You are on page 1of 10

YTAAP-12021; No.

of pages: 10; 4C:


Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

Contents lists available at ScienceDirect

Toxicology and Applied Pharmacology


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / y t a a p

Utilizing toxicogenomic data to understand chemical mechanism of action in


risk assessment☆
Vickie S. Wilson a,⁎, Nagalakshmi Keshava b, Susan Hester a, Deborah Segal b, Weihsueh Chiu b,
Chad M. Thompson c, Susan Y. Euling b
a
National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
b
National Center for Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, 1200 Pennsylvania Ave., NW, Washington, DC 20460, USA
c
ToxStrategies, Inc., 23501 Cinco Ranch Blvd., Suite G265, Katy, TX 77494, USA

a r t i c l e i n f o a b s t r a c t

Article history: The predominant role of toxicogenomic data in risk assessment, thus far, has been one of augmentation of
Received 12 October 2010 more traditional in vitro and in vivo toxicology data. This article focuses on the current available examples of
Revised 25 January 2011 instances where toxicogenomic data has been evaluated in human health risk assessment (e.g., acetochlor and
Accepted 25 January 2011
arsenicals) which have been limited to the application of toxicogenomic data to inform mechanism of action.
Available online xxxx
This article reviews the regulatory policy backdrop and highlights important efforts to ultimately achieve
Keywords:
regulatory acceptance. A number of research efforts on specific chemicals that were designed for risk
Mechanism of action assessment purposes have employed mechanism or mode of action hypothesis testing and generating
Toxicogenomics strategies. The strides made by large scale efforts to utilize toxicogenomic data in screening, testing, and risk
Risk assessment assessment are also discussed. These efforts include both the refinement of methodologies for performing
toxicogenomics studies and analysis of the resultant data sets. The current issues limiting the application of
toxicogenomics to define mode or mechanism of action in risk assessment are discussed together with
interrelated research needs. In summary, as chemical risk assessment moves away from a single mechanism
of action approach toward a toxicity pathway-based paradigm, we envision that toxicogenomic data from
multiple technologies (e.g., proteomics, metabolomics, transcriptomics, supportive RT-PCR studies) can be
used in conjunction with one another to understand the complexities of multiple, and possibly interacting,
pathways affected by chemicals which will impact human health risk assessment.
Published by Elsevier Inc.

Introduction of action (Hamadeh et al., 2002a, 2002b) and, in general terms, screen for
adverse effects on human health of pharmaceutical drugs under
The ability to assess a collection of biological changes in response to development. Screening and prioritization data can certainly complement
exposure to environmental chemicals in toxicological evaluations and risk other toxicological evaluations in risk assessment especially as an aid to
assessments is an attractive promise of genomic and related “-omics” early problem formulation. The goals of screening applications, however,
technologies. If this potential was fully realized, toxicogenomic data could ultimately have a different purpose than risk assessment. In a screening
contribute to identification of the multiple modes of action (MOA) and the and prioritization context, genomic data interpretation can be less precise
underlying detailed mechanistic steps involved in chemical toxicity. Omic than would be required for risk assessment. For example, a relatively high
technologies have been successfully applied in screening and prioritiza- false positive rate and low false negative rate in a genomic assay may be
tion programs, particularly in the pharmaceutical industry. For example, acceptable for screening purposes (e.g., clinical screens, chemical
microarrays, reverse transcriptase-polymerase chain reaction (rt-PCR), screening programs, early stages of drug development) because false
proteomics, and metabolomics data have been used to inform mechanism positives would be discovered and eliminated in subsequent “testing”
phases. Assays with high false positive rates and low false negative rates,
however, are not as useful when performing a risk assessment. Use of
☆ Disclaimer: This article has been reviewed by the U.S. Environmental Protection genomic data in risk assessment requires that the data and statistical
Agency and is approved for publication. The views expressed in this article are those of analysis be reproducible and sensitive (with a low false positive rate) to
the authors and do not necessarily reflect the policies of the U.S. Environmental confirm genomic changes as well as to evaluate the magnitude of the
Protection Agency. health risk associated with the genomic change after exposure. The more
⁎ Corresponding author at: National Health and Environmental Effects Research
Laboratory, Office of Research and Development, U.S. Environmental Protection Agency,
firmly a change in gene expression or other cellular level alteration can be
MD-72, Research Triangle Park, NC 27711, USA. Fax: + 1 919 541 4017. linked to an adverse health outcome or malformation (e.g. phenotypic
E-mail address: wilson.vickie@epa.gov (V.S. Wilson). anchoring), the more useful it is to risk assessment.

0041-008X/$ – see front matter. Published by Elsevier Inc.


doi:10.1016/j.taap.2011.01.017

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
2 V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

