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.

2006, 40:6269-74). Rats received either a low (11.2 mg/kg) or high (27.4 mg/kg)<br />

pyrethroid mixture dose and were sacrificed at 1, 2, 4, 8 or 24 h after dosing. Blood,<br />

liver, fat and brain were removed. Tissues were extracted and analyzed for parent<br />

chemical by LC/MS/MS. For both doses, pyrethroid residues were greater in fat relative<br />

to blood, brain and liver, based on area under the curve from 0 to 24 h (AUC0- 24h ) and maximal concentration (Cmax ). With dose, the AUC0-24h increased 4- to 9fold<br />

in blood, 2- to 3-fold in brain, 2-fold in fat and 4- to 6-fold in liver. <strong>The</strong>re were<br />

similar increases in pyrethroid tissue Cmax with dose. Cypermethrin and cis-permethrin<br />

consistently had the highest AUC0-24h and Cmax in all tissues. trans-<br />

Permethrin consistently had lower AUC0-24h and Cmax in all tissues, which may be<br />

due to increased metabolism relative to the other pyrethroids. Determining the<br />

pharmacokinetics <strong>of</strong> a mixture <strong>of</strong> environmental chemicals such as the pyrethroids<br />

is important when assessing the risk to chemicals that have a similar mode <strong>of</strong> action.<br />

(This abstract does not represent US EPA policy.)<br />

2263 EVALUATING PHARMACOKINETICS AND<br />

BIOAVAILABILITY OF LEVOFLOXACIN IN RATS.<br />

A. Banerjee 1 , K. Kabirov 1 , T. Martín-Jiménez 2 and A. Lyubimov 1 . 1 <strong>Toxicology</strong><br />

Research Laboratory, University <strong>of</strong> Illinois at Chicago, Chicago, IL and 2 College <strong>of</strong><br />

Veterinary Medicine, University <strong>of</strong> Tennessee, Knoxville, TN.<br />

Drug exposure <strong>of</strong> the antibiotic lev<strong>of</strong>loxacin was examined in Sprague Dawley rats<br />

following oral and intravenous administration. Six female and six male cannulated<br />

rats were randomly assigned to two treatment groups (3 males + 3 females/ group).<br />

One group received a single IV dose <strong>of</strong> lev<strong>of</strong>loxacin at 25 mg/kg and the other received<br />

a single oral dose at 50 mg/kg. Blood samples were collected, analyzed and<br />

the data used for pharmacokinetic analysis. Intravenous pharmacokinetic pr<strong>of</strong>iles<br />

were very similar in males and females, with the exception <strong>of</strong> systemic clearance,<br />

which was 2222 and 2667 ml/hr/kg in males and females, respectively. Accordingly,<br />

the AUC0-inf values were higher in males (11264 vs. 9464). <strong>The</strong> estimated values<br />

<strong>of</strong> C0, Cmax, Vss and terminal elimination half-life were very similar across genders.<br />

<strong>The</strong> volume <strong>of</strong> distribution was rather large (Vss: 3.2-3.6 L/kg), which indicates<br />

wide distribution and high affinity <strong>of</strong> the drug for proteins and perhaps other<br />

molecules in tissue. <strong>The</strong> absolute oral bioavailability <strong>of</strong> lev<strong>of</strong>loxacin in rats was 34%<br />

in males and 47% in females which is relevant for a concentration-dependent antibiotic<br />

such as lev<strong>of</strong>loxacin. Accordingly, the AUC0-inf after the oral administration<br />

was lower in males than in females (7668 and 8952 ng*hr/mL, respectively).<br />

<strong>The</strong> larger gender-related difference in oral pharmacokinetics was in Cmax and average<br />

values in males and females were 1370 and 2733 ng/mL, respectively.<br />

Coefficients <strong>of</strong> variability for this parameter were rather high (43-59%). <strong>The</strong> halflife<br />

