 30/88675 from Huntingdon Research Centre Inc, Huntingdon, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Harling, R.J., Barker, M.H., Buist, D.P., Crook, D., Majeed, S., Gopinath, C., Dawe, I.S. & Anderson, A. (1989b) Chlorpyrifos oral toxicity study in beagle dogs (repeated daily dosage for 13 weeks). Unpublished report No. MBS 31/88999 from Huntingdon Research Centre Inc, Huntingdon, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Hazleton Laboratories (1968) Short term (subacute) dietary administration -- Rats organophosphate insecticide Dursban. Final report. Unpublished report No. 174-114 from Hazleton Laboratories, Falls Church, Virginia, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Henck, J.W. & Kociba, R.J. (1980) Three samples of Dursban insecticide: Acute oral toxicity and acute percutaneous absorption potential. Unpublished report No. A1A-130 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Hoberman, A.M. (1998) Developmental neurotoxicity study of chlorpyrifos administered orally via gavage to Crl:CD.BR VAF/Plus presumed pregnant rats. Unpublished report No. K-044793109 from Argus Research Laboratories, Horsham, PA, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Hooser, S.B., Beasley, V.R., Sundberg, J.P. & Harlin, K. (1988) Toxicologic evaluation of chlorpyrifos in cats. Am. J. Vet. Res., 49, 1371-1375. Humbert, R., Adler, D.A., Distechhe, C.M., Hassett, C., Omiecinski, C.J. & Furlong, C.E. (1993) The molecular basis of the human serum paraoxonase activity polymorphism. Nature Genet., 3, 73-76 Jackson, D. & Ogilvie, S.W. (1994a) Chlorpyrifos tech: Acute dermal toxicity (LD50) test in rats. Unpublished report No. 10439 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Jackson, D. & Ogilvie, S.W. (1994b) Chlorpyrifos tech: Primary eye irritation test in rabbits. Unpublished report No. 10441 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Jackson, D. & Ogilvie, S.W. (1994c) Chlorpyrifos tech: Primary skin irritation test in rabbits. Unpublished report No. 10440 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Jackson, D. & Ogilvie, S.W. (1994d) Chlorpyrifos tech: Magnusson-Kligman maximisation test in guinea pigs. Unpublished report No. 10442 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Jai Research Foundation (1996a) Reverse gene mutation study of chlorpyrifos tech (95%) in bacteria by Ames Salmonella/microsome test. Unpublished report No. 5/JRF/MichiganC/96 from Department of Microbiology, Jai Research Foundation, Gujarat, India. Submitted to WHO by Mitsu Industries, Gujarat, India. Jai Research Foundation (1996b) Neurotoxicity study of chlorpyrifos technical to hen. Unpublished report No. 1539/JRF/TOX/96 from Department of Toxicology, Jai Research Foundation, Gujarat, India. Submitted to WHO by Mitsu Industries, Gujarat, India. James, P., Stubbs, A., Parker, C.A., Offer, J.M. & Anderson, A. (1988) The effect of Pyrinex (chlorpyrifos) on reproductive function of two generations in the rat. Unpublished report No. MBS 29/881452 from Huntingdon Research Centre Ltd, Huntingdon, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Jeffrey, M.M., Battjes, J.E. & Eisenbrandt, D.L. (1986) Dursban F (tech): Acute dermal toxicity study in Fischer 344 rats. Unpublished report No. HET-K 044793-083 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Jones, J.R. (1985a) Dursban F: Eye irritation study in the rabbit. Unpublished report No. DET 424 from Hazleton Laboratories Europe, Harrogate, North Yorkshire, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Jones, J.R. (1985b) Dursban F: Primary skin irritation and corrosivity study in the rabbit. Unpublished report No. DET 421 from Hazleton Laboratories Europe, Harrogate, North Yorkshire, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Jones, J.R. (1985c) Dursban F: Delayed contact