In light of the recent National Academy of Science (NAS) report (NRC, this issue), intra-species (Mortensen and Euling, 2010—this issue) and
2007b) that developed a long-term vision for moving to a risk inter-species differences in MOA, and toxicokinetics. A number of issues,
assessment paradigm based on identifying toxicity pathways, it is however, must be surmounted before genomic technologies can be fully
important to determine how to best utilize and/or mine toxicogenomic utilized in risk assessment (Pettit et al., 2010) and, therefore, approaches
data for risk assessment. The NAS report pointed to the strong potential to using these datasets in a risk assessment scenario continue to be
for genomic technologies to contribute to the risk assessment process explored (Hackett and Lesko, 2003; Boverhof and Zacharewski, 2006).
but acknowledged that neither its full impact has been realized nor has it
been broadly implemented in the field.
As definitions vary, the toxicogenomic terms used in both this article Use of toxicogenomic data in risk assessment
and this special issue are described. -Omics is the broad discipline that
analyzes genome-wide interactions within a biological system. Various Standardization and validation of techniques
subsets of -omics have developed including genomics, epigenomics,
transcriptomics, proteomics, and metabolomics. Genomics is the study A significant challenge in the use of microarray technologies is the lack
of an organism's DNA sequence (i.e., genome). Epigenomics or of standards for presenting and exchanging such data. As a result, a
epigenetics relates to the mechanisms by which parts of the genome consortium of scientists from the Microarray Gene Expression Data
are switched off or on at strategic times and locations without actual (MGED) Society developed and agreed upon the Minimum Information
modification of gene sequence. Transcriptomics measures genome-wide Standards for Microarray Experiments (MIAME) for publications and
mRNA expression or, in other words, genes that have been transcribed at submission to public repositories (Brazma et al., 2001; Ball et al., 2004).
a particular point in time (NRC, 2007a). Proteomics is the study of a Since its inception, this database has become the “gold-standard” for
broad spectrum of proteins within an organism, which might include generating, analyzing, sharing, and publishing microarray-based data
their expression at a particular point in time, structural status (e.g., (Table 1).
phosphorylated or dephosphorylated), functional states (i.e., specificity A more recent large-scale standardization effort is the MicroArray
and activity level), and their interactions with other cellular components Quality Control (MAQC) Consortium, a scientific community-wide
(Pandey and Mann, 2000). Metabolomics and metabonomics both study effort, spearheaded by FDA scientists, which brought together
low molecular weight (LMW) molecules that are produced as the net researchers from government, industry, and academia to address
result of cellular function (NRC, 2007a), but the former refers to the study some of the issues of historical variability and to contribute to the
of LMW molecules within cells, whereas the latter refers to a more standardization of microarray procedures. This consortium in its first
systemic and complex change in tissues and body fluids (Ekins et al., two phases, through various experimental design modifications and
2005). Toxicogenomics (TGx) is the application of any of these -omics- technical discussions, has contributed significantly towards providing
based techniques to characterize responses to a toxic stimulus (NRC, reproducible results within sites, between sites, and among the
2007a). various platforms (Canales et al., 2006; Casciano and Woodcock,
One key strength of microarray and related -omics technologies is the 2006; Dix et al., 2006; Frueh, 2006; Guo et al., 2006; Ji and Davis, 2006;
ability to generate information about thousands of genes or proteins (or Patterson et al., 2006; Shi et al., 2006; Shippy et al., 2006; Tong et al.,
entire genomes or proteomes) in a single assay. Transcriptomics uses high 2006). The group's goal is to reach consensus among stakeholders on
density and/or high-through-put methods of assessing mRNA expression the best practices for the development and validation of predictive
and new, second-generation microarrays are designed for the maximum models based on microarray gene expression as well as genotyping
coverage of genes across the genome. However, the evaluation, data for personalized medicine and safety assessment (Table 1).
confirmation and interpretation of such a large, high density data set Briefly, in MAQC I, four pools of RNA samples were evaluated using
presents additional challenges (Pettit et al., 2010). Specialized quantitative seven microarray platforms (6 commercially available and one
real-time reverse transcriptase-polymerase chain reaction (qrt-PCR)- National Cancer Institute produced microarray spotted with Operon
based approaches that focus on specific genes or specialized sub-sets of oligonucleotides) by three independent laboratories per platform
gene expression changes have also been developed. Qrt-PCR has with five replicates per RNA sample. The working list of genes was
developed into a powerful, preferred confirmatory tool for quantifying refined to include 12,091 reference genes that were detected on each of
gene expression with improvements in sensitivity and specificity and the six high density platforms. Generally there was good concordance
efficiency for signal detection. across the various platforms. For example, all one-color microarray
Pathway-level analysis usually requires the data analyst or bioinfor- platforms (such as Affymetrix) had median coefficients of variation (CVs)
matician to employ several tools to adequately mine the data set, as each of 5–15% and a concordance rate of 80–95% for their qualitative detection
software tool provides a different perspective and evaluation of the data. calls (detected or non-detected) between sample replicates. When
Bioinformatics is an overarching term referring to the creation of archival comparing variation within and between test sites, the one-color
databases, statistical algorithms, and specific analytical techniques for platforms demonstrated 80−95% agreement (Shi et al., 2006). Further,
both management and analysis of biological data. The primary goal of the results from three qrt-PCR-based platforms were evaluated against
bioinformatics is to increase the understanding of biological processes five of the commercial microarray platforms. Linear regression analysis
through the application of computationally intensive techniques such as indicated good agreement across the three qrt-PCR platforms with R2
pattern recognition and visualization of high-dimensional data (http:// values from 0.81 to 0.88. There was also very good correlation of fold
www.ncbi.nlm.nih.gov/Tools/). Challenges to risk assessment include the change data between microarray results and quantitative gene expression
selection of appropriate data analysis tools as well as the interpretation of results. Several sources for limited incongruence were identified such as
the results. decreased sensitivity for low expression genes in the microarray platforms
The goal of this article is to review advances in the application of as compared to the qrt-PCR-based technologies and differences in probe
toxicogenomic technologies to inform mechanism of action for risk location (Canales et al., 2006).
assessment, including strengths and limitations regarding the use of -omic In MAQC-II the capabilities and limitations of various data analysis
technologies and data analysis tools in risk assessment. As such, this article methods were assessed. The information and conclusions have been
focuses on the largely qualitative application of toxicogenomic data to recently published in a series of articles in the Pharmacogenomics
informing mechanism of action for risk assessment. Journal (Aug 2010) and Nature Biotechnology (Shi et al., 2010). The
Further, toxicogenomic data have the potential to inform other third phase, MAQC-III aims to evaluate the technical performance of
areas of risk assessment which are addressed in other articles within next-generation sequencing platforms and phase IV, MAQC-IV, is
this edition, including dose response assessment (Chiu et al., 2010- being planned with an objective of assessing inter-individual

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx 3

Table 1
Some of the major activities, including documents, tools, and workshops, in the field of toxicogenomics that have significantly impacted the acceptance and use of toxicogenomic
data in research and regulatory arenas.

Activity (source) Description Key point(s) References

Research and resources


MicroArray Quality Control MAQC is a scientific community-wide research ▪ MAQC I resulted in demonstration of http://www.fda.gov/ScienceResearch/
Consortium (MAQC) (multi-stakeholder effort, led by U.S. FDA, including MAQC I, II, and III relatively high reproducibility within and BioinformaticsTools/
scientific and regulatory community) (to date). The goal of the project is to assess between laboratories and among microarray MicroarrayQualityControlProject/
microarray study variability and develop platforms (but not different biological sample default.htm
standards and quality measures in order for preparations).
microarray and next generation sequencing data ▪ MAQC II assessed predictive models among 36 MAQC Consortium (Shi et al., 2006)
to be used in a regulatory context. teams, for microarray data analysis. The
conclusions are that model performance
depended on the endpoint and the team and that
the performance of models generated using
different methods were quite similar.
▪ The goal of MAQC-III (or Sequencing Quality MAQC Consortium (Shi et al., 2010)
[SEQC]) is to assess the performance of next-
generation sequencing platforms by developing
benchmark datasets with reference samples and
assessing advantages and limitations of various
RNA and DNA bioinformatic approaches.
Application of toxicogenomics The document, aimed at regulatory decision- Overall conclusions regarding risk NRC, 2007a
technologies to predictive toxicology makers, presents the potential benefits and assessment include:
and risk assessment (NRC) limitations to utilizing genomic data and ▪ The reproducibility issues surrounding
committee recommendations to address toxicogenomic tools have been largely resolved
challenges. but regulatory validation of technologies needed;
▪ Specialized bioinformatic, statistical, and
computational methods are needed;
▪ Potential of toxicogenomic data to improve
risk assessment is just beginning and is not
ready to replace current toxicity testing;
▪ Need to improve govt agencies' ability to
integrate toxicogenomic data into the
different risk assessment steps/areas by
investing in research and personnel.
Toxicity testing in the 21st century: a The document presents a future vision of a Toxicity tests would be based on identifying NRC, 2007b
vision and a strategy (NRC) toxicity-pathway based toxicity testing and risk perturbations in key human toxicity pathways
assessment, representing a paradigm shift. from a combination of human in vitro tests and
computational tools.

