Loading...
HomeMy WebLinkAboutDEQ-CFW_00004207Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx �1? 2 Evaluation and prediction pharmacokinetics of PFOA and PFOS using3 and human a PBPKmodel 4 Anne E. Loccisano *, Jerry L. Campbell Jr., Melvin E. Andersen, Harvey J. Clewell III 5 Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park; NC 27709, United States 6 2§ ARTICLE I NFO 9 Article history: 10 Received 18 August 2010 11 Available online xxxx 12 Keywords: 13 Physiologically -based pharmacokinetic 14 modeling 15 PBPK 16 Toxicokmetic modeling 17 Perfluoroalkyls 18 Pharmacokinetics 19 PFOA 20 PFOS 21 V 1.Introduct1on A B S T R A C T Perfluoroalkyl acid carboxylates and sulfonates;,(PFAAs) have many consumer and industrial applications. 23 The persistence and widespread distribution of these compounds in humans have brought them under 24 intense scrutiny. Limited pharmacokinetic data is available in humans; however, human data exists for 25 two communities with drinking water contaminatedby PFAAs. Also, there is toxicological and pharma- 26 cokinetic data for monkeys, which can be quite useful for cross -species extrapolation to humans. The goal 27 of this research was to develop a physiologically -based pharmacokinetic (PBPK) model for PFOA and PFOS 28 for monkeys and then scale this model to humans in order to describe available human drinking water 29 data. The monkey model simulations were consistent with available PK data for monkeys. The monkey 30 model was then extrapolated to the human and then used to successfully simulate the data collected 31 from residents of two communities exposed to PFOA in drinking water. Human PFOS data is minimal; 32 however, using the half-life estimated from occupational exposure, our model exhibits reasonable agree- 33 ment with the available human serum PFOS data. It is envisioned that our PBPK model will be useful in 34 supporting human health risk assessments for PFOA and PFOS by aiding in understanding of human 35 pharmacokinetics. 36 © 2010 Published by Elsevier Inc. 37 38 39 42 Perfluorooctanoate (PFOA) and perfluorooctane sulfonate 43 (PFOS) are fully fluorinated man-made chemicals that have wide 44 spectrum of industrial and consumer uses. These compounds are 45 used as surfactants in situations requiring stability towards heat 46 and chemical degradation or other unique ,properties imparted 47 Ql by the perfluorinated chain (Lau et al., 2007). They can also be 48 formed from the degradation (environmental' or metabolic) of cer- 49 tain fluorinated materials (Olsen et al., 2007, 2003a). Due to the 50 high electronegativity of the fluorine atom, the C-F bond has a sig- 51 nificant dipole moment, imparting ionic character to the bond 52 through partial charges; the partial charges are attractive, making 53 the C-F bond one of the strongest in organic chemistry (Lau et al., 54 2007). The stability imparted to these compounds by the C-F bond 55 is very desirable industrially; they are stable in air at high temper- 56 ature, nonflammable, and not readily degraded by strong acids, 57 bases, or oxidizers (Lau et al., 007). The C-F bond also provides 58 these compounds with low surface tension making them ideal Abbreviations: PFAAs, collectively refers to perfluoroalkyl acid carboxylates and sulfonates; PBPK model, physiologically based pharmacokinetic model; PFOA, perfluorooctanoate; PFOS, perfluorooctane sulfonate. * Corresponding author. Fax: +1 919 558 1300. E-mail address: ALoccisano@thehamner.org (A.E. Loccisano). 0273-2300,'S - see front matter 2 2010 Published by Elsevier Inc. doi:10.1016(j.yrtph.2010.12.004 surfactants (Lau et al., 2004). However, the same properties that 59 make these compounds so useful make them resistant to break- 60 down by metabolism, hydrolysis, photolysis, or biodegradation 61 and thus they can be persistent in the environment indefinitely. 62 Industrial use of PFOA began in the late 1940s, and since then it 63 has been estimated that cumulative global emissions are between 64 2400 and 5200 metric tons (Prevedouros et al., 2006). Production 65 and use of PFOS and materials which can form PFOS were initially 66 greater than that of PFOA, but since the phaseout of PFOS by its 67 major manufacturer between 2000 and 2002, global production 68 has dropped (175 metric tons in 2003) (3M, 2003). 69 Although exposure sources and routes for humans are not well 70 understood, perfluoroallyl carboxylates and sulfonates (collec- 71 tively perfluoroalkyl acids or PFAAs) escape to the environment 72 through production, manufacture, disposal, and degradation of 73 other fluoropolymers (Andersen et al., 2008). These compounds 74 are now widespread throughout the global ecosystem. They have 75 been found in wildlife samples from remote ocean sites and the 76 Arctic, although concentrations are usually greater in animals liv- 77 ing in more populated and industrial areas (Lau et al., 2007). These 78 compounds have also been identified in parts -per -billion levels in 79 human serum from the general population and in parts -per -mil- 80 lion levels for occupationally exposed workers and other popula- 81 tions with high exposure to these compounds (Calafat et al., 82 2007b; Emmett et al., 2006; Olsen et al., 2007). The long plasma 83 DEQ-CFW 00004207 2 A.E. Loccisano el al./Regulatory Toxicology and Phonnocology xxx (2010) xxx-xxx 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 ill 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 half-lives of PFOA and PFOS (3-5 years) observed in humans is of particular concern because this indicates that they can accumulate, which may result in higher body burdens and potential adverse health effects. Possible exposure sources for humans include drink- ing water, dust in homes, food and food packaging, fabrics, carpet- ing, and cookware (Fromme et al., 2009; Ostertag et al., 2009; Sinclair et al., 2007; Skutlarek et al., 2006; Strynar and Lindstrom, 2008; Vestergren and Cousins, 2009; Washburn et al., 2005; Wil- helm et al., 2008). The pharmacokinetic properties of PFOA and PFOS have been studied. In summary, animal studies have shown that these com- pounds are well absorbed orally, poorly eliminated, and not metabolized (Johnson et al., 1984; Kuslikis et al., 1992; Ophaug and Singer, 1980; Vanden Heuvel et al., 1991). They are distributed mainly to the serum, liver, and kidney, usually with liver concen- trations being 3-5 times higher than serum concentrations in the rat, while liver concentrations in humans and non -human primates are only -1.3-2 times higher than in serum (DePierre, 2009; Hundley et al., 2006; Johnson et al., 1979; Kemper, 2003; Kudo et al., 2007; Olsen et al., 2003b; Seacat et al., 2002). Distribution at steady state is mainly extracellular (Butenhoff et al., 2004a; No- ker and Gorman, 2003). Both compounds have a high affinity for binding to albumin and are thus highly bound in plasma, and to a lesser extent, P-lipoproteins and liver fatty acid binding protein (Han et al., 2003; Kerstner-Wood et al., 2003; Luebker et al., 2002). Both compounds have been found in rodent fetuses as well in as umbilical cord blood of humans and in breast milk indicating that they can cross the placenta and partition into milk, exposing the fetus and neonate (Apelberg et al., 2007; Hinderliter et al., 2005; Karrman et al., 2007; Kuklenyik et al., 2004; Thibodeaux et al., 2003). The most notable aspect of PFOA and PFOS PK is that they exhibit major differences in plasma half-lives across species, and for PFOA, a sex difference in elimination is observed in rats, The elimination half-life for PFOA in female rats is 2-4 h; it is 4- 6 days in male rats (Hanhijarvi et al., 1987; Kemper, 2003; Kudo Both compounds are agonists for peroxisome proliferator activated receptor alpha (PPAR-a) and the tumor induction has been attrib- uted to this activation (Elcombe et al., 2010). However, PPAR-OE activation may not be of human relevance because humans express much lower levels of these receptors than do rodents and human liver cells have been shown to be much less responsive to the ef- fects of ammonium PFOA than rat liver cells (13jork and Wallace, 2009; Lake, 2009). Due to their prevalence and stability in the environment, toxic effects observed in animal studies, and long half-life in humans, PFAAs have drawn considerable attention from public and regula- tory agencies with regard to health risks that they may present. Risk assessments and interpretation of available human data are slowed due to lack of framework to understand and estimate hu- man pharmacokinetics. Previously, a biologically based compart- mental model was developed by our group that was able to describe the complex PK of PFOA and PFOS in rats and monkeys (Andersen et al., 2006; Tan et al,, 2008). The key process required to describe kinetics was resorption by renal transporters in the fil- trate compartment, The goal of the present work was to develop a complete PBPK model for the monkey for PFOA and PFOS and then scale up to the human, The consistency of renal resorption to de- scribe the kinetics in both the rat and monkey suggests the exis- tence of a saturable, high -affinity resorption process that governs the kinetics of these compounds in other species, including hu- mans (Tan et al., 2008). If confirmed, once the transporter protein that is responsible for renal resorption of PFOA and PFOS is identi- fied, information on the activity of that transporter can then be used to identify susceptible sub -populations due to polymorphic variation. The human model was used to successfully simulate available data and also to predict and thus examine variability in human half-lives of PFOA. The result of these efforts is a human PBPK model with reasonable predictive ability that can be used to aid in risk assessment for perfluorinated compounds. et al., 2002). The gender difference in elimination is age -dependent 2. Methods and the slower elimination in males is observed starting around 3- 5 weeks in age (Hinderliter et al., 2006). This gender difference in 11 11 F_ Dune ; 11 IF I�F_ ; '.� 1) 2. 1. Monkey model development elimination s no o serve r n rats (half S - - 50 days, depending on the isomer) or mice (half-life is -40 days) (Benskin et al., 2009; Butenhoff, 2010). Monkeys have half-lives of 21-30 days for PFOA and 4-6 months for PFOS while human half-lives are even longer for both compounds (3-5 years) (Bartell et al., 2010; Butenhoff et al., 2004a; Noker and Gorman, 2003; Ol- sen et al., 2007; Seacat et al., 2002). Neither the monkey nor human appears to have any major sex differences in elimination of either compound, and this appears to be true for the mouse also (Buten- hoff et al., 2004a; Noker and Gorman, 2003; Olsen et al., 2007; Rodriguez et al., 2009; USEPA, 2005). The cause of the differential elimination across sexes and species has not been conclusively demonstrated, but there is evidence from studies performed both in rats and in vitro with rat and human transporters that transport- ers in the proximal tubule of the kidney may be responsible (Andersen et al., 2006; Kudo et al., 2002; Tan et al., 2008; Weaver et al., 2009; Yang et al., 2010, 2009). Potential toxicities in animals have been characterized. Sub - chronic exposure to PFOS led to significant weight loss, reductions in serum cholesterol and thyroid hormones, and hepatotoxicity. Developmental toxicity studies with PFOS in rats, mice, and rabbits reveal reduction of fetal weight, cleft palate, and reduced neonatal survival (Lau et al., 2004). Repeated -dose studies with PFOA in ro- dents showed induction of liver tumors, Leydig cell tumors, and pancreatic acinar cell tumors (Andersen et al., 2008; Biegel et al., 200 1; USEPA, 2005). Reproductive toxicity studies with PFOA in ro- dents showed increased post weaning mortality, decreased body weight, and delayed sexual maturation (Butenhoff et al., 2004b). The structure for the monkey model was developed from a bio- logically based compartmental model for PFOA and PFOS in rats and monkeys (Fig. 1) (Andersen et al., 2006; Tan et al., 2008). The compartmental model contained compartments for plasma, li- ver, tissues, and filtrate, and it was developed to examine the role of renal resorption through a saturable transport process that is thought to be responsible for the long half-life of PFOA and PFOS in rats and monkeys. Free chemical in the central (plasma) com- partment can move to the liver compartment, tissue compartment, or be filtered by the filtrate (i.e., kidney) compartment. Once in the filtrate compartment, the chemical can be resorbed back to the plasma compartment by a saturable process with transporter max- imum Tm and affinity constant Kt. This model was able to success- fully describe PFOA and PFOS kinetics from oral and IV dosing in rats and monkeys, but in order to extrapolate the model to the hu- man to aid in risk assessment, a physiologically based model with realistic tissue volumes and blood flows and physicochemical and biochemical properties of the chemical under study were needed. Thus, a physiologically -based pharmacokinetic (PBPK) model was developed for PFOA and PFOS in the monkey for use in extrapola- tion to the human. 