27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

data, both in terms <strong>of</strong> the chemical space (industrial/agro chemicals vs. drugs) and<br />

in terms <strong>of</strong> the hCYP activity pr<strong>of</strong>iles, which for ToxCast include a large proportion<br />

(80%) <strong>of</strong> chemicals inactive in all 3 hCYP inhibition assays. <strong>The</strong> ToxCast inhibition<br />

assay results and the isoCYP metabolic activity data together afford the opportunity<br />

to explore SAR associations for a significantly expanded chemical space,<br />

using both features and chemical property descriptors to discern requirements for<br />

qualitative hCYP activity, as well as hCYP is<strong>of</strong>orm specificity. ToxCast Phase II is<br />

screening an additional 700 chemicals, including expanded sets <strong>of</strong> drugs and<br />

agro/industrial chemicals, in the same 3 hCYP inhibition assays, providing an ideal<br />

opportunity to prospectively evaluate the performance <strong>of</strong> the new models, as well as<br />

to iteratively improve upon these models.<br />

This abstract does not necessarily reflect U.S. EPA policy.<br />

142 EFFECTS OF GENETIC VARIATION IN ENZYME<br />

CYTOCHROME P450 2D6 ON XENOBIOTIC<br />

METABOLISM THROUGH IN SILICO MOLECULAR<br />

DOCKING MODELS.<br />

C. B. Tolson 1, 2 , Y. Tie 1 and E. Demchuk 1 . 1 Division <strong>of</strong> <strong>Toxicology</strong> &<br />

Environmental Medicine, ATSDR/CDC, Atlanta, GA and 2 Biomedical Engineering,<br />

Georgia Institute <strong>of</strong> Technology, Atlanta, GA. Sponsor: B. Fowler.<br />

Most drug metabolism performed in the liver is a result <strong>of</strong> enzymatic activity <strong>of</strong> the<br />

cytochrome P450 family <strong>of</strong> enzymes, especially cytochrome P450 3A4 and 2D6.<br />

Genetic variation is a large source <strong>of</strong> enzyme activity variability. This project determined<br />

whether methods <strong>of</strong> computational molecular docking can be used to detect<br />

the effects <strong>of</strong> genetic variation on substrate binding. <strong>The</strong> project is part <strong>of</strong> a large<br />

joint program <strong>of</strong> ATSDR/CDC and NCTR/FDA aimed at modeling liver toxicity.<br />

<strong>The</strong> project concentrates on enzymatic activity in CYP2D6 variants with the 120<br />

residue substitution (CYP2D6*53 and CYP2D6*49). Computational molecular<br />

docking modeling using FRED, Induced-Fit, and GLIDE was applied. <strong>The</strong> in silico<br />

docking models analyzed the interactions between individual ligands and the active<br />

sites <strong>of</strong> the CYP2D6 enzyme variants. <strong>The</strong> results suggested that computational<br />

molecular docking models could be used as accurate predictors in pre-clinical drug<br />

testing, but may have limited sensitivity to genetic variations beyond the active site.<br />

<strong>The</strong> FRED docking model best suited for the CYP2D6 enzyme-substrate system.<br />

Since data on human cytochrome P450 metabolism and associated genetic susceptibility<br />

have been obtained almost exclusively in clinical drug testing, better understanding<br />

<strong>of</strong> the molecular interactions between xenobiotics and the cytochrome<br />