Policies and standards


Minimum Information About a Established to provide guidance for a minimum The MIAME standards were developed in order http://www.mged.org/
Microarray Experiment (MIAME) amount and type of microarray information to 1) allow for the interpretation of the results of Brazma et al., 2001
(multistakeholder; The Functional needed to publish microarray papers. The goal the experiment unambiguously; and 2) to
Genomics Data (FGED) Society) was to have uniformity among scientists for the reproduce the experiment.
amount of microarray data needed for
publication. Currently, several public
repositories exist such as ArrayTrack™ at the
FDA (U.S.), ArrayExpress at the European
Bioinformatics Institute (EBI) (U.K.), GEO at
NCBI (U.S.), and CIBEX at DDBJ (Japan), which
are designed to accept, hold, and distribute
MIAME-compliant microarray data.
Guidance for industry to The guidance provides recommendations to FDA is requiring for some cases, and accepting, in FDA, 2005; http://www.fda.gov/
incorporate submission of industry with investigational new drug other voluntary cases, genomic data for some downloads/RegulatoryInformation/
pharmacogenomic data (FDA) applications, new drug applications, and drug applications. Guidances/ucm126957.pdf
biologics license applications on when, during
product development and review processes,
the pharmacogenomic data should be
submitted; the submission format; and how
and in what cases the data will be used in
regulatory decision making.
Interim policy on genomics (EPA) Outlines the strengths and limitations to using Genomic data can be used on a case-by-case basis U. S. EPA, 2002; http://www.epa.gov/
genomic data for EPA regulatory decision- in a weight-of-evidence approach in EPA risk spc/pdfs/genomics.pdf
making. Encourages the submission and assessments.
evaluation of genomic data, when available, as
supporting material, particular for
characterizing a chemical's MOA.
Draft interim guidance for microarray- Document developed to respond to the need Regarding submission of microarray data to EPA, U. S. EPA, 2007 http://www.epa.gov/
based assays: data submission, quality, for a framework for analysis and acceptance the document recommends that submissions spc/pdfs/
analysis, management, and training criteria for genomic data for use in regulatory include sufficient information to allow an epa_interim_guidance_for_microarray-
considerations (EPA) decision-making. The document covers the independent reviewer to reconstruct how the based_assays-external-review_draft.
performance of assays across genomic data were collected and analyzed and further, pdf
platforms (e.g., reproducibility, sensitivity, proposes a slight modification to MIAME format
pathway analysis tools) and the criteria for for EPA submissions. Regarding data analysis, the
accepting genomic data for use in a risk document presents a systematic approach, a
assessment (e.g., assay validity, biologically genomics Data Evaluation Record template to
meaningful response). present and organize data from genomics studies.

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
4 V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

variability in response to treatment with an emphasis on idiosyncratic Genomics Task Force that produced a white paper, Potential Implications of
adverse drug reactions. Genomics for Regulatory and Risk Assessment Applications at EPA, which
The MAQC project evaluated intrasite, intersite and interplatform identified four areas of oversight likely to be influenced by genomic data
differences, and has contributed much valuable information for and identified needs for (a) analysis and acceptance criteria for genomic
evaluation of TGx data. There are several issues of value to the risk information in scientific and regulatory applications, (b) methods for
assessment process, however, that were not addressed. Because all interpreting genomic information for risk assessment, and (c) deter-
test sites used the same RNA samples, protocol, and generated mining a relationship between genomic changes and adverse outcomes
replicate data at approximately the same time (Shi et al., 2006), the (U.S. EPA, 2004b). In response, the Genomics Technical Framework and
evaluation does not address different sample preparation methods, Training Workgroup, developed an Interim Guidance for Microarray-
variation in protocols, effect of time differences or other technical Based Assays: Data Submission, Quality, Analysis, Management, and
variables. It also did not include more “biology-based” performance Training Considerations (U. S. EPA, 2007). This document made specific
metrics such as pathway analysis. Further, while providing valuable recommendations for the submission, quality assurance, analysis, and
information, the MAQC efforts are all technique and technology focused. management of genomics data in the context of current possible
Little attention has been dedicated to other needs for the use of TGx data in applications.
risk assessment such as study design including dose–response, length of The NRC report, Applications of Toxicogenomic Technologies to Predictive
exposure and adequate statistical power especially at low doses. These Toxicology and Risk Assessment (NRC, 2007a) and Toxicity Testing in the 21st
factors all currently limit the application of TGx data to the risk assessment Century: a Vision and a Strategy (NRC, 2007b) recommends a toxicity-
process. pathway perturbation approach to toxicity testing and risk assessment
The development of improved toxicogenomic tools for the and making full use of the rapidly evolving scientific understanding of
technologies and data analysis, including publicly-available resources, molecular pathways that maintain cell function and influence the causal
has led to increased sensitivity and reproducibility of the resulting pathway to disease. The NAS vision (NRC, 2007b) to move to a process
toxicogenomic data, and as a result, improves the ability to evaluate that relies more on in vitro assays has been adopted by EPA (U. S. EPA,
and interpret these data for risk assessment. For example, multiple 2009).
species now have complete genome sequence data available. The
availability of the DNA genome sequences for multiple species used in Using toxicogenetics data to inform mechanism of action for risk
toxicity testing and risk assessment should enhance the development assessment purposes
of more complete microarrays. The availability of the human genome
sequence allows for comparisons to be made between the function of Toxicogenomic data can be used in risk assessment of a given chemical
genes and pathways between test species and humans. Another at multiple levels (e.g., toxicokinetic, toxicodynamic), depending on the
development that has improved toxicogenomic tools is the develop- available data, to inform the exposure to outcome continuum. Using data
ment of “2nd” generation microarrays that represent a majority of the from multiple genomic approaches (genome, transcriptome, proteome,
species’ genes and microarrays that represent genes from particular and metabolome) is promising for defining the temporal steps in the
pathways or processes with enhanced capability to provide informa- process of toxicity and for identifying precursor events. To do so, however,
tion about altered gene expression and affected pathways. As would require the integration of multiple disciplines and require expertise
mentioned earlier, the development of quantitative RT-PCR allows from multiple fields. Further, studies would need to be designed with risk
for assessment of the quantity and timing of expression of genes of assessment requirements for dose response information and linkages to
interest. There are numerous repositories for microarray data (e.g., adverse outcome. The predominant role of toxicogenomic data in risk
Array Express) that are consistent with MIAME compliance that have assessment thus far has been one of augmentation of other more
improved transparency of toxicogenomic data. Further, the develop- traditional in vitro and in vivo toxicology data. Nevertheless, considera-
ment and testing of data analysis tools, including publicly-available tools tions and approaches for using toxicogenomic data sets in risk assessment
(National Center for Biotechnology Information, U.S. National Library of continue to be explored (Waters and Fostel, 2004; Chan and Theilade,
Medicine, NIH EntrezGene, http://www.ncbi.nlm.nih.gov/gene; NIEHS 2005; Oberemm et al., 2005; Boverhof and Zacharewski, 2006; NRC,
Environmental Genome Project, http://www.niehs.nih.gov/research/ 2007a, 2007b; Thybaud et al., 2007; Daston, 2008; Carlson and Silkworth,
supported/programs/egp/; National Human Genome Research Institute, 2009; Van Aggelen et al., 2010). This section provides an overview of how
http://www.genome.gov/; Wellcome Trust's The Human Genome, toxicogenomic data, depending on the type, could potentially be used in
http://genome.wellcome.ac.uk), have provided the scientific and regu- screening and prioritization programs and risk assessment to inform a
latory communities with multiple tools for different applications, some chemical's mechanism of action, defined here as toxicokinetics and
of which are more appropriate for risk assessment than others (see toxicodynamics steps (Fig. 1).
(Ovacik et al., 2010—this issue). Toxicokinetics (TK) is the rate at which a chemical enters the body and
what happens to the chemical within the body. Pre-and post-exposure
Regulatory guidance information on gene and protein expression, protein function, and LMW
molecules (e.g., GSH) can provide important information about the
Many regulatory agencies such as the Food and Drug Administration absorption, distribution, metabolism, and excretion (ADME) capabilities
(FDA) and Environmental Protection Agency (EPA) are evaluating -omics- of an organism or tissue. In this regard, -omics technologies are similar to
generated data for risk assessment. An important benchmark in the use of more traditional in vitro and in vivo studies. However, these newer
toxicogenomic technologies is its inclusion and acceptance in regulatory technologies can provide information on activities of key proteins such as
submission packages and its use in the risk assessment process. While at transporters and metabolic enzymes in tissues on a larger scale. For
this point in time, there is no requirement for the submission of example, high-throughput approaches have been used to broadly assess
toxicogenomic data to major regulatory agencies, many agencies metabolic differences between cell types of normal, malignant, and
including the EPA and FDA encourage voluntary submission and have immortalized phenotypes (Hedberg et al., 2001; Vondracek et al., 2002;
drafted guidance documents for those submissions (FDA, 2007, 2009; Staab et al., 2008). Furthermore, metabolomic/metabonomic data may
European Medicines Agency, 2009; U. S. EPA, 2009). indicate depletion, consumption, or production of LMW molecules
The EPA's Science Policy Council Interim Policy on Genomics instructs involved in metabolism of a chemical. Such changes in some tissues, but
EPA to use genomics data on a case-by-case basis in a weight-of-evidence not others, may inform the disposition of a toxicant of interest. In addition,
approach in risk assessment (U. S. EPA, 2002). Following the release of the pre- and post-exposure differences in transcripts, proteins, and LMW
Interim Policy, the EPA's Science Policy Council (SPC) created a cross-EPA molecules in vitro have potential to be useful for toxicokinetic

