Loading...
HomeMy WebLinkAboutDEQ-CFW_00000137Available online at www.sciencedirect.com ///--- Toxicology SCIENCE DIRECT- and ApIt plied ���•� Pharmacology ELSEVIER Toxicology and Applied Pharmacology 198 (2004) 231-241 www.elsevier.com/locate/ytaap Review The developmental toxicity of perfluoroalkyl acids and their derivatives Christopher Lau, a,* John L. Butenhoff,b and John M. Rogersa "Reproductive Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA 63M Medical Department, St. Paul, MN 55144, USA Received 20 August 2003; accepted I 1 November 2003 Available online 8 April 2004 Abstract Perfluoroalkyl acids such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) have applications in numerous industrial and consumer products. Although the toxicology of some of these compounds has been investigated in the past, the widespread prevalence of PFOS and PFOA in humans, as demonstrated in recent bio-monitoring studies, has drawn considerable interest from the public and regulatory agencies as well as renewed efforts to better understand the hazards that may be inherent in these compounds. This review provides a brief overview of the perfluoroalkyl chemicals and a summary of the available information on the developmental toxicity of the eight -carbon compounds, PFOS and PFDA. Although the teratological potentials of some of these chemicals had been studied in the past and the findings were generally unremarkable, results from recent postnatal studies on developmental and reproductive indices have prompted consideration of their relevance to human health risk. Based on current understanding of the developmental effects of PFOS and PFOA in rodents, several avenues of research are suggested that would further support the risk assessment of these perfluorinated organic chemicals. © 2004 Elsevier Inc. All rights reserved. Keywords: PFOS; PFOA; PFBS; PFDA; Developmental toxicity; Rodents; Review Environmental exposures of perfluoroalkyl acids In recent years, perfluoroalkyl acids have been identi- fied at low parts per billion (ppb) concentrations in samples of human serum taken from the general popula- tion as well as certain wildlife samples (Giesy and Kannan, 2001, 2002; Hansen et al., 2001- Hoff et al., 2003a, 2003b; Kannan et al., 2001a, 2001b, 2002a, 2002b, 2002c, 2002d; Martin et al., 2004; Olsen et al., 2002, 2003a, 2004; Sottani and Minoia, 2002). The Abbreviations: POSF, perfluorooctane sulfonyl fluoride; PFOS, per- fluorooctane sulfonate; PFBS, perfluorobutane sulfonate; PFOA, perfluor- ooctanoic acid; PFDA, perfluorodecanoic acid. 'a This document has been reviewed in accordance with the U.S. Environmental Protection Agency policy and approved for publication. Approval does not signify that the contents reflect the views of the Agency, nor does mention of trade names or commercial products constitute endorsement or recommendation for use. * Corresponding author, Reproductive Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Mail Drop 67, Research Triangle Park, NC 27711. Fax: +1-919-541-4017. E-mail address: lau.christopher@epa.gov (C. Lau). 0041-008X/$ - see front matter © 2004 Elsevier Inc. All rights reserved. doi:10.1016/j. taap.2003.11.031 presence of organic fluoride in humans was actually first reported by Taves (1968) and Shen and Taves (1974) over 30 years ago and the organic form was originally suggested to be perfluorooctanoate (PFDA) or, possibly, perfluorooctane sulfonate (PFOS) (Taves et al., 1976). It is only in the past several years that significant advances in analytical chemistry have enabled routine detection of individual perfluoroalkyl acids in various matrices in the ppb or lower range by high-performance liquid or gas chromatography followed by electrospray tandem mass spectrometry (ES/MS/MS) (Hansen et al., 2001; Hebert et al., 2002; Martin et al., 2002; Sottani and Minoia, 2002). Equipped with such improvement in detection sensitivity, Olsen et al. (1999, 2001, 2003b, 2003c) reported mean serum PFOS levels of 1-2 ppm (range 0.1-13 ppm) and mean PFOA serum concentrations (Olsen et al., 1998, 2000, 2003b) of approximately 5 ppm (range <0.1-114 ppm) in 3M fluorochemical production workers. A broad survey of individual blood samples from adult Red Cross blood donors (Olsen et al., 2003a), children from a clinical trial (Olsen et al., 2002), and elderly subjects enrolled in a longitudinal study (Olsen et al., 2004) DEQ-CFW 00000137 232 C. Lau et al. / Toxicology and Applied Pharmacology 198 (2004) 231-241 indicated geometric mean serum concentrations of PFOA, perfluorohexanesulfonate (PFHS), and PFOS in these populations of approximately 4-5, 2-5, and 30-40 ppb, respectively. More recently, Olsen et al. (2003d) reported liver concentrations of PFOS ranging from <4.5 to 57 ppb in human organ donors from the general population. The hepatic levels of PFOA in most of the organ donor subjects were below current detection limits. Several recent studies by Giesy and Kannan (2001, 2002), Kannan et al. (2001a, 200Ib, 2002a, 2002b, 2002c, 2002d), and others (Hoff et al., 2003a, 2003b; Martin et al., 2004) have reported detection of PFOS in a variety of wildlife species including fresh water and marine mammals, fish, birds, and shellfish. Although distribution of the chemical appears to be widespread in the northern hemisphere, including remote locations in the Arctic and North Pacific Oceans, concentrations of PFOS in these animals are generally greater in the more popu- lated and industrial regions. These investigators and others (Martin et al., 2003b) also suggested that PFOS can be bio-magnified in the top levels of the food chain. Although PFOA was also found occasionally in the serum and liver of various wildlife species at different geographic loca- tions, concentrations are generally lower than those of PFOS (Kannan et al., 2002a, 2002b, 2002d). The prevalence of these perfluoroalkyl acids in humans and wildlife populations has thus drawn considerable attention from regulatory agencies (US EPA, 2003a, 2003b) and the public (Renner, 2001). Largely because of the observed broad distribution and persistence of PFOS and PFOA in humans and the environment, 3M Company (one of the chief manufacturers) phased out production of these chemicals by the end of 2002; although other perfluoroalkyl acid products are poised to take up the void left by PFOS in the commercial markets. In addition, the Center of Disease Control and Prevention (CDC) has recently nominated PFOS and PFOA to be included in their National Health and Nutrition Examination Survey (NHANES) for future exposure reports, in order to provide a better assessment of the distribution of these chemicals among human populations. It is therefore timely to review the toxicology of these perfluorochemicals, and this paper will focus on the current understanding of developmental toxicity of these compounds. A search of the extant literature revealed only limited peer -reviewed publications on this topic. Because the issues related to developmental toxicity of these chemicals arise from recently completed studies, manuscripts are still in preparation and substantial toxicity data are available only in the form of reports submitted to the U.S. EPA and other regulatory agencies. Information from the latter source is included in this review and details of these studies are generally accessible to the public upon request from these agencies. As a class, these perfluorochemicals share unique phys- ical and chemical properties that are related to their com- mercial applications as well as potentials for toxicity. A clear understanding of these properties will be useful for toxicologists and risk assessors in evaluating the toxic potentials of these compounds. Thus, although the focus of this review is directed toward the developmental toxicity of perfluoroalkyl acids, a brief background on their chem- istry, environmental distribution, and pharmacokinetic pro- files is also provided for the readers' purview. Chemistry, production, and uses of perfluoroalkyl acids Organic fluorochemicals are compounds in which one or more carbon —hydrogen (C-H) bonds are replaced by carbon —fluorine (C-F) bonds. Fluorine is one of the most reactive elements in ionic form and one of the most stable in bound form. Hence, the fully fluorinated hydrocarbons, or perfluorocarbons, are stable in air even at high temper- atures (in excess of 150 °C), nonflammable, not readily degraded by strong acids, alkalis or oxidizing agents, and not subject to photolysis. The unique stability of these chemicals thus renders them practically nonbiodegradable and thus, very persistent in the environment (Key et al., 1997, 1998; Prescher et al., 1985). Naturally occurring fluorinated organic chemicals are rare in biological systems due to the high energy required to form or break the C-F bonds, and all known biologically produced fluorinated organics contain only one fluorine atom (review: Key et al., 1997). However, partially or fully fluorinated (perfluorinated) organic molecules can be syn- thesized in the laboratory and in full-scale manufacturing operations. One method for production employs the Simons Elec- trochemical Fluorination (ECF) process that was discovered in 1937. The ECF process is based on the reaction between organic feedstocks such as 1-octanesulfonyl fluoride (C8H17SO2F) and anhydrous hydrogen fluoride (HF), which is fueled by an electrical current, causing the hydrogen atoms on the carbon backbone to be replaced by fluorine atoms, with resultant products such as perfluorooctane sulfonyl fluoride (POSF, C$F17SO2F). However, during this process, fragmentation and rearrangement of the carbon skeleton can occur, producing fluorinated organic molecules of various carbon chain lengths (less than eight) and a mixture of linear, branched, and cyclic isomers of the chemical. Typically 70-85% of the mixture was linear and 15-301X0 was a mixture of branched isomers. 