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.

containing substituents and/or hydrophobic ring systems as being significantly correlated<br />

with PL activity. Model features and scaffolds were organized using a hierarchical<br />

scaffold tree to illustrate their relatedness and their ability to predict PL independently.<br />

Closer examination <strong>of</strong> subsets <strong>of</strong> related scaffolds show the effects <strong>of</strong><br />

various molecular substitution patterns on the correlation with PL activity, as well<br />

as underscore the need to be judicious in selecting molecular features that best predict<br />

the training set without overly compromising the model’s domain <strong>of</strong> applicability<br />

by defining the local structure environment too narrowly.<br />

137 IN SILICO PREDICTIVE MODEL FOR DRUG-<br />

INDUCED PHOSPHOLIPIDOSIS USING BIOEPISTEME<br />

SOFTWARE.<br />

S. S. Choi, L. G. Valerio and N. Sadrieh. U.S. FDA, Silver Spring, MD.<br />

Drug-induced Phospholipidosis (DIPL) is a recognized finding in pharmaceutical<br />

drug development. DIPL is characterized by accumulation <strong>of</strong> drugs and phospholipids<br />

in lysosomes. Pathologically, DIPL manifests foamy macrophages or cytoplasmic<br />

vacuoles in various tissues <strong>of</strong> both animals and humans. <strong>The</strong>se pathologic<br />

findings can be confirmed by the appearance <strong>of</strong> lamellar inclusion bodies by electron<br />

microscopy. CADs (cationic amphiphilic drugs) are known to induce PL and<br />

share common structural features <strong>of</strong> containing hydrophobic ring structure and hydrophilic<br />

amine portion. This has led FDA to investigate the structural similarities<br />

and differences for active and inactive compounds. FDA has been developing a PL<br />

database that currently contains over 700 PL positive and negative drugs. Using this<br />

database and various computational programs, FDA has developed predictive<br />

QSAR models for DIPL. In this study, new In silico models were generated and validated<br />

using BioEpisteme, a toxicity screening and QSAR model development program,<br />

developed by the Prous Institute for Biomedical Research. BioEpisteme facilitates<br />

creation and application <strong>of</strong> In silico toxicity predictions based on molecular<br />

descriptors. In addition, it provides predictions on mechanism <strong>of</strong> action (MOA)<br />

(mechanism <strong>of</strong> action) for 432 different pharmacological targets. Preliminary performance<br />

statistics <strong>of</strong> a new DIPL QSAR model using a genetic algorithm <strong>of</strong><br />

BioEpisteme show 87% specificity, 73% sensitivity, and 82% accuracy. <strong>The</strong>se results<br />

were compared with other available FDA QSAR models. In addition, the<br />

MOA model was investigated to predict possible pharmaceutical MOA targets <strong>of</strong><br />

the DIPL data set. <strong>The</strong>se highly performing In silico predictive models will help to<br />

predict DIPL early in drug development and will subsequently serve to support regulatory<br />

decisions.<br />

138 PREDICTION OF DRUG-INDUCED LIVER INJURY IN<br />

HUMANS WITH TOXICOGENOMICS.<br />

M. Zhang, Q. Shi, M. Chen and W. Tong. NCTR, U.S. FDA, Jefferson, AR.<br />

Drug induced liver injury (DILI) is a leading cause <strong>of</strong> liver failure and the withdrawal<br />

<strong>of</strong> drugs from the market. However, the mechanism <strong>of</strong> DILI is still not well<br />

understood, and the prediction <strong>of</strong> the DILI potential for specific drugs remains a<br />

challenge. Over the past few years, large-scale microarray experiments testing DILI<br />

have been performed on animal models and have provided a good opportunity to<br />

understand DILI on the gene expression level. Combining our recently developed<br />

liver toxicity knowledge base- benchmark database (LTKB-DB), which groups<br />

drugs by DILI potential based on their drug labels, we used the microarray data<br />

from a previous toxicogenomic study to distinguish the DILI potentials <strong>of</strong> drugs by<br />

analyzing gene expression pr<strong>of</strong>iles <strong>of</strong> rats treated by those drugs. We focused on<br />

eighteen drugs with different DILI potentials and achieved a classification accuracy<br />