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx 5

Fig. 1. Potential uses of toxicogenomic data in understanding a chemical's mechanism of action for application to human health risk assessment of environmental chemicals. The
exposure–outcome continuum for environmental chemical exposures is presented. In between exposure and outcome are all of the steps of the mechanism of action, including TK as
well as TD steps. TGx data can inform one or more steps in this continuum depending on the available TGx data information. Arrows with “TGx” indicate the potential types of
information these data can provide. For example, data from direct TGx measures of gene expression changes after chemical exposure can inform ADME (e.g., by identifying metabolic
pathways/liver enzyme effects) which could be used, in turn to inform internal dose. Direct TGx measurement data can provide information about altered molecular events within TD. TGx
data associated with either exposure or a particular outcome can be used as a biomarker of exposure or outcome. TGx data from multiple species can be used to inform interspecies
differences. Or, if TGx data are only available for one species, sequence data for multiple species could be used to make inferences about interspecies differences in the mechanism of action.
TGx data from multiple individuals can inform intraspecies differences (see Mortensen and Euling, 2010—this issue). ADME, absorption, distribution, metabolism, and excretion; TD,
toxicodynamics; TK, toxicokinetics; TGx, toxicogenomic. This figure is adapted from US EPA (2009).

extrapolations to in vivo settings for risk assessment purposes (Kedderis cytotoxicity with regenerative cell proliferation as the MOA for acetochlor
et al., 2006). Conceivably, toxicogenomic techniques could also be used to in the assessment (Genter et al., 2002). The genomic data enabled the
similarly examine in vivo differences in metabolic competency across identification of processes and pathways affected by alachlor exposure
species although, at the current level of understanding, these would not be including oxidative damage and the generation of reactive oxygen species,
substitutes for traditional cross-species toxicokinetic evaluations. as possible mechanisms underlying cytotoxicity (Genter et al., 2002).
Within humans, inter-individual differences in chemical or drug Specifically, based on genomic analysis of alachlor-induced tumors in the
disposition arise in part from genetic variability within the population. olfactory mucosa, a four-step carcinogenesis model was proposed.
Genetic polymorphisms in phase I and phase II enzymes can alter Second, a recent EPA risk assessment of DMA evaluated available
responses to xenobiotics (Ginsberg et al., 2002, 2009a, 2009b, 2009c; genomic and toxicity studies as part of their evaluation (U.S. EPA, 2006).
Neafsey et al., 2009a, 2009b; Walker et al., 2009; Ginsberg et al., 2010). The DMA data suggest that urothelial cytotoxicity and regeneration, key
Copy number polymorphisms in phase I enzymes such as that seen with events in rat bladder tumor formation, are relevant to humans. Pathway-
the metabolic enzyme, CYP2D6, can result in some individuals being level analysis revealed that DMA affects both common and unique
characterized as either “poor” or “ultrarapid” metabolizers of certain pathways in the bladder transitional cells of rats and an immortalized
xenobiotics (Bodin et al., 2005; Buckley et al., 2005) Toxicogenomic human cell line (non-availability of primary human cells influenced the
techniques can be used to assess the frequency of genetic and copy choice of an immortalized cell line (Sen et al., 2007). This UROtsa cell line
number polymorphisms within populations. When the impact of these was derived from the normal urothelium lining the ureter and was
polymorphisms on enzyme function is established, the information could immortalized (simian virus (SV40) large T antigen). The authors
be used to characterize the differences in predicted internal dose across recognized that immortalized cell lines have inherent limitations, but in
the population, and potentially be used in probabilistic risk assessment. lieu of primary cells, human immortalized cell lines can provide relevant
information. The finding of common pathways in the rat urothelium (Sen
Example uses of toxicogenomic data to inform mechanism of action in et al., 2005) and human cells provides evidence of concordant biological
EPA risk assessments processes affected by DMA, thereby supporting the conclusion of
biological plausibility in humans. However, the finding of other pathways
Toxicogenomic data have been used in several different ways for indicated that there are some species differences in response. Thus, the
informing mechanism of action. EPA has evaluated toxicogenomic data genomic data, in conjunction with toxicology studies, provided both
qualitatively in two final assessments of environmental chemicals, mechanism of action and human relevance information to the assessment.
acetochlor and dimethylarsenic acid (DMA). The EPA's Office of Pesticide In both cases, the toxicogenomic data informed the mechanism of action
Programs (OPP) Cancer Assessment Review Committee (CARC) evaluated in the weight of evidence analysis.
genomic data in the cancer assessment of acetochlor (U.S. EPA, 2004a).
OPP considered acetochlor, alachlor, and butachlor as a group with a Hypothesis generation for mechanism of action and testing studies
common mechanism of action based on the observation that exposure to
each of the three pesticides induced nasal turbinate tumors via Policy language (U. S. EPA, 2002) clearly indicates that EPA is
cytotoxicity and cell proliferation. Genomic data from studies with interested in utilizing toxicogenomic data in the risk assessment of
olfactory mucosa from rats treated with alachlor, were used to support environmental chemicals. Some available toxicogenomic studies are being