2.1.1. Study designs and data - PFOA in monkeys Pharmacokinetic data for PFOA in the monkey have been re- ported by Butenhoff et al. (2004a) for oral and IV dose routes. Both data sets were used to develop the current model. In the IV dosing 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 DEQ-CFW-00004208 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx Oral dose Liver compartment F---> feces gof m Central Compartment IV- ___ (vol. of distr., free fraction of chemical in plasma) k1 3, k31 QFill Tin, Kt Tissue Filtrate Compartment compartment (vol. of renal filtrate, renal filtrate rate, saturable resorption) storage > urine to develop the current monkey model. The IV data has been re- ported by Noker and Gorman (2003). Male and female monkeys (3(sex) were administered a single IV dose of 2 mg/kg PFOS and then monitored for 161 days after dosing. PFOS concentrations in plasma and urine of the monkeys were measured 0.5, 2, 4, 8, and 24 h post dose and on days 2, 7, 14, 21, 28, 42, 56, 70, 105, and 161 post dose. The other data used for model development was from a subchronic toxicity study of PFOS reported by Seacat et a]. (2002). Male and female monkeys received daily oral doses (by capsule) of 0.03, 0.15, or 0.75 mg/kg PFOS for 26 weeks. Two mon- keys per sex in the 0.15 and 0.75 mg/kg dose groups were allowed to recover and monitored for a year. Plasma concentrations of PFOS were measured throughout the dosing period for all 3 dose groups and during the recovery period for the mid- and high -dose groups. Significant adverse effects were observed only in the high -dose group, which included death of 2 of 6 male monkeys for which PFOS exposure could not be ruled out as a contributing cause. The monkey model was used to simulate the data from both the IV and oral studies with PFOS in the cynornoIgus monkey. Fig. 1. Structure of biologically motivated model for PFOA and PFOS in rats and monkeys. Figure adapted from Tan et al. (2008). Free chemical in the central 2.2. Model structure - monkeys compartment can move to the liver compartment based on flow rate OL and partition coefficient PL, to the tissue compartment based on the rate constants k13 The PBPK model structure for PFOA and PFOS in the monkey is and k31, or be filtered by the filtrate compartment with flow rate Qfil. Chemical can resorbed back to the plasma compartment with transporter maximum Trn and the same (Fig, 2). The model contains compartments for plasma, affinity constant Kt. gut, liver, kidney, filtrate, fat, skin, and a lumped compartment for remaining body tissues. The plasma and liver are the primary target tissues for PFOA and PFOS with possible involvement of 211 study, male and female cynornoIgus monkeys (3/sex) received a enterohepatic circulation (Butenhoff et al., 2004a; Johnson et al., 212 single IV dose of 10 mg/kg PFOA and were then monitored for 1979; Kemper, 2003; USEPA, 2005). These compounds also activate 213 123 days after dosing. PFOA concentration in plasma was mea- nuclear receptors PPAR-ot and CAR/PXR known to be associated 214 sured at 0.5, 2, 4, 8, and 24 h post dose and on days 2, 4, 7, 11, with a hypertrophic response in liver tissue (Elcombe et al., 215 14, 21, 28, 57, 79, 87, and 123. No adverse effects related to PFOA 2010; Rosen et al., 2010). The liver in some cases is the site ofaccu- 216 were reported (Butenhoff et al., 2004a). In the oral dosing study, mulation (Andersen et al., 2008; Kennedy et al., 2004; Lau et al., 217 male cynornolgus monkeys were administered a daily oral dose 218 by capsule of 0 (n = 6),3 (n = 4), 10 (n = 6), or 20 mg/kg (n = 6) PFOA 219 for 6 months. Several of the animals were then allowed to recover k Oral dose., drinking water 220 and were monitored for 90 or more days after dosing cessation Gut 221 (n = 2 for the 10 mg/kg dose group and n = 2 in the 20 mg/kg dose QGUt 222 group). Monkeys in the 20 mg/kg dose group were originally 223 started on 30 mg/kg PFOA; however, dosing had to be stopped after giver j 224 12 days due to toxicity. Dosing began again at day 21, and the dose QLiv 225 was lowered to 20 mg/kg. Dosing again had to be discontinued for Fat 226 three of the monkeys due to toxicity on day 43, 66, and 81. Only Plasma U-al - -------------------------------------- 227 two monkeys in the high -dose group received treatment for the IV Skin 228 entire 6 months. During the dosing period, PFOA concentration in Free QS!kn ------------------------------------- 229 plasma and urine of the monkeys still receiving doses were taken fraction 230 at weeks 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, and 26. During Best of body 231 recovery, plasma and urine PFOA concentrations were measured QR 232 for only the 10 and 20 mg/kg dose groups; these were measured Kidney K,'-'ne 233 once every two weeks from weeks 28 through 40 of the study. Be- QIK i d T rn, M 234 cause there was no recovery data for the 3 mg/kg dose group, and 235 the monkeys in the 30/20 mg/kg dose group exhibited signs of tox- Filtrate QH Filtraa te 236 icity and had an irregular dosing regimen, only the plasma and ur- ------------ ---- 237 ine data from the 10 mg/kg dose group was used for development 238 of the monkey model for PFOA by the oral route. Also, although the storarle 239 10 mg/kg dose would be a much higher concentration than hu- kurine 240 mans would be exposed to environmentally, the lower dose and urine 241 the effects and PK associated with it would be more relevant for 242 development of a human model. The monkey model was used to Fig. 2. Structure of PBPK model for PFOA and PFOS in monkeys and humans. 243 simulate the data from both the IV and oral studies with PFOA in Chemical is taken up into the plasma (iv) or into the gut (oral). From the gut, 244 the cynomolgus monkey. chemical is transported to the liver by the portal blood. Only the free fraction of chemical in plasma is assumed to be available for partitioning into tissues. Chemical is eliminated through the filtrate compartment to storage into urine. While in the 245 2.1.2. Study designs and data - PFOS in monkeys filtrate compartment, chemical can be reabsorbed back into the plasma through a 246 Similar to PFOA, there is existing PK data for PFOS in cynornol- .saturable process with a transporter maximum Tm and affinity constant Kt. The Qs indicate blood flows into and out of tissues. Qfil is not a blood flow; it is a clearance 247 gus monkeys for IV and oral dose routes. Both data sets were used (L/h) from the plasma to the filtrate compartment. DEQ-CFW-00004209 A.E. Loccisano el al./Regulatory Toxicology and Phonnocology xxx (2010) xxx-xxx 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 2004; Luebker et al., 2002; USEPA, 2005). The PBPK model retains the same features that were important for describing kinetics of PFOA and PFOS in the biologically based model of Andersen and Tan, namely, a free fraction of chemical in the plasma and renal resorption in the filtrate compartment (Andersen et al., 2006; Tan et al., 2008). The fat and skin compartments were built into the monkey model because these compartments will be of interest in the human. PFOA and PFOS are lipophobic, thus fat would serve as an exclusionary volume for these chemicals, and dermal absorp- tion is a possible exposure route for the human (USEPA, 2005). An IV dose to the monkey enters the plasma, and oral doses enter the gut and are then passed to the liver by the portal blood. Only the free fraction of chemical in plasma is assumed to be available to partition into tissues. Chemical is eliminated through the filtrate compartment to storage and subsequently to urine. The clearance from plasma to the filtrate compartment is described by a flow rate, QFil. In the filtrate compartment, chemical can be reabsorbed by a saturable process with a transporter maximum (Tm) and affinity constant (Kt) into the kidney and back into systemic circu- lation. Coding and simulations for both the PFOA and PFOS monkey models was performed in the Berkeley -Madonna program (Macey et al., 2000). Model code can be found in the Supplementary Material. from tissue concentration data from male C57131/6 mice (mice were fed PFOS in their diet from one to five days) (DePierre, 2009). PFOA and PFOS have been shown to be >97% bound to plasma proteins such as albumin in the rat, monkey, and human (Han et al., 2003; Jones et al., 2003; Kerstner-Wood et al., 2003); a constant free fraction of each chemical (<0.03) was used. Because of the lim- ited evidence of extensive binding for PFOA and PFOS, it was nec- essary to estimate the free fraction of chemical in plasma (Free) for each compound to provide the best fit the plasma concentration data from the IV and oral studies while adhering to the aforemen- tioned constraint. It is hypothesized that the long plasma half-lives of PFOA and PFOS are due to renal resorption by transporters in the proximal tubule of the kidney (Andersen et aL, 2006; Tan et al., 2008). In fact, more recent research has suggested that renal tubular resorption transporters are capable of transporting PFOA (Nakagawa et al., 2009; Weaver et al., 2009; Yang et al., 2010, 2009). In order to de- scribe the kinetics of these chemicals, a description of saturable re- nal resorption was implemented in the filtrate compartment in the model. The parameters used to describe renal resorption are the transporter maximum (Tm) and the transporter affinity constant (Kt). In the rat, a significant sex difference is observed in the clear- ance of PFOA but not PFOS (Andersen et al.. 2008: Kemper. 2003: Kennedy et al., 2004; Kudo et al., 2002). It has been shown that 302 303 304 305 306 307 308 309 2.3. Model parameterization 2.3.1. Chemical -specific parameters for PFOA and PFOS Chemical -specific parameters used for the monkey PFOA and PFOS models are presented in Table 1. Tissue:plasma partition coefficients for PFOA for all tissues in the model were estimated from tissue concentration data reported by Kudo et al. (2007) for a single IV dose of PFOA to male Wistar rats. Tissue:blood partition coefficients for PFOS for all tissues in the model were estimated Table I Chemical parameters for PBPK models for PFOA and PFOS in monkeys and humans. male rats exhibit higher expression of a particular organic anion transporter, Oatplal, that is thought to play a role in renal resorp- tion, thus extending the half-life of PFOA in the male rat (Weaver et al., 2009; Yang et al., 2009). However, no significant sex differ- ences are observed in elimination of either PFOA or PFOS in the monkey (Butenhoff et al., 2002, 2004a; Seacat et al., 2002), both chemicals have long half-lives in both sexes of the monkey (20-30 days for PFOA; 5-6 months for PFOS). Therefore, in the monkey model, the male and female monkeys have the same PFOA Definition and units Value Source Tmc Resorption maximum 0.15 (monkey); 10 (human; 3.8 yrs); 6 Fit to plasma concn. in monkey; estimated to give desired half-life in (mg/h(kg075)a (human (2.3 yrs) human (3.8 or 2.3 yrs) Kt Affinity constant (mg/L) 0.055 Fit plasma and urine concn. in monkey Free Free fraction of chemical in 0.02 Fit to plasma concn. in monkey plasma PL Liver:plasma partition 2.2 Rat tissue data (Kudo et al., 2007) coefficient PF Fat:plasma partition coefficient 0.04 Rat tissue data (Kudo et al., 2007) PK Kidney:plasma partition 1.05 Rat tissue data (Kudo et al., 2007) coefficient PSk Skin:plasma partition coefficient 0A Rat tissue data (Kudo et al., 2007) PR Rest of body:plasma partition 012 Rat tissue data (Kudo et al., 2007) coefficient PG Gut:plasma partition coefficient 0.05 Rat tissue data (Kudo et al., 2007) Kurine Urinary elimination rate (/h/ 50 (monkey); 3e-' (human) Fit to urine concn. in monkey and human kg -0.25) PFOS Tmc Resorption maximum (mglhl 1.3 (monkey); 3.5 (human) Fit to plasma concn. data in monkey; estimated to give desired half-life in kg 0.75r human (5.4 yrs) Kt Affinity constant (mg(L) 0.023 Fit to plasma concn. data in monkey Free Free fraction of chemical in 0.025 Fit to plasma concn. in monkey plasma PL Liver:plasma partition 3.72 Mouse tissue data (DePierre) coefficient PF Fat:plasma partition coefficient 0.14 Mouse tissue data (DePierre) PK l(idney:plasma partition 0.8 Mouse tissue data (DePierre) coefficient PSk Skin:plasma partition coefficient 0.29 Mouse tissue data (DePierre) PR Rest of body:plasma partition 0.2 Mouse tissue data (DePierre) coefficient PG Gut:plasma partition coefficient 0.57 Mouse tissue data (DePierre) Kurine Urinary elimination rate (/h/ 0.003 (monkey); 0.001 (human) Fit to urine concn. in monkey and human kg --0.25) 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 DEQ-CFW-0000421 0 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 61 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 values for Tm and Kt; i.e., we assume that the transporter(s) responsible for resorption of PFOA would be the same in both sexes. The same goes for PFOS; however, we assume that either the transporter(s) responsible for resorption of PFOS is different from that of PFOA or simply that the activity of each compound with the transporter(s) is different. PFOS has a longer half-life; the capacity and affinity of the transporter are thus higher than those for PFOA. The transporter maxima and affinity constants for each chemical were estimated by fitting them to the plasma and urine concentration data for PFOA and PFOS in both sexes. The transporter maximum was scaled as BW°.75. The urinary elim- ination rate constant (Kurine) was estimated to fit the urine con- centration data from the IV doses of chemicals. 2.3.2. Physiological parameters for the monkey model - PFOA and PFOS The physiological parameters for the monkey can be found in Table 2. Body weights for the monkeys were obtained from Buten- hoff et al. (2004a) and Seacat et al. (2002). Fractional tissue vol- umes were the values reported from Brown et al. (1997) and Forsyth et al. (1968) for rhesus monkeys. Tissue volumes were scaled linearly to body weight. Cardiac output and fractional blood flows to tissues were obtained from Fisher et al. (2000) and Forsyth et al. (1968) for rhesus monkeys. Blood flows were scaled as BW0.75. Because PFOA and PFOS do not partition into red blood cells, (Ehresman et al., 2007) blood flows were adjusted for plasma flows to tissues by multiplying the blood flow by 1-hematocrit. sure that the simulations were consistent with both the plasma and urine concentrations. The shapes of the simulated curves were assessed to make sure that they were in line with the trends ob- served in the experimental data. Agreement between the experi- mental data and the model simulations was good overall. Further efforts to refine the model parameters will require more detailed studies (i.e., plasma protein binding and transporter elucidation in the monkey). A normalized local sensitivity analysis was performed on the model to examine the influence of each model parameter on the model output. Sensitivity coefficients were calculated for the pre- dicted plasma area under the curve (AUC; total concentration) with the original parameters and for that resulting from a 1% change in each parameter value. The normalized sensitivity coefficients were calculated by the following equation: A B Sensitivity coefficient - rB A is the AUC resulting from the 1 % increase in parameter value, B is the AUC resulting from the original parameter value, C is the param- eter value increased by 1%,'and D is the original parameter value. Plasma AUC predictions for the monkeys were run at 3 and 10 mg1kg doses for PFOA and 0.03 and 0.75 mg/kg doses for PFOS. 2.5. Human model development 2.5.1. Study designs and data - human PFOA and PFOS health and pharmacokinetic data in humans is 2.4. Model evaluation available from occupational and environmental and epidemiologi- cal investigations and worker medical surveillance. Developmental For PFOA, we used the monkey model to simulate plasma and and immunotoxic effects have been reported in studies with rats urine concentrations of the chemical resulting from IV and oral and mice exposed to PFOA and PFOS. However, fetal effects in dose routes. The simulated concentrations were compared to the the offspring of treated animals were observed only at high doses experimental data of Butenhoff et al. (2002, 2004a). For PFOS, the that caused maternal toxicity (Lau et al., 2004). Similarly, the inter - monkey model was used to simulate plasma concentrations of pretation of decreased spleen and thymus weights and altered nat- PFOS from a single IV dose and