<strong>of</strong> over 80% using the leave one compound out cross validation process, which not<br />

only provided in silico evidence for drug categories in LTKB-DB, but also suggested<br />

the feasibility <strong>of</strong> predicting drug induced liver injury using microarray expression<br />

data. Additionally, the effects <strong>of</strong> the ALT/TBL level in the treated samples<br />

on the DILI potentials <strong>of</strong> the drugs were also discussed.<br />

139 PREDICTIVE VALUE OF CHEMICAL AND<br />

TOXICOGENOMIC DESCRIPTORS FOR DRUG-<br />

INDUCED HEPATOTOXICITY.<br />

Y. Low 1, 5 , T. Uehara 1, 2 , Y. Minowa 2 , H. Yamada 2 , Y. Ohno 3 , T. Urushidani 2, 4 ,<br />

A. Sedykh 5 , D. Fourches 5 , H. Zhu 5 , I. Rusyn 1 and A. Tropsha 5 . 1 Environmental<br />

Sciences & Engineering, University <strong>of</strong> North Carolina, Chapel Hill, NC,<br />

2 Toxicogenomics Informatics Project, National Institute <strong>of</strong> Biomedical Innovation,<br />

Osaka, Japan, 3 National Institute <strong>of</strong> Health Sciences, Tokyo, Japan, 4 Doshisha<br />

Women’s College <strong>of</strong> Liberal Arts, Kyoto, Japan and 5 Eshelman School <strong>of</strong> Pharmacy,<br />

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

Quantitative Structure-Activity Relationship (QSAR) modeling and toxicogenomics<br />

have been extensively explored independently as predictive toxicology tools.<br />

In this study, we employed a rigorous QSAR modeling workflow to predict the he-<br />

patotoxicity for 127 well-known drugs in the Toxicogenomics Informatics Project<br />

using both approaches. Predictor variables included chemical descriptors as well as<br />

biological descriptors represented by the short-term gene expression pr<strong>of</strong>ile for rat<br />

liver (24h after dosing). <strong>The</strong> prediction target, hepatotoxicity, was defined by<br />

histopathology and serum chemistry results after 28 days <strong>of</strong> treatment. We first developed<br />

conventional QSAR models using a comprehensive set <strong>of</strong> chemical descriptors<br />

and several classification methods. Using only chemical descriptors, external<br />

predictivity expressed as Correct Classification Rate (CCR) and evaluated by<br />

5-fold external cross-validation was as high as 59%. In parallel, models were built<br />

using only gene expression data. Optimal models comprising <strong>of</strong> 85 genes had CCR<br />

<strong>of</strong> 76%. Finally, hybrid models combining both chemical descriptors and gene expression<br />

data were also developed but their predictivity was under 76%. Although<br />

the accuracy <strong>of</strong> the QSAR models involving only chemical descriptors was limited<br />

due to the high chemical diversity <strong>of</strong> the small dataset, using both chemical and biological<br />

descriptors enriched the interpretation <strong>of</strong> the modeling results: the 85 predictor<br />

genes were mechanistically relevant, involving hepatotoxicity-related pathways,<br />

and structural alerts were identified among hepatotoxicants. This study shows<br />

that concurrent exploration <strong>of</strong> chemical features and early treatment-induced<br />

changes in gene expression affords predictive and interpretable models <strong>of</strong> subacute<br />

hepatotoxicity.<br />

140 CLASSIFICATION OF HUMAN CYTOCHROME 3A4<br />

LIGANDS BY MEANS OF MOLECULAR DOCKING.<br />

Y. Tie 1 , B. T. McPhail 1 , H. Hong 2 , R. D. Beger 2 , B. A. Fowler 1 and E.<br />

Demchuk 1 . 1 Division <strong>of</strong> <strong>Toxicology</strong> & Environmental Medicine, ATSDR/CDC,<br />

Atlanta, GA and 2 Division <strong>of</strong> Systems Biology, NCTR/U.S. FDA, Jefferson, AR.<br />

An interagency collaboration <strong>of</strong> ATSDR/CDC and NCTR/FDA was developed to<br />

model adverse drug-drug interactions mediated by CYP3A4 and 2D6. <strong>The</strong> modeling<br />