hypersensitivity study in the guinea pig (Buehler Test). Unpublished report No. DET 475 from Hazleton Laboratories Europe, Harrogate, North Yorkshire, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Kamataki, T. & Neil, R.A. (1976) Metabolism of diethyl A-nitrophenyl phosphorothienate (parathion) by a reconstituted mixed-function oxidase enzyme system: Studies of covalent binding of the sulfur atom. Mol. Pharmacol., 12, 933-944. Kaplan, J.G., Kessler, J., Rosenberg, N., Pack, D. & Schaumburg, H.H. (1993) Sensory neuropathy associated with Dursban (chlorpyrifos) exposure [published erratum appears in Neurology 1994 44, 367]. Neurology, 43, 2193-2196. Kenaga, E.E. (1967) Comments on data concerning cholinesterase effects of Dursban in man following US Public Health Service experimental yellow fever mosquito eradication tests in Florida during 1966 and 1967. Unpublished report No.A1A-394, TK 2-1684 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Kenny, T.J., Coombs, D.W. & Hardy, C.J. (1987) Chlorpyrifos technical acute inhalation toxicity in rats 4-hour exposure. Unpublished report No. MBS 20/87575 from Huntingdon Research Centre, Huntingdon, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Kenny, T.J., Coombs, D.W., Hardy, C.J. & Crook, D. (1988) Chlorpyrifos inhalation toxicity in the rat. 14-Day preliminary dose range finding study. Unpublished report No. MBS 21/871329 from Huntingdon Research Centre Ltd, Huntingdon, United Kingdom. Submitted by Makteshim Chemical Works, Beer-Sheva, Israel. Kilian, D.J., Edwards, H.N., Benge, M.C. & Tabatabai, Z. (1970) Results of human skin exposure to Dowco 179. Unpublished report No. TMD-70-2 from Dow Chemical Co., Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Kisicki, J.C., Seip, C.W. & Combs, M.L. (1999) A rising dose toxicology study to determine the no-observable-effect-levels (NOEL) for erythrocyte acetylcholinesterase (AChE) inhibition and cholinergic signs and symptoms of chlorpyrifos at three dose levels. Unpublished report No. LLC Study ID: DR#K-044793-284 dated 19 April 1999, from MDS Harris, Lincoln, NE, USA. Submitted to WHO by Dow Agrosciences, Indianapolis, Indiana, USA. Kociba, R.J., McCollister, S.B., Keyes, D.G. & Dittenber, D.A. (1985) Supplement to original report entitled 'Results of two-year dietary feeding studies on Dowco 179 in beagle dogs'. Unpublished report No. NBT 3512-44793-18 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Lackenby, F. (1985a) Dursban F and Dursban F (OP1): Acute oral toxicity studies in the rat. Unpublished report No. 4703-50/390 & 591 from Hazleton Laboratories Europe, North Yorkshire, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Lackenby, F. (1985b) Dursban F: Acute dermal median lethal dose in the rat. Unpublished report No. 4206-50/391 from Hazleton Laboratories Europe, Harrogate, North Yorkshire, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Landry, T.D., Streeter, C.M. & Lomax, L.G. (1985) Chlorpyrifos: 2-week vapor inhalation toxicity study in Fischer 344 rats. Protocol. Unpublished report No. A1A-714 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Landry, T.D., Dittenber, D.A., Lomax, L.G., Calhoun, L.L. & Morabito, P. (1986) Chlorpyrifos: 2 week nose-only vapor inhalation exposure study in Fischer 344 rats. Unpublished report No. HET K-44793-81 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Liberacki, A.B., Breslin, W.J., Dittenber, D.A. & Quast, J.F. (1990) Chlorpyrifos: Two week dietary probe study in Sprague-Dawley rats. Unpublished report No. HET K 044793090 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Linscombe, V.A. & Gollapudi, B. (1986) Evaluation of 3,5,6-trtichloro-2-pyridinol (TCP) in the Chinese hamster ovary cell/hypoxanthine-guanine-phosphoribosyl transferase (CHO/HGPRT) forward mutation assay. Unpublished report