27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>of</strong> chemical/dietary induced changes in serum thyroxine (T 4 ) concentrations in either<br />

euthyroid or sensitive populations, such as the developing fetus. Collaborative<br />

researchers are examining the effects <strong>of</strong> iodide deficiency on CNS development in<br />

the rat pup at the USEPA using propylthiouracil as a positive control.<br />

Electrophysiological, biochemical and molecular studies <strong>of</strong> the pup brain combined<br />

with behavioral studies are used for evaluating HPT mediated developmental neurotoxicity.<br />

A Biologically Based Dose Response (BBDR) HPT axis model for the<br />

lactating rat and nursing pups predicts the relationship between brain concentration<br />

<strong>of</strong> T 3 and serum concentration <strong>of</strong> T 4 as a function <strong>of</strong> iodide intake. <strong>The</strong> BBDR<br />

HPT axis model describes two negative feedback loops: 1) TSH production controlled<br />

by the brain concentration <strong>of</strong> T 3 , and 2) TSH stimulation <strong>of</strong> thyroid iodide<br />

uptake and T 3 and T 4 synthesis and secretion. <strong>The</strong> model is calibrated to predict dietary<br />

iodine deficiency-induced perturbations in serum and brain thyroid hormones.<br />

<strong>The</strong> model successfully described published data sets in the dam and pup<br />

for both euthyroid and iodide deficient conditions. After successful validation for<br />

iodide deficiency, the model will be expanded to include thyroid active chemicals.<br />

<strong>The</strong> model will allow for simulation <strong>of</strong> complex dose response and exposure conditions<br />

and serve as a template for the future development <strong>of</strong> human gestation and<br />

lactation BBDR HPT axis models for use in risk assessment.<br />

703 AGE-DEPENDENT METABOLISM OF CHLOPYRIFOS<br />

AND CHLORPYRIFOS-OXON IN HUMAN PLASMA<br />

AND HEPATIC MICROSOMES.<br />

T. S. Poet 1 , C. Timchalk 1 , M. J. Bartels 2 and J. N. Smith 1 . 1 Biological Monitoring<br />

& Modeling, Battelle, Pacific Northwest Division, Richland, WA and 2 <strong>The</strong> Dow<br />

Chemical Company, Midland, MI.<br />

Young animals and, potentially, children may have increased sensitivity to chlorpyrifos<br />

(CPF) exposure compared to adults at higher exposure levels, which could<br />

be in part due to age differential metabolism <strong>of</strong> CPF and CPF-oxon (ultimate toxicant).<br />

In humans, there have not been studies characterizing CPF or CPF-oxon metabolism<br />

over various ages. Thus, age-dependent enzymatic metabolism <strong>of</strong> CPF and<br />

CPF-oxon were quantified in vitro using human donor hepatic microsomes (n =<br />

30, ages 13 d-75 y) and plasma (n = 20, ages 3 d-43 y) by measuring product formation<br />

<strong>of</strong> 3,5,6-trichloro-2-pyridinol (TCPy) and CPF-oxon using gas chromatography/mass<br />

spectrometry (GC/MS). Mean hepatic CPF desulfation and dearylation<br />

Vmax values across ages were 0.35 and 0.73 nmol/min/mg microsomal<br />

protein, respectively. <strong>The</strong> mean hepatic CPF-oxon hydrolysis Vmax value was 78<br />

nmol/min/mg microsomal protein. All hepatic measures <strong>of</strong> metabolism were consistent<br />

across ages on a per microsomal protein basis using a linear regression model<br />

(p = 0.90, 0.60, and 0.17, respectively). Ratios <strong>of</strong> desulfation to dearylation also did<br />

not show any age dependent relationships (p = 0.87). Pearson product-moment<br />

correlation coefficients <strong>of</strong> pseudo first order rate estimates and Vmax values with<br />

substrate marker activities provided by the vendor implicated CYP2B6, 3A4/5,<br />

2C8, and 2D6 with CPF desulfation and CYP2C19 and 3A4/5 with CPF dearylation.<br />