<strong>of</strong> CYP3A4 was carried out using several computational methods, including<br />

molecular docking. Molecular docking is a technique frequently used to estimate<br />

the binding interactions between ligands and a protein. Initially, 121 drugs were<br />

classified as strong and weak binders <strong>of</strong> CYP3A4. Docking studies were then performed<br />

by eHiTS, FRED, Glide, and SYBYL. Docking scores were used to develop<br />

multiple logistic regression (LR) models. <strong>The</strong> model with the least number <strong>of</strong> parameters<br />

(specificity <strong>of</strong> 100%, sensitivity <strong>of</strong> 42.4%, and overall accuracy <strong>of</strong> 84.3%<br />

at a LR probability cut<strong>of</strong>f <strong>of</strong> 0.5) was selected for final modeling and cross-validation<br />

(CV). A 10-fold CV was used. <strong>The</strong> average accuracy at the CV stage was<br />

83.8% and 79.3% for the training and CV sets, respectively. <strong>The</strong>se results were<br />

compared to those <strong>of</strong> two other models: SDAR (spectrum-data-activity relationship)<br />

and a DF-SAR (decision forest structure-activity relationship). Although the<br />

accuracy <strong>of</strong> classification was comparable among the modeling methods, the number<br />

<strong>of</strong> parameters, and consequently the robustness <strong>of</strong> the models, was different.<br />

While only 7 parameters were used in the LR docking, 27 descriptors were used in<br />

SDAR, and even a greater number in the DF-SAR: 5 trees ranging from 5 to 17 descriptors.<br />

All three models were used to predict which ligands in a test set <strong>of</strong> 120<br />

were strong or weak binders. In summary, molecular docking provides a unique<br />

way <strong>of</strong> CYP3A4 ligand classification, which is complementary to traditional<br />

S(D)AR modeling. While S(D)AR techniques are focused at intra-molecular properties,<br />

molecular docking takes account inter-molecular interactions. <strong>The</strong> three<br />

modeling methods combined provide an a priori assessment <strong>of</strong> hepatic drug-drug<br />

interactions and could serve as an adjunct to the conventional FDA drug approval<br />

process and to the public health review process <strong>of</strong> chemical safety by ATSDR.<br />

141 BUILDING STRUCTURE FEATURE-BASED MODELS<br />

FOR PREDICTING ISOFORM-SPECIFIC HUMAN<br />

CYTOCHROME P450 (HCYP 3A4, 2D6 AND 2C9)<br />

INHIBITION ASSAY RESULTS IN TOXCAST.<br />

P. Volarath 1 , B. Bienfait 2 , J. Gasteiger 2 , K. Houck 1 and A. Richard 1 . 1 NCCT,<br />

U.S. EPA, Research Triangle Park, NC and 2 Molecular Networks GmbH, Erlangen,<br />

Germany.<br />

EPA’s ToxCast project is using high-throughput screening (HTS) to pr<strong>of</strong>ile and prioritize<br />

chemicals for further testing. ToxCast Phase I evaluated 309 unique chemicals,<br />

the majority pesticide actives, in over 500 HTS assays. <strong>The</strong>se included 3<br />

human cytochrome P450 (hCYP3A4, hCYP2D6, hCYP2C9) inhibition assays<br />

that are routinely used to evaluate drug candidates for potential drug-drug interactions.<br />

isoCYP is a structure-activity relationship (SAR) prediction model built on a<br />

training set <strong>of</strong> hCYP is<strong>of</strong>orm-specific metabolism data that uses a decision-tree approach<br />

to predict which <strong>of</strong> the these 3 CYP is<strong>of</strong>orms is preferentially involved in<br />

drug metabolism. <strong>The</strong> published isoCYP models were derived from a training set <strong>of</strong><br />

146 drugs for which the particular single metabolizing CYP is<strong>of</strong>orm is known in<br />

each case (Terfloth et al., 2007, J.Chem. Inf. Model., 47:1688-1701). ToxCast results<br />

for the 3 hCYP inhibition assays differ significantly from the isoCYP model<br />

SOT 2011 ANNUAL MEETING 29

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

Saved successfully!

Ooh no, something went wrong!