<strong>of</strong> the drug after intravenous administration was short (1.5-2.0 hr) but it increased<br />

after oral administration (3.9-4.1 hr). Mean terminal elimination half-lives<br />

were very similar in males and females (3.9 and 4.1, respectively). In conclusion,<br />

bioavailability seen in rats is about 2-fold lower than in humans indicating species<br />

based differences in absorption pr<strong>of</strong>ile <strong>of</strong> lev<strong>of</strong>loxacin.<br />

2264 ROLE OF INTESTINAL P450 AND P-GLYCOPROTEIN<br />

IN MODULATING THE BIOAVAILABILITY OF ORAL<br />

LOVASTATIN.<br />

Y. Zhu. Environmental Health Sciences, Wadsworth Center, New York State<br />

Department <strong>of</strong> Health, and School <strong>of</strong> Public Health, State University <strong>of</strong> New York at<br />

Albany, Albany, NY. Sponsor: X. Ding.<br />

Variations in drug metabolism and transport can affect drug bioavailability, and potentially<br />

cause drug toxicity. However, the roles <strong>of</strong> intestinal enzymes or drug transporters<br />

in controlling the bioavailability <strong>of</strong> oral drugs are not well defined for many<br />

drugs. Our aim was to determine the specific roles <strong>of</strong> small intestinal (SI) P450 enzymes<br />

and the P-glycoprotein (P-gp) in the first-pass clearance <strong>of</strong> lovastatin (LVS),<br />

an anticholesterol drug. In an intestinal epithelium (IE)-specific P450 reductase<br />

(CPR) knockout (IE-Cpr-null) mouse model, which has little P450 activities in the<br />

IE, we found that the rates <strong>of</strong> LVS metabolism by SI microsomes were reduced by<br />

>90%, compared with wild-type (WT) mice. In vivo clearance <strong>of</strong> oral LVS was<br />

much slower in IE-Cpr-null than in WT mice; consequently, the bioavailability <strong>of</strong><br />

LVS was 14% in IE-Cpr-null mice, but only 5% in WT mice. In contrast, the<br />

clearance rates for intraperitoneally dosed LVS were not different between the two<br />

mice strains. <strong>The</strong>se results demonstrate a predominant role <strong>of</strong> SI P450 enzymes in<br />

the first-pass clearance <strong>of</strong> LVS. This conclusion was further supported by additional<br />

data, obtained through pharmacokinetic studies on a liver-specific Cpr knockout<br />

(liver-Cpr-null) mouse model, indicating minimal contribution <strong>of</strong> hepatic P450<br />

enzymes to the first-pass clearance <strong>of</strong> oral LVS. <strong>The</strong> specificity <strong>of</strong> the impact <strong>of</strong> the<br />

CPR loss on SI P450 activity was confirmed by the results from pharmacokinetic<br />

studies for pravastatin (PVS), a drug which is similar to LVS, but is not a P450 sub-<br />

486 SOT 2011 ANNUAL MEETING<br />

strate. We then determined whether P-gp contribute to LVS clearance, by using cyclosporin<br />

A (CsA), an inhibitor <strong>of</strong> both P450 and P-gp, and by taking advantage <strong>of</strong><br />

the absence <strong>of</strong> P450 activities in the SI <strong>of</strong> the IE-Cpr-null mice. We found that CsA<br />

treatment (oral) significantly enhanced the systemic exposure <strong>of</strong> LVS in both IE-<br />

Cpr-null and WT mice, a result strongly suggest that P-gp also influences the<br />

bioavailability <strong>of</strong> oral LVS in mice. (Supported in part by NIH grant GM082978)<br />

2265 PARAQUAT DISPOSITION: A POTENTIAL ROLE FOR P-<br />

GLYCOPROTEIN TRANSPORT.<br />

S. Lacher, F. Cardozo-Pelaez and E. L. Woodahl. University <strong>of</strong> Montana,<br />

Missoula, MT.<br />

P-glycoprotein (P-gp) is an efflux drug transporter expressed in many tissues important<br />

in xenobiotic disposition. At the blood brain barrier (BBB), P-gp is one <strong>of</strong><br />

the mechanisms that protects the brain from toxic substances circulating in the<br />

blood. P-gp has been proposed to play a role in the susceptibility to Parkinson’s disease<br />

(PD), possibly by altering accumulation <strong>of</strong> neurotoxicants in the brain.<br />

However, evidence is missing as to whether neurotoxicants associated with PD are<br />

P-gp substrates and if P-gp alters their ability to cross the BBB. Our goal is to measure<br />

P-gp transport <strong>of</strong> neurotoxic xenobiotics associated with PD across the BBB.<br />

Our recent studies focus on the pesticide paraquat with the goal <strong>of</strong> measuring<br />

paraquat toxicokinetics, paraquat brain accumulation, and markers <strong>of</strong> neurodegeneration<br />

in FVB mice. We administered paraquat to FVB mice to measure brain accumulation<br />

and estimate paraquat toxicokinetics. Two different groups <strong>of</strong> FVB<br />

mice were administered a single oral dose <strong>of</strong> paraquat (10mg/kg and 25mg/kg),<br />

plasma samples were collected at 1, 2, 4, and 8 hours, and the brains were harvested<br />

at 8 hours. Plasma and brain samples were analyzed for total paraquat concentration<br />

using high performance liquid chromatography. We were able to detect<br />

paraquat in the plasma and brain samples in both treatment groups. We found that<br />

the average elimination half-lives <strong>of</strong> the 10mg/kg and the 25mg/kg treatment<br />

groups were similar at approximately 8 hours, which suggests that there is no dose<br />

effect on half-life. Average brain accumulations were 1.02 +/- 0.21μg and 0.95 +/-<br />

0.44μg after 10mg/kg and 25mg/kg doses, respectively. Our future studies will examine<br />

the toxicokinetics <strong>of</strong> paraquat disposition in P-gp knock-out (mdr1a-/-<br />

/mdr1b-/-) mice, as well as identifying markers <strong>of</strong> neurodegeneration. This study<br />

will provide the missing data to measure P-gp-mediated transport <strong>of</strong> paraquat and<br />

the susceptibility to Parkinson’s disease.<br />

2266 THE IMPORTANCE OF EFFLUX TRANSPORTERS IN<br />

CNS EXPOSURE TO AVERMECTIN INSECTICIDES.<br />

A. M. Dalzell 1 , F. M. Williams 2 , P. Mistry 3 , J. Wright 3 and C. D. Brown 1 . 1 Cell<br />

& Molecular Biosciences, University <strong>of</strong> Newcastle, Newcastle-Upon-Tyne, United<br />

Kingdom, 2 Medical <strong>Toxicology</strong> Centre, University <strong>of</strong> Newcastle, Newcastle-Upon-<br />

Tyne, United Kingdom and 3 Syngenta, Bracknell, United Kingdom. Sponsor: R.<br />

Peffer.<br />

Avermectins are used as agricultural insecticides and as anti-helminthic treatments<br />

in man. Avermectins have good safety pr<strong>of</strong>iles in a variety <strong>of</strong> species; however neurotoxicities<br />

observed in some CF-1 mice initiated investigations into the relevance<br />

<strong>of</strong> this mouse model for human risk assessment. Susceptible CF-1 mice lack the<br />

functional xenobiotic efflux transporter, P-glycoprotein (mdr1a/P-gp), demonstrating<br />

its importance in neuroprotection. In humans a non-functional MDR1 has yet<br />

to be identified, although the importance <strong>of</strong> MDR1 in xenobiotic efflux in man<br />

needs to be better understood. In this study we compared two neuroblastoma cell<br />

lines; human SH-SY5Y cells, characterised for uptake and efflux transporter expression,<br />

with mouse N2a cells to identify species differences in avermectin handling.<br />

Intracellular levels <strong>of</strong> the fluorescent MDR1-substrate H33342 dye in the presence<br />

<strong>of</strong> avermectins (0-7μM) were compared to the increase (p emamectin > abamectin, in human and mouse<br />

cells. Parallel experiments using a CMFDA dye assay suggest that the avermectins<br />

may be substrates for MRP efflux transporters expressed in both cell types

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

Saved successfully!

Ooh no, something went wrong!