CPF-oxon metabolism in plasma showed age-dependent increases over two<br />

factors: (1) metabolism on a per protein basis (p = 0.03) and (2) total plasma protein<br />

levels (p = 0.005). <strong>The</strong> mean overall CPF-oxon metabolism Vmax value for<br />

children < 6 mo <strong>of</strong> age was 1901 nmol/min/mL vs. 6829 nmol/min/mL for adults,<br />

> 3.5 times higher. <strong>The</strong>se results are useful when evaluating potential response <strong>of</strong><br />

different age individuals to CPF and will be integrated into a lifestage physiologically<br />

based pharmacokinetic and pharmacodynamic (PBPK/PD) model for CPF.<br />

704 DEVELOPMENT OF A LIFESTAGE PHYSIOLOGICALLY<br />

BASED PHARMACOKINETIC AND<br />

PHARMACODYNAMIC (PBPK/PD) MODEL FOR<br />

CHLORPYRIFOS IN RATS AND HUMANS.<br />

J. N. Smith 1 , P. M. Hinderliter 1 , C. Timchalk 1 , M. J. Bartels 2 and T. S. Poet 1 .<br />

1 Biological Monitoring & Modeling, Battelle, Pacific Northwest Division, Richland,<br />

WA and 2 <strong>The</strong> Dow Chemical Company, Midland, MI.<br />

Young animals and, potentially, children may have increased sensitivity to chlorpyrifos<br />

(CPF) exposure compared to adults at higher exposure levels. Thus, an agedependent,<br />

lifestage PBPK/PD model was developed to computationally predict<br />

disposition <strong>of</strong> CPF and its metabolites chlorpyrifos-oxon (CPF-oxon) and 3,5,6trichloro-2-pyridinol<br />

(TCPy) as well as corresponding cholinesterase (ChE) inhibition<br />

in rats and humans. In this model, age-dependent body weight was calculated<br />

from a generalized Gompertz function, and compartments (liver, brain, fat, blood,<br />

diaphragm, rapid, and slow) were scaled based on body weight from polynomial<br />

functions on a fractional body weight basis. Blood flows among compartments<br />

were calculated as a constant flow per compartment volume. Carboxylesterase activity,<br />

hepatic microsomal protein, and CPF-oxon metabolism in plasma have all<br />

demonstrated age-dependency and were scaled as such. PK <strong>of</strong> TCPy was handled as<br />

a one compartment model. Consistent with literature results, simulations <strong>of</strong> rats<br />

152 SOT 2011 ANNUAL MEETING<br />

dosed with a single dose <strong>of</strong> 10 mg/kg CPF orally at 5, 12, and 45 days <strong>of</strong> age<br />

demonstrate increased peak CPF concentrations in blood (0.72, 0.49, and 0.40<br />

μM) and greater maximal inhibition <strong>of</strong> plasma ChE activity (97, 93, and 77%), red<br />

blood cell ChE activity (82, 67, and 34%), and brain ChE activity (80, 56, and<br />

25%), respectively. In humans, PBPK/PD model simulations predict similar agedependent<br />

outcomes. For humans 6 mo, 3 y, and 30 y <strong>of</strong> age, peak CPF concentration<br />

in blood were (0.20, 0.18, and 0.17 μM), maximal inhibition <strong>of</strong> plasma ChE<br />

activity were (98, 91, and 85%), and red blood cell ChE activity were (19, 11, and<br />

9%), respectively, after receiving a single oral dose <strong>of</strong> 0.5 mg/kg CPF. This model<br />

provides a computational framework for age comparative simulations that can be<br />

utilized to evaluate age-related responses resulting from CPF exposure.<br />

705 EFFECT OF PARAMETER VARIABILITY AND<br />

UNCERTAINTY IN A LIFESTAGE PBPK MODEL FOR<br />

CHLORPYRIFOS.<br />

P. M. Hinderliter 1 , J. N. Smith 1 , M. J. Bartels 2 , P. S. Price 2 and T. S. Poet 1 .<br />

1 Biological Monitoring & Modeling, Battelle, Pacific Northwest Division, Richland,<br />

WA and 2 <strong>The</strong> Dow Chemical Company, Midland, MI.<br />

Physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) models<br />

are important tools in risk assessment, allowing for the evaluation <strong>of</strong> predicted effects<br />

<strong>of</strong> chemical exposures on humans that occur as the result <strong>of</strong> real world exposures.<br />

Human populations are unique in their differences and more specifically are<br />

sexually dimorphic and exhibit a wide range in heights and weights and body composition.<br />

A lifestage model has been constructed to include growth from birth to<br />

adulthood and inter-individual variability in organ sizes and metabolic parameters.<br />

<strong>The</strong> model incorporates measured body weights (from NHANES or other published<br />

datasets) and calculates the necessary model body compartments (i.e., liver,<br />

brain, fat, blood, rapid, and slow) which are scaled based on body weight from<br />

polynomial functions on a fractional body weight basis. Variability in enzymatic parameters<br />

is included, based on measured chlorpyrifos bioactivation and detoxification<br />

in humans <strong>of</strong> different ages. To validate the model, human exposure data were<br />

simulated and matched from two controlled human oral studies. Variability was determined<br />

by calculating confidence intervals around in vitro-derived metabolic rate<br />

constants for age-dependent plasma PON1 activity. <strong>The</strong> activities measured in vitro<br />

were bootstrapped into the PBPK/PD model to account for variability and uncertainty<br />

in these parameters. As an example, RBC AChE was examined as a function<br />

<strong>of</strong> metabolic and physiological parameters using daily oral exposures from diet<br />

(CARES). <strong>The</strong> results indicate that there are no trends between RBC AChE inhibition<br />

with BMI, cardiac output, liver PON1, blood PON1, liver P450, or brain<br />

P450 in adults. Simulation <strong>of</strong> higher doses does show a trend between PON1 activities<br />

and AChE inhibition. This suggests that human variability will not substantially<br />

impact predictions <strong>of</strong> inhibition at real-world low exposures. At higher doses<br />

reduced levels <strong>of</strong> PON1 may be associated with increased inhibition.<br />

706 DEVELOPMENT OF HUMAN GESTATION AND<br />

LACTATION PBPK MODELS FOR<br />

PERFLUOROOCTANOATE (PFOA) AND<br />

PERFLUOROOCTANESULFONATE (PFOS).<br />

A. E. Loccisano 1 , M. E. Andersen 1 , J. Butenh<strong>of</strong>f 2 and H. J. Clewell 1 . 1 <strong>The</strong><br />

Hamner Institutes for Health Sciences, Research Triangle Park, NC and 2 3M<br />

Corporation, St. Paul, MN.<br />

Conflicting results have been reported for associations between maternal<br />

serum/plasma concentrations <strong>of</strong> perfluorooctanoate (PFOA) and perfluorooctanesulfonate<br />

(PFOS) and human birth outcomes. Physiological changes during pregnancy<br />

such as plasma volume expansion can affect pharmacokinetics and can also<br />

be associated with birth outcomes. We have developed PBPK models for PFOA<br />

and PFOS for the gestation and lactation life stages in humans to understand more<br />

fully how the physiological changes associated with development affect tissue distributions<br />

<strong>of</strong> these compounds in the mother, fetus, and neonate. <strong>The</strong>se models were<br />

derived from PBPK models for PFOA and PFOS that we previously developed for<br />

adult humans and for rats during gestation and lactation. <strong>The</strong> same model structure<br />

and parameters used in the adult human model could be used to simulate plasma<br />

concentrations in pregnant and lactating women, indicating that pharmacokinetics<br />

is not appreciably affected. Transfer <strong>of</strong> both chemicals to the human fetus was described<br />

by simple diffusion, while transfer <strong>of</strong> chemical from plasma to milk was assumed<br />

to be flow-limited. <strong>The</strong> models simulated PFOA and PFOS concentrations<br />

in maternal and fetal plasma and milk and were compared to available epidemiologic<br />

data and also used to estimate maternal exposure in the populations studied.<br />

Another application was to estimate maternal and fetal PFOA plasma concentrations<br />

in communities with contaminated drinking water. <strong>The</strong> model development<br />

identified several research needs, including the identification <strong>of</strong> transporters involved<br />

in renal resorption to explain the multi-year half-lives <strong>of</strong> these compounds<br />

in humans, factors affecting the clearance <strong>of</strong> PFOA and PFOS during gestation,<br />

and data to estimate clearance <strong>of</strong> PFOA and PFOS in the human neonate.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!