P450 family will aid in the development <strong>of</strong> better predictive computational models<br />

to be used during pre-clinical drug development. Once a robust human model is<br />

developed, it can be applied in other areas, including chemical and biological defense<br />

and environmental health problems, such as estimation <strong>of</strong> metabolic parameters<br />

in physiologically-based pharmacokinetic modeling (PBPK) or more specific<br />

recommendations for toxic substance dosages based on genetic variants within a<br />

particular population.<br />

143 INCORPORATION OF TOXCAST IN VITRO ASSAY<br />

DATA AND RELEVANT TOXICITY PATHWAY<br />

INFORMATION IMPROVES THE EXTERNAL<br />

PREDICTION ACCURACY OF QUANTITATIVE<br />

STRUCTURE-ACTIVITY RELATIONSHIP (QSAR)<br />

MODELS OF CHEMICAL HEPATOTOXICITY.<br />

H. Zhu, L. Zhang, J. Staab, A. Sedykh, H. Tang, S. Gomez, I. Rusyn and A.<br />

Tropsha. University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC.<br />

<strong>The</strong> US EPA ToxCast program provides data for 610 in vitro assays, in which 320<br />

substances with known in vivo toxicity measured in 76 assays were tested. We have<br />

explored this complex data with novel computational approaches aimed to link<br />

chemical structures, in vitro responses, pathway information, and in vivo toxicity<br />

effects. We have used data from 22 ToxCast assays linked to Peroxisome<br />

Proliferator-Activated Receptor (PPAR) pathway to build biologically inferred<br />

QSAR models <strong>of</strong> six chronic animal hepatotoxicity endpoints. <strong>The</strong> datasets included<br />

228 and 235 compounds for mouse and rat hepatotoxicity subsets, respectively,<br />

and both conventional QSAR models and those using the following novel hierarchical<br />

QSAR modeling workflow were built. First, all chemicals were<br />

partitioned into two classes based on whether a compound was active (Class I) or<br />

inactive (Class II) in any <strong>of</strong> the PPAR-relevant assays; we found that there were 100<br />

Class I compounds and 128 Class II compounds for the mouse data and 106 Class<br />

I compounds and 129 Class II compounds for the rat data. Second, classification<br />

hepatotoxicity models for each endpoint were developed using both random forest<br />

and MOE binary QSAR approaches for Class I and II compounds, independently.<br />

30 SOT 2011 ANNUAL MEETING<br />

<strong>The</strong> prediction for external compounds is realized by initially placing them into<br />

Class I or II based on in vitro responses and then using class-specific QSAR models<br />

for in vivo toxicity prediction. <strong>The</strong> external prediction accuracy <strong>of</strong> models built<br />

with this two-step workflow was in the range <strong>of</strong> 57-76% for all six hepatotoxicity<br />

endpoints, while that achieved by conventional QSAR models was only 50-65%<br />

for the same external set. We conclude that methods discussed herein <strong>of</strong>fer novel<br />

ways <strong>of</strong> exploiting biological screening data to improve the accuracy <strong>of</strong> predictive<br />

toxicology models.<br />

144 COMPUTATIONAL DOCKING OF THE ISOMERS OF<br />

NONYLPHENOL TO THE LIGAND BINDING DOMAIN<br />

OF THE ESTROGEN RECEPTOR.<br />

J. Rabinowitz and S. Little. NCCT/ORD/Environmental Protection Agency, Research<br />

Triangle Park, NC.<br />

Nonylphenols are environmentally persistent endocrine disrupting chemicals. <strong>The</strong>y<br />

exist in the environment as complex mixtures containing many nonylphenol isomers.<br />

Environmental mixtures <strong>of</strong> nonylphenols, along with a few single isomers<br />

have been tested for their capacity to interact with the estrogen receptor (ER) and<br />

have been shown to be weakly estrogenic. <strong>The</strong> few individual isomers tested have<br />

only in rare examples been shown to be more estrogenic than environmental mixtures<br />

<strong>of</strong> undetermined isomers. <strong>The</strong> capacity <strong>of</strong> a molecule to bind to ER depends<br />

on the three dimensional structure <strong>of</strong> the potential ligand. <strong>The</strong>re are 211 geometric<br />

isomers and 551 stereo isomers <strong>of</strong> nonylphenol. <strong>The</strong> potential for each isomer to<br />

bind to ER will depend on the energetics the interaction. Computational molecular<br />

docking has been used to examine the capacity <strong>of</strong> each nonylphenol isomer to<br />

bind to ER. Computational targets have been created by removing the ligand<br />

atoms from crystal structures <strong>of</strong> ER-ligand complexes. <strong>The</strong> isomers <strong>of</strong> nonylphenol<br />

have been docked sucessfully into these targets. <strong>The</strong> results indicate that the potential<br />

for binding will be greatest when the molecular structure maximizes both the<br />

strength <strong>of</strong> the hydrogen bonds between the receptor and the phenolic hydroxyl<br />

group, and the steric interaction between the alkyl group and the hydrophobic core<br />