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
6 V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

evaluated and incorporated in ongoing EPA risk assessments, particularly a more scientifically defensible approach to determining whether the
by providing information to further define a chemical's mechanism of mechanism of tumorigenesis of these pesticides are unlike PB, or like
action. Conazoles, phenobarbital, and formaldehyde are examples of PB, and thus potentially relevant or not relevant to humans. Moreover,
environmental agents with toxicogenomic data that informs the these data may be informative for the assessment of human relevance.
mechanism/mode of action. A comprehensive research study was Another example of toxicogenomic studies that were used to
undertaken to examine the toxicity and transcriptional effects of inform the MOA is a transcriptional study related to nasal carcinoge-
hepatotumorigenic conazoles, a group of fungicides used in agriculture nicity that compared gene expression changes after formaldehyde vs.
and medicine, in mouse (Ward et al., 2006; Wolf et al., 2006; Nesnow et al., after glutaraldehyde exposure (Hester et al., 2005). Human exposure
2009). These papers, of gene transcriptional and in-life evaluation studies, to formaldehyde and glutaraldehyde occurs in a number of workplace
related the alterations at both the gene expression and pathway level to and environmental settings. Formaldehyde is a known carcinogen in
tumorigenesis leading to the identity of potential modes of tumorigenic the rat nose, while glutaraldehyde, though cytotoxic, is not known to
action. The mouse studies of Ward et al. (2006) and Allen et al. (2006) are induce cancer. Both compounds induce similar tissue responses,
briefly reviewed first. Ward et al. (2006) conducted a microarray study to including inflammation and cellular damage, and cannot be distin-
identify transcriptional changes provide clues for potential mechanisms of guished by commonly used toxicological endpoints. Given that the
tumorigenic action. Parallel in-life studies were conducted to measure risk to humans is uncertain, a series of genomic studies were
conventional toxicological endpoints (Allen et al., 2006). Briefly, CD-1 undertaken at the U.S. EPA's Office of Research and Development
mice received triadimefon, propiconazole, or myclobutanil in the feed for utilizing genomic profiling to discriminate between the two com-
4, 30, or 90 days. Combining apical endpoints described in Allen et al. pounds (Hester et al., 2003, 2005). The studies were conducted using
(2006) to the transcriptional profiles defined in Ward et al. (2006) F344 rats. Treatments included 1-, 5-, or 28-day exposure by nasal
allowed a richer understanding of conazole-induced toxicity. The three instillation of formaldehyde solution (400 mM) or glutaraldehyde
conazoles shared common treatment-related responses for the following: solution (20 mM). Total RNA was extracted from respiratory cells for
induction of hepatomegaly, high levels of hepatic pentoxyresorufin-O- subsequent genomic profiling. Given that both aldehydes induced
dealkylase activity, increased hepatic cell proliferation, and decreased similar acute and subchronic histopathology and given that early
serum cholesterol. GeneChip arrays were used to generate the transcrip- aldehyde-induced lesions are microscopically similar, the authors
tional measurements. As expected all tested conazoles had effects on investigated whether transcriptional patterns using cDNA technology
nuclear receptors, however, a subset of altered genes and pathways could explain the different cancer outcomes.
distinguished the three conazoles. In summary, while triadimefon, Results showed that the carcinogenic formaldehyde could be
propiconazole, and myclobutanil had similar non-genomic effects, distinguished from the noncarcinogenic glutaraldehyde based on the
genomic analyses showed differences in their gene expression profiles. different patterns of genomic responses. Glutaraldehyde's lack of
Moreover, a series of potential key events leading to conazole-induced carcinogenicity may be more related to the pattern of transcriptional
hepatocarcinogenesis were proposed from the transcriptomic and profiles showing greater toxicity from the observed lack of DNA-
toxicological data. These data have the potential to support a future repair, greater mitochondrial damage, and increased apoptosis
conazole mechanism of action risk assessment review. compared to profiles associated with formaldehyde exposure. These
In a second mouse toxicogenomic study, Nesnow et al. (2009) two environmental chemical examples illustrate how microarray and
noted that some hepatotumorigenic conazoles display many of the RT-PCR studies, by examining molecular pathway alterations associ-
same hepatic toxicologic responses as the prototypical mouse liver ated with exposure to environmental chemicals, can be used to
carcinogen phenobarbital (PB) and therefore the authors examined generate and test mode and mechanism of action hypotheses to
whether these conazoles were like or unlike PB using toxicogenomics. further understand the mechanism of action that, in turn, could be
This study was relevant because epidemiologic studies on patients utilized in risk assessments.
who received long-term PB therapy showed no increase in the
incidence of hepatic cancer, even though their PB plasma levels were New methods to analyze toxicogemomics data in risk assessment
close to those found in susceptible rodents (IARC, 2001). Chemicals
that are mouse liver tumorigens and produce PB-associated responses As mentioned in earlier sections, to fully utilize toxicogenomic
in mouse liver are considered to be “PB-like” and thus their data in understanding mechanism of action, new data analysis and
tumorigenic responses have been viewed as not relevant to humans management methods are needed. This is in line with the recom-
(Holsapple et al., 2006). Therefore, it was important to compare the mendations put forth in the NAS report Toxicity Testing in the 2st
molecular changes induced by conazoles to those change induced by Century (NRC, 2007b). The NAS report stated that in order to
PB using detailed transcriptomic analyses. Nesnow et al., 2009 applied implement its vision, regulatory agencies will need to utilize high
transcriptional analyses to hepatic tissues from mice exposed to and medium-throughput assays, many of which provide -omics data.
tumorigenic doses of PB, propiconazole (Pro) or triadimefon (Tri) The NAS report added that the development of methods to
(same genomic data generated in mouse liver from Ward et al., 2006). understand the circuitry of toxicity pathways will involve the use of
This was done to probe whether gene changes induced by the “functional genomics techniques for integrating and interpreting
tumorigenic conazoles shared similarities or differences to those various data types…It will most likely require improved methods in
generated by PB treatments. Mice were administered feed containing bioinformatics, systems biology, and computational toxicology” (page
PB, Pro or Tri for 4 and 30 days. Targeted transcriptomic analyses were 129) (NRC, 2007b). To this end, several institutions have been
conducted at many levels including genes, canonical pathways, engaged in developing such methods and tools for analyzing
toxicity pathways, and gene networks. In addition, subsets of genes microarray data and merging data from genomics, quantitative
including cell cycle genes, transcription factors, and genes reported to structure–activity relationship (QSAR), and other medium to high
be associated with human hepatocellular cancer were compared. The throughput assays. This section highlights cutting edge techniques
authors concluded that although PB and the two conazoles induced under development at the EPA-funded Science To Achieve Results
mouse liver tumors and exhibited similar toxicological responses, (STAR) Computational Toxicology Research Centers.
their transcriptional profiles were significantly different and thus The STAR New Jersey Research Center for Environmental Bioinfor-
their mechanisms of tumorigenic action were highly likely to differ. matics and Computational Toxicology (ebCTC) is a research consor-
The authors posited that the transcriptional profiles of tissues exposed tium of the University of Medicine and Dentistry — Robert Wood
to toxic chemicals inherently contain their mechanisms of toxicity. Johnson Medical School, Princeton University, Rutgers University, and
The authors suggest, therefore, that the results of this study provided the FDA's Center for Toxicoinformatics. This center is developing tools