No. TexasT:K-038278-013 from Dow Chemical Co., HES, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Linscombe, V.A., Mensik, D.C. & Clem, B.A. (1992) Evaluation of chlorpyrifos in an in vitro chromosomal aberration assay utilizing rat lymphocytes. Unpublished report No. TexasT:K 044793-092 from Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Lotti, M., Moretto, A., Zoppellari, R., Dainese, R., Rizzuto, N. & Barusco, G. (1986) Inhibition of lymphocytic neuropathy target esterase predicts the development of organophosphate-induced delayed polyneuropathy. Arch. Toxicol., 59, 176-179. Loveday, K.S. (1987) In vitro chromosomal aberration assay on Pyrinex (chlorpyrifos). Unpublished report No. R4660 from Arthur D. Little Inc., Cambridge, Massachusetts, USA. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Loveday, K.S., Findlen, K.M.B. & Yadlon, S. (1987) Evaluation of Pyrinex in the Ames mutagenesis assay. Unpublished report No. 59487-00 from Arthur D. Little Inc., USA. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Ma, T. & Chambers, J.E. (1994) Kinetic parameters of desulfuration and dearylation of parathion and chlorpyrifos by rat liver microsomes. Food Chem. Toxicol., 32, 763-767. Maita, K., Hirano, M., Harada, T., Mitsumori, K., Yoshida, A., Takahashi, K., Nakashima, N., Kitazawa, T., Enomoto, A. & Inui, K. (1988) Mortality, major cause of moribundity aqnd spontaneous tumours in CD-1 mice. Toxicol. Pathol., 16, 340-349. Mattsson, J.L., Wilmer, J.W., Shankar, M.R., Berdasco, N.M., Crissman, J.W., Maurissen, J.P. & Bond, D.M. (1996) Single-dose and 13-week repeated-dose neurotoxicity screening studies of chlorpyrifos insecticide. Food Chem. Toxicol., 34, 393-405. Maurissen, J.P., Shankar, M.R. & Mattsson, J.L. (1996) Chlorpyrifos: Cognitive study in adult Long-Evans rats. Unpublished report No. K-044793-096 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McClintock, M.L. & Gollapudi, B.B. (1989a) Evaluation of chlorpyrifos in the mouse bone marrow micronucleus test. Unpublished report No. TexasT:K 044793-067A from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McClintock, M.L. & Gollapudi, B.B. (1989b) Evaluation of 3,5,6-trichloro-2-pyridinol in the mouse bone marrow micronucleus test. Unpublished report No. K-038278-008A from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McCollister, S.B., Kociba, R.J., Gehring, P.J. & Humiston, C.G. (1971a) Results of two-year dietary feeding studies on Dowco 179 in beagle dogs. Unpublished report No. T35.1244793-18 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McCollister, S.B., Kociba, R.J., Gehring, P.J. & Humiston, C.G. (1971b) Results of two-year dietary feeding studies on Dowco 179 in rats. Unpublished report No. NBT35.12-4479321 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McCollister, S.B., Kociba, R.J. & Keyes, D.G. (1985) Supplement to original report entitled 'Results of two-year dietary feeding studies on Dowco 179 in rats'. Unpublished report No. NBT35.12-44793-21 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. McKellar, R.L., Wetters, J.H. & Dishburger, H.J. (1972) Residues of chlorpyrifos and 3,5,6trichloro-2-pyridinol in tissues of swine fed chlorpyrifos. Unpublished report No. GHC549 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Mendrala, A.L. (1985) Evaluation of chlorpyrifos in the Chinese hamster ovary cellhypoxanthine (guanine) phosphoribosyl transferase (CHO/HGPRT) forward mutation assay. Unpublished report No. HET-K-044793-072 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Mendrala, A.L. & Dryzaga, M.D. (1986) Evaluation of chlorpyrifos in the rat hepatocyte unscheduled DNA synthesis (UDS) assay. Unpublished report No. HET K 044793-073 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Moretto, A. & Lotti, M. (1998) Poisoning by organophosphorusinsecticides