daily oral doses; the simulated con ural killer cell activity in the animals in the immunotoxicity studies centrations were then compared to the experimental data of Noker were complicated by the high doses used in the studies and also re - and Gorman (2003) and Seacat et al. (2002). duced food intake (DeWitt et al., 2009; Dong et al., 2009; Qazi The adequacy of the monkey models and the corresponding et al., 2010; Yang et al., 2000). Repeat -dose studies in rodents parameters were assessed based on whether the model was able and monkeys with PFOS have shown reduced body weight, to consistently describe the available pharmacokinetic data on increased liver weight, reduced cholesterol, and a steep dose- PFOA and PFOS in cynomolgus monkeys. To evaluate the current response curve for mortality (Goldenthal, 1978; Seacat et al., monkey models, the correspondence between the data points for 2002). A chronic bioassay with PFOS in SD rats showed an increase tissue concentrations of the chemicals and the model -predicted in hepatocellular adenomas at only the highest dose of 20 ppm in values at different time points were examined visually to make the diet (Lau et al., 2007). Two separate chronic bioassays have Table 2 Physiological parameters for monkey and human PBPI{ models Physiological parameters Monkey Human Source Cardiac output (L(h/kg075), QCC 15 12.5 Forsyth et al. (1968), Brown et al. (1997) Blood flow, fraction of CO Fat, QFC 0.02 0.052 Fisher et al. (2000), Brown et al. (1997) Liver, QLC 0.2 0.25 Fisher et al. (2000), Brown et al. (1997) Kidney, QKC 0.123 0.175 Forsyth et al. (1968), Brown et al. (1997) Filtrate, QFiIC 0.0246 0.035 20% of kidney blood flow; GFR Gut, QGC 0.096 0.181 Forsyth et al. (1968), Brown et al. (1997) Skin, QSkC 0.048 0.058 Forsyth et al. (1968), Brown et al. (1997) Hematocrit, Htc 0.41 0.44 Davies and Morris (1993), Brown et al. (1997) BW (kg, average) 3.5-5.5 70 Butenhoff et al. (2004a,b), Seacat et al. (2002), Brown et al. (1997) Tissue volume, fraction of BW Liver, VLC 0.026 0.026 Brown et al. (1997) Fat, VFC 0.05 0.214 Fisher et al. (2000), Brown et al. (1997) Kidney, VKC 0.004 0.004 Brown et al. (1997) Filtrate, VFilC 0.0004 0.0004 10 0 of kidney volume Gut, VGC 0.017 0.017 Brown et al. (1997) Plasma, VP1asC 0.0448 0.0428 Davies and Morris (1993), Brown et al. (1997) DEQ-CFW 00004211 6 A.E. Loccisano el al./Regulatory Toxicology and Phonnocology xxx (2010) xxx-xxx 428 examined the turnorigenecity of PFOA. In the study by Biegel et al., measured from a cross-section (divided into public water users 494 429 PFOA was found to increase the incidence of benign hepatocellular, and bottled water users) of the Little Hocking and Lubeck popula- 495 430 Leydig cell, and pancreatic acinar cell tumors at only a high dose of tions by Bartell et al. (2010). Pre -filtration concentrations were 496 431 300 ppm in the diet (Biegel et al., 2001). In a Riker Pharmaceuticals measured in May/June 2007; filtration systems were installed in 497 432 study, PFOA increased Leydig cell hyperplasia only at a high dietary Lubeck in June 2007 and in Little Hocking in November 2007. 498 433 dose of 300 ppm (Riker, 1987). Post -filtration concentrations were measured in May/June 2008. 499 434 However, the PPAR-a mode of action that is responsible for tu- Although the population size for this study was small (N = 132 500 435 mor development in rodents is most likely not relevant for humans for Lubeck; N = 40 for LH), the model was used to simulate the 501 436 because PPAR-a activation in humans does not lead to a hyperplas- pre- and post -filtration serum PFOA concentrations. When simu- 502 437 tic response (Gonzalez and Shah, 2008; Kennedy et al., 2004; Lake, lating the LH population, the pre -filtration drinking water concen- 503 438 2009). PPAR-ot activators are non-genotoxic carcinogens, and they tration was set at 3.55 ppb; for the Lubeck population the drinking 504 439 include hypolipidemic drugs and cholesterol -lowering drugs. In water PFOA concentration ranged from 0.41 to 1 ppb (Bartell et al., 505 440 contrast to the liver tumors observed when these agents are chron- 2010), and for the simulations, 0.7 ppb (middle of the range) was 506 441 ically administered to rats and mice, there is no evidence that used. 507 442 administration over an extended period of time in humans are Another study performed by Holzer et al. examined the serum 508 443 associated with liver or any other types of tumors in humans, PFOA concentrations in a population in Arnsberg, Germany in July 509 444 which indicates a species difference in turnorigenic response (Ash- 2006 (Holzer et al., 2008). As in Little Hocking, the drinking water 510 445 by et al., 1994; Gonzalez and Shah, 2008). Epidemiology studies of had been contaminated with PFOA, and this is thought to have aris- 511 446 exposure to PFOA and PFOS and adverse health events are incon- en from farmers dumping soil conditioner mixed with industrial 512 447 elusive; in fact, no causal associations of adverse health effects to waste into nearby rivers. The average concentration of PFOA in 513 448 PFOA or PFOS exposure have been observed (Lau et al., 2007; USEP- the German public water was found to be approximately seven 514 449 A, 2009). Most of the human data available for PFOA is for a popu- times lower than that measured in Little Hocking public water 515 450 lation in Little Hocking, Ohio. The residents of this community get (0.519 ppb versus 3.55 ppb) (Holzer et al., 2008). The data in the 516 451 their drinking water from the Ohio River, which is adjacent to a HbIzer et al. study was from approximately 355 people (n = 101 517 452 plant in Washington, West Virginia that has manufactured fluoro- for men, n = 164 for women, n = 90 for kids); as in the Emmett 518 453 polymers since 1951. The highest levels of PFOA in public water et al. study, only a mean and standard deviation for the population 519 454 supplies in the US have been found in the Little Hocking water sys- surveyed was available, and it is not known how long the people 520 455 tem (Emmett et al., 2006). Emmett et al. measured the serum PFOA had been exposed to PFOA; however, it is thought to be only a 521 456 levels in residents of the Little Hocking population as well as the few years in contrast to the Little Hocking population that has been 522 457 concentration of PFOA in the public drinking water (mean concen- exposed for over 30 years (Holzer et al., 2008). After the study was 523 458 tration = 3.55 ppb), which is the primary route of exposure for conducted in July 2006, a filtration system was installed to remove 524 459 these residents (Emmett et al., 2006). The available data from this the PFOA in the drinking water; the PFOA concentrations in the 525 460 study for Little Hocking was from 291 individuals whose primary drinking water were reduced to concentrations below the detec- 526 461 water source was the public water system. Only a mean, median, tion limit (LOD was 10 ng/Q. Hblzer et al. measured the serum 527 462 and interquartile range is available for the population's serum PFOA concentrations again in July 2007 to quantify the changes 528 463 PFOA concentration; we were not able to obtain individual data. in serum concentration (Holzer et al., 2009). 288 participants from 529 464 Also, it is unknown for how long the residents were exposed to the original 2006 study were included in the 2007 study. In addi- 530 465 the contaminated water. No data was collected on serum PFOS tion to Little Hocking, we used the human model to simulate the 531 466 concentrations in this study. Aside from Emmett's data, more data serum PFOA concentrations in Arnsberg in both 2006 and 2007. 532 467 for the Little Hocking population is available on the Little Hocking The half-lives of PFOA and PFOS are much longer in the human 533 468 Water Association (LHWA) website (LHWA, 2005), This data con- than in laboratory animals; the half-lives in humans are reported 534 469 tains serum PFOA, PFOS, and several other perfluorinated chemical to be on the order of several years (Andersen et al., 2008; Olsen 535 470 concentrations for some residents (N = 25) as of May 2005 in addi- et al., 2007). Two studies have been performed to investigate the 536 471 tion to gender, age, and exposure duration. Both data sets were half-life of PFOA, and the gender difference in elimination of PFOA 537 472 used to evaluate the predictive ability of the human model. in the rat has not been observed in humans (Olsen et al., 2007). The 538 473 Another data set from the Little Hocking population used to test first, reported by Olsen et al. (2007) examined the half-lives of sev- 539 474 the model was from the CS Health Project, which was conducted as eral perfluorinated chemicals, including PFOA and PFOS, in 26 re- 540 475 part of a settlement of a class-action lawsuit (Leach v. E.I. du Pont tired fluorochernical workers. The average serum half-life 541 476 de Nemours & Co., No. 01C-608 (W. Va., Wood County Cir. Ct. filed estimated for PFOA was 3.8 years and that for PFOS was 5.4 years. 542 477 Apr. 10, 2002)) from August 2005-July 2006. The participants in The second study was performed by Bartell et al. (2010) using the 543 478 the study were from Little Hocking and surrounding communities pre- and post -filtration serum PFOA concentration of 200 residents 544 479 that were served with public drinking water, and the purpose of of Little Hocking and the nearby town of Lubeck. The average ser- 545 480 the study was to assess the serum PFOA levels in the residents, um half-life of PFOA estimated from their study was 2.3 years. We 546 481 The results of the CS Health Project were published by Steenland considered both half-life estimates when running the model 547 482 et al. (2009), and the average serum PFOA concentration was simulations. 548 483 -227 ppb, which is approximately half of what was measured by PFOA and PFOS have been detected in the serum of the US gen- 549 484 Emmett et al. (2006) (mean -448 ppb). The Steenland et al. study eral population; however, the chemicals are present in much lower 550 485 did not give a drinking water concentration of PFOA; thus, the levels than what has been found in workers and people drinking 551 486 range from the Emmett study (1.5-7.2 ppb) was tested for simulat- the contaminated water. Exposure pathways for the general popu- 552 487 ing the serum PFOA concentrations. The concentration chosen from lation are not well characterized (Andersen et al., 2008; Calafat 553 488 the Emmett study to simulate the Steenland et al. data was 1.5 ppb et al., 2007a; Paustenbach et al., 2007). Calafat et al. reported and 554 489 as this concentration gave the best fit to the data when a 3.8 year compared the serum concentrations of several perfluorinated 555 490 half-life was assumed. chemicals of the general population from 1999-2000 and 2003- 556 491 To reduce exposure to PFOA, activated carbon filters were in- 2004 National Health and Nutrition Examination Survey (NHANES) 557 492 stalled in Little Hocking and the surrounding communities in (Calafat et al., 2007b). Due to reduction in production and manu- 558 493 2007. Pre- and post -filtration serum PFOA concentrations were facturing emissions of these chemicals by 2002, the serum concen- 559 DEQ-CFW-00004212 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx—xxx 7 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 trations of PFOA and PFOS were found to have decreased in the general population (Calafat et al., 2007b). We used the human model to estimate the exposures of the general population to PFOA and PFOS from both 1999-2000 and 2003-2004. Olsen et al. also examined the decline in serum PFOA and PFOS concentrations (Olsen et al., 2003a, 2008). In these studies, the ser- um of Red Cross adult blood donors in six geographic locations (100 donors from each location) around the US was analyzed for PFOA, PFOS, and five other fluorochemicals. The first study was conducted in 2000-2001; the follow-up study was performed in 2006. As with the NHANES participants, a decrease in the levels of PFOA and PFOS was observed in the serum of the Red Cross do- nors. The human model was used to estimate the exposure of all Red Cross donors to PFOA and PFOS in 2000-2001 and in 2006. 2.6. Model structure - human The PBPK model structure for PFOA and PFOS in the human is the same as that for the monkey (Fig. 2); extrapolation to the hu- man was the principal reason for development of the monkey model. As mentioned in the methods section for the monkey mod- el, the skin compartment was built in because dermal uptake is of interest for the human, and the fat compartment serves as a restriction volume; a larger fat compartment would result in a smaller volume of distribution for these chemicals. Coding and simulations for both the PFOA and PFOS human models was per- formed in the Berkeley -Madonna program (Macey et al., 2000). Model code can be found in the Supplementary Material. resorption process that is hypothesized to be responsible for the kinetics and long half-life in the monkey is also responsible for the persistence in humans (Andersen et al., 2006). If this is con- firmed and the transporter protein(s) that is/are responsible for re- nal resorption of these chemicals is/are identified, information on the activity of that transporter can then be used to identify suscep- tible sub -populations due to polymorphic variation. The human PBPK model thus includes the description of renal resorption that we included in the monkey to allow for eventual incorporation of data describing human population variability in renal transporter activities. We assumed that the transporter protein would be the same one in the monkey and the human; studies on the cloning and function of renal transporters in the monkey suggest that the monkey is a good predictor of renal uptake of organic anions in the human (Tahara et al., 2005). Therefore, the transporter affinity constant (Kt) is the same in the human as was estimated for the monkey. We then adjusted the transporter capacity (Tm) for each chemical in the human to get the correct plasma half-lives. Since the clearance of both chemicals is so much slower in the human, the transporter capacity is greater than in the monkeys. 2.7.2. Physiological parameters for the human model - PFOA and PFOS The physiological parameters used for the human models can be found in"rable 2. Body weight, cardiac output, fractional tissue volumes, and fractional blood flows to tissue were the values re- ported by Brown et al. (1997). Tissue volumes were scaled linearly to body weight and blood flows were scaled as BW1_75. To adjust for plasma flows to tissues tissue blood flows were multiplied by I- hematocrit. 