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx 7

and methods for the analysis or prediction of molecular events and The Texas-Indiana Virtual Star Center (TIVS), the newest STAR
the evaluation of changes to genes and proteins in an organism computational toxicology center, is a collaborative effort between the
following exposure to toxicants (Ovacik et al., 2010—this issue). These University of Houston, Texas A&M, and Indiana University. This center
tools and methods include ebTrack and the environmental bioinfor- began its research to develop methods and tools for assessing
matics Knowledge Base. developmental toxicity in November 2009. Through medium and
ebTrack, an integrated bioinformatics system for environmental high throughput assays, utilizing mouse embryonic stem cells and
research and analysis, is being built upon the framework of the FDA's zebrafish, the Center plans to generate data that will be used to build
ArrayTrack™ . ebTrack will provide (1) centralized data management, predictive in silico models for developmental toxicity that are also
(2) rapid data analysis capability via an intuitive user interface, and relevant to humans. First, in silico models will be built to replicate
(3) informed data interpretation through interfaces to relevant normal developmental processes and then they will be developed to
knowledge bases of gene annotation, protein function and pathways elucidate the mechanisms by which chemical exposures perturb the
(Chen et al., 2009). By enhancing ArrayTrack™, which manages and pathways (U.S. EPA, 2009). The major challenge to be addressed by
interprets microarray data, ebTrack will be more applicable to the in silico models is how to move from events taking place on the
environmental health research data and will not be limited to the cellular level to tissue-level patterning (U. S. EPA, 2002).
management and analysis of data from DNA microarray experiments
for humans, mice, and rats. In addition to gene expression data from Discussion
DNA microarrays, it will be able to handle proteomics data,
metabonomics data, chemical structure information, and in vitro/in Current issues and research needs in utilizing toxicogenomics to define
vivo toxicological data (Chen et al., 2009). mode and mechanism of action in chemical risk assessment
The environmental bioinformatics Knowledge Base is a comprehen-
sive compendium of tools, databases, and literature under development The genome-wide coverage of genomic technologies as compared
by the Center. Currently, the Knowledge Base is undergoing alpha testing to the toxicological evaluations including histology, organ weights,
(Chen et al., 2009). This Knowledge Base will support ebTrack, and will and single gene expression measures offers great potential for
eventually be a public resource. It will provide resources on research areas advancing chemical risk assessment by providing a much richer
that are relevant to the development of methods and tools for improving picture of the biological state of the an organism at the mechanistic
linkages in the source-to-outcome paradigm, hazard identification and level across, for example, life stage, duration of exposure, and organs
quantitative risk assessment. Specifically, it will support the following and tissues. However, there are also a number of limitations to
bioinformatics areas: physiomics/histomics, cytomics, metabolomics, applying the technology to human health chemical risk assessment.
proteomics/interactomics, and genomics/transcriptomics (http://dido. Here we highlight some of the current issues and key research needs
rutgers.edu:4000/enviroinformatics). in utilizing toxicogenomic data to define a chemical's mode or
The STAR Carolina Environmental Bioinformatics Center (CEBC) mechanism of action within a risk assessment context, some of which
established to support the field of computational toxicology through are related to regulatory acceptance of these data within a changing
expertise in -omics and the “elucidation of genetic variation” (U.S. EPA, risk assessment landscape.
2009), is developing many tools and methods to assist in the The Health and Environmental Sciences Institute's (HESI) Committee
interpretation of toxicogenomics. FastMap and SAFEGUI are two on the Application of Genomics to Mechanism-based Risk Assessment
examples. FastMap is a software package for gene Expression Quantitative multi-sector scientist survey identified three areas that toxicogenomic
Trait Locus (eQTL) mapping (Gatti et al., 2009a). The increase in gene data would have the greatest potential utility to risk assessment:
expression and high-density genotype data has improved the ability of understanding biological mechanisms, biomarker candidates, and species
eQTL mapping to identify genomic loci associated with phenotypic differences (Pettit et al., 2010). Regarding understanding biological
variation in homozygous populations. However, existing tools cannot mechanism, which is the focus of this discussion, the survey identified
accurately calculate associations between tens of thousands of transcripts some key challenges, including the currently incomplete biological
and thousands to millions of single nucleotide polymorphisms (SNPs). context in which to understand toxicogenomic data and variability in
FastMap organizes SNPs into what is called a “Hamming distance-based data interpretation. For instance, a particular chemical's toxicogenomic
tree” that reduces the number of arithmetic operations needed for such data set may identify some genes lacking annotation; some genes with
calculations by taking advantage of the association between the SNP and data indicating involvement in multiple pathways; and pathways with
its parent SNP in the tree (Gatti et al., 2009a). The graphical user interface, limited information about pathway relationships to one other and to the
SAFEGUI, for the Significance Analysis of Function and Expression (SAFE) toxicological outcome. This lack of information about the complex inter-
software assesses the significance of biological categories in microarray relationships among chemicals, genes, pathways, and toxicological
studies, while accounting for the effects of expression correlations among outcome is exacerbated by two other overarching problems: 1) data
mRNA transcripts. This is accomplished through the incorporation of interpretation variability; 2) insufficient data to determine causal
permutation and bootstrap algorithms that are better able to detect relationships between toxicogenomic effects and toxicological outcomes
differential expression across categories of genes (Gatti et al., 2009b). (i.e., phenotypic anchoring). While these issues apply to the evaluation of
Although only nearing the end of its second year, the STAR Carolina any data type, they present a particular challenge for toxicogenomic data
Center for Computational Toxicology (CCCT), also located at the University because of the high density and quantity of data.
of North Carolina, is developing novel methods, including one for the With respect to data interpretation variability, because data are
analysis of metabolomics data, Progressive Consensus Alignment of NMR generated from thousands of genes, the probability increases of
Spectra—PCANS (Staab et al., 2010). Just as microarrays and other incorrectly interpreting the data for any particular gene and
technological advances are enabling the evaluation of thousands of mRNA generating either false-positive or false-negative results. Thus, many
transcripts, techniques have been developed that allow for quantitative algorithms such as multiple test corrections have been developed in
measurements of metabolites in a large number of biological samples. an attempt to control for this issue. Improved methods are greatly
However, when trying to measure large numbers of samples using NMR needed, particularly those that err conservatively to further reduce
spectroscopy, peak location can vary as a function of different sample identifying genes and pathways by chance. On the other hand, there is
conditions. PCANS aligns NMR spectra, through the progressive integra- also the potential for missing larger patterns of more modest gene
tion of many pairwise comparisons (Staab et al., 2010). This approach changes that may be filtered out by stricter statistical cut-offs on
should improve the ability to analyze metabolomic data that will likely be individual genes. Methodologies are currently being developed for
useful in evaluating the toxicity of chemicals. such “pathway-based” analyses to estimate the degree of toxicity

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
8 V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