3M has produced fluorochemicals by the ECF process for industrial and consumer use since the 1950s, although this manufac- turer has now phased out essentially all production of perfluorooctanyl (PFOA and POSF-based) chemistries. An alternative production method for the functionalized perfluoroalkyl acids involves telomerization of tetrafluoro- ethylene units, and many manufacturers employ this tech- nology to produce materials that, in some cases, have similar properties to the ECF-based materials. In this pro- DEQ-CFW 00000138 C. Lau et al. / Toxicology and Applied pharmacology 198 (2004) 231-241 cess, a telogen such as pentafluoroethyl iodide (F(CF2CF2)I) polymerizes with terafluoro ethylene (CF2CF2) to form telomer intermediates of desired carbon chain length (typ- ically up to 12); the resultant telomer iodides are then reacted with ethylene via free radical addition. The telome- rization process thus yields straight -chain alcohols (F(CF2CF2) CH2CH2OH) that can be converted into final products for commercial application. In general, these telomer-derived products may consist of a distribution of various carbon chain lengths. Hence, the fluorinated organic chemicals found in the environment represent a family of compounds of various sizes and isomeric forms. The high ionization potential and low polarizability of fluorine lead to weak inter- and intramolecular interactions that are reflected by the extremely low surface tension of the perfluoroalkyl acids. The partitioning behavior of perfluor- oalkyl acids is also unique; when they are mixed with water and hydrocarbons, three immiscible phases are formed, indicating the hydrophobic and oleophobic nature of these compounds. By attaching a charged moiety such as carbox- ylic acid, sulfonic acid, or phosphate to the perfluorinated chain, the molecule becomes more water soluble due to the hydrophilic nature of the added functional group. Together, the physical properties of the functionalized perfluoroalkyl acids render these compounds ideal surfactants (Kassa, 2001). Although all amphoteric perfluoroalkyl acids share some surfactant properties, the eight -carbon chemicals such as POSF-based materials, PFOS and PFOA have been most effective in commercial uses. Indeed, POSF- and telomer- related products are found in over 200 known industrial and consumer applications ranging from water-, soil-, and stain - resistant coatings for clothing fabrics, leather, upholstery, and carpets, to oil -resistant coatings for paper products approved for food contact, electroplating, and electronic etching bath surfactants, photographic emulsifier, aviation hydraulic fluids, fire -fighting foams, floor polishes, and insecticide formulations (Renner, 2001; Seacat et al., 2002). According to 3M, the global production of POSF for 2000 (the last year of active production) was estimated at about 3,545 metric tons, and in 2001, the worldwide production was 175 metric tons (3M, 2003). Information on PFOA production by various manufacturers is not readily available, but can be estimated in excess of 500 metric tons per year. In commercial applications, POSF was converted into other intermediates such as acids, alcohols, and sulfona- mides for further polymerization and csterification, but some of these compounds and their intermediates have the potential to break down metabolically or in the environment to PFOS as an end -stage metabolite and product. PFOA (primarily ammonium salt) can be used as a surfactant and an emulsifier in the production of polytetrafluoroethylene as well as other fluoropolymers and fluoroelastomers. Telomer-based molecules may have the potential to degrade metabolically to perfluorinated acids (Hagen et al., 1981). Most of the polymers based on fluorochemistry tend to be 233 relatively stable and resistant to degradation. Current under- standing of fluorochemistry suggests that PFOS and PFOA are substantially different compounds, and that toxicological evaluation of these two chemicals should be considered separately. Distribution of perfluoroalkyl acids in the environment POSF is a volatile liquid at normal temperature and pressure; however, PFOS and PFOA have very low volatil- ity and vapor pressure. Certain sulfonamide intermediary compounds derived from POSF, such as alkyl amides, alkyl alcohols, and alkyl acids, may sublime (3M, 2003; Schal, 1992). However, little is known about the transport and fate of these fluorochemicals in the environment. Recent studies reported <0.017 to 2,260 µg/1 for perfluoroalkane sulfonate (C-6—C-8) and <0.009 to 11.3 µg/1 for PFOA in the surface water of a Canadian tributary after an accidental release of fire -fighting foams (Moody et al., 2002). Both PFOS and PFOA were detected in water samples from the Tennessee River near a 3M fluorochemical manufacturing site (Hansen et al., 2002). Concentrations ranging from a low of approx- imately 17 ppt (below quantitation limit) upstream of the facility to a high of 144 ppt downstream of the site for PFOS, and a low of 25 ppt to a high of approximately 600 ppt for PFOA were reported. Monitoring of drinking water sources near a production plant in West Virginia indicated the presence of PFOA at below 3 µg/l, substantially below the interim risk screening of 150 µg/1 established by the State and US EPA region (West Virginia Department of Environmental Protection, 2002). Martin et al. (2002) also reported airborne levels of certain perfluorooetanesulfona- mide-based chemistries and telomer alcohols ranging from approximately 10 to 400 pg/m3 in Ontario (Toronto and Long Point), Canada. Pharmacokinetics of PFOS and PFOA PFOS and PFOA have been observed in retired fluro- chemical workers to have mean elimination half-lives of several years (Burris et al., 2002). PFOS is readily absorbed and distributed primarily in the serum and liver (most likely through enterohepatic circulation), but poorly eliminated (Burris et al., 2002; Johnson and Ober, 1979; Johnson et al., 1979, 1984; Nokcr and Gorman, 2003a; Seacat et al., 2002). The elimination half-life of PFOS in rats and monkeys has been estimated at >90 days for male rats (Johnson et al., 1979) and approximately 100--200 days for male and female cynomolgus monkeys (Noker and Gorman, 2003a; Seacat et al., 2002). PFOA is also readily absorbed (Kemper and Jepson, 2003); however, notable differences in elimination between species have been reported (Kennedy et al., 2004; Kojo et al., 1986; Kudo and Kawashima, 2003; US EPA, 2003a). In the rat, a major DEQ-CFW 00000139 234 C. Lau et al. / Toxicology and Applied Pharmacology 198 (2004) 231-241 gender difference exists in the elimination of PFOA, with half-lives estimated at 1.9-24 h for females and 4.4-9 days for males (Hanhijarvi et al., 1982; Kemper and Jepson, 2003; Kudo et al., 2002; Ophaug and Singer, 1980; 1987; Vanden Heuvel et al., 1991a; Ylinen et al., 1990). The gender difference in rats appears to be under steroid hor- monal control (Kawashima et al., 1991; Kudo et al., 2002; I-lanhijarvi et al., 1982; Vanden Heuvel et al., 1991a, 1992; Ylinen et al., 1989). Gibson and Johnson (1983) reported that hormonal changes during pregnancy did not appear to alter the rate of PFOA elimination. A smaller sex -related difference in PFOA elimination was found in beagle dogs (Hanhijarvi et al., 1988), with plasma half-lives of 202 and 473 h estimated for females and males, respectively. How- ever, it is noteworthy that striking gender differences in PFOA