586 2.7. Model parameterization - human 587 2.7.1. Chemical -specific parameters for PFOA and PFOS 3. Results 588 Chemical -specific parameters used for the human PFOA model 589 are presented in Table 1. The tissue -plasma partition coefficients 3.1. Monkey model for PFOA 590 used for both chemicals were the same as those used for the mon- 591 key. Fiserova-Bergerova (1975) examined partition coefficients for 3.1.1. IV data 592 two compounds (Forane and methylene chloride) in several spe- The simulated plasma and urine PFOA concentrations for a sin- 593 cies (man, monkey, dog, and rat). Tissue:blood partition coeffi- gle IV dose of 10 mg/kg PFOA to the monkey were in good agree- 594 cients were very similar for the tissues examined across species; ment with the experimental data (Fig. 3). There appears to be 595 thus, it is reasonable to use the same partition coefficients in both some variability in the plasma clearance of PFOA for the monkeys; 596 the human and monkey models. For PFOA, the PCs were estimated at about 60 days post dose, some monkeys have eliminated most of 597 from tissue concentration data reported by Kudo et al. (2007) for a the chemical while some still have higher plasma concentrations. 598 single IV dose of PFOA to male Wistar rats. For PFOS, PCs were esti- Adjustment of the transporter maximum (Tm) will affect how 599 mated from tissue concentration data from male C57131/6 mice slowly or quickly the chemical is eliminated in the model. The 600 (DePierre, 2009). Only the free concentration of chemical is as- I'm estimated to the average plasma concentration provided the 601 sumed to be available to partition into tissues. As in the monkey best fit to the data. 602 and rat, PFOA and PFOS are highly bound to plasma proteins in 603 the human. The species differences in half-lives of these com- 3.1.2. Oral data 604 pounds does not appear to be due to differences in protein binding, As mentioned in Section 2, only the data from the 10 mg/kg dai- 605 (Han et al., 2003) so we used the same free fraction of chemical ly oral dose of PFOA to the monkeys was used for model develop- 606 (Free) estimated for the monkeys for PFOA and PFOS in the human ment. The simulated plasma and urine concentrations are in good 607 models. Urinary elimination rate constants were scaled as BW-0-25 agreement with the observed data from the 10 mg/kg dose (Fig. 4). 608 and estimated by fitting the model to the data of Harada et al. The model simulates the fast rise to steady state in the plasma and 609 (2005), where serum and urine PFOA and PFOS concentrations the slower terminal half-life. The model was used to simulate the 610 were measured in residents in Kyoto City, Japan. 30/20 mg/kg daily dose, and the irregular dosing regimen reported 611 Similar to the monkey, humans have long half-lives for these in the study was used. However, the observed experimental data 612 chemicals; however, instead of months as for the monkey, the hu- for the 30/20 mg/kg dose produced different shapes of plasma 613 man plasma half-life is on the order of several years. Based on a and urine concentration curves from the 10 mg/kg dose during 614 study of retired fluorochemical workers, the estimated arithmetic the elimination phase. Tm could be increased to fit the high dose 615 mean serum elimination half-life in the human is approximately data (data not shown); however, because the monkeys in the 616 3.8 years for PFOA (geometric mean was 3.5 years) and 5.4 years high -dose group exhibited toxicity, the data observed may be 617 for PFOS (geometric mean was 4.8 years) (Olsen et al., 2007). A due other effects, such as impaired renal function (decreasing the 618 more recent study conducted on a population that had previous rate of flow to the filtrate compartment also allowed for fitting of 619 exposure to PFOA in drinking water estimated a plasma half-life the high dose data). The lower dose is also more relevant for devel- 620 for PFOA of2.3 years (Bartell et al., 2010). The long half-lives across opment of the human model, so we chose to use the parameters 621 species (monkey to human) suggests that the same saturable renal that best fit the 10 mg/kg dose. DEQ-CFW-00004213 8 A.E. Loccisano el al./Regulatory Toxicology and Phonnocology xxx (2010) xxx-xxx 100 0.01 0 20 40 60 80 days after dosing 100 0.01 4 --------------------------------------- 7 ------------------- I ------------------- I ------------------- 7 ------------------- 7 ------------------- I 0 5 0 15 20 25 30 35 days after dosing Fig. 3. Comparison of model simulations (lines) with experimental data (points) from Butenhoff et al. (2004). The plots contain PFOA plasma (top) and urine (bottom) concentrations in monkeys administered a single 10 mg/kg iv dose of PFOA and then allowed to recover. Females in triangles; males in squares. 100 0.01 0 10 0.01 0,001 50 '100 150 200 250 7300 350 400 thne (days of study) 0 50 100 150 200 250 300 350 400 time (days of study) Fig. 4. Comparison of model simulations (lines) with experimental data (points) from Butenhoff et al. (2004). The plots contain PFOA plasma (top) and urine (bottom) concentrations in male monkeys administered daily gavage doses of 10 mg/kg PFOA for 26 weeks and were then allowed to recover. 682 3.2. Monkey model for PFOS 683 3.2.1. IV data 684 The simulated plasma and urine PFOS concentrations for a 685 single IV dose of 2 mg/kg PFOS to monkeys exhibits good correspondence with the observed data of Noker and Gorman (Fig. 5). Up to about 70 days post dose the monkeys exhibit some variability in plasma elimination of PFOS, but at the last two time points, the animals have similar plasma PFOS levels. Because PFOS appears in the urine at a slower rate than it disappears from the plasma, a storage compartment was added to the model. This "de- lay" compartment was also used in the compartmental model of Andersen et al. (2006) and Tan et al. (2008) and its use allows for a better fit to the urine data. The biologically based model by Tan et al. (2008) for PFOS in the monkey incorporated a time -depen- dent description for the free fraction of chemical in the plasma (this was necessary to describe the kinetics resulting from the IV dose). This description was not necessary to implement in the PBPK model in order to describe PFOS pharmacokinetics; thus, the free fraction (Free) was kept constant. 3.2.2. Oral data The simulated PFOS plasma concentrations resulting from an oral dose of 0.03, 0.15, or 0.75 mg/kg daily to the monkeys are shown in Fig. 6. Overall, the simulated concentrations are in good agreement with the experimental data of Seacat et al. (2002). The 0.15 mg/kg dose lines up well with the experimental data while the highest dose is slightly overestimated by the model, especially during the dosing period. The model simulated a sharper rise in plasma concentration and faster approach to steady state during the dosing period that what was observed in the experimental data. Adjustment of Tm and Kt allowed for better fitting of the high dose curve; however, the best overall fit for all three doses was with the Tm and Kt for the 0.15 mg/kg dose. 3.3. Sensitivity analysis The normalized sensitivity coefficients for the model parame- ters for PFOA and PFOS in the monkey with respect to serum PFOA _J E -a Z W 0 LL Q. W 1 4 50 100 150 200 days after dosing 0 o'c"'... 0 20 do 60 80 100 days after dosing Fig. 5. Comparison of model simulations (lines) with experimental data of Noker and Gorman (2003). The plots contain PFOS plasma (top) and urine (bottom) concentrations after a single iv dose of 2 mg/kg PFOS. Male and female monkeys were administered an iv dose of PFOA and allowed to recover for 5 months. Squares -males, triangles -females. 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 DEQ-CFW-00004214 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 9 350 300 250 _J E 200 0 150 E E 0 100 50 0 0 100 200 300 400 600 700 time (days) Fig. G. PFOS in monkey plasma during and after oral closing for 6 months. Male and female monkeys were administered 0.03, 0.15, or 0.75 mg/kg PFOS orally for 6 months and monitored for a vear after dosing stopped. 717 or PFOS AUC are shown in Fig. 7. Only parameters with sensitivity 718 coefficients greater than 0.1 are shown. For PFOA, sensitivity anal- 719 ysis was performed at 3 and 10 mg/kg daily doses. There do not ap- 720 pear to be any dose -dependent differences in model sensitivity to 0.5 .2 0 0 U _0.5 IL 0.5 .2 0 0 W ] 0 * CC QRIC BW VUvC Htc Tmc Kt Free PLiv PTis -2 Fig. 7. Calculated sensitivity coefficients for PBPK model parameters for PFOA (top) and PFOS (bottom) in monkeys with respect to serum PFOA or PFOS AUC. Only parameters with SCs > 0.1 are shown on the plots. any of the parameters shown. Serum PFOA is primarily dependent on the free fraction of chemical in the plasma and the parameters governing elimination, which are the flow to the filtrate compart- ment and the transporter maximum. Cardiac output and hemato- crit will both influence the plasma flow to the filtrate compartment since blood flows are dependent on cardiac output and also must be adjusted for plasma flows because PFOA does not partition into red blood cells. For PFOS, sensitivity analysis was performed at 0.03 and 0.75 mg/kg daily doses. Unlike PFOA, there appear to be some dose -dependent differences in model sen- sitivity at the low and high doses of PFOS. At 0.03 mg/kg/day, ser- um PFOS appears to be more dependent on liver volume, liver:plasma partitioning, and rest of body:plasma partitioning than at the higher dose. At 0.75 mg/kg, the free fraction of PFOS in plasma and the elimination parameters (QFiIC, Tmc) appear to be more important than at 0.03 mg/kg. At the higher dose, uptake into tissues may be limited and clearance parameters show a greater influence on serum PFOS. 3.4. Human model 3.4.1. Comparison with available data For PFOA, we used the human model to simulate plasma con- centrations of the chemical resulting from exposure to contami- nated drinking water in both the Little Hocking, Ohio and Arnsberg, Germany populations. The adequacy of the human PFOA model and the corresponding parameters were assessed based on whether the model was able to describe the available plasma con- centration data on PFOA in humans, which exhibits high variabil- ity. Overall, agreement between the experimental data and the model simulations was good. Further efforts to refine the model parameters will require more detailed kinetic studies in the human. The exposure conditions (drinking water rate, concentra- tion of PFOA on drinking water) used to simulate the different populations are in Table 3. 3.4.1.1. Little Hocking, Ohio. Since it is not known how long the peo- 754 ple had been exposed (i.e., drinking and using the water), the mod- 755 el was run until the plasma PFOA concentration reached steady 756 state and until the model predictions were within the measured 757 DEQ-CFW-00004215 10 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 Table 3 Exposure estimates used in simulations with human PBPK model for PFOA. Parameters Values Sources PFOA in water Mean: 3.55 ppb (Little Hocking)Mean: 0.519 ppb(Arnsberg) Emmett et al. (2006) and Holzer et al. (2008) Intake Mean: 11 mL/kg/day(range:1-95) EPA-822-R-00-008 PFOA in serum (Little Hocking)(N = 291) Mean: 448 µg/LMedian: 374 µg/Llnterquartile range:221-576 µg(L Emmett et al. (2006) PFOA in serum Mean: 28.5 µg/LSD: 12.9 Holzer et al. (2008) (Arnsberg, Germany)(N = 101: men only) mean and standard deviation. For the Little Hocking population, the model had to be run for approximately 30 years before steady state and the mean PFOA plasma concentration was reached. In addition to malting sure the model reached steady state and the mean/SD, we made sure that we had adjusted the transporter max- imum (Tm) to yield the proper half-life. When simulating the Little Hocking population, we assumed that their only exposure to PFOA was through the drinking water as this was their primary exposure source. Because there are two different plasma half-life estimates that we found in the literature, (Bartell et al., 2010; Olsen et al., 2007) the model was run assuming both half-lives. Fig. 8 shows the plot of simulated plasma concentrations with the measured data from Emmett's study. From the plot, it can be seen that either half-life (3.8 versus 2.3 years) is possible; both yield simulated concentrations that are within the mean and SD. The Little Hocking Water Association (LHWA) website contains plasma PFOA concen- tration data for 25 residents. The average and standard deviation calculated from this data were slightly different that reported in Emmett's 2006 study, so for comparison with the simulations, both data sets (mean and standard deviations for LHWA website data (triangles) and mean and interquartile range for Emmett data (squares)) are shown in Fig. 8. In addition to the data from Emmett et al. and the LHWA web site, data collected from the participants in the C8 Health Project was also simulated with the human model. The purpose of the C8 Health Project was to assess the serum PFOA levels of the LH and surrounding area residents that were served with water con- taining PFOA. This data was collected from August -2005 July 2006 and was published by Steenland et al. (2009). As with the Em- mett data, the model was run for 30 years of exposure to order to r�tiir 600 500 o)400 Lo 300 a- 200 100 0 reach steady state, and both available estimates of the plasma half- life for PFOA were tested (2.3 versus 3.8 years). Fig. 9 shows the plot of the simulated plasma concentrations along with the mea- sured data from both Emmett's study and the C8 Health Project study. For this plot, a drinking water concentration of 3.55 ppb was used. The mean serum PFOA concentration measured from the LH residents participating, in the C8 Health Project is —227 ppb, which is approximately half that measured in the Em- mett et al. study (-448 ppb); however, the mean value from the C8 Health Project overlaps with the lower end of the interquartile range (221-576 ppb) measured in Emmett's study. The serum half-life of PFOA is not the only factor that probably has variability for this population. No drinking water concentra- tions of PFOA were listed in the Steenland et al. study; however, in the Emmett et al. study, the measured drinking water concentra- tions ranged from 1.5 to 7.2 ppb (mean = 3.55 ppb). In Fig. 9, the 3.8 year half-life appeared to overestimate the serum PFOA con- centrations measured from the C8 Health Project participants, so the model was then run with a reduced drinking water concentra- tion (1.5 ppb) but the half-life was kept at 3.8 years. Fig. 10 shows the results of this (bottom) and also the model predictions plotted with the Emmett et al. data (top) and the 3.55 ppb intake. Lowering the drinking water concentration allowed for good predictions with the 3.8 year half-life. The people in the Emmett cohort were from LH only, which was the area with the highest water concen- tration of PFOA, while the participants in the C8 Health Project were from LH and surrounding communities where the water con- centration of PFOA was lower; thus, it is reasonable that the people from the C8 Health Project have lower serum PFOA concentrations. In addition to variability in serum half-life and the concentration of 0 5 10 15 20 25 30 35 40 45 50 time exposed (years) Fig. 8. Comparison of model simulations (lines) with experimental data from Emmett et al. (2006) (squares) and the Little Hocking Water Association website in 2005 (triangles). The plot contains PFOA plasma concentrations for the Little Hocking, OH population that was exposed to drinking water contaminated with PFOA (3.55 ppb). The simulations were run for an exposure period of 30 years assuming two different half-lives: 3.8 years (red curve) from a study of occupational workers by Olsen et al. (2007), and 2.3 years (blue curve) from a study by Bartell et al. (2010). Both half-lives appear to be probable. 788 789 790 791 792 793 794 795 796 797 798 799 Soo 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 DEQ-CFW 00004216 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 191 818 819 820 700 600 500 a 400 d 0 300 LL 200 100 0 0 ;> 10 15 20 25 30 35 40 45 50 time exposed (years) Fig. 9. Comparison of model simulations (lines) with experimental data from Emmett et al. (2006) (squares) and Steenland et al. (2009) (triangles). The plot contains PFOA plasma concentrations for the Little Hocking, OH population that was exposed to drinking water contaminated with PFOA. Both data sets were collected in 2005-2006. The simulations were run for an exposure period of 30 years assuming a half-life of 3.8 years (top curve) and 2.3 years (bottom curve). The data from Steenland et at. (2009) is from the participants in the C8 Health Panel. The mean serum concentration is —227 ppb, about half of that measured by Emmett et al. in 2006 (mean —448 ppb). A drinking water concentration of 3.55 ppb PFOA was assumed. 700 600 500 400 0 300 u_ o. 200 100 0 350 300 250 u 200 CO 1;>0 o< 100 50 0 0 10 20 30 40 50 time exposed (years) 0 10 20 30 40 50 time exposed (years) Fig. 10. Comparison of model simulations (tines) with experimental data from Emmett et al. (2006) (top, squares) and Steenland et at. (2009) (bottom, triangles). The plots contain PFOA plasma concentrations for the Little Hocking, OH population that was exposed to drinking water contaminated with PFOA. Both data sets were collected in 2005-2006. The simulations were run for an exposure period o 30 years and a serum half-life of 3.8 years was assumed. The water concentration of PFOA for simulating the Emmett data sets was set at 3.55 ppb; however, for the Steenland data, the drinking water concentration had to be reduced to 1.5 ppb if a 3.8 year half-life is used. The water concentrations in LH between 2002 and 2005 were found to range from 1.5 to 7.2 ppb (Emmett et at., 2006), so the people studied by Steenland et al. simply may have been in a lower exposure region. Variable water concentrations and/or half-lives of PFOA in individuals is likely. PFOA in drinking water in various regions, drinking water rates and how much exposure comes from drinking water versus other sources most likely differ among the populations examined as well. To reduce exposure to PFOA in the drinking water, an activated carbon filtration system was installed in Little Hocking and the nearby community of Lubeck in 2007 (Bartell et al., 2010). Pre - and post filtration serum PFOA serum concentrations were mea- suredby Bartell et al. in a cross-section of the populations, and the participants were divided into public water or bottled water users (Bartell et al., 2010). The model was used to simulate the ser- um PFOA concentrations of the public water users in both Lubeck and LH (Fig. 11). For both Lubeck and LH, both available serum half-lives of PFOA were taken into consideration when running the simulations. For LH public water users (top), exposure was as- sumed to be 30 years (to reach steady-state) plus 6 months, as fil- tration systems were installed 6 months after the pre -filtration serum concentrations were measured. The pre -filtration water concentration was assumed to be 3.55 ppb; when filtration started, the exposure was simply shut off, as the PFOA water concentration after the filtration system was installed was nonquantifiable or nondetectable (Bartell et al., 2010). For Lubeck public water users (bottom), exposure was assumed to be 30 years and then it was shut off; filtration systems were installed in Lubeck right after the pre -filtration serum concentrations were measured. PFOA in drinking water samples in Lubeck ranged from 0.41 to 1 ppb be- tween 2001 and 2006; 0.7 ppb (middle of this range) was used in running the simulations. Due to the small population sizes used in the Bartell et al. study, the ± SD from the mean for the post -fil- tration serum PFOA concentrations are quite small. However, from the pre -filtration concentrations, it appears from Fig. 11 that either the 2.3 or 3.8 year half-life is possible. A larger population size would be helpful in examining the extent of the variability in ser- f um PFOA concentration. 3.4.1.2. Arnsberg, Germany. As with the people of Little Hocking, it is not known how long the people of Arnsberg were exposed to PFOA in their drinking water. It is believed that the contamination occurred in the time period between 2000 and 2006; this popula- tion was therefore not exposed as long as the people of Little Hock- ing (Holzer et al., 2008). The concentration of PFOA in the drinking water was approximately 7 times lower than that measured in Lit- tle Hocking (0.519 ppb versus 3.55 ppb). For the Arnsberg popula- tion, the plasma PFOA concentration reached measured mean DEQ-CFW 00004217 12 A.E. Loccisano et al./Regulatory Toxicology and Phonuacology xxx (2010) xxx-xxx 860 861 862 863 800 700 600 < 500 - 0 400 E 300 �y 200 - 100 0 25 3.8 year haif-life 2.3 year nalf-life spring 2007 n spring 2008 LH .................................................................:::..>. 27 250 3.8 year half-life 200 2.3 year half-life spring2007 spring2008 0 150 uv Ei E 100 - 50 0 1- 0 0 0 25 27 29 31 33 35 0 10 20 30 40 50 time exposed (years) time (years) Fig. 11. Comparison of model simulations (lines) with experimental data from Fig.13. Comparison of model simulations (lines) with serum PFOA concentration of Bartell et al. (2010) (points). The plots contain PFOA plasma concentrations for the general population (points). The plots contain the simulated serum PFOA concen- Little Hocking (top) and Lubeck (bottom), OH populations that were exposed to trations using a half-life of 2.3 years (top) and 3.8 years (bottom). Estimated general drinking water contaminated with PFOA. Serum PFOA was measured in spring 2007 population exposure was simulated as a direct input to blood. For a 2.3 year half - (before filtration systems were installed; diamonds) and in spring 2008 (-6 months life, the estimated exposure range from 1999 to 2000 was 4.25-5.2e-4 ug(kg/day after filtration started; squares). The simulations were run for an exposure period of and 3.3-3.9e-4 ug(kgjday for 2003-2004. For a 3.8 year half-life, the estimated 30 years assuming a half-life of 3.8 years (top curves) and 2.3 years (bottom curves). exposure range was 2.6-3.2e 4 ug(kgjday for 1999-2000 and 2.0-2.4e -4 ug(kg/day Average drinking water concentration of PFOA in LH was 3.55 ppb and 0.7 ppb in for 2003-2004. Lubeck The population size for this study was small (N = 132 for Lubeck; N = 40 for LH); however, both half-lives appear to be likely. installed by the waterworks. The drinking water concentration within a few years (Fig. 12); however, the plasma concentration was lowered considerably by filtration (mostly below the LOD, does not reach steady state in this time. Two studies were per which was 10 ng/L). The follow-up study was then performed in formed on this population. The initial study was performed in July' July 2007 to examine the effects of the filtration on serum PFOA 2006, (Holzer et al., 2008) after which a filtration system was concentration (Holzer et al., 2009). The authors of the studies 10 20 30 40 50 time (years) qr 30 25 ra 20 0 tL a- 15 E as 10 5 0 0 0.5 1 1,5 2 2.5 3 3.5 4 4.5 5 time Gears exposed) Fig. 12. Comparison of model simulations (tines) with experimental data from Holzer et al. (2008) and Holzer et al. (2009) (points). The plot contains PFOA plasma concentrations for a population in Arnsberg, Germany that was exposed to drinking water contaminated with PFOA (0.519 ppb). A filtration system was installed right after the initial serum concentrations were measured (July 2006), and the serum PFOA concentrations were measured a year after this (July 2007). The exposure duration is unknown, but it appears to be on order of a few years. The simulations pictured were run for two years assuming two different half-lives: 3.8 years (blue curve) from Olsen et al. (2007) and 2.3 years (red curve) from Bartell et al. (2010). As for the Little Hocking population, both half-lives appear to be likely. 864 865 866 867 868 DEQ-CFW 00004218 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 13 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 measured the plasma concentration of PFOA in men, women, and F5 10 children; however, since the means were similar for all groups 70 (28.5 ppb for men, 26.7 ppb for women, and 24.6 ppb for kids in 60 the initial study; 23.4 ppb for men, 18.8 ppb for women, and 17.4 for kids in the follow-up study), only the simulations for the men are shown in Fig. 12. As with the Little Hocking simulations, we as- 40 0 surned that drinking water was the only source of exposure to LL 30 CL PFOA and the simulations were run using both a 3.8 and 2.3 year 20 half-life. From Fig. 12, it can be seen that either half-life fits with 10 the data (although the 3.8 year half-life yields a slightly better fit). 3.4.1.3. General population. PFOA and PFOS have also been mea- 0 10 20 30 40 50 time exposed (years) sured in the blood of the general population (Calafat et al., 2007a; Calafat et al., 2007b). These levels are much lower than 40 b those found in the serum of the residents of Little Hocking and 35 Arnsberg; however, the exposure sources are unknown and could 2 31) 1999-2000 .................... potentially be from a number of sources, including drinking water, 25 food and food packaging, consumer products, and in the air. Serum 0 20 LL 2003-2004 concentrations of PFOA and PFOS from the general population are available from 1999-2000 to 2003-2004 from the National Health E 5 and Nutrition Examination Survey (NHANES). We used the model 10 to simulate the serum concentrations and to estimate how much 5 PFOA and PFOS the general population might be exposed to daily. 0 ----------------------- T ----------------------------------- I ----------------------------------- I ----------------------------------- 1 Figs. 13 and 14b show the comparison of the model simulations 0 20 40 60 80 with the measured concentrations reported by Calafat et al. time exposed (years) (2007b). The top plot in Fig. 13 shows the simulation for PFOA assuming a 2.3 year half-life and the bottom plot assumes a Fig. 14. (a) Comparison of model simulations (lines) of serum PFOS concentration 3.8 year half-life. Fig. 14b shows the simulation for PFOS assuming with Little Hocking Water Association website data (squares). The data from the webSiLe is the average and SO of the plasma concentrations of residents in Little a 5.4 year average serum elimination half-life (Olsen et al., 2007). Hocking (N = 25). The concentration of PFOS in the drinking water was not reported Because the exposure sources of PFOA and PFOS are unknown, (or unknown) at this time. As for PFOA, the simulation for PFOS was run to reach the simulations were performed with chemical exposure being a steady state (30 years) and to reach the average concentration measured in plasma direct input to blood (effectively an IV dose). The major US manu- (the concentration of PFOS in the drinking water was estimated). A half-life of facturer of PFOS and compounds that can degrade to PFOS ceased 5.4 years (from occupational data of Olsen et al.) was assumed for the simulation. (b) Comparison of model simulations (lines) with serum PFOS concentration of all production by the end of 2002, and, while PFOA continues to general population (points). The plots contain the simulated serum PFOS concen- be produced and used in the US, emissions have been reduced. This trations using a half-life of 5.4 years. Estimated general population exposure was is apparently reflected in the serum concentrations of both PFOA simulated as a direct input to blood. The estimated exposure range was 2.3- 3.Oe -' ug/kg/day for 1999-2000 and 1.6- 1.9e -' ug/kg/day for 2003-2004. and PFOS: the geometric mean of the serum PFOA concentration decreased from 5.2 ppb in 1999-2000 to 3.1 ppb in 2003-2004 and from 30.4 to 20.7 for PFOS. As far as exposure estimates go, if a 2.3 year half-life is assumed for PFOA the exposure range is chemicals was assumed to be constant (the simulation was run 4.25-5.2e 4 ug/kg/day in 1999-2000 and 3.0-3.9e -A ug/kg/day in so that concentrations reached steady state, but exposure was 2003-2004. If a 3.8 year half-life is assumed, the exposure range not stopped; Figs. 15 and 16, top panels). For PFOS, the half-life is 2.6-3.2e --4 ug/kg/day in 1999-2000 and 2.0-2.15e -4 ug/k,g/day was assumed to be that estimated from retired fluorochernical in 2003-2004. For PFOS, the exposure range is 2.3-3.0e-ug/kg/ workers (arithmetic mean of 5.4 years) by Olsen et al., for PFOA, day in 1999-2000 and 1.6-1.9e --3 ug/kg/day in 2003-2004. These both the 3.8 year arithmetic mean half-life estimated by Olsen et ranges were estimated using the 95% confidence intervals for the al., and 2.3 year half-life estimated by Bartell et al. were considered geometric means reported by Calafat et al. (2007b). For PFOA, if (Bartell et al., 2010; Olsen et al., 2007). The estimated daily expo - we compare these estimates to what the average person in Little sure to PFOS in 2000-2001 was 2.2-4.5e -3 ug/kg BW; in 2006, it Hocking ingested daily in 2006 (assuming consumption of 11 mL was estimated between 8.5e --4 and 1.8e --3 ug/kg BW. The esti- water/kg BW/day and 3.55 ppb PFOA in drinking water), the gen- mated daily exposures for PFOA in 2000-2001 using the model eral population exposure is approximately 195 times less than that are 3.2-6.Oe -4 u,u/kg BW (2.3yr half-life) and 1.9-3.7e -4 u,u/kg of a person in Little Hocking. For PFOS (if the estimate of 0.34 ppb BW (3.8 yr half-life); the estimated daily exposures in 2006 are PFOS in drinking water in LH is used; see section on PBPK human 1.3-2.65e -4 (2.3 year half-life) and 2.2-4.4e -4 (3.8 year half-life). model for PFOS), if the general population estimates are compared These estimates were made using the 95% confidence intervals to that of LH (assuming consumption of 11 mL/kg BW/day of for the geometric means and also the interquartile ranges reported water), the general population exposure is approximately 2.3 times by Olsen et al. (2003a, 2008) and are close to the values estimated less than for the people in LH. for the participants in the NHANES study, which ranged from In addition to the general population data from the NHANES, Ol- 1.6e --3 to 3.Oe --3 ug/kg/day for PFOS and 3.0-5.2e --4 ug/kg/day for sen et al. reported the serum PFOA and PFOS concentrations in PFOA. In the second set of simulations, the concentrations were al - adult Red Cross blood donors from 2000-2001 to 2006 in six geo- lowed to reach steady state and then exposure was shut off two graphic locations around the US (N = -600 in both studies; -100 years after the initial measurements were taken. We assumed that samples from each clinic) (Olsen et al., 2003a, 2008). We used steady state was reached when the initial measurements were ta- the model to simulate the serum concentrations of PFOA and PFOS ken (2000-2001); 3 M completely ceased production of PFOS-pro- at both time periods in the donors and also the decline in serum ducing chemicals in 2002. For both PFOA and PFOS, the exposure concentrations of these chemicals after the phase -out of PFOS-pro- was estimated for all donors in the study in both 2000 and 2006. ducing chemicals. In the first set of simulations, exposure to both The estimated exposure in 2000 was then used to examine the de- 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 DEQ-CFW-00004219 14 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 60 50 ra CL a. 40 93 0 -0 n. 20 U1 Ij 0 M 70 � 60 en 50 u- 40 CL E 30 En 20 10 0 10 20 30 40 50 60 70 80 years 80 70®rn0dEll de 60 <, Ilage.r c; en -' SC 0 Boston u. 40 CL E 30 <; 2 20 10 0 --------------------------------------------- 0 20 40 60 80 0 20 40 60 80 years years Fig. 15. Comparison of model simulations (lines) with serum PFOS concentration of Red Cross adult blood donors in 2000 and 2006 (points). A serum half-life of 5.4 years for PFOS was assumed. The top panel shows the serum PFOS concentrations from donors in all six locations examined, assuming constant exposure in both 2000 (estimated exposure was 0.0022-0.0045 ug/kg) and 2006 (estimated) exposure was 0.00085-0.0118 ug/kg). The bottom panel shows the serum PFOS concentrations with exposure shut off 2 years after the initial measurements were taken to simulate the phase -out of PFOS-producing chemicals. The points are the serum PFOS concentrations measured in each of the six cities. The exposure estimated for all donors and the 5.4-year half-life yield good agreement for the individual cities. r 7 2000-2001 5 2flflfl-2001 4 4 LL 9L 2006 a - ::::. 2006 J E 2 2 " 3.8 yr half-life � 2.3 yr half-life n C 0 19 20 39 40 59 0 10 20 30 40 50 years years 6 12 5 10 - model 3.8 yr. half-life Charlotte e � 4 Portland c( Boston —rru i-3.8 yr half- �y av 3 ife u. 0 rnodel 2.3 yr. half-life CL LA M � E E 2 4 . Portland ro 1 hf Eapolis 2 ----------- 0 1ti 20 39 40 59 0 10 20 30 40 50 years years Fig. 16. Comparison of model simulations (lines) with serum PFOA concentration of Red Cross adult blood donors in 2000 and 2006 (points). A serum half-life of 3.8 and 2.3 years for PFOA was assumed. The top panel shows the serum PFOA concentrations from donors in all six locations examined, assuming constant exposure in both 2000 and 2006. The bottom panel shows the serum PFOS concentrations with exposure shut off 2 years after the initial measurements were taken to simulate the phase -out of perfluoroalkyl compounds. The points are the serum PFOA concentrations measured in each of the six cities. The exposure estimates for all donors and both half-lives yield good agreement for the individual cities. 961 cline in serum concentration that was observed in 2006 for each of Although the concentration ranges for the donors in each city 963 962 the six cities in the study (Figs. 15 and 16, bottom panels). may vary, the estimated exposure for all donors in 2000 appears 964 DEQ-CFW 00004220 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 15 965 to yield good model predictions for each city and for the decrease 3.4.1.5. Predicted serum PFOA half-lives. Only two studies have been 966 in concentration observed in 2006. conducted that have estimated the half-life of PFOA in humans (Bartell et al., 2010; Olsen et al., 2007). The average half-life esti- mated from the 26 retired fluorochernical workers (3.8 years) 967 3.4.1.4. PBPK human model for PFOS. Data in the human is extremely was longer compared to the environmentally exposed populations 968 limited for PFOS. The largest US manufacturer of PFOS completed its in Lubeck and Little Hocking (2.3 years). From running the human 969 phaseout of the chemical between 2000 and 2002 (Butenhoff et al., model for the Little Hocking and Arnsberg populations and taking 970 2006). As for PFOA, no definite association between exposure to both half-lives into consideration, it was observed that both esti- 971 PFOS and adverse health effects in humans has been established. mated half-lives appear likely; both allowed for the predicted PFOA 972 Serum PFOS concentrations have been measured in residents from serum concentrations to fall within the measured means and stan- 973 Little Hocking; however, PFOS was present in levels similar to those dard deviations (Figs. 8-12). In addition to running the model to 974 measured from the general US population (Frisbee et al., 2009; compare the simulations with available data, we used the model 975 LHWA, 2005). The study conducted by Olsen et al. (2007) with re- to predict the serum half-lives of the 25 individuals for whom data 976 tired fluorochernical workers estimated an average half-life of was provided on the Little Hocking Water Association website. The 977 5.4 years for PFOS, and the LHWA website included serum PFOS con- data on the individuals and their serum PFOA concentration is gi- 978 centrations of 25 individuals. Using this data, we were able to esti- ven in Table 4. For simplicity, the drinking water concentration 979 mate a transporter maximum for the human (Tmc = 3.5) that was assumed to be 3.55 ppb and the drinking water rate was set 980 would yield the estimated 5.4 year half-life and run the model to at 11 mL/kg BW/day for each person unless noted in the table 981 reach the measured mean serum PFOS concentration in the Little (USEPA, 1997). For men, a standard BW of 70 kg was used; for wo- 982 Hocking people. Fig. 14a shows the comparison of the simulated ser- men, a BW of 58 kg was used in the model. To predict the serum 983 um PFOS concentration compared to the measured average and half-life for each person in the table, the exposure period was mod- 984 standard deviations from the LHWA website data. No PFOS concen- ified to reflect how long they had been drinking the water (the ser- 985 tration in the drinking water could be found for Little Hocking; um concentration given in the table was assumed to be the 986 therefore, we had to estimate this to achieve the correct serum con- maximum serum concentration and the end -of -exposure concen- 987 centration. The mean estimated concentration was 0.34 ppb, which tration). To get the correct serum PFOA concentration for each indi- 988 is ten times less than the PFOA concentration in the drinking water. vidual, the transporter maximum Jrn) was adjusted. The predicted 989 The water levels are most likely lower because the nearby produc- half-lives can be found in Table 4. The mean half-life of the 25 990 tion plant did not produce PFOS. As for PFOA, it took about 30 years people is approximately 2.6 years, and they range from 0.8 to 991 of exposure to reach steady state and it was assumed that drinking 8 years. There were two individuals in the table for whom the max- 992 water was the only source of PFOS exposure. Although the existing imum serum PFOA concentration could not be reached simply by 993 data for human PK for PFOS is scarce, we were able to make a human adjusting Tm. In order to reach their serum concentrations in the 994 model for this chemical that works reasonably well. Further refine- time period that they were exposed, their drinking water intake 995 ment of the model will require that more data for PFOS in humans rates had to be increased from 11 mL/kg BW/day to 29 and 996 becomes available. 22 mL/kg BW/day. These increased drinking water rates are not Table 4 Model prediction of serum PFOA half-lives in the Little Hocking population. LHWA Sex May-05 Age ppb PFOA Years drinking H2O Tmc (PFOA) PFOA Half-life (years) F 46-55 281 28 5.5 2.04 M 46-55 395 51 7.5 2.89 F 46-55 248 55 4.9 1.82 F 26-35 228 27 4.5 1.67 M <15 629 3 10 3.7 a (29 mL/kg BW/day) F >65 442 35 8.7 3.23 M 36-45 220 35 4.1 1.61 F 36-45 112 9 2.2 0.82 M 56-65 532 34 10 3.87 F 26-35 116 4 2.3 0.86 F 26-35 211 6 4.3 1.61 M <15 268 4 7.1 2.93 F <15 436 3 10 3.81' (22 mL,'kg BW/day) M 26-35 216 4 4.7 1.86 M 36-45 325 25 6.2 2.4 F 46-55 475 33 9.25 3.44 M 46-55 432 33 8.2 3.15 F >65 358 25 7 2.64 M >65 346 20 6.5 2.52 M 36-45 1040 37 20 7.71 F 36-45 240 35 4.7 1.75 M 56-65 204 16 3.8 1.46 M 26-35 176 24 3.3 1.28 F 16-25 488 16 9.7 3.59 M >65 238 34 4.5 1.74 Mean 346.24 23.84 2.576 SD 196.350087 Median 281 a Indicates that serum PFOA concentration could not be attained by adjusting only Tmc. Drinking water rate had to be increased from 11 mL/ kg BW/day to the amount indicated. Drinking water concentration was set at 3.55 ppb for each individual. DEQ-CFW-00004221 16 A.E. Loccisano el al./Regulatory Toxicology and Phonnocology xxx (2010) xxx-xxx unreasonable; these individuals may have had higher activity lev- els (USEPA, 1997), simply drank more water, or other physiological factors could be involved. The estimated half-lives from retired workers and the environ- mentally exposed population obviously fit into the predicted range of half-lives from the people on the LHWA website (3.8 and 2.3 years). The retired workers in the study were no longer occupa- tionally exposed, but their serum concentrations were higher than the general population (Calafat et al., 2007b; Olsen et al., 2007). Fig. 17 compares the distributions of the predicted half-lives from Little Hocking with those calculated for the retired workers. The workers' average initial serum concentration was higher than that of the average concentration in Little Hocking (691 ppb for workers versus 346 ppb for LH), but the half-lives for both groups are com- parable. Little Hocking has a higher frequency of shorter half-lives (in the 1-2 year range) while the workers have a higher frequency of half-lives in the 3-4 year range. Neither group has many individ- uals with a half-life longer than 4 years. Prediction of individual half-lives gives an idea of the variability in how long different people retain or eliminate PFOA. From the half-lives predicted by the model, the half-life does not appear to be dependent on how long the people were exposed to PFOA in the drinking water (Fig. 18, top). There is no correlation between length of exposure and half-life; the person with the longest half-life (8 years) was drinking the water for 37 years, but people who drank the water for over 50 years had shorter half-lives (2- 3 years). The same was found for the fluorochernical workers; there does not appear to be a correlation between how long they worked in the plant and their half-life of PFOA (Fig. 18, bottom). If renal resorption is the main process responsible for the long half-lives observed in the human, the variability in elimination of PFOA could be due to polymorphisms in renal transporters or var- iability in expression of the transporter(s). Once the transporter responsible for retention of PFOA in human is identified, informa- tion on the activity and expression of the transporter can then be used to determine if there are potentially sensitive individuals. 4. Discussion Previously, a biologically motivated compartmental model was developed that was able to describe the PK of PFOA and PFOS in the cynomolgus monkey. However, to extrapolate the model to a hu- man to aid in risk assessment, a physiologically, based model with realistic tissue volumes, blood flows, and biochemical properties of PFOA and PFOS were needed. We have therefore developed a PBPK model for PFOA and PFOS in the cynomolgus monkey, and the model is able to describe the available pharmacokinetic data in monkeys exposed to these chemicals via IV and oral dosing. 12 - workers 10 LHWA B- ea ..... ..... ..... ...... ..... ..... ..... ...... ..... ..... ..... ...... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... 2 . ..... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ...... ..... ..... ..... ..... ..... ..... ..... ..... 0 1 2 3 4 5 7 8 10 half-life (years) Fig. 17. Distribution of half-lives for LH residents and retired fluorochemical workers. 41 7 -1 6 Z' 5 Al 41 ,5 3 2 0 ------------------------------------------------ 7 ------------------------ r ----------------------- - ------------------------------------------------ 1 0 10 20 750 40 50 60 years drinking H2O 10 5 4 3 0 41 2 41 a. J 0 19 24 29 734 39 years worked Fig. 18. Half-lives of residents of LH vs. exposure period (top) and retired workers (bottom). There is no correlation between their time drinking the water or working with fluoropolymers and their half-lives of PFOA The monkey model was extrapolated to the human based on the hypothesis that the long -half lives in humans for PFOA and PFOS, like the monkey, can be attributed to saturable renal resorption in the proximal tubule of the kidney. We used the human model to successfully describe the available PFOA data from two commu- nities exposed to PFOA in their tap water and also the general pop- ulation. In addition, we used the model to predict the serum half- lives of 25 residents of the Little Hocking community. Obviously, just because models that incorporate saturable renal resorption are able to describe the existing body of kinetic infor- mation does not mean that this process controls the serum half- lives of these chemicals in the animal or that the same process is responsible in the human. However, existing data for renal trans- porters in the rat suggest a tubular resorptive role for at least one transporter (Oatplal), and we were able to achieve good agreement between the observed data experimental data and the model predictions for both monkeys and humans. In addition, we have developed a PBPK model for these compounds in the rat using the renal resorption description, and this model successfully de- scribes kinetics in the rat (manuscript in preparation). This is evi- dence that the saturable resorption process is responsible for the kinetics of these chemicals across species. If this process is con- firmed to the be the process responsible for the long half life in hu- mans and the transporter proteins are identified, information on the activity of the transporters can be used to identify susceptible individuals due to polymorphic variation. A recent study by Yang et al. showed that two human renal or- ganic anion transporters on the apical membrane side of proximal tubule cells are capable of transporting PFCs and thus may play a key role in tubular resorption of these compounds (Yang et al., 2010). This further suggests that renal resorption is at least partly responsible for the long plasma half-life of these chemicals. Two other human OATs, OAT1 and OAT3, which are located on the baso- lateral membrane of proximal tubule cells, can also transport PFCs (tubular secretion) (Weaver et al., 2009); OAT1 and OAT3 thus ex- crete the chemicals from the blood to the urine side, while OAT4 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 DEQ-CFW-00004222 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx—xxx 17 1113 1114 1115 1116 1117 ills 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 and URAT1 bring the chemicals back into the cell. In the rat, OAT1 and OAT3 have been shown to secrete PFCs; the higher expression of OATplal (apical side) in the male rat is related to the longer half-life observed in the male rat. The human orthologs of OAT1 and OAT3 have similar affinities for PFCs and due to the high expression levels of these transporters in the human, tubular secretion is expected to be similar to that in the rat (Nakagawa et al., 2008) (Hilgenclorf et al., 2007; Nakagawa et al., 2009). How- ever, because PFCs do not accumulate in the kidney, Yang, et al. hypothesize that there may be an efflux transporter localized to the basolateral membrane that could aid in transporting PFCs back into systemic ci rcu lation. (Yang et al., 2010) If identified, this efflux transporter(s) would, in addition to the apical transporters respon- sible for tubular resorption, probably contribute to extending the half-life of PFCs in humans. 4.1. Monkey PBPK models In the oral dosing study with PFOA, it was observed that there is a rapid rise to steady state plasma concentrations of PFOA with a much slower terminal half-life (Butenhoff et al., 2002, 2004a). In the studies with PFOS, a similar trend is observed, and the terminal half-life is even slower (Seacat et al., 2002). In addition, the differ- ences observed in elimination with increasing dose are indicative that a capacity -limited, saturable process must be involved in the pharmacokinetic behavior of these compounds (Andersen et al., 2006; "ran et al., 2008). It is hypothesized that saturable renal resorption is the process responsible for the kinetics and long half-lives of these chemicals; thus, this description was incorpo- rated into both the compartmental model of Andersen and Tan and the current PBPK model for PFOA and PFOS. This process was described with a transporter maximum (Tm) and transporter affin- ity constant (Kt) where chemical was reabsorbed from the filtrate compartment back to the kidney into the plasma. The longer half- life of PFOS compared to PFOA was accounted for by using a higher capacity and higher affinity for PFOS in the model. The biologically motivated model required a time -dependent feature to better characterize the kinetics of these chemicals, which was a changing free fraction of chemical in the plasma. This feature was not neces- sary for description of kinetics in the PBPK model; a constant free fraction (Free) was sufficient. Because both PFOA and PFOS are highly bound to plasma proteins (>97%), a free fraction of <0.03 (0.02 for PFOA and 0.025 for PFOS) was estimated to fit the plasma time course data for the PBPK model. The monkey models success- fully describe the available PK data, and further improvement to the monkey PBPK model will rely on additional data. information on plasma binding proteins in the monkey (dissociation constants, number of binding sites for PFOA and PFOS) and elucidation and information (any sex differences in expression, transporter capac- ities and affinity constants) on renal transporters responsible for tubular resorption in the monkey would aid in refinement of the model. The goal of developing a PBPK model for the monkey was to extrapolate to the human to in order to enhance the scientific basis for human health risk assessment of PFOA and PFOS. Due to the physiological structure of the monkey model, it was possible and easy to extrapolate the model to the human. Epidemiological data suggest that the half-life of PFOA and PFOS is on the order of years in the human in contrast to months in the monkey. If renal resorp- tion is the main process responsible for the long half-life of these compounds in both species, the slower clearance in the human can now be easily described in the model by a higher transporter capacity. In addition to extrapolation to the human, the PBPK mod- el for the monkey could be extended to describe the kinetic data available for other perfluoroalkyl acids. 4.2. Human PBPK models Available human data for PFOA and PFOS suggest that humans are slow eliminators of these chemicals. It is hypothesized that these long half-lives are due to saturable renal resorption as they are in the monkey and rodent. The human PBPK model thus incor- porated saturable renal resorption in the kidney and exhibits good agreement with available data for PFOA in Little Hocking and Arns- berg residents and also in the general population. Human data on PFOS is even sparser than that for PFOA, but using the half-life esti- mate from retired workers and the data available on serum PFOS concentrations from Little Hocking, the model shows reasonable agreement with the data. Plasma protein binding is another possi- ble cause of the long plasma half-lives in humans. PFOA and PFOS have been found to be >90% bound to plasma proteins in rats, mon- keys, and humans; however, binding is not well -characterized (Han et al., 2003; Jones et al., 2003; Kerstner-Wood et al., 2003). For example, it may be difficult to distinguish between 90% and 99% bound, but if humans have a higher bound fraction, this would mean a 10-fold difference in the free fraction and therefore a slower clearance and longer half-life would be possible. Further refinement will depend on future studies of both renal transporters and plasma protein binding. Even though the model exhibits good predictive ability, we had only means, medians, and standard deviations of human serum measurements at our disposal. We were not able to obtain the data on the individuals in the populations in the studies, so we were not able to incorporate factors that may be important to determining variability in serum concentration, plasma half-life, or susceptibil- ity to toxic effects. These factors would include, but may not be limited to: gender, age, how long each person had been drinking the water, how much tap water each person drinks daily, the PFOA concentration measured in different wells or water supplies, and any preexisting medical conditions, such as compromised renal function. The more data we can obtain on individuals in exposed populations, the better we can characterize population variability in a risk assessment context. Several years ago, residents from Lit- tle Hocking and the surrounding communities of the fluoropoly- mer production plant filed a class-action lawsuit alleging adverse health effects due to consumption of water containing PFOA. As part of the settlement, the CS Health Project, which is an indepen- dent panel of researchers investigating the potential link between PFOA exposure and adverse health effects, was created. We believe that using this information when it becomes available to us would greatly improve the value of the model for describing the variabil- ity in the human p harm acoki netics of PFOA. When running simulations, we took into consideration both serum elimination half-life estimates available in the literature for PFOA. From the model predictions, both half-lives appear to be equally likely. Both populations used to estimate the half-lives were small (26 for workers and 200 for environmental exposure); however, the 26 workers exhibited half-lives from 1.5 to 9 years (the environmental study did not give the individuals' half-lives). From the half-lives that the model predicted from the 25 residents in Little Hocking, the half-lives range from 0.9 to 8 years. Even in small populations a wide spectrum is observed, indicating that there is probably a lot of variability in how individuals retain, han- dle, and eliminate PFOA. The average half-life from the model pre- dictions was 2.7 years, which is between that measured from the workers (3.8 years) and the environmentally exposed people (2.3 years). Future refinement of the human model will depend on addi- tional human studies and information on activity and variation in renal transporters. Use of the human PBPK model along with indi- vidual data and information on exposure routes and sources of PFAAs will enable a better understanding of pharmacokinetics of DEQ-CFW-00004223 is A.E. Loccisano el al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 1241 PFAAs in humans, link possible exposure routes and external doses 1242 to blood concentrations, aid in understanding interindividual vari- 1243 ability, and ultimately aid in human health risk assessment for 1244 PFAAs. 1245 5. Conflict of interest statement 1246 The authors declare that there are no conflicts of interest. 1247 1248 1249 1250 1251 1252 1253 1254 1255 Acknowledgments We would like to thank Dr. John Butenhoff at 3M Company and Mr. Gerald Kennedy, Jr. at DuPont Company for helpful discussion and comments. Financial support was supported by an EPA STAR Grant (No. R833450) and the 3M and DuPont Companies. Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.yrtph.2010.12.004. References 3M, 2003. Environmental and Health Assessment of Perfluorooctane Sulfonate and its Salts. USEPA Administrative Record AR 226-1486. Andersen, M.E., Butenhoff, J.L., Chang, 3­C_ Farrar, D.G., Kennedy Jr., ".L., Lau, C., Olsen, G.W., Seed, J., Wallace, K.B., 2008. Perfluoroalkyl acids and related chemistries-toxicokinetics and modes of action. Toxicol. Sci. 102, 3-14. Andersen, M.E., Clewell, H.J., Tan, Y.-M., Butenhoff, J.L., Olsen, G., 2006. Pliarmacokinetic modeling of saturable, renal absorption of perfluoroalkylacids in monkeys -probing the determinants of long plasma half-lives. Toxicology 227, 156-164. Apelberg, B.J., Witter, F.R., Herbstman, J.B., Calafat, A.M., Halclen, R.U., Needham, L.L., Goldman, L.R., 2007. Cord serum concentrations of perfluorooctane sulfonate (pfos) and perfluorooctanoate (pfoa) in relation to weight and size at birth. Environ. Health Perspect. 115, 1670-1676. Ashby, J., Brady, A., Elcombe, C.R., Elliott, B.M., Ishmael, J., Odum, J., Tugwood, J.D_ Kettle, S., Purchase, I.F.H., 1994. Mechanistically -based human hazard assessment of peroxisome proliferator-induced hepatocarcinogenesis, Hum. Exp. Toxicol. 13 (Suppl. 2), S1 -Sl 17. Bartell, S.M., Calafat, A.M., Lyu, C., Kato, K., Ryan, P.B., Steenland, K, 2010, Rate of decline in serum pfoa concentrations after granular activated carbon filtration at two public water systems in Ohio and West Virginia. Eaviron- Health PerspeCL. 118, 222-228. Benskin, J.P., De Silva, A.C., Martin, L.J., Arsenault, G., McCrindle, R., Riddell, N., Mabury, S.A., Martin, J.W., 2009. Disposition of perfluorinated acid isomers in sprague-dawley rats: part 1: single dose. Environ. Toxicol. Chem. 28, 542-554. Biegel, L.B., Hurtt, M.E., Frame, S.R., O'Connor, J.C., Cook, J.C., 2001, Mechanisms of extrahepatic tumor induction by peroxisome proliferators in male cd rats. Toxicol. Sci. 60, 44-55. Bjork,J.A., Wallace, K.B., 2009. Structure -activity relationships and human relevance for perfluoroalkyl acid -induced transcriptional activation of peroxisome proliferation in liver cell cultures. Toxicol. Sci. 111, 89-9.9. Brown, R.P., Delp, M.D., Lindstedt, M.D., Rhomberg, LP,, Beliles, R.P., 1997. Physiological parameter values for physiologically based pharmacokmetic models. Toxicol. Ind. Health 13, 407-484. Butenhoff, J.L, 2010. Personal communication. Butenhoff, J.L., Costa, G., Elcombe, C., Farrar, E., Hansen, K., Iwai, H., Jung, R., Kennedy Jr., G., Lieder, P., Olsen, G., Thomford, P., 2002. Toxicity of ammonium perfluorooctanoate in male cynomolgus monkeys after oral dosing for 6 months. Toxicol. Sci. 69, 244-257. Butenhoff, J.L., Kennedy Jr., G., Hinderliter, P.M., Lieder, P., Jung, R., Hansen, K, Gorman, G.S., Noker, P.E., Thornford, P., 2004a. Pharmacokinetics of perfluorooctanoate in cynomolgus monkeys. Toxicol. Sci. 82, 394-406. Butenhoff, J.L., Kennedy Jr., G.L., Frame, S.R., O'Conner, J.C., York, R.G., 2004b. The reproductive toxicology of ammonium perfluorooctanoate (apfo) in the rat. Toxicology 196, 95-116. Butenhoff, J.L, Olsen, G.W., Pfahles-Hutchens, A., 2006. The applicability of biomonitoring data for perflurooctanesulfonate to the environmental public health continuum. Environ. Health Perspect. 114, 1776-1782. Calafat, A.M., Kuklenyik, Z., Reidy, J.A, Caudill, S.P., Tully, J.S., Needham, LL., 2007a. Serum concentrations of 11 polyfluoroalkyl compounds in the US population: data from the national health and nutrition examination survey (nhanes) 1999- 2000. Environ. Sci. Technol. 41, 2237-2242. Calafat, A.M., Wong, L.-Y., Kuklenyik, Z., Reidy, J.A., Needham, L.L., 2007b. Polyfluoroalkyl chemicals in the US population: data from the national health and nutrition examination survey (nhanes) 2003-2004 and comparisons with nhanes 1999-2000. Environ. Health Perspect. 115, 1596-1602. DePierre, J.W., 2009. Personal communication. DeWitt, J., Shnyra, A., Badr, M., Loveless, S., Hoban, D., Frame, S.R., Cunard, R., Andersen, S., Meade, BJ., Peden -Adams, M., Luebke, R., Luster, M., 2009. Immunotoxicity of perfluorooctanoic acid and perfluorooctane sulfonate and the role of peroxisome proliferator-activated receptor alpha. Crit. Rev. Toxicol. 39,76-94. Dong, G.-H., Zhang, Y.H., Zheng, L., Liu, W., Jin, Y.-H., He, Q.-C., 2009. Chronic effects of perfluorooctanesulfonate exposue on immunotoxicity in adult male c57b1/6 mice. Arch. Toxicol. 83, 805-815. Eliresman, D.J., Froehlich, J.W., Olsen, G.W., Chang, S.-C., Butenhoff, J.L, 2007. Comparison of human whole blood, plasma, and serum matrices for the determination of perfiLiOrOOCLaiiesulfonate (pfos), perfluorooctanoate (pfoa), and other fluorochemicals. Environ. Res. 103, 176-184. Elcombe, C.R., Elcombe, B.M., Foster, J.R, Farrar, D.G., Jung, R., Chang, S.-C., Kennedy Jr., G.L., Butenhoff, J.L., 2010. Hepatocellular hypertrophy and cell proliferation in sprague-dawley rats following dietary exposure to ammonium perfluorooctanoate occurs through increased activation of the xenosensor nuclear receptors ppar (alpha) and car/pxr. Arch. Toxicol. 84, 787-798. Emmett, E.A., Shofer, F.S., Zhang, H_ Freeman, D., Desai, C., Shaw, L.M., 2006. Community exposure to perfluomoctanoate: relationships between serum concentrations and exposure sources. J. Occup. Environ. Med. 48, 759-770. Fiserova-Bergerova, V., 1975. Biological -mathematical modeling of chronic toxicity. Wright -Patterson Air Force Base, OH: Aerospace Medical Research Laboratory, AMRL-TR-75-5. Fisher, J.W., Dorman, D.C., Medinsky, M.A., Welsch, F., Conolly, R.B., 2000. Analysis of respiratory exchange of methanol in the lung Of the monkey using a physiological model, Toxicol. Sci. 53, 185-193. Forsyth, R.P., Nies, AS_ Wyler, F., Neutze, J., Melmon, K.L., 1968. Normal distribution of cardiac output in the unanesthetized, restrained rhesus monkey. J. Appl. Physiol. 25, 736-741. Frisbee, S., Brooks, A.P_ Maher, A., Flensborg, P., Arnold, S., Fletcher, T., Steenland, K., Shankar, A., Knox, S,S., Pollard, C., Halverson, J.A., Vieira, V.M., Jin, C., Leyden, K.M., Ducatman, A. 2009. The c8 health project: design, methods, and participants, Environ. Health Perspect. 117, 1783-1882. Fromme, H., Tittlemier, S.A., Volkel, W., Wilhelm, M., Twardella, D., 2009. Perfluorinated compounds - exposure assessment for the general population in western countries. hit. J. Hyg. Environ. Health 212, 239-270. Goldenthal, E.I., 1978. Final report, ninety day subacute rat toxicity Study on fluorad fluorochemical fc-143. International Research and Development Corporation, Study No. 137-089, 3M Reference No. T-3141. Gonzalez, 'F.J., Shah, Y.M., 2008. Ppar-o,.: mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators. Toxicology 246, 2-8. Han, X, Snow, T.A., Kemper, RA, Jepson, G.W., 2003. Binding of perfluorooctanoic acid to rat and human plasma proteins. Chem. Res. Toxicol. 16, 775-781. Hanhijarvi, H., Ylinen, M., Kojo, A., Kosma, V.-M., 1987. Elimination and toxicity of perfluorooctanoic acid during subchronic administration in the Wistar rat. Pharm. Toxicol. 61, 66-68. Harada, K., Inoue, K., Morikawa, A., Yoshinaga, T., Saito, N., Koizurni, A., 2005. Renal clearance of perfluorooctane sulfonate and perfluorooctanoate in humans and their species -specific c excretion. Environ. Res. 99, 253-261. Hilgenclorf, C., Ahlin, G., Seithel, A., Artursson, P., Ungell, A.L., Karlsson, J., 2007. Expression of thirty-six drug transporter genes in human intestine, liver, kidney, and organotypic cell lines. Drug Metab. Dispos. 35, 1333-1340. Hinderliter, P.M., Han, X., Kennedy Jr., G.L., Butenhoff, J.L., 2006. Age effect on perfluorooctanoate (pfoa) plasma concentration in post -weaning rats following oral gavage with ammonium perfluorooctanoate (apfo). Toxicology 225, 195- 203. Hinderliter, P.M., Mylchreest, E., Gannon, S.A., Butenhoff, J.L., Kennedy Jr., G.L., 2005. Perfluorooctanoate: placental and lactational transport pharmacokinetics in rats. Toxicology 211, 139-148. H61zer, J., Goen, T., Rauchfuss, K., Kraft, M., Angerer, J., Kleeschulte, P., Wilhelm, M., 2009. One-year follow-up of perfluorinated compounds in plasma of german residents from arnsberg formerly exposed to pfoa-contaminated drinking water. Int. J. Hyg. Environ. Health 212, 499-504. H61zer, J., Miclasch, 0., Rauclifuss, K., Kraft, M., Reupert, P_ Angerer, J., Kleeschulte, P., Marschall, N., Wilhelm, M., 2008. Bionionitoring of perfluorinated compounds in children and adults exposed to perfluorooctanoate- contaminated drinking water. Environ. Health PerspeCL. 116, 651-657. Hundley, S.G., Sari -if, A.M., Kennedy Jr., G.L., 2006. Absorption, distribution, and excretion of ammonium perfluorooctanoate (apfo) after oral administration to various species. Drug Chem. Toxicol. 29, 137-145. 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 Intravenous Dose of fc-95- 14c. Riker Laboratories, Inc., St. Paul, MN, USEPA Docket No. 8(e)HQ-1180- 00374. Johnson, J.D., Gibson, SJ., Ober, R.E., 1984. Cholestyramine-enhanced fecal elimination of carbon-14 in rats after administration of ammonium [ 14C jperfluorooctanoate or potassium [ 14C jperfluorooctanesulfonate. Fundam. Appl. Toxicol. 4, 972-976. Jones, P.D., Hu, W., De Coen, W., Newsted, J.L., Giesy, J.P., 2003. Binding of perfluorinated fatty acids to serum Proteins. Environ. Toxicol. Chem. 22, 2639- 2649. Karrman, A., Ericson, L, Van Bavel, B., Darnerud, P.O., Aune, M., Glynn, A., Lignell, S., Lindstrom, G., 2007. Exposure of perfluorinated chemicals through lactation: Levels of matched human milk and serum and a temporal trend, 1996-2004, in Sweden. Environ. Health Perspect. 115, 226-230. DEQ-CFW-00004224 A.E. Loccisano et al./Regulatory Toxicology and Pharmacology xxx (2010) xxx-xxx 19 Kemper, R., 2003. Perfluooctanoic acid: toxicokinetics in the rat. DuPont Haskell Laboratories, Project ID: DuPont-7473. USEPA Administrative Record AR-226. Kennedy Jr., G.L., Butenhoff, J.L, Olsen, G.W., O'Conner, J.C., Seacat, A.M., Perkins, R.G., Biegel, L.B., Murphy, S.R., Farrar, D.G., 2004. The toxicology of perfluorooctanoate. Crit. Rev. Toxicol. 34, 351-384. Kerstner-Wood, C., Coward, L., Gorman, G., 2003. Protein binding of perfluorobutane sulfonate, perfluorohexane sulfonate, perfluorooctane sulfonate, and perfluorooctanoate to plasma (human, rat, and monkey) and various human - derived plasma Protein fractions. Southern Research Corporation, Study 9921.7. USEPA Docket AR-226-1354. Kudo, N., Katakura, M., Sato, Y., Kawashima, Y., 2002. Sex hormone -regulated renal transport of perfluorooctanoic acid. Chem.-Biol. Int. 139, 310-316. Kudo, N., Sakai, A., Mitsurnoto, A., Hibino, Y., Tsuda, T., Kawashima, Y., 2007. Tissue distribution and hepatic subcellular distribution of perfluorooctanoic acid at low doses are different from those at high doses. Biol. Pharm. Bull. 30, 1535- 1540. Kuklenyik, Z., Reich, J.A., Tully, J.S., Needham, L.L, Calafat, AM., 2004. Automated solid -phase extraction and measurement of perfluorinated organic acids and amides in human serum and milk. Environ. Sci. Technol. 38, 3698-3704. Kuslikis, B.I., Vanden Heuvel, J.P., Peterson, R.E., 1992. Lack of evidence for perfluorodecanoyl- or perfluorooctanoyl-coenzyme a formation in male and female rats. J. Biochem. Toxicol. 7, 25-29. Lake, B.G., 2009. Species differences in the hepatic effects of inducers of cyp2b and cyp4a subfamily forms: relationship to rodent liver turnout formation. Xenobiotica 39, 582-596. Lau, C., Anitole, K, Hodes, C., Lai, D., Pfahles-Hutchens, A., Seed, J., 2007. Perfluoroalkyl acids: a review of monitoring and toxicological findings. Toxicol. Sci. 99, 366-394. Lau, C., Butenhoff, J.L, Rogers, J.M., 2004. The developmental toxicity of perfluoroalkyl acids and their derivatives. Toxicol. Appl. Pharmacol. 198, 231- 241. Lhwa Member Blood Testing Results (May 2005), 2005. Little Hocking Water Board, <http://www.littlebockingwater.org,, 2005 (accessed May 2009). Luebker, D.J., Hansen, K.J., Bass, N.M., Butenhoff, J.L, Seacat, A.M., 2002. Interactions of fluorochemicals with rat liver fatty acid -binding protein. Toxicology 176, 175-185. Macey, R., Oster, G., Zalmley, T., 2000. Berkeley-Madomia. University of California, Department of Cellular and Molecular Biology. Berkeley, CA. Nakagawa, H., Hirata, T., Terada, T., JULabha, P., Miura, D., Harada,J.H., Inoue, K., Anzai, N., Endou, H., Inui, K, 2008. Roles of organic anion transporters in the renal excretion of perfluorooctanoic acid. Basic Clin. Pharmacol. Toxicol. 103, 1- 8. Nakagawa, H., Terada, T., Harada, K.H., Hitorm, T., Inoue, K, Inui, K., Koizumi, A_ 2009. Human organic anion transporter hoat4 is a transporter of perfluooctanoic acid. Basic Clin. Pharmacol. Toxicol. 105,136-138. Noker, P., Gorman, G., 2003. A pharmacokinetic study of potassium perfluorooctanesulfonate in the cynomolgus monkey. Southern Research Institute, Study ID: 9921.6, USEPA Public Docket, Administrative Record AR- 226-1356. Olsen, G.W., Burris, J.M., Ehresman, D.J., Froehlich, J.W., Seacat, AM., Butenhoff, J,L, Zobel, I-R., 2007. Half-life of serum elimination of perfluorooctanesulfonate, perflLiorohexanesulfonate, and perfluorooctanoate in retired fluorochemical production workers. Environ. Health Perspect. 115, 1298-1305. Olsen, G.W., Church, T.R., Miller, J.P., Burris, J.M., Hansen, K.J., Lundberg, JX, Armitage, J.B., Herron, R.M., Medlidizadehkashi, Z., Nobiletti, J.B., O'Neill, E.M., Mandel, J.H., Zabel, L.R., 2003a. Perfluorooctanesulfonate and other fluorochernicals in the serum of American red cross adult blood donors. Environ. Health Perspect. 111, 1892-1901. Olsen, G.W., Hansen, K.J., Stevenson, LA., Burris, J.M., Mandel, J.H., 2003b. Human donor liver and serum concentrations of perfluooctane sulfonate and other perfluorochemicals. Environ. Sci. Technol. 37, 888-891. Olsen, G.W., Mair, D.C., Church, T.R., Ellefson, M.E., Reagen, W.K., Boyd, T.M., Herron, R.M., Medhdizadehkashi, Z., Nobiletti, J.B., Rios, J.A., Butenhoff, J.L., Zobel, L.R., 2008. Decline in perfluorooctanesulfonate and other polyfluoroalkyl chemicals in American Red Cross adult blood donors, 2000-2006. Environ. Sci. Technol. 42, 4989-4995. Ophaug, R.H., Singer, L., 1980. Metabolic handling of perfluorooctanoic acid in rats. Proc. Soc. Exp. Biol. Med. 163, 19-23. Ostertag, SX, Chan, H.M., Moisey, J., Dabeka, R., Tittlernier, S.A., 2009. Historic dietary exposure to perfluorooctane sulfonate, perfluorinated carboxylates, and fluorotelemer unsaturated carboxylates from the consumption of store-bought and restaurant foods for the Canadian population. J. Agric. Food Chem. 57, 8534-8544. Paustenbach, D.J., Panko, J.M., Scott, P.K., Unice, K.M., 2007. A methodology for estimating human exposure to petfluorooctanoic acid (pfoa): a retrospective exposure assessment of a community (1951-2003). J. Toxicol. Environ. Health, A 70, 28-57. Prevedouros, K., Cousins, I.T., Buck, R.C., Korzeniowski, S.H., 2006. Sources, fate, and transport of perfluorocarboxylates. Environ. Sci. Technol. 40, 32-44. Qazi, M.R., Nelson, B.D., Depierre, J.W., Abedi-Valugerdi, M., 2010. 28-Day dietary exposure of mice to a low total dose (7 mg/kg) of perfluorooctane sulfonate (pfos) alters neither the cellular compositions of the thymus and spleen nor Immoral immune responses: does the route of administration play a pivotal role in pfos-inducted immunotoxicity? Toxicology 267, 132-139. Riker, 1987. Two year oral (diet) toxicity/carcinogenicity study offluorochemical fc- 143 in rats. Riker Laboratories, Inc.. Experiment No. 0281CR0012.. Rodriguez, C.E., Setzer, R.W., Barton, H.A., 2009. Pharmacokinetic modeling of perfluorooctanoic acid during gestation and lactation in the mouse. Reprod. Toxicol. 27, 373-386. Rosen, M.B., Schmid, j.R., Carton, J.C., Zehr, R.D., Das, K.P., Abbott, B.D., Lau, C., in press. Gene expression profiling in wild -type and ppara-mill mice exposed to perfluorooctane sulfonate reveals pparcx-independent effects. PPAR Res. Seacat, AM., Thomford, P.J., Hansen, K.J., Olsen, G.W., Case, M.T., Butenhoff, J.L., 2002. Subchronic toxicity studies on perfluorooctanesulfonate potassium salt in cynomolgus monkeys. Toxicol. Sci. 68, 249-264. Sinclair, E., Kim, SX, Akinleye, H,B., Kannan, K, 2007. Quantitation of gas -phase perfluoroalkyl surfactants and fluorotelomer alcohols released from nonstick cookware and microwave popcorn bags. Toxicol. Sci. 41, 1180-1185. Skutlarek, D., Exner, M., Farber, H., 2006. Perfluorinated surfactants in surface and drinking waters. Environ. SCI. PCIILIL. Res. 13, 299-307. Steenland, K., Jin, C., MacNeil, J., Lally, C., Ducatman, A., Vieira, V., Fletcher, T., 2009. Predictors of pfoa levels in a community surrounding a chemical plant. Environ. Health Perspect. 117, 1083-1088. Strynar, M.J., Lindstrom, A.B., 2008. Perfluorinated compounds in house dust from Ohio and North Carolina, USA Environ. Sci. Technol. 42, 3751-3756. Tahara, H., Shona, M., Kusuhara, H., Kinoshita, H., Fuse, E., Takadate, A., Otagiri, M., Sugiyama, Y., 2005. Molecular cloning and functional analyses of oatl and oat3 from cynomolgus monkey kidney. Pharm. Res. 22, 647-660. Tan, Y.-M., Clewell, H.J., Andersen, M.E., 2008. Time dependencies in perfluorooctylacids disposition in rat and monkeys: a kinetic analysis. Toxicol. Lett. 177, 38-47. Thibodeaux, J.R., Hanson, R.G., Rogers, J.M., Grey, B.E., Barbee, B.D., Richards, J.H., Butenhoff, J.L., Stevenson, LA., Lau, C., 2003. Exposure to perflU01`00CLane sulfonate during pregnancy in rat and mouse. 1: maternal and prenatal evaluations. Toxicol. Sci. 74, 369-381. USEPA, 1997. Exposure Factors Handbook. <http://www.epa.gov/ncea/efh/pdfs/eth- front-gloss.pdf>. USEPA, 2005. Draft Risk Assessment of the Potential Human Health Effects Associated with Exposure to Perfluorooctanoic Acid and its Salts. <http:/'/ www.epa.gov/opptintr/pfoa/pubs/pfoarisk.pdf>. USEPA, 2009. Provisional health advisories for perfluorooctanoic acid (pfoa) and perfluorooctanesulfonate (pfos). http://www.epa.gov/waterscience/criteria/ drinking/pba-PFOA PFOS.pdf. Vanden Heuvel, J.P., Kuslikis, B.I., Rafelghem, M.J., Peterson, R.E., 1991. Tissue distribution, metabolism, and elimination of perfluorooctanoic acid in male and female rats. J. Biochem. Toxicol. 6, 83-92. Vestergren, R., Cousins, I.T., 2009. Tracking the pathways of human exposure to perfluorocarboxylates. Environ. Sci. Technol. 43, 5565-5575. Washburn, S.T., Bingman, T.S., Braithwaite, S.K., Buck, R.C., Buxton, LW., Clewell 111, H.J., Haman, L.A., Kester, J.E., Rickard, R.W., Shipp, A.M., 2005. Exposure assessment and risk characterization for perfluorooctanoate in selected consumer articles. Environ. Sci. Technol. 39, 3904-3910. Weaver, Y.M., Ehresman, D.J., Butenhoff, J.L., Hagenbuch, B., 2009. Roles of rat renal organic anion transporters in transporting perfluorinated carboxylates with different chain lengths. Toxicol. Sci. 113, 305-314. Wilhelm, M., Kraft, M., Rauchfuss, K, Holzer, J., 2008. Assessment and management of the first german case of a contamination with perfluorinated compounds (pfc) in the region Sauerland, North Rhine-Westphalia. J. Toxicol. Environ. Health, A 71, 725-733. Yang, C.-H., Glover, K.P., Hall, X., 2010. Characterization of cellular uptake of perfluoroctanoate via organic anion -transporting polypeptide 1a2, organic anion transporter 4, and urate transporter I for their potential roles in mediating human ren], reabsorption of perfluorocarboxylates. Toxicol. Sci. 117,294-302. Yang, C., Glover, K.P., Han, X., 2009. Organic anion transporting polypeptide (oatp) lal-mediated perfluorooctanoate transport and evidence for a renal reabsorption mechanism of oatplal in renal elimination of perfluorocarboxylates in rats. Toxicol. Lett. 190, 163-171. Yang, Q., Xie, Y., Depierre, J.W., 2000. Effects of peroxisome proliferators on the thymus and spleen of mice. Clin. Exp. Immunol. 122, 219-226. DEQ-CFW-00004225