and sensory neuropathy. J. Neurol. Neurosurg. Psych., 64, 463-468. Mortensen, S.R., Chanda, S.M., Hooper, M.J. & Padilla, S. (1996) Maturational differences in chlorpyrifos-oxonase activity may contribute to age-related sensitivity to chlorpyrifos. J. Biochem. Toxicol., 11, 279-287. Moser, V.C. (1995) Comparisons of the acute effects of cholinesterase inhibitors using a neurobehavioral screening battery in rats. Neurotoxicol. Teratol., 17, 617-625. Moser, V.C. & Padilla, S. (1998) Age- and gender-related differences in the time course of behavioral and biochemical effects produced by oral chlorpyrifos in rats. Toxicol. Appl. Pharmacol., 149, 107-119. Muscarella, D.E., Keown, J.F. & Bloom, S.E. (1984) Evaluation of the genotoxic and embryotoxic potential of chlorpyrifos and its metabolites in vivo and in vitro. Environ. Mutag., 6, 13-23. Nakatsugawa, T. (1992) Hepatic disposition of organophosphorus insecticides: A synthesis of in vitro, in situ and in vivo data. In: Chambers, J.E., Levi, P.E. & Levi, A.P., eds, Organophosphates, San Diego, Academic Press, Chapter 10. Newton, P.E. (1988a) A 5-day nose-only inhalation toxicity study of chlorpyrifos technical (Pyrinex) in the rat. Unpublished report from Bio/Dynamics Inc., East Millstone, NJ, USA. Submitted by Makteshim Chemical Works, Beer-Sheva, Israel. Newton, P.E. (1988b) A thirteen week nose-only toxicity study of chlorpyrifos technical (Pyrinex) in the rat. Unpublished report No. R-4750 from Bio/dynamics Inc., East Millstone, NJ, USA. Makteshim Chemical Works, Beer-Sheva, Israel. Nissimov, S. & Nyska, A. (1984a) Pyrinex tech: Acute oral toxicity in the rat. Unpublished report No. MAK/056/PYR from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Nissimov, S. & Nyska, A. (1984b) Pyrinex tech: Acute dermal toxicity in rabbits. Unpublished report No. MAK/059/PYR from Life Science Research Israel Inc., Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Nolan, R.J. (1997) Proposed reference dose for acute and chronic exposure to chlorpyrifos based on the criteria described by the Acute Cholinesterase Risk Assessment Task Force and the available animal and human data. Unpublished report No. K-044793-110 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Nolan, R.J., Rick, D.L., Freshour, N.L. & Saunders, J.H. (1982) Chlorpyrifos: pharmacokinetics in human volunteers following single oral and dermal doses. Unpublished report No. HEB-DR-0043-4946-4 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Nolan, R.J., Rick, D.L., Freshour, N.L. & Saunders, J.H. (1984) Chlorpyrifos: pharmacokinetics in human volunteers. Toxicol. Appl. Pharmacol., 73, 8-15. Nolan, R.J., Streeter, C.M. & Kastl, P.E. (1986) Chlorpyrifos: Absorption by female Fischer 344 rats exposed for 6 hours to vapours of chlorpyrifos in a nose-only or a whole-body inhalation chamber. Unpublished report No. HET K-044793-82 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Nolan, R.J., Dryzga, M.D., Landenberger, B.D. & Kastl, P.E. (1987) Chlorpyrifos: Tissue distribution and metabolism of orally administered 14C labelled chlorpyrifos in Fischer 344. Unpublished report No. HET K-044793-(76) from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Nostrandt, A.C., Padilla, S. & Moser, V.C. (1997) The relationship of oral chlorpyrifos effects on behaviour, cholinesterase inhibition, and muscarinic receptor density in rat. Pharmacol. Biochem. Behav., 58, 15-23. Omenn, G.S. (1987) The role of genetic differences in human susceptibility to pesticides. In: Costa, L.G., Galli, C.L. & Murphy, S.D., eds, Toxicology of Pesticides: Experimental, Clinical and Regulatory Perspectives, Berlin, Springer-Verlag. Osterloh, J., Lotti, M. & Pond, S.M. (1983) Toxicologic studies in a fatal overdose of 2,4-D, MCPP, and chlorpyrifos. J. Anal. Toxicol., 7, 