<strong>of</strong> the receptor. For some isomers the formation <strong>of</strong> the necessary hydrogen bonds<br />

forces the ligand-receptor complex into a structure with less favorable steric interactions.<br />

For example, branching at the alpha position decreases the interaction while<br />

branching in the beta and gamma positions <strong>of</strong>ten increases the interaction. Analysis<br />

<strong>of</strong> these considerations can provide insight for the design and manufacture <strong>of</strong><br />

nonylphenol (and other alkylphenol) mixtures that minimize their potential estrogenicity.<br />

[This abstract does not necessarily reflect U.S. EPA policy.]<br />

145 ANALYSIS OF RELATIVE ROSIGLITAZONE,<br />

PIOGLITAZONE, AND EPIRUBICIN CARDIOTOXICITY<br />

IN RATS USING HISTORICAL MICROARRAY DATA.<br />

H. Zhou and T. J. Colatsky. CDER/OPS/OTR/DAPR, U.S. FDA, Silver Spring, MD.<br />

Concerns have been raised about increased cardiovascular risk in patients treated<br />

with rosiglitazone (ROSI), but ROSI’s cardiovascular safety pr<strong>of</strong>ile remains an open<br />

question because <strong>of</strong> conflicting data on the existence and magnitude <strong>of</strong> the risk. We<br />

evaluated rat heart microarray data extracted from the Iconix DrugMatrix (IDM)<br />

database for ROSI (5d 1800 mg/kg/day; IDM high dose) and pioglitazone (PIO;<br />

5d 1500 mg/kg/day; IDM high dose), and compared the results to similar data obtained<br />

for epirubicin (EPI; 5d 2.7mg/kg/day; IDM high dose), a drug with well<br />

characterized cardiotoxicity. ROSI treatment induced 1002 differentially expressed<br />

genes (DEGs) in rat heart compared to 360 DEGs for PIO and 986 DEGs for EPI.<br />

Further analysis using Ingenuity Pathways Analysis revealed that ROSI and EPI induced<br />

more than twice the number <strong>of</strong> cardiotoxicity-related DEGs than PIO treatment<br />

but fewer than EPI (EPI:ROSI:PIO = 100:88:40). All 3 drugs were associated<br />

with NRF-2-mediated Oxidative Stress Response (EPI>ROSI>PIO), while ROSI<br />

and EPI both impacted Oxidative Stress and VDR/RXR activation. In addition,<br />

ROSI treatments altered 20 cardiac hypertrophy related molecules while PIO altered<br />

only 6. <strong>The</strong>se results could be correlated with the increase in heart weight produced<br />

by ROSI and PIO in these studies. Among the cardiotoxicity related molecules<br />

demonstrating altered expression, ROSI and PIO shared 17/111 common<br />

DEGs, ROSI and EPI shared 22/166 common DEGs, and PIO and EPI shared<br />

13/127 common DEGs while all the three shared only 6 common DEGs: CCNA2,<br />

CD36, FOS, HOPX, NCALD and PRKCH. In conclusion, these data suggest<br />

ROSI may be more cardiotoxic than PIO in rats, but the large number <strong>of</strong> unique<br />

cardiotoxicity related DEGs suggested multiple pathways exist for the cardiovascular<br />

effects <strong>of</strong> ROSI, PIO and EPI treatment. However, the relevance <strong>of</strong> these analyses<br />

to the more complex clinical situation still needs to be assessed.

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

Saved successfully!

Ooh no, something went wrong!