pathway perturbations, some of which do not involve “pre-filtering” the appropriate cutoffs for differentially expressed gene identification,
that may increase false negatives (see Ovacik et al., 2010—this issue). developing appropriate pathway analyses to estimate the degree of
However, no method or set of methods has reached sufficient toxicity pathway perturbations, and the determination of biological
maturity and undergone sufficient testing for general regulatory significance of affected genes or pathway. Each of these requires
acceptance and the determination of biological significance of affected placing the data in a more biological context specifically related to
genes or pathway remains a challenge. Further, data analysis and toxicity — a context for which the current state of knowledge remains
interpretation methods considered appropriate for screening pur- sparse. Therefore, a key parallel research need is developing a base of
poses, which have been the focus of most existing toxicogenomic knowledge as to the web of biological pathways, their interrelations,
applications and research, differs significantly from those appropriate and how they interact to the give occurrence to adverse effects. Part of
for risk assessment purposes. Thus, a key research need is the this effort also necessarily involves relating pathway perturbations to
development of data analysis and interpretation methods specifically human outcome data as opposed to experimental animal model data.
geared towards testing hypotheses related to risk assessment. This can be accomplished only through enhanced collaboration
One example of a risk assessment need related to the interpreta- between toxicology and other biological sciences, particularly those
tion of toxicogenomic data is the issue of establishing causation rich in human data.
between gene expression changes and toxicological outcomes. Part of
the difficulty stems from the fact that, even with traditional data, Summary
different scientists may put the mechanism of action puzzle together
differently based on the same data set. However, with respect to Despite the challenges and limitations in utilizing toxicogenomic
toxicogenomic data, variability in scientific judgment is exacerbated data in risk assessment, these data can currently be informative to risk
by the complexity and incompleteness of the biological understanding assessment by corroborating the existing information base on mode
of how genes relate to one another, and how they relate to toxicity. In or mechanism of action. In addition, toxicogenomic data can provide
addition, most studies are not specifically designed to test causation. supportive information for the acceptance of a related critical effect,
Ideally, experiments for establishing causation would involve evalu- including providing some information about human relevance. For
ation of genomic data and data on the toxicological outcome of example, if the human relevance of a particular toxicological endpoint
interest in the same, or at least the same set of, experimental animals. considered the critical effect is debated, and toxicogenomic data
Better yet would be longitudinal data in which early (in time) related to the endpoint indicates effects on pathways known to be
genomic markers could be linked to subsequent development of a related to human disease, then the toxicogenomic data may provide
toxicological outcome in the same experimental unit. Thus, stronger support for human relevance. This is one example of how toxicoge-
experimental designs that include linkages to adverse toxicological nomic data can be used in a weight-of evidence approach in a
outcome are a key research need, as determining associations or qualitative manner to impact decisions within the risk assessment.
causal links between the alterations in gene expression and in vivo Data from well-designed microarray studies have been used in
toxicological endpoints is difficult with the currently available data. hypothesis generating and testing of a mechanism of action and have
Other concerns with the use of toxicogenomics in risk assessment been used as supporting information for a particular mode of action in
reach beyond the use of these data to establish a mechanism or mode some EPA risk assessments. Many challenges and research needs
of action. While the MAQC I project results demonstrated good remain in terms of experimental designs, data analysis, and
reproducibility when using the same biological sample and platform, interpretation appropriate for risk assessment. Nonetheless, as
other issues leading to variability in response (e.g., biological sample chemical risk assessment moves away from a single mechanism of
preparation and intrinsic heterogeneity) remain to be addressed. To action approach toward a toxicity pathway-based paradigm, we
resolve these issues, continued iterative and collaborative efforts envision that toxicogenomic data from multiple technologies (e.g.,
between research scientists of various institutions, industry that proteomics, metabolomics, transcriptomics, supportive RT-PCR stud-
develops and improves the technology, and risk assessors would be ies) can be used in conjunction with one another to understand the
beneficial. Additional concerns include the lack of available toxicoge- complexities of TK and TD steps in multiple, and possibly interacting,
nomic data for most environmental chemicals; lack of maturity of pathways affected by a single chemical or mixture in human health
some of the technologies for use as toxicology testing and risk risk assessment.
assessment assays (including the establishment of reproducibility,
sensitivity, and specificity); concern about whether there is a return Conflict of interest statement
on such a large investment; and the need for criteria specifically for
inclusion of toxicogenomic data in risk assessment. The lack of The authors declare that there are no conflicts of interest.
available toxicogenomic data is due in part to the newness and cost of
conducting toxicogenomic studies. Studies become even more costly Acknowledgments
when designing specific comparative studies assessing multiple
tissues or time points, for example. While the cost has been declining The authors would like to thank US EPA scientists, Drs. Babasaheb
and may be less of an issue in the future, this limitation has resulted in Sonawane, Sury Vulimiri, Doug Wolf, and William Ward, for their
investigations with single-dose, single-time point of exposure or helpful, constructive review and comments.
fewer samples/replicates. For example, the dibutyl phthalate (DBP)
case study example discussed in this issue (Euling et al., 2010—this References
issue) has such a limited data set. More complex study designs may
Allen, J.W., Wolf, D.C., George, M.H., Hester, S.D., Sun, G., Thai, S.F., Delker, D.A., Moore,
also be needed for MOA hypothesis testing, as described above.
T., Jones, C., Nelson, G., Roop, B.C., Leavitt, S., Winkfield, E., Ward, W.O., Nesnow, S.,
In sum, toxicogenomics is continuing to be refined with new 2006. Toxicity profiles in mice treated with hepatotumorigenic and non-
methodologies being developed for both performing toxicogenomic hepatotumorigenic triazole conazole fungicides: propiconazole, triadimefon, and
myclobutanil. Toxicol. Pathol. 34, 853–862.
studies and analyzing data, particularly for microarray studies. A
Ball, C.A., Brazma, A., Causton, H., Chervitz, S., Edgar, R., Hingamp, P., Matese, J.C.,
number of major efforts have addressed some of the technical Parkinson, H., Quackenbush, J., Ringwald, M., Sansone, S.A., Sherlock, G., Spellman,
challenges to providing reliable, reproducible data. The next step in P., Stoeckert, C., Tateno, Y., Taylor, R., White, J., Winegarden, N., 2004. Submission of
this regard is greater standardization in data analysis, particularly the microarray data to public repositories. PLoS Biol. 2, E317.
Bodin, L., Beaune, P.H., Loriot, M.A., 2005. Determination of cytochrome P450 2D6
development of analyses geared towards risk assessment rather than (CYP2D6) gene copy number by real-time quantitative PCR. J. Biomed. Biotechnol.
screening and prioritization. Examples of these include determining 2005, 248–253.