125-129. Ouelette, J.H., Dittenber, D.A., Kociba, R.J. & John, J.A. (1983a) Chlorpyrifos: Oral teratology probe study in rats. Unpublished report No. HET K-44793-(46) from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Ouelette, J.H., Dittenber, D.A., Kloes, P.M. & John, J.A. (1983b) Chlorpyrifos: Oral teratology study in Fischer 344 rats. Unpublished report No. HET K-44793-(47) from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Pennington, J.Y. & Edwards, N.H. (1971) Comparison of cholinesterase depression in humans and rabbits following exposure to chlorpyrifos. Unpublished report no. TA-477 from Dow Chemical Company USA. Submitted to WHO by Dow Agrosciences, Indianapolis, Indiana, USA. Phillips, J.E. & Lomax, L.G. (1989) 1% aqueous dilution of Dursban acute aerosol inhalation toxicity study in Fischer 344 rats. Unpublished report No. M-004803-004A from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Poole, D.C., Simmon, V.F. & Newell, C.W. (1977). In vitro mutagenic activity of fourteen pesticides. Toxicol. Appl. Pharmacol., 41, 196. Pope, C.N. & Chakraborti, T.K. (1992) Dose related inhibition of brain and plasma cholinesterase in neonatal and adult rats following sub-lethal organophosphate exposure. Toxicology, 73, 35-43. Pope, C.N., Chakraborti, T.K., Chapman, M.L., Farrar, J.D. & Arthun, D. (1991) Comparison of in vivo cholinesterase inhibition in neonatal and adult rats by three organophosphorothioate insecticides. Toxicology, 68, 51-61. Pope, C.N., Chakraborti, T.K., Chapman, M.L. & Farrar, J.D. (1992) Long-term neurochemical and behavioral effects induced by acute chlorpyrifos treatment. Pharmacol. Biochem. Behav., 42, 251-256. Richardson, R.J., Moore, T.B., Kayyali, U.S. & Randall, J.C. (1993a) Chlorpyrifos: Assessment of potential for delayed neurotoxicity by repeated dosing in adult hens with monitoring of brain acetylcholinesterase, brain and lymphocyte neurotoxic esterase and plasma butyrylcholinesterase activities. Fundam. Appl. Toxicol., 21, 89-96. Richardson, R.J., Moore, T.B., Kayyali, U.S., Fowke, J.H. & Randall, J.C. (1993b) Inhibition of hen brain acetylcholinesterase and neurotoxic esterase by chlorpyrifos in vivo and kinetics of inhibition by chlorpyrifos oxon in vitro: Application to assessment of neuropathic risk. Fundam. Appl. Toxicol., 20, 273-279. Roberts, N.L., Phillips, C.N.K., Gopinath, C., Begg, S., Anderson, A. & Dawe, I.S. (1987) Acute delayed neurotoxicity study with chlorpyrifos in the domestic hen. Unpublished report No. MBS 16/87764 from Huntingdon Research Centre Ltd, Huntingdon, United Kingdom. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Ross, D.B. & Roberts, N.L. (1974) The acute oral toxicity (LD50) of Dursban (DOWCO 179) to the chicken (DWC/234/74877). Unpublished report from Huntingdon Research Centre, Huntingdon, United Kingdom. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Rowe, L.D., Warner, S.D. & Johnston, R.V. (1978) Acute delayed neurotoxicologic evaluation of chlorpyrifos in white Leghorn hens. Unpublished report No. A1A-715 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Rubin, Y.,. Gal, N., Waner, T. & Nyska, A. (1987a) Pyrinex. Teratogenicity study in the rat. Unpublished report No. MAK/101/PYR from Life Science Research Israel Ltd, Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Rubin, Y., Nyska, A. & Waner, T. (1987b) Pyrinex. Pyrinex teratogenicity study in the rabbit. Unpublished report No. MAK/103/PYR from Life Science Research Israel Ltd, Ness Ziona, Israel. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Scortichini, B.H., Lomax, L.G., Wolfe, E.L. & Hanley, T.R., Jr (1986a) 3,5,6-Trichloro-2pyridinol: Oral teratology probe study in Fischer 344 rats. Unpublished report No. HETK-038278-006 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Scortichini, B.H., Lomax, L.G., Wolf, E.L. & Hanley, T.R., Jr (1986b) 3,5,6-Trichloro-2pyridinol: Oral teratology probe study in New Zealand white rabbits. Unpublished report No. HET-K-038278-007 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Shankar, M.R., Bond, D.M. & Crissman, J.W. (1993) Chlorpyrifos: 13-week neurotoxicity study in Fischer 344 rats. Unpublished report No. K-044793-094 from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Sharp, L.D. & Warner, S.D. (1968) The clinical toxicity of Dursban in the dog after multiple applications of an aerosol formulation. Unpublished report from Dow Chemical Co., USA. Submitted to WHO by Dow Agrosciences, Indianapolis, Indiana, USA. Shemesh, I., Bourvin, A., Gold, D. & Kutscherowsky, M. (1988) Chlorpyrifos poisoning treated with ipratropium and dantrolene: a case report. J. Toxicol. Clin. Toxicol., 26, 495-498. Sherman, J.D. (1996) Chlorpyrifos (Dursban)-associated birth defects: Report of four cases. Arch. Environ. Health, 51, 5-8. Sherman, J.D. & Herrick, R.B. (1973) Fly control and chronic toxicity from feeding Dursban (O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphorothioate) to laying hens. Poultry Sci., 52, 741-747. Sherman, J.D., Herrick, R.B., Ross, E. & Chang, M.T.Y. (1967) Further studies on the acute and sub-acute toxicity of insecticides to chicks. Toxicol. Appl. Pharmacol., 11, 49-67. Shirasu, Y., Moriya, M. & Ohta, T. (1980) Mutagenicity test of chlorpyrifos in bacteria. Unpublished report No. GHF-R-47 from Dow Chemical Pacific, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Shurdut, B.A., Chen, W.L., Burns, C.J., McCormick, R.A., Nolan, R.J. & Racke, K.D. (1997) Critical assessment of report entitled 'Review of chlorpyrifos poisoning data' (by J. Blondell and V. Dobozy, January 14, 1997). Unpublished report No. GH-C 4359, by Dow Chemical Co., USA. Submitted to WHO by Dow Agrosciences, Indianapolis, Indiana, USA. de Silva, H.J., Sanmuganathan, P.S. & Senanayake, N. (1994) Isolated bilateral recurrent laryngeal nerve paralysis: A delayed complication of organophosphorus poisoning. Hum. Exp. Toxicol., 13, 171-173. Smith, G.N., Watson, B.S. & Fischer, F.S. (1967) Investigations on Dursban insecticide. Metabolism of [36Cl] O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphorothiate in rats. Agric. Food Chem., 15, 132-138. Smolen, A., Eckerson, H.W., Gan, K.N., Hailat, N. & LaDu, B.N. (1991) Characteristics of the genetically determined allozymic forms of human paraoxonase/artylesterase. Drug Metab. Disposition, 19, 107-112 Sobti, R.C., Krishan, A. & Pfaffenberger, C.D. (1982) Cytokinetic and cytogenetic effects of some agricultural chemicals on human lymphoid cells in vitro: Organophosphates. Mutat. Res., 102, 89-102. Song, X., Seidler, F.J., Saleh, J.L., Zhang, J., Padilla, S. & Slotkin, T.A. (1997) Cellular mechanisms for developmental toxicity of chlorpyrifos: Targeting the adenylyl cyclase signaling cascade. Toxicol. Appl. Pharmacol., 145, 158-174. Stevenson, G.T. (1963) An LD50 toxicity study of Dursban in Leghorn chickens. Unpublished report from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Stevenson, G.T. (1966a) A single oral dose LD50 toxicity study of Dursban in broiler type chickens. Unpublished report No. A1A-364 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Stevenson, G.T. (1966b) A neurotoxicity study of Dursban in laying hens. Unpublished report No. A1A-390 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Stevenson, G.T. (1967) A single oral dose LD50 toxicity study of Dursban in turkey/poultry. Unpublished report No. A1A-362 from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Streeter, C.M., Lomax, L.G., Landry, T.D. & Dittenber, D.A. (1987) Chlorpyrifos: 2-week whole-body vapor inhalation toxicity study in Fischer 344 rats. Unpublished report No. HET K 044793-078 from Dow Chemical Co., Midland, Michigan, USA Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Sultatos, L.G. & Murphy, S.D. (1983) Hepatic microsomal detoxification of the organophosphates paraoxon and chlorpyrifos oxon in the mouse. Drug Metab. Disposition, 11, 232-238. Szabo, J.R., Young, J.T. & Granjean, M. (1988) Chlorpyrifos: 13-week dietary toxicity study in Fisher 344 rats. Unpublished report No. TexasT: K-044793-071 from Lake Jackson Research Center, Health and Environmental Sciences, Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Taylor, M.L. & Olson, K.J. (1963) Toxicological properties of O,O-diethyl-O-3,5,6trichloro-2-pyridyl phosphorothioate. Unpublished report. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Thompson, D.J., Gerbig, C.C. & Warner, S.D. (1971) Three generation reproduction and teratology study in the rat following prolonged dietary exposure to Dursban O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphorothioate. Unpublished report from Dow Chemical Co., USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Tu, A.S. (1987) CHO/HGPRT in vitro mammalian cell mutation assay on Pyrinex (chlorpyrifos). Unpublished report No. 59487 from Arthur D. Little Inc., Cambridge, Massachusetts, USA. Submitted to WHO by Makteshim Chemical Works, Beer-Sheva, Israel. Warner, S.D., Gerbig, C.G., Strebing, R.J. & Molello, J.A. (1980) Results of a two-year toxicity and oncogenic study of chlorpyrifos administered to CD-1 mice in the diet. Unpublished report from Dow Chemical Co., Indianapolis, Indiana, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Whitney, K.D., Seidler, F.J. & Slotkin, T.A. (1995) Developmental neurotoxicity of chlorpyrifos: Cellular mechanisms. Toxicol. Appl. Pharmacol., 134, 53-62. WHO (1999) Recommended Classification of Pesticides by Hazard and Guidelines to Classification 1998-1999 (WHO/PCS/98.21/Rev. 1), Geneva, International Programme on Chemical Safety. Wilmer, J.W. (1992) Acute neurotoxicity study in Fischer 344 rats. Unpublished report No. K-044793-093B from Dow Chemical Co., Midland, Michigan, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Wilson, J.A. & MacBeth, D. (1994) Chlorpyrifos tech: Acute oral toxicity (LD50) test in rats. Unpublished report No. 10521 from Inveresk Research International, Tranent, Scotland. Submitted to WHO by Luxembourg Industries, Tel Aviv, Israel. Wolcott, R.M., Vaughn, W.K. & Neal, R.A. (1972) Comparison of the mixed function oxidase-catalyzed metabolism of a series of dialkyl p-nitrophenyl phosphorothionates. Toxicol. Appl. Pharmacol., 22, 213-220. Yang, R.S.H., Hodgson, E. & Dauterman, W.C. (1971) Metabolism in vitro of diazinon and diazoxon in rat liver. J. Agric. Food Chem., 19, 10-13. Young, J.T. & Grandjean, M. (1988) Chlorpyrifos: 2 year dietary chronic toxicity-oncogenicity study in Fischer 344 rats. Unpublished report No. K-044793-079 from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Zempel, J.A. & Bruce, R.J. (1986) 3,5,6-Trichloro-2-pyridinol: Evaluation in the Ames Salmonella/mammalian-microsome mutagenicity assay. Unpublished report No. TexasT:K038278-010 from Lake Jackson Research Center, Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA. Zempel, J.A., Rachunek, B.L. & Szabo, J.R. (1987) 3,5,6-Trichloro-2-pyridinol: Results of a one-year dietary study in male and female beagle dogs. Unpublished report No. TexasT:K038278-009 from Dow Chemical Co., Freeport, Texas, USA. Submitted to WHO by Dow AgroSciences, Indianapolis, Indiana, USA.
See Also: Toxicological Abbreviations Chlorpyrifos (ICSC) Chlorpyrifos (WHO Pesticide Residues Series 2) Chlorpyrifos (WHO Pesticide Residues Series 5) Chlorpyrifos (Pesticide residues in food: 1977 evaluations) Chlorpyrifos (Pesticide residues in food: 1981 evaluations) Chlorpyrifos (Pesticide residues in food: 1982 evaluations) Chlorpyrifos (Pesticide residues in food: 1983 evaluations)