elimination have not been observed in primates and humans (Burris et al., 2002; Noker and Gorman, 2003b), and appear to be unlikely in the mouse (Sohlenius et al., 1992; Uy-Yu et al., 1990; US EPA, 2003a), Toxicity of perfluorooctane sulfonate The potential toxicities of PFOS in monkeys (Seacat et al., 2002), rats (Austin et al., 2003; Seacat et al., 2003; Thibodeaux et al., 2003), fish (Hoff et al., 2003b: Martin et al., 2003a, 2003b), and humans (Olsen et al., 1999, 2003b, 2003c) have been characterized and summarized in recent reviews (3M, 2003; US EPA, 2003b; link to OECD: http://www.oecd.org/dataoeedi23/18/2382880.pclo. In brief, subchronic exposure to PFOS led to significant weight loss accompanied by hepatotoxicity and reductions of serum cholesterol and thyroid hormones. Teratological studies have been conducted in rat, rabbit, and mouse with PFOS (potassium and lithium salts) (Case et al., 2001b; Christian Rat 0 5 10 15 20 Postnatal Age (days) et al., 1999a; Gortner, 1980; Henwood et al., 1994; Thibodeaux et al., 2003; Wetzel, 1983). The findings are in agreement between laboratories and across species examined, and are generally unremarkable when maternal effects are taken into consideration. Observed developmen- tal effects include reduction of fetal weight, cleft palate, anasarca (edema), delayed ossification of bones (sternebrae and phalanges), and cardiac abnormalities (ventricular septal defects and enlargement of the right atrium). None- theless, it should be noted that a preponderance of these structural abnormalities was found in the highest PFOS dose groups, where significant reductions of weight gain and food consumption were also observed in the pregnant dams. On the other hand, maternal malnutrition is not likely the sole factor accounting for the induction of birth defects. Indeed, Thibodeaux et al. (2003) have shown that equivalent or higher incidence of malformations was seen in the mouse, compared to the rat; yet, the deficits of maternal weight gain and food consumption in the mouse were much less extensive than those in the rat. When rodent dams exposed to PFOS throughout preg- nancy were allowed to give birth, dose -dependent deleteri- ous effects were seen in the newborns (Lau et al., 2003; Fig. ). As a reference to body burdens of the chemical, serum levels of PFOS in the pregnant dams at term and in the newborns are summarized in Table 1. All rat pups were born alive, pink, and active. However, in the highest dose (10 mg/ kg) group evaluated, the neonates became pale, inactive, and moribund within 30-60 min, and all died soon after- ward. In the next highest dose group (5 mg/kg), the neonates also became moribund but survived for a longer period of time (8-12 h). Over 95% of these animals did not survive the first day of postnatal life, and only a few pups reached puberty. Survival improved with lower PFOS exposure, and was about 50% at 3 mg/kg. The morbidity and mortality of Mouse 0 5 10 15 20 25 Postnatal Age (days) -a-Control -0-1 mg/kg t2 mg/kg -v-3 mg/kg --F-5 mg/kg -a-10 mg/kg -*-15 mg/kg -*-20 mg/kg Fig. 1. Effects of prenatal exposure to PFOS on postnatal survival in rats and mice. Each data point represent means ± SE of 9-18 litters for rats and 7-18 litters for mice. Results are excerpted and slightly modified from the primary study that contained the experimental details (Lau et al., 2003). DEQ-CFW 00000140 C. Lau et at. / Toxicology and Applied Pharmacology 198 (2004) 231-241 Table 1 Concentrations of PFOS in rat maternal serum at near -term (GD 21) and in newborn pups Treatment dose Maternal serum Newbom serum (mg/kg) (pPm) (pp-) 0 0.244 ± 0.044 0.188 ± 0.070 1 19.6±1.8 35.9±0.6 2 45.0±1.6 71.9±4.1 3 71.9±6.6 86.5±10.0 5 80.6±3.0 108.2±4.6 10 189.9 ± 6.6 — Data represent means ± SE of 9-14 dams and 3-5 newborns, and are excerpted from primary studies that contained the experimental details (Lau et al., 2003; Thibodeaux et al., 2003). the newborn rats appeared to be correlated to their body burden of the fluorochemical. After the first week of postnatal life, the mortality rate was not different between control and treated groups. Cross -fostering the PFOS-ex- posed rat pups (5 mg/kg) to control nursing dams immedi- ately after birth failed to improve survival of the neonates, thus ruling out a role for abnormal maternal behaviors associated with the fluorochemical treatment. The maternal dose corresponding to the BMDL5 (lower limit of the 95% confidence interval on the Benchmark Dose that would predict a 5% increase in response above background inci- dence) for survival of rat pups at postnatal day 8 was estimated at 0.58 mg/kg. Postnatal growth of surviving rat pups was somewhat stunted by in utero exposure to PFOS. Body weights of pups in the 2 mg/kg and higher dose groups significantly lagged behind those in controls, and this effect persisted past wean- ing. Development of the PFOS-exposed pups was also significantly hindered as the average age at eye opening was delayed by a day, although no significant changes in the onset of puberty were noted. Hypothyroxinemia was observed in PFOS-exposed neonates. Because thyroid hor- mones are known to regulate brain development, the ontog- eny of neurochemical and neurobehavioral markers was evaluated. Prenatal exposure to PFOS produced only mar- ginal, but statistically significant deficits in the developmen- tal patterns of choline acetyltransferase activity (an enzyme marker sensitive to thyroid hormone status), and did not affect learning and memory behaviors determined by T-maze delayed alternation. This lack of notable changes in the CNS in these studies may have been related to the extent of thyroid hormone imbalance (reductions of thyroxine but not triiodo- thyronine or TSII) induced by the prenatal PFOS treatment. A similar profile of postnatal mortality was observed in the mouse (Fig. 1), although the maternal dosage required for the effect was higher than that in the rat. Most mouse offspring of dams exposed to 15 or 20 mg/kg PFOS did not survive for 24 h after birth. The LD50 was estimated at 10 mg/kg (compared to approximately 3 ing/kg in the rat), whereas survival of the lower dose groups (1 and 5 mg/kg) was not different from that of controls. The maternal dose corresponding to the BMDL5 for survival of mouse pups at 235 postnatal day 6 was estimated at 3.88 mg/kg, approximately six times higher than that of the rat. Among survivors, a trend toward growth deficit was evident. Compared to the rat, a more pronounced increase of liver weight was noted in PFOS-exposed neonatal mice. Hepatic enlargement per- sisted through the duration of the study (to PD 35). Serum thyroxine levels in the developing mice exposed to PFOS prenatally tended to be lower, but less consistently so than in PFOS-exposed rat pups. A significant delay in eye opening was also detected in treated mouse neonates. Similar effects of PFOS on newborn rodents can be achieved by different treatment paradigms. Butenhoff et al. (2002b) treated female rats with PFOS for 42 days before mating and throughout pregnancy at lower doses (0.4-2 mg/kg/day) and reported marked reduction of viability in pups in the two highest dose groups (49% and 17%, respectively, for the 1.6 and 2 mg/kg groups), as well as significant retardation in postnatal growth among the survivors, even in the lower dose groups. These results confirmed those from a two -generation reproductive study with the same treatment period for parental rats at doses of 0.1, 0.4, 1.6, and 3.2 mg/kg/day PFOS. In this study, the 1.6 and 3.2 mg/kg doses produced 34% and 100% Ft pup mortality within the first days postpartum, respectively, transient reductions in weight gain in F2 pups during the lactation period at maternal doses of 0.4 mg/kg/day, and no effects at 0.1 mg/kg/day (Christian et al., 1999a). In a cross -foster study with the same treatment paradigm, postpartum transfer of the PFOS-exposed pups (1.6 mg/ kg) to control nursing dams failed to reduce mortality (Case et al., 2001a). Grasty et al. (2003b) examined critical windows of PFOS toxicity and reported a similar increase of neonatal mortality in the rat with exposure of pregnant dams to high doses of PFOS (25 mg/kg/day) for a 4-day period during various stages of pregnancy. Neonatal mor- tality occurred after dosing in all time periods, but the incidence of neonatal death increased as the exposure period fell later during gestation, reaching 100% in the treatment group of GD 17-20. In another experiment, dosing with 50 mg/kg/day of PFOS on GD 19 and 20 only was sufficient to produce almost 100% mortality. Hence, the neonatal mortality resulting from PFOS admin- istration to pregnant rats does not require PFOS exposure before day 19 of gestation, suggesting that the critical period is late -gestational or perinatal. Interestingly, al- though the incidence of neonatal mortality was correspond- ingly lower in the early exposure groups, body weights of the newborns in these groups (4-day treatment on GD2-5, 6-9, or 10-14) were significantly lower than those of controls or late exposure groups Grasty et al. (2003b). The pathophysiology underlying PFOS-induced neonatal mortality is largely unknown at present. Results from Grasty et al. (2003b) suggest that organ systems developing late in gestation may be targets for PFOS insult. This hypothesis is also consistent with the relatively unremarkable teratologi- cal findings. Considering that PFOS-induced organ toxicity DEQ-CFW 00000141 236 C. Lau et al. / Toxicology and Applied pharmacology 198 (2004) 231-241 is incompatible with postnatal survival, maturation of the lung and pulmonary function is a plausible target. Numerous chemicals have previously been shown to interfere with fetal lung development (Lau and Kavlock, 1994). The profile of neonatal mortality induced by PFOS is in fact reminiscent of the developmental toxicity of nitrofen, a herbicide that perturbs fetal lung maturation, ultimately leading to compromised cardiopulmonary function and death in newborn rats (Lau et al., 1986, 1988, Stone and Manson, 1981). Indeed, the most recent findings reported by Grasty et al. (2003a, 2003b) are promising in support of this contention. These investigators described significant differ- ences in histological and morphometric evaluations of the lung between control and PFOS-treated newborns that suggest PFOS-induced inhibition of or delayed perinatal lung maturation. Nevertheless, the involvement of other possible mech- anisms of PFOS toxicity such as interference with mito- chondria) bioenergetics (Berthiaume and Wallace, 2002; Starkov and Wallace, 2002), impedance of cell -cell com- munication through gap junctions (Hu et al., 2002), inter- actions with fatty -acid binding proteins (Luebker et al., 2002a), hepatotoxicity (peroxisome proliferation) (Ikeda et al., 1987; Sohlenius et al., 1993), and alterations of maternal and fetal thyroid hormone economy ('Thibodeaux et al., 2.003) warrants consideration, On the other hand, a recent report did not provide evidence to support PFOS-induced interference with cholesterol synthesis (through inhibition of HMG CoA reductase activity, Haughom and Spydevold, 1992) as a potential mechanism for developmental toxicity of the chemical because cholesterol or mevalonic acid supplementation failed to ameliorate the adverse effects of PFOS (Luebker et al., 2002b). Toxicity of N-ethyl-N-(2-hydroxyethyl)- p erf luoroo ctan esulfon am ide N-ethyl-N-(2-hydroxyethyl)-perfluoro octanesulfonami de (N-EtFOSE) represents a building block for POSF-related products for commercial applications and can be degraded metabolically and in the environment to PFOS. Case et al. (2001b) evaluated its potential developmental effects in rats (with doses of 1, 5, 10, and 20 mg/kg) and rabbits (with doses of 0.1, 1, 2.5, and 3.75 mg/kg). In the rat, maternal body weight gains were reduced in the high dose groups, where reductions of fetal weight were also evident; but teratological findings were unremarkable. In the rabbit, dose -dependent reductions of maternal weight gain were seen and late -term resorptions were increased at the highest dose. No structural aberrations were noted in fetal examinations. A two -generation reproductive study in rats was con- ducted with N-EtFOSE (Christian et al., 1999b) at doses of 1, 5, 10, and 15 mg/kg/day, and the profile of developmental toxicity was quite similar to that described with PFOS (Case et al., 2001a, 2001b; Christian et al., 1999a; Lau et al., 2003; Thibodeaux et al., 2003). Doses of the fluorochemical as high as 15 mg/kg/day had no effect on estrous cycling, mating, or fertility of the F0 generation rats, although a decrease in numbers of implantations and viable embryos was noted. Postnatal survival of the F1 generation pups was significantly reduced in the 10 and 15 mg/kg dose groups, characterized by stillbirth and mortality during the first 72 h of life; in addition, weight gain deficits were evident among the survivors whose reflex and physical development were slightly delayed. These adverse effects were less pronounced in the lower dose groups, and their reproductive performances were not ad- versely affected. However, the number of F1 dams in the 5 mg/kg dose group that had stillborns or neonatal mortality (F2) was slightly, but significantly increased, and the weight gain of F2 pups was also reduced. There were no significant adverse effects at 1 mg/kg/day. Toxicity of perfluorooctanoic acid The general toxicity profile of PFOA has been charac- terized in monkeys (Butenhoff et al., 2002a; Griffith and Long, 1980), rats (Abdellatif et al., 1991; Biegel et al., t 995, Cook et al_, 1992; Diaz et al., 1994; Griffith and Long, 1980; Kennedy, 1985; Kennedy et al., 1986; Kudo et al., 1999; IJu et al., 1996; Ohmori et al., 2003; Olson and Andersen, 1983; Pastoor et al., 1987; Reo et al., 1994), mice (Yang et al., 2000, 2001), rabbits (Griffith and Long, 1980; Kennedy, 1985), fish (Martin et al,, 2003a, 2003b), and humans (Gilliland and Mandel, 1993, 1996, Olsen et al., 1999, 2000, 2003b), and summarized in a recent review (Kennedy et al., 2004; Kudo and Kawashima, 2003). A preliminary risk assessment of the developmental toxicity of PFOA was recently conducted by the Office of Pollution Prevention and Toxic Substances of the U.S. Environmental Protection Agency (US EPA, 2003a). Hence, only an abbreviated summary of the animal studies mentioned in the risk assessment document will be provided here. Because of the peculiar gender difference in the clearance and excretion of PFOA in some animal species (vide supra), the choice of animal models is critical when evaluating the reproductive and developmental toxicity of this chemical. Thus far, only information pertaining to the rat and the rabbit is available. Teratological studies of PFOA have been conducted by Gortner (1981) with rats and rabbits (Gortner, 1982), and Staples et al. (1984) with rats, but neither laboratory reported any significant findings with adminis- tered doses up to 100-150 mg/kg/day for rats and 50 mg/ kg/day for rabbits. More recently, Butenhoff et al. (2004) undertook a two - generation study of ammonium PFOA where rats were given 1, 3, 10, or 30 mg/kg daily. In the F, generation, observations during the lactational period were unremark- able, with the exception of a significant lag of weight gain in the offspring of the 30 mg/kg PFOA-treated group noted during the first postnatal week and a statistically significant DEQ-CFW 00000142 C. Lau el al. / Toxicology and Applied pharmacology 198 (2004) 231-241 decrease in the lactation index. A statistically significant increase of mortality was seen in both male and female pups at 30 mg/kg, especially in the first week after initiating gavage dosing at weaning. Nine of the thirteen neonatal deaths in the postweaning period involved the lightest pups and occurred in the first few days postweaning, suggesting that low body weight or immaturity at weaning was a likely factor in the mortality of these pups. Among the survivors, significantly reduced weight gain was observed in males, reaching statistical significance in all treated groups by termination of the study at 113 days postweaning. F, females in the 30 mg/kg dose group had significant reduc- tions in body weight after weaning and these deficits persisted through the remainder of the study. Both Fl- generation males and females, at the highest dose (30 mg/ kg/day) evaluated, experienced statistically significant delays (by 2 and 4 days for females and males, respectively) in reaching sexual maturity. However, no effects on mating or fertility parameters were apparent, and findings in the F2 generation were unremarkable. Histological examination of tissues by light microscopy did not reveal any abnormalities in sex organs. However, hypertrophy and vacuolation of the zona glomerulosa of the adrenals of P-generation (10 and 30 mg/kg) and Fl-generation (30 mg/kg) males were seen, and hepatocellular hypertrophy was present in F, males at doses of 3 mg/kg and higher. Interpretation of these data and their extrapolation for human health risk assessment are quite challenging because sexually mature female rats possess an innate ability to excrete PFDA rapidly, with an elimination half-life of 2-3 h, as opposed to days or weeks in male rats, dogs, and monkeys, or years in humans. Preliminary results from a pharmacokinetic study (Kemper and Jepson, 2003) further complicate the issue. These investigators reported that plasma kinetics in adult female rats appeared to be biphasic at the two highest administered single oral doses of 5 and 25 mg/kg (unlike the monophasic profile in male rats), and terminal elimination half-lives were dependent on the dose level. Thus, the toxic manifestation of PFDA in female pregnant rats, where steady state of the chemical is unlikely to be reached on a daily gavage dosing regimen and significant variations in the serum concentration of PFOA occur between the treatment intervals, may be quite different from those species (including humans) where the steady- state kinetics is reached gradually. The postweaning mortality and delays in pubertal onset in the F, generation at 30 mg/kg (Butenhoff ct al., 2004) reflect potential developmental effects of PFOA that may be partially explained by reduced body weight at weaning or weight gain thereafter. Interestingly, the fact that these effects occurred in both genders suggests a lack of the sex hormone -mediated differences in excretion. This contention would be consistent with the observation that the organic anion transporter protein (OAT2) that has been proposed to be responsible for the high PFOA excretion rate in adult female rats (Kudo et al., 2002) does not begin to be 237 expressed until sometime between postnatal days 35 and 40 (Buist et al., 2002). These data suggest that pharmaco- kinetic profiles in sexually immature rats may be different from those in the adults. Toxicity of other perfluoroalkyl acids Compared to the C-8 perfluorochemicals, less is known about perfluoroalkyl acids of other carbon chain lengths. Perfluorohexanesulfonate (PFHS) has been found in serum samples from the general population including children, adult Red Cross blood donors, and elderly subjects enrolled in a prospective study of cognitive disorders (Olsen et al., 2002, 2003a, 2004), with geometric means of 0.002-0.005 µg/ml. In the same studies, geometric mean serum concentrations for PFOA were approximately 0.005 µg/ml, and, for PFOS, they ranged from approximately 0.03-0.04 µg/ml. Perfluorobu- tane sulfonate (PFBS) is known to have a much shorter half- life than PFOS (Paul Lieder, 3M, personal communication); in contrast, perfluorodecanoate (PFDA) appears to have a low rate of elimination in the rat (Vanden Heuval et al., 199lb). In this regard, it will be of interest in future pharmacokinetic and structure -activity studies to determine to what extent the rate of elimination for these fluorochemicals is directly related to their carbon chain length. Like PFDA, both perfluorononanoate (PFNA) and PFDA are peroxisome proliferators (Harrison et al., 1988; Ikeda et al., 1985; Kinney et al., .1989). The toxic potency of PFDA is quite high, with an LD50 4.6-fold lower than that estimated for PFOA (Olson and Andersen, 1983). PFDA has also been reported to produce hepatotoxicity, anorexia, alteration of fatty acid metabolism, reduction of circulating thyroid hormones and androgen, bradycardia, and hypother- mia in the rat (Bookstaff et al., 1990; George and Andersen, 1986; Gutshall et al., 1988, 1989, Langley and Pitcher, 1985; Pitcher and Langley, 1986; Pilcher et al., 1987; Singer et al., 1990). Both potassium PFBS and potassium PFHS have re- cently been assessed for developmental and reproductive effects. Maternal exposure to PFBS did not produce any adverse effects on embryo/fetal development, and no significant alterations were noted in a two -generation study in rats at doses as high as 1 g/kg (Paul Lieder, 3M, personal communication). PFHS did not produce develop- mental or reproductive effects in rats at doses up to 10 mg/ kg in a onc-gencration reproductive and developmental toxicity screening test (Hoberman and York, 2003). In addition, Harris and Birnbaum (1989) reported a lack of teratogenic effects of PFDA in the mouse. Future research needs and directions The industrial and consumer uses of perfluoroalkyl acids and their derivatives have increased tremendously over the DEQ-CFW 00000143 238 C. Lan et al. / Toxicology and Applied Pharmacology 198 (2004) 231-241 past half century, and the unique physical and chemical properties of these materials makes substitution by other organic materials difficult for some of these products and applications. On the other hand, several of these fluorochem- icals have become ubiquitous in the environment and the presence of PFOS and PFOA in humans in particular has drawn considerable attention in the public arena. Although the toxicology of some of these compounds has been inves- tigated in the past, the widespread exposure demonstrated in recent bio-monitoring studies has renewed efforts to under- stand the hazards that may be associated with these chem- icals, and new information has come to light in recent studies. This review has summarized recent findings from several laboratories on the developmental toxicity of perfluoroalkyl acids. Although the teratological potentials of some of these chemicals had been studied in the past and the findings were generally unremarkable, information from postnatal studies on developmental and reproductive indices have been pub- lished or submitted to the U.S. EPA public docket recently, and these findings have prompted further consideration of their relevance to human health risk. Confirmation and elucidation of the pharmacokinetic and mechanistic bases of these findings by different experimental approaches and study designs will be important to establish the context of these findings for human health risk assessment. This goal should be attainable by active investigation within the scien- tific community in the next few years. Other research avenues can be useful in defining the potential health risks of fluorochemicals. At present, little is known about the environmental exposure, transport, and fate of these compounds, particularly PFOA. In June 2003, the U.S. Environmental Protection Agency began an En- forceable Consent Agreement process with industry to explore the environmental fate and transport of the PFOA, telomer-based materials, and related compounds, and to enhance understanding of the sources of PFOA in the environment, as well as the pathways by which human exposure to this chemical can occur. Information gathered under this agreement will complement the data collected in bio-monitoring programs and may lead to predictive models of exposure. Because of their physical properties (being hydrophobic, lipophobic and protein -bound; Ilan et al., 2003), perfluoroalkyl chemicals may have unique profiles of distribution in the body. Thus, pharmacokinetic charac- terization of these compounds, especially during pregnancy and early postnatal ages (before sexual maturity), will greatly facilitate our understanding of chemical disposition and potential cellular targets of PFOS and PFOA. In addition, as information pertaining to body burden of these fluorochemicals comes to light in the next few years (from bio-monitoring surveys by the CDC, for instance), this "internal dosimetric" can be used readily for cross -species (from laboratory animal models, such as rodents and monkeys to humans) as well as low -dose (from experimen- tal doses to exposure levels) extrapolations for risk analysis. More in-depth descriptive information will undoubtedly provide additional clues to the modes and mechanisms of PFOS and PFOA developmental toxicity in laboratory animal models. Cellular and molecular mechanistic findings will be instrumental in extrapolating the health risk potential of these compounds for humans. For PFOA, potential disparities between genders and species (i.e. pharmacoki- netic parameters between rats and humans) have to be reconciled to aid interspecies extrapolation. As other per- fluoroalkyl acids are slowly coming onto the commercial market to replace PFOS, the potential developmental toxic- ity of these compounds should be investigated thoroughly. References 3M, 2003. Environmental and Health Assessment of Perfluorooctane Sulfonate and its Salts. Available on US EPA Administrative Record AR-226. Abdellatif, A.G., Preat, V, Taper, H.S., Roberfroid, M., 1991. The modu- lation of rat liver carcinogenesis by perfluorooctanoic acid, a peroxi- sorne proliferator. Toxicol. Appl. Pharmacol. 111, 530-537. Austin, M.E., Kasturi, B.S., Barber, M., Kannan, K., MohanKumar, P.S., MohanKumar, S.M.J., 2003. Neuroendocrine effects of perfluorooctane sulfonate in rats. Environ. Health Perspect. 111, 1485-1489. Berthiaume, J., Wallace, K.B., 2002. Perfluorooctanoate, perfluorooctane- sulfonate, and N-ethyl perfluorooetanesulfonamido ethanol; peroxi- some proliferation and mitochondrial biogenesis. Toxicol. Lett. 129, 23 — 32. Biegel, L.B., Liu, R.C.M., Hurtt, M.E., Cook, J.C., 1995. Effects of am- monium perfluoroctanoate on Leydig cell function: in vitro, in vivo, and ex vivo studies. Toxicol. Appl. Pharmacol. 134, 18-25. Bookstaff, R.C., Moore, R.W., Ingall, G.B., Peterson, R.E., 1990. Andro- genic deficiency in male rats treated with perfluorodecanoic acid. Tox- icol. Appl. Pharmacol. 104, 322-333. Buist, S.C.N., Cherrington, N.J., Choudhuri, S., Hartley, D.P., Klaassen, C.D., 2002. Gender -specific and developmental influences on the ex- pression of rat organic anion transporters. J. Pharmacol. Exp. Ther. 301, 145-151. Burris, J.M., Lundberg, J.K., Olsen, G., Simpson, C., Mandel, J., 2002. Interim report No. 2, Determination of serum half-lives of several fluorochemicals. St. Paul (MN), 3M Company. US EPA Docket AR-226-1086. Butenhoff, J.L., Costa, G., Elcombe, C., Farrar, D., Hansen, K., Iwai, H., Jung, R., Kennedy, G., Lieder, P., Olsen, G., Thomford, P., 2002a. . Toxicity of ammonium perfluorooctanoate in male Cynomolgus monkeys after oral dosing for 6 months. Toxicol. Sci. 69, 244-257. Butenhoff, J.L., York, R., Seacat, A., Luebker, D., 2002b. Perfluoroocta- nesulfonate-induced perinatal mortality in rat pups is associated with a steep dose -response. Toxicologist 66, 25. Butenhoff, J.L., Kennedy, G.L., Frame, S.R., O'Connor, J.C., York, R.G., 2004. The reproductive toxicology of ammonium perfluorooctanoate (APFO) in the rat. Toxicology 196, 95-116. Case, M.T., York, R.G., Butenhoff, J.L., 2001 a. Oral (gavage) cross-£os- teiing study of potassium perfluorooctane sulfonate (PFOS) in rats. Toxicologist 60, 221. Case, M.T., York, R.G., Christian, M.S., 2001b. Rat and rabbit oral devel- opmental toxicology studies with two perfluorinated compounds. Int. J. Toxicol. 20, 101-109. Christian, M.S., Hoberman, A.M., York, R.G., 1999a. Combined Oral (gavage) Fertility, Developmental and Perinatal/Postnatal Reproduction Toxicity Study of PFOS in Rats. Argus Research Laboratories, Inc., Horsham, PA. US EPA Docket 8EHQ-0200-00374. Christian, M.S., Hoberman, A.M., York, R.G., 1999b. Combined Oral (gavage) Fertility, Developmental and Perinatal/Postnatal Reproduction DEQ-CFW 00000144 C. Lau et at. / Toxicology and Applied Pharmacology 198 (2004) 231-241 Toxicity Study of N-EtFOSE in Rats. Argus Research Laboratories, Inc., Horsham, PA. US EPA Docket AR-226-0552. Cook, J.C., Murray, S.M., Frame, S.R., Hurtt, M.E., 1992. Induction of Leydig cell adenomas by ammonium perfluorooctanoate: a pos- sible endocrine -related mechanism. Toxicol. Appl. Pharmacol. 113, 209-217. Diaz, M.J., Chinje, E., Kentish, P., Jamot, B., George, M., Gibson, G., 1994. Induction of cytochrome P4504A by the peroxisome proliferators perfluoro-n-octanoic acid. Toxicology 86, 109-122. George, M.E., Andersen, M.E., 1986. Toxic effects of nonadecafluoro-n- decanoic acid in tat. Toxicol. Appl. Pharmacol. 85, 169-180. Gibson, S.J., Johnson, J.D., 1983. Extent and Route of Excretion of Total Carbon-14 in Pregnant Rats after it Single Oral Dose of Ammonium 14C-Perfluorooctanoate. Riker Laboratories, Inc., St. Paul, MN. US EPA AR-226-0458. Giesy, J.P., Kannan, K., 2001. Global distribution of perfluorooctane sul- fonate in wildlife. Environ. Sci. Technol. 35, 1339-1342. Giesy, I.P., Kannan, K., 2002. Perfluorochemical surfactants in the envi- ronment. Environ. Sci. Technol. 36, 146A-152A. Gilliland, F.D., Mandel, J.S., 1993. Mortality among employees of a per- fluorooctanoic acid production plant. J. Occup. Med. 35, 950-954. Gilliland, F.D., Mandel, J.S., 1996. Serum perfluorooctanoic acid and he- patic enzymes, lipoproteins, and cholesterol: a study of occupationally exposed men. Am. J. Ind. Med. 29, 560-568. Gortner, E.G., 1980. Oral Teratology Study of FC-95 in Rats Riker Labo- ratories, Inc. Experiment Number: 0680TR0008, December, 1980. US EPA AR-226 226-0016. Gortner, E.G., 1981. Oral teratology study of T-2998CoC in rats. Exper- iment Number 0681TR0110. Safety Evaluation Laboratory and Riker Laboratories, Inc., St. Paul, MN. USEPA Public Docket, AR-226 226-0463. Gortner, E.G., 1982. Oral teratology study of T-3141CoC in rabbits. Experiment Number 0681TB0398. Safety Evaluation Laboratory and Riker Laboratories, Inc., St. Paul, MN. USEPA Public Docket AR-226-0465. Grasty, R.C., Brey, B.E., Lau, C., Rogers, J.M., 2003a. Window of sus- ceptibility to perfluorooctane sulfonate (PFOS)-induced neonatal mor- tality in the rat. Birth Defects Res. 68 (Part B), 219. Grasty, R.C., Grey, B.E., Lau, C., Rogers, J.M., 2003b. Prenatal window of susceptibility to perfluorooctane sulfonate-induced neonatal mortality in the Sprague Dawley rat. Birth Defects Res. 68 (Part B), 465-471. Griffith, F.D., Long, J.E., 1980. Animal toxicity studies with ammonium perfluorooctanoate. Am. Ind. Hyg. Assoc. J. 41, 576-583. Gutshall, D.M., Pilcher, G.D., Langley, A.E., 1988. Effect of thyroxine supplementation on the response to perfluoro-n-decanoic acid (PFDA) in rats. J. Toxicol. Environ. Health 24, 491-498. Gutshall, D.M., Pilcher, G.D., Langley, A.E., 1989. Mechanism of the serum thyroid hormone lowering effect of perfluoro-n-decanoic acid (PFDA) in rats. J. Toxicol. Environ. Health 28, 53-65. Hagen, D.F., Belisle, J., Johnson, J.D., Venkateswarlu, P., 1981. Charac- terization of fluorinated metabolites by a gas chromatographic -helium microwave plasma detector -The biotransformation of 1H, 1H, 2H, 2H-perfluorodecanol to perfluorooctanoate. Anal. Biochem. 118, 336-343. Han, X., Snow, T.A., Kemper, R.A., Jepson, G.W., 2003. Binding of per- fluorooctanoic acid to rat and human plasma proteins. Chem. Res. Toxicol. 16, 775-781. Hanhijarvi, H., Ophaug, R.H., Singer, L., 1982. The sex -related difference in perfluorooctanoate excretion in the rat. Proc. Soc. Exp. Biol. Med. 171, 50-55. Hanhijarvi, H., Ylinen, M., Kojo, A., Kosma, V.M., 1987. Elimination and toxicity of perfluorooctanoic acid during subchronic administration in the Wistar rat. Pharmacol. Toxicol. 61, 66-68. Hanhijarvi, H., Ylinen, M., Haaranen, T., Nevalainen, T., 1988. A proposed species difference in the renal excretion of perfluorooctanoic acid in the beagle dog and rat. In: Beynen, A.C., Solleveld, H.A. (Eds.), New Developments in Biosciences: Their Implications for Laboratory Am- 239 mal Science. Martinus Nijhoff Puublishers, Dordrecht, The Netherlands, pp. 409-412. Hansen, K.J., Clemen, L.A., Ellefson, M.E., Johnson, H.O., 2001. Com- pound -specific, quantitative characterization of organic fluorochemicals in biological matrices. Environ. Sci. Technol. 35, 766-770. Hansen, K.J., Johnson, H.O., Eldridge, J.S., Butenoff, J.L., Dick, L.A., 2002. Quantitative characterization of trace levels of PFOS and PFOA in the Tennessee River. Environ. Sci. Technol. 36, 1681-1685. Hams, M.W., Birnbaum, L.S., 1989. Developmental toxicity of perfluoro- decanoic acid in C57BL/6N mice. Fundam. Appl. Toxicol. 12, 442-448. Harrison, E.H., Lane, J.S., Luking, S., Van Rafelghem, M.J., Andersen, M.E., 1988. Perfluoro-n-decanoic acid: induction of peroxisomal beta - oxidation by a fatty acid with dioxin -like toxicity. Lipids 23, 115-119. Haughom, B., Spydevold, 0., 1992. The mechanism underlying the hypo- lipemic effect of perfluorooctanoic acid (PFOA), perfluorooctane sul- phonic acid (PFOSA) and clofibric acid. Biochim. Biophys. Acta 1128, 65-72. Hebert, G.N., Odom, M.A., Craig, P.S., Dick, D.L., Strauss, S.H., 2002. Method for the determination of sub-ppm concentration of perfluoroal- kylsulfonate anions in water. Environ. Monit. 4, 90-95. Henwood, S.M., McKee-Pesick, P., Costello, A.C., Osmitz, T.G., 1994. Developmental toxicity study with lithium perfluorooctane sulfonate in rats. Teratology 49, 398. Hoberman, A.M., York, R.G., 2003. Oral (gavage) combined repeated dose toxicity study of T-7706 with the reproduction/developmental toxicity screening test. Argus Research Protocol No. 418-028. Argus Research Charles Rivers Laboratory. USEPA Public Docket AR-226. Hoff, P.T., Van de Vijver, K., Van Dongen, W., Esmans, E.L., Blust, R., De Coen, W.M., 2003a. Perfluorooctane sulfonic acid in bib (Trisopterus luscus) and plaice (Pleuronectes platessa) from the Western Scheldt and the Belgian North Sea: distribution and biochemical effects. Environ. Toxicol Chem. 22, 608-614. Hoff, P.T., Van Dongen, W., Esmans, E.L., Blust, R., De Coen, W.M., 2003b. Evaluation of the toxicological effects of perfluorooctane sul- fonic acid in the common carp (Cyprinus carpio). Aquat. Toxicol. 62, 349-359. Hu, W., Jones, P.D., Upham, B.L., Trosko, J.E., Lau, C., Giesy, J.P., 2002. Inhibition of gap junctional intercellular communication by perfluori- nated compounds in rat liver and dolphin kidney epithelial cell lines in vitro and Sprague-Dawley rats in vivo. Toxicol. Sci. 68, 429-436. Ikeda, T., Aiba, K., Fukuda, K., Tanaka, M., 1985. The induction of per- oxisome proliferation in rat liver by perfluorinated fatty acids, metabol- ically inert derivatives of fatty acids. J. Biochem. 98, 475-482. Ikeda, T., Fukuda, K., Mori, I., Enomoto, M., Kornai, T., Suga, T., 1987. Induction of cytochrome P-450 and peroxisome proliferation in rat liver by perfluorinated octanesulfonic acid. In: Fahimi, H.D., Sies, H. (Eds.), Peroxisomes in Biology and Medicine. Springer Verlag, New York, pp. 304-308. Johnson, J.D., Ober, R.E., 1979. Absorption of FC-95-14C in rats after a single oral dose. Project No. 8900310200. Riker Laboratories, Inc., St. Paul, MN. US EPA Docket No. 8(e)HQ-1180-00374. Johnson, J.D., Gibson, S.J., Ober, R.E., 1979. Extent and route of excretion and tissue distribution of total carbon-14 in rats after a single i.v. dose of FC-95-14C. Project No, 8900310200. Riker Laboratories, Inc., St. Paul, MN. US EPA Docket No. 8(e)HQ-1180-00374. Johnson, J.D., Gibson, S.J., Ober, R.E., 1984. Cholestramine-enhanced fecal elimination of carbon-14 in rats after administration of ammonium [14C]perfluorooctanoate or potassium [14C]perfluoronctanesulfonate. Fundam. Appl. Toxicol. 4, 972-976. Kannan, K., Franson, J.C., Bowerman, W.W., IIansen, K.J., Jones, P.D., Giesy, J.P., 2001a. Perfluorooctane sulfonate in fish -eating water birds including bald eagles and albatrosses. Environ. Sci. Technol. 35, 3065-3070. Kannan, K., Koistinen, J., Beckmen, K., Evans, T., Gorzelany, J.F., Han- sen, K.J., Jones, P.D., Helle, E., Nyman, M., Giesy, J.P., 2001b. Accu- mulation of perfluorooctane sulfonate in marine mammals. Environ. Sci. Technol. 35, 1593-1598. DEQ-CFW 00000145 240 C. Lau et al. / Toxicology and Applied Pharmacology 198 (2004) 231-241 Kannan, K., Choi, J.W., Iseki, N., Senthilkumar, K., Kim, D.H., Giesy, J.P., 2002a. Concentrations of perfluorinated acids in livers of birds from Japan and Korea. Chemosphere 49, 225-231. Kannan, K., Corsolini, S., Falandysz, J., Oehme, G., Focardi, S., Giesy, J.P., 2002b. Perfluorooctanesulfonate and related fluorinated hydrocar- bons in marine mammals, fishes, and birds from coasts of the Baltic and the Mediterranean Seas. Environ. Sci. Technol. 36, 3120-3126. Kannan, K., Hansen, K.J., Wade, T.L., Giesy, J.P., 2002c. Perfluorooctane sulfonate in oysters, Crassostrea viiginica, from the Gulf of Mexico and the Chesapeake Bay, USA. Arch. Environ. Contam. Toxicol. 42, 313-318. Kannan, K., Newsted, J., Halbrook, R.S., Giesy, J.P., 2002d. Perfluorooc- tanesulfonate and related fluorinated hydrocarbons in mink and river otters from the United States. Environ. Sci. Technol. 36, 2566-2571. Kawashima, Y, Matsunaga, T., Uy-Yu, N., Kozuka, H., 1991. Induction by perfluorooctanoic acid ofmicrosomal 1-acyglycerophosphocholine ace- tyltransferase in rat kidney. Sex -related difference. Biochem. Pharma- col. 42, 1921-1926. Kemper, R.A., Jepson, G.W., 2003. Pharmacokinetics of pei-fluorooctanoic acid in male and female rats. Toxicologist 72, 148. Kennedy, G.L., 1985. Dermal toxicity of ammonium perfluorooctanoate. Toxicol. Appl. Pharmacol. 81, 348-355. Kennedy, G.L., Hall, G.T., Brittelli, M.R., Barnes, J.R., Chen, H.C., 1986. Inhalation toxicity of ammonium perfluorooctanoate. Food Chem. Tox- icol. 24, 1325-1329. Kennedy, G.L., Butenhoff, J.L., Olsen, G.W., O'Connor, J.C., Seacat, A.M., Perkins, R.G., Biegel, L.B., Murphy, S.R., 2004. The toxi- cology of perfluorooctanoate. Crit. Rev. Toxicol. (in press). Key, B.D., Howell, R.D., Criddle, C.S., 1997. Fluorinated organics in the biosphere. Environ. Sci. Technol. 31, 2445-2454. Key, B.D„ Howell, R.D., Criddle, C.S., 1998. Defluorination of organo- fluorine sulfur compounds by Pseudomonas sp. strain D2. Environ. Sci. Technol. 32, 2283-2287. Kinney, L.A., Chromey, N.C., Kennedy, G.L., 1989. Acute inhalation toxicity of ammonium perfluorononanoate. Food Chem. Toxicol. 27, 465 -468. Kissa, E., 2001. Fluorinated Surfactants and Repellents, second edition. Marcel Dekker, New York. Kojo, A., Hanhijarvi, H., Ylinen, M., Kosma, V.M., 1986. Toxicity and kinetics of perfluoro-octanoic acid in the Wistar rat. Arch. Toxicol., Suppl. 9, 465-468. Kudo, N., Kawashima, Y., 2003. Toxicity and toxicokinetics of perfluor- ooctanoic acid in humans and animals. J. Toxicol. Sci. 28, 49-57. Kudo, N., Miztiguchi, H., Yamamoto, A., Kawashima, Y., 1999. Altera- tions by perfluorooctanoic acid of glycerolipid metabolism in rat liver. Chem.-Biol. Interact. 118, 69-83. Kudo, N., Katakura, M., Sato, Y, Kawashima, Y., 2002. Sex hormone - regulated renal transport of perfluorooctanoic acid. Chem.-Biol. Inter- act. 139, 301-316. Langley, A.E., Pilcher, G.D., 1985. Thyroid, bradycardic and hypothermic effects of perfluoro-n-decanoic acid in rats. J. Toxicol. Environ. Health 15, 485-491. Lau, C., Kavlock, R.J., 1994. Functional toxicity in the developing heart, lung, and kidney. In: Kimmel, C.A., Buelke-Sam, J. (Eds.), Developmental Toxicology, second edition. Raven Press, New York, pp. 119-188. Lau, C., Cameron, A.M., Irsula, O., Robinson, K.S., 1986. Effects of prenatal nitrofen exposure on cardiac structure and function in the rat. Toxicol. Appl. Pharmacol. 86, 22-32. Lau, C., Cameron, A.M., Irsula, O., Antolick, L.L., Langston, C., Kavlock, R.J., 1988. Teratogenic effects of nitrofen on cellular and functional maturation of the rat lung. Toxicol. Appl. Pharmacol. 95, 412-422. Lau, C., Thibodeaux, J.R., Hanson, R.G., Rogers, J.M., Grey, B.E., Stanton, J.L., Butenhoff, J.L., Stevenson, L.A., 2003. Exposure to perfluorooc- tane sulfonate during pregnancy in rat and mouse: II. Postnatal evalua- tion. Toxicol. Sci. 74, 382-392. Liu, R.C.M., Hurtt, M.E., Cook, J.L., Biegel, L.B., 1996. Effect of the peroxisome proliferators, ammonium perfluorooctanoate (C8), on he- patic aromatase activity in adult male CrI:CD BR (CD) rats. Fundam. Appl. Toxicol. 30, 220-228. Luebker, D.J., Hansen, K.J., Bass, N.M., Butenhoff, J.L., Seacat, A.M., 2002a. Interactions of fluorochemicals with rat liver fatty acid -binding protein. Toxicology 176, 175-185. Luebker, D., York, R., Seacat, A., Butenhoff, J., 2002b. Perfluorooctane- sulfonate-induced perinatal mortality in rat pups is not a result of re- duced serum lipids. Toxicologist 66, 26. Martin, J.W., Muir, D.C., Moody, C.A., Ellis, D.A., Kwan, W.C., Solomon, K.R., Mabury, S.A., 2002. Collection of airborne fluorinated organics and analysis by gas chromatography/chemical ionization mass spec- trometry. Anal. Chem. 74, 584-590, Martin, J.W., Mabury, S.A., Solomon, K.R., Muir, D.C., 2003a. Dietary accumulation of perfluorinated acids in juvenile rainbow trout (Onco- orhynchus.nrykiss). Environ. Toxicol. Chem. 22, 189-195. Martin, J.W., Mabury, S.A., Solomon, K.R., Muir, D.C., 2003b. Biocon- centration and tissue distribution of perfluorinated acids in rainbow trout (Oncorhynchus mykiss). Environ. Toxicol. Chem. 22, 196-204. Martin, J.W., Smithwick, M.M., Braune, B.M., Hoekstra, P.F., Muir, D.C., Mabury, S.A., 2004. Identification of long -chain perfluorinated acids in biota from the Canadian Arctic. Environ. Sci. Technol. 38, 373-380. Moody, C.A., Martin, J.W., Kwan, W.C., Muir, D.C., Mabury, S.A., 2002. Monitoring perfluorinated surfactants in biota and surface water sam- ples following an accidental release of fire -fighting foam into Etobicoke Creek. Environ. Sci. Technol. 36, 545-551. Noker, P.E., Gorman, G.S., 2003a. A pharmacokinetic study of potassium perfluorooctanesulfonate in the cynomolgus monkey. Southern Re- search Institute. Unpublished report. Available on US EPA Administra- tive Record 226. Noker, P.E., Gorman, G.S., 2003b. A pharmacokinetic study of potassium perfluorooctanoate in the cynomolgus monkey. Southern Research In- stitute. Unpublished report. Available on US EPA Administrative Re- cord 226. Ohmori, K., Kudo, N., Katayama, K., Kawashima, Y, 2003. Comparison of the toxicokinetics between perfluorocarboxylic acids with different carbon chain length. Toxicology 184, 135-140. Olsen, G.W., Gilliland, F.D., Burlew, M.M., Burris, J.M., Mandel, J.S., Mandel, J.H., 1998. An epidemiologic investigation of reproductive hormones in men with occupational exposure to perfluorooctanoic acid. J. Occup. Environ. Med. 40, 614-622. Olsen, G.W., Burris, J.M., Mandel, J.H., Zobel, L.R., 1999. Serum per- fluorooctane sulfonate and hepatic and lipid clinical chemistry tests in fluorochemical production employees. J. Occup. Environ. Med. 41, 799-806. Olsen, G.W., Burris, J.M., Burlew, M.M., Mandel, J.H., 2000. Plasma cholecystokinin and hepatic enzymes, cholesterol and lipoproteins in ammonium perfluorooctanoate production workers. Drug Chem. Tox- icol. 23, 603-620. Olsen, G.W., Schmickler, M.N., Tierens, J.M., Logan, P.W., Burris, J.M., Burlew, M.M., Lundberg, J.K., Mandel, J.H., 2001. Descriptive summary of serum fluorochemical levels among employee partici- pants of the year 2000 Antwerp fluorochemical medical surveillance program. St. Paul (MN), 3M Company. U.S. EPA Docket AR-226- 1030-0206. Olsen, GN, Burris, J.M., Lundberg, J.K., Hansen, K.J., Mandel, J.H., Zobel, L.R., 2002. Identification of Fluorochemicals in Human Sera: III. Pediatric Participants in a Group A Streptococci Clinical Trial Investigation. US EPA Docket AR-226-1085. Olsen, G.W., Church, T.R., Miller, J.P., Burris, J.M., Hansen, K.J., Lund- berg, J.K., Armitage, J.M., Herron, R.M., Medhdizadehkashi, Z., Nobi- letti, J.B., O'Neill, E.M., Mandel, J.H., Zobel, L.R., 2003a. Perfluorooctanesulfonate and other fluorochemicals in the serum of American Red Cross adult blood donors. Environ. Health Perspect. 111, 1892-1901. Olsen, G.W., Burris, J.M., Burlew, M.M., Mandel, J.H., 2003b. Epidemi- ologic assessment of worker serum perfluorooctanesulfonate (PFOS) DEQ-CFW 00000146 C. Lau et al. / Toxicology and Applied Pharmacology 198 (2004) 231-241 and perfluorooctanoate (PFOA) concentrations and medical surveil- lance examinations. J. Occup. Environ. Med. 45, 260-270. Olsen, G.W., Logan, P.W., Hansen, K.J., Simpson, C.A., Burris, J.M., Burlew, M.M., Vororath, P.P., Venkateswarlu, P., Schumpert, J.C., Mandel, J.H., 2003c. An occupational exposure assessment of a per- fluorooctanesulfonyl fluoride production site: biomonitoring. Am. Ind. Hyg. Assoc. J. 64, 651-659. Olsen, G.W., Hansen, K.J., Stevenson, L.A., Barris, J.M., Mandel, J.H., 2003d. Human donor liver and serum concentrations of perfluoroocta- nesulfonate and other perfluorochemicals. Environ. Sci. Technol. 37, 888-891. Olsen, G.W., Church, T.R., Larson, E.B., van Belle, G., Lundberg, J.K., Hansen, K.J., Burris, J.M., Mandel, J.H., Zobel, L.R., 2004. Serum concentrations of perfluorooctanesulfonate and other fluorochemicals in an elderly population from Seattle, Washing- ton. Chemosphere. 54, 1599-1611. Olson, C.T., Andersen, M.E., 1983. The acute toxicity of perfluorooctanoic and perfluorodecanoic acid in male rats and effects on tissue fatty acids. Toxicol. Appl. Pharmacol. 70, 362-372. Ophaug, R.H., Singer, L., 1980. Metabolic handling of perfluorooctanoic acid in rats. Proc. Soc. Exp. Biol. Med. 163, 19-23. Pastoor, T.P., Lee, K.P., Perri, M.A., Gillies, P.J., 1987. Biochemical and morphological studies of ammonium perfluorooctanoate-induced hepa- tomegaly and peroxisome proliferation. Exp. Mol. Pathol. 47, 98-109. Pilcher, G.D., Langley, A.E., 1986. The effects of perfluoro-n-decanoic acid in the rat heart. Toxicol. Appl. Pharmacol. 85, 389-397. Pilcher, G.D., Gutshall, D.M., Langley, A.E., 1987. The effects ofperfluoro- n-decanoic acid (PFDA) on rat heart beta-receptors, adenylate cyclase, and fatty acid composition. Toxicol. Appl. Pharmacol. 90, 198-205. Prescher, D., Gross, U., Wotzka, J., Txcheu-Schlueter, M., Starke, W., 1985. Environmental behavior of fluoro surfactants: Part 2. Study on biochemical degradability. Acta Hydrochim. Hydrobiol. 13, 17-24. Renner, R., 2001. Growing concern over perfluorinated chemicals. Envi- ron. Sci. Technol. 35, 154A-160A. Reo, N.V., Goecke, C.M., Narayanan, L., Jamot, B.M., 1994. Effects of perfluoro-n-octanoic acid, perfluoro-n-decanoic acid, and clofibrate on hepatic phosphorus metabolism in rats and guinea pigs in vivo. Tox- icol. Appl. Pharmacol. 124, 165-173. Seacat, A.M., Thomford, P.J., Hansen, K.J., Olsen, G.W., Case, M.T., Butenh- off, J.L., 2002. Subchronic toxicity studies on perfluoroctanesulfonate potassium salt in Cynomolgus monkeys. Toxicol. Sci. 68, 249-264. Seacat, A.M., Thomford, P.J., Hansen, K.J., Clemen, L.A., Eldridge, S.R., Elcombe, C.R., Butenhoff, J.L., 2003. Sub -chronic dietary toxicity of potassium perfluorooctanesulfonate in rats. Toxicology 183, 117-131. Schal, C., 1992. Sulfluramid resistance and vapor toxicity in field -collected German cockroaches (Dictyoptera: Blattellidae). J. Med. Entomol. 29, 207-215. Shen, Y.W., Taves, D.R., 1974. Fluoride concentrations in the human placenta and maternal and cord blood. Am. J. Obstet. Gynecol. 119, 205-207. Singer, S.S., Andersen, M.E., George, M.E., 1990. Perfluoro-n-decanoic acid effects on enzymes of fatty acid metabolism. Toxicol. Lett. 54, 39-46. Sohlenius, A.K., Andersson, K., DePierre, J.W., 1992. The effects of per- fluoro-octanoic acid on hepatic peroxisome proliferation and related parameters show no sex -related differences in mice. Biochem. J. 285, 779-783. Sohlenius, A.K., Eriksson; A.M., Hogstrom, C., Kimland, M., DePierre, J.W., 1993. Perfluorooctane sulfonic acid is a potent inducer of per- oxisornal Catty acid (3-oxidation and other activities known to be af- fected by peroxisome proliferators in mouse liver. Pharmacol. Toxicol. 72, 90-93. 241 Sottani, C., Minoia, C., 2002. Quantitalive determination ofperfluoroocta- noic acid ammonium salt in human serum by high-performance liquid chromatography with atmospheric pressure chemical ionization tandem mass spectrometry. Rapid Commun. Mass Spectrum. 16, 650-654. Staples, R.E., Burgess, B.A., Kerns, W.D., 1984. The embryo -fetal toxicity and teratogenic potential of ammonium perfluorooctanoate (APFO) in the rat. Fundam. Appl. Toxicol. 4, 429-440. Starkov, A.A., Wallace, K.B., 2002. Structural determinants of fluorochem- ical-induced mitochondrial dysfunction. Toxicol. Sci. 66, 244-252. Stone, L.C., Manson, J.M., 1981. Effects of the herbicide 2,4-dichloro- phenyl p-nitrophenyl ether (nitrofen) on fetal lung development in rats. Toxicology 20, 195-207. Taves, D.R., 1968. Evidence that there are two forms of fluoride in human semm. Nature 16, 1050-1051. Taves, D., Guy, W., Brey, W., 1976. Organic fluorocarbons in human plasma: prevalence and characterization. In: Filler, R. (Ed.), Biochem- istry Involving Carbon -Fluorine Bonds. American Chemical Society, Washington, DC, pp. 117-134. Thibodeaux, J.R., Hanson, R.G., Rogers, J.M., Grey, B.E., Barbee, B.D., Richards, J.H., Butenhoff, J.L., Stevenson, L.A., Lau, C., 2003. Expo- sure to perfluorooctane sulfonate during pregnancy in rat and mouse: I. Maternal and prenatal evaluations. Toxicol. Sci. 74, 369-381. U.S. Environmental Protection Agency (US EPA), 2003a. Preliminary risk assessment of the developmental toxicity associated with exposure to perfluorooctanoic acid and its salts. US EPA, OPPT-2003-0012; FRL-7303-8. U.S. Environmental Protection Agency (US EPA), 2003b. Toxicological review of perfluorooctane sulfonate (PFOS). In support of summary information on the Integrated Risk Information System (IRIS). Submit- ted for review. Uy-Yu, N., Kawashima, Y., Kozuka, H., 1990. Comparative studies on sex -related difference in biochemical responses of livers to perfluor- ooctanoic acid between rats and mice. Biochem. Pharmacol. 39, 1492 -1495. Vanden Heuvel, J.P., Kuslikis, B.I., Van Rafelghem, M.J., Peterson, R.E., 1991a. Tissue distribution, metabolism, and elimination of perfluorooc- tanoic acid in male and female rats. Biochem. Toxicol. 6, 83-92. Vanden Heuval, J.P., Kuslikis, B.I., Van Rafelghem, M.J., Peterson, R.E., 1991b. Disposition of perfluorodecanoic acid in male and female rats. Toxicol. Appl. Pharmacol. 107, 450-459. Vanden Heuvel, J.P., Davis, J.W., Sommers, R., Peterson, R.E., 1992. Renal excretion of perfluorooctanoic acid in male rats: inhibitory effect of testosterone. Biochem. Toxicol. 7, 31 -36. West Virginia Department of Environmental Protection, 2002. Final Am- monium Perfluorooctanoate (C8) Assessment of Toxicity Team (CATT) Report. August, 2002. Wetzel, L.T., 1983. Rat teratology study, T-3351, final report. Hazelton Laboratories America, Inc. Project Number: 154-160, December 19, 1983. US EPA AR-226 226-0014. Yang, Q., Xie, Y., DePierre, J.W., 2000. Effects ofperoxisome proliferators on the thymus and spleen of mice. Clin. Exp. Immunol. 122, 219-226. Yang, Q., Xie, Y., Eriksson, A.M., Nelson, B.D., DePierre, J.W., 2001. Further evidence for the involvement of inhibition of cell proliferation and development in thymic and splenic atrophy induced by the perox- isome proliferator perfluorooctanoic acid in mice. Biochem. Pharmacol. 62, 1133-1140. Ylinen, M., Hanhijarvi, H., Jaakonaho, J., Peura, P., 1989. Stimulation by oestradinl of the urinary excretion of perfluorooctanoic acid in the male rat. Pharmacol. Toxicol. 65, 274-277. Ylincn, M., Kojo, A., Hanhijarvi, H., Peura, P., 1990. Disposition of per fluorooctanoic acid in the rat after single and subchronic administration. Bull. Environ. Contain. Toxicol. 44, 46-53. DEQ-CFW 00000147