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx 9

Boverhof, D.R., Zacharewski, T.R., 2006. Toxicogenomics in risk assessment: applica- Hamadeh, H.K., Bushel, P.R., Jayadev, S., Martin, K., DiSorbo, O., Sieber, S., Bennett, L., Tennant,
tions and needs. Toxicol. Sci. 89, 352–360. R., Stoll, R., Barrett, J.C., Blanchard, K., Paules, R.S., Afshari, C.A., 2002b. Gene expression
Brazma, A., Hingamp, P., Quackenbush, J., Sherlock, G., Spellman, P., Stoeckert, C., Aach, analysis reveals chemical-specific profiles. Toxicol. Sci. 67, 219–231.
J., Ansorge, W., Ball, C.A., Causton, H.C., Gaasterland, T., Glenisson, P., Holstege, F.C., Hedberg, J.J., Grafstrom, R.C., Vondracek, M., Sarang, Z., Warngard, L., Hoog, J.O., 2001.
Kim, I.F., Markowitz, V., Matese, J.C., Parkinson, H., Robinson, A., Sarkans, U., Micro-array chip analysis of carbonyl-metabolising enzymes in normal, immortalised
Schulze-Kremer, S., Stewart, J., Taylor, R., Vilo, J., Vingron, M., 2001. Minimum and malignant human oral keratinocytes. Cell. Mol. Life Sci. 58, 1719–1726.
information about a microarray experiment (MIAME)-toward standards for Hester, S.D., Benavides, G.B., Yoon, L., Morgan, K.T., Zou, F., Barry, W., Wolf, D.C., 2003.
microarray data. Nat. Genet. 29, 365–371. Formaldehyde-induced gene expression in F344 rat nasal respiratory epithelium.
Buckley, P.G., Mantripragada, K.K., Piotrowski, A., Diaz de Stahl, T., Dumanski, J.P., 2005. Toxicology 187, 13–24.
Copy-number polymorphisms: mining the tip of an iceberg. Trends Genet. 21, Hester, S.D., Barry, W.T., Zou, F., Wolf, D.C., 2005. Transcriptomic analysis of F344 rat nasal
315–317. epithelium suggests that the lack of carcinogenic response to glutaraldehyde is due to its
Canales, R.D., Luo, Y., Willey, J.C., Austermiller, B., Barbacioru, C.C., Boysen, C., greater toxicity compared to formaldehyde. Toxicol. Pathol. 33, 415–424.
Hunkapiller, K., Jensen, R.V., Knight, C.R., Lee, K.Y., Ma, Y., Maqsodi, B., Papallo, A., Holsapple, M.P., Pitot, H.C., Cohen, S.M., Boobis, A.R., Klaunig, J.E., Pastoor, T., Dellarco, V.
Peters, E.H., Poulter, K., Ruppel, P.L., Samaha, R.R., Shi, L., Yang, W., Zhang, L., L., Dragan, Y.P., 2006. Mode of action in relevance of rodent liver tumors to human
Goodsaid, F.M., 2006. Evaluation of DNA microarray results with quantitative gene cancer risk. Toxicol. Sci. 89, 51–56.
expression platforms. Nat. Biotechnol. 24, 1115–1122. IARC, 2001. Phenobarbital and its sodium salts. IARC Monographs, Some Thyrotropic
Carlson, E.A., Silkworth, J.B., 2009. Toxicogenomics in human health risk assessment. Agents. IARC Press, Lyon, France, pp. 161–288.
Toxicol. Appl. Pharmacol. 236, 254–256 author reply 257–260. Ji, H., Davis, R.W., 2006. Data quality in genomics and microarrays. Nat. Biotechnol. 24,
Casciano, D.A., Woodcock, J., 2006. Empowering microarrays in the regulatory setting. 1112–1113.
Nat. Biotechnol. 24, 1103. Kedderis, G.L., Elmore, A.R., Crecelius, E.A., Yager, J.W., Goldsworthy, T.L., 2006. Kinetics of
Chan, V.S., Theilade, M.D., 2005. The use of toxicogenomic data in risk assessment: a arsenic methylation by freshly isolated B6C3F1 mouse hepatocytes. Chem. Biol. Interact.
regulatory perspective. Clin. Toxicol. (Phila) 43, 121–126. 161, 139–145.
Chen, M., Martin, J., Fang, H., Isukapalli, S., Georgopoulos, P.G., Welsh, W.J., Tong, W., Mortensen, H.M., Euling, S.Y., 2010. Genomic and Informatic Approaches to Characterize
2009. ebTrack: an environmental bioinformatics system built upon ArrayTrack. Human Genetic Susceptibility for Application to Risk Assessment. Toxicol. Appl.
BMC Proc. 3 (Suppl 2), S5. Pharmacol. This issue.
Chiu, W.A., Euling, S.Y., Scott, C.S., Subramaniam, R.P., 2010-this issue. Approaches to Neafsey, P., Ginsberg, G., Hattis, D., Johns, D.O., Guyton, K.Z., Sonawane, B., 2009a.
advancing quantitative human health risk assessment of environmental chemicals Genetic polymorphism in CYP2E1: population distribution of CYP2E1 activity. J.
in the post-genomic era. Toxicol. Appl. Pharmacol. Toxicol. Environ. Health B Crit. Rev. 12, 362–388.
Daston, G.P., 2008. Gene expression, dose–response, and phenotypic anchoring: Neafsey, P., Ginsberg, G., Hattis, D., Sonawane, B., 2009b. Genetic polymorphism in
applications for toxicogenomics in risk assessment. Toxicol. Sci. 105, 233–234. cytochrome P450 2D6 (CYP2D6): population distribution of CYP2D6 activity. J.
Dix, D.J., Gallagher, K., Benson, W.H., Groskinsky, B.L., McClintock, J.T., Dearfield, K.L., Toxicol. Environ. Health B Crit. Rev. 12, 334–361.
Farland, W.H., 2006. A framework for the use of genomics data at the EPA. Nat. Nesnow, S., Ward, W., Moore, T., Ren, H., Hester, S.D., 2009. Discrimination of
Biotechnol. 24, 1108–1111. tumorigenic triazole conazoles from phenobarbital by transcriptional analyses of
Ekins, S., Nikolsky, Y., Nikolskaya, T., 2005. Techniques: application of systems biology mouse liver gene expression. Toxicol. Sci. 110, 68–83.
to absorption, distribution, metabolism, excretion and toxicity. Trends Pharmacol. NRC, 2007a. Applications of toxicogenomic technologies to predictive toxicology and
Sci. 26, 202–209. risk assessment. (N. R. Council, Ed.). National Academy Press, Washington, DC.
Euling, S.Y., Thompson, C.M., Benson, R., Chiu, W.A., 2010. An approach to using NRC, 2007b. Toxicity testing in the 21st century. (N. R. Council, Ed.). National Academy
toxicogenomic data in risk assessment: dibutyl phthalate (DBP) case study. Toxicol. Press, Washington, DC.
Appl. Pharmacol. This issue. Oberemm, A., Onyon, L., Gundert-Remy, U., 2005. How can toxicogenomics inform risk
European Medicines Agency, 2009. Scientific Guidelines for Human Medicinal Products. assessment? Toxicol. Appl. Pharmacol. 207, 592–598.
eEuropean Medicines Agencyhttp://www.emea.europa.eu/htms/human/human- Ovacik, M.A., Sen, B., Euling, S.Y., Gaido, K.W., Ierapetritou, M.G., Androulakis, I.P., 2010.
guidelines/multidiscipline.htm. Pathway modeling of microarray data: a case study of pathway activity changes in the
FDA, 2005. Guidance for industry: Pharmacogenomic Data Submissions. eFood and Drug testis following in utero exposure to dibutyl phthalate (DBP). Toxicol. Appl. Pharmacol.
Administration. http://www.fda.gov/downloads/RegulatoryInformation/Guidances/ This issue.
ucm126957.pdf. Pandey, A., Mann, M., 2000. Proteomics to study genes and genomes. Nature 405, 837–846.
FDA, 2007. Guidance for Industry: Pharmacogenomic Data Submissions — Companion Patterson, T.A., Lobenhofer, E.K., Fulmer-Smentek, S.B., Collins, P.J., Chu, T.M., Bao, W.,
Guidance. eFood and Drug Administration, Rockville, MDhttp://www.fda.gov/ Fang, H., Kawasaki, E.S., Hager, J., Tikhonova, I.R., Walker, S.J., Zhang, L., Hurban, P.,
downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ de Longueville, F., Fuscoe, J.C., Tong, W., Shi, L., Wolfinger, R.D., 2006. Performance
ucm079855.pdf. comparison of one-color and two-color platforms within the MicroArray Quality
FDA, 2009. Genomic Data Submission. eFood and Drug Administrationhttp://www.fda. Control (MAQC) project. Nat. Biotechnol. 24, 1140–1150.
gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083641.htm. Pettit, S., des Etages, S.A., Mylecraine, L., Snyder, R., Fostel, J., Dunn II, R.T., Haymes, K.,
Frueh, F.W., 2006. Impact of microarray data quality on genomic data submissions to Duval, M., Stevens, J., Afshari, C., Vickers, A., 2010. Current and future applications of
the FDA. Nat. Biotechnol. 24, 1105–1107. toxicogenomics: results summary of a survey from the HESI Genomics State of
Gatti, D.M., Shabalin, A.A., Lam, T.C., Wright, F.A., Rusyn, I., Nobel, A.B., 2009a. FastMap: Science Subcommittee. Environ. Health Perspect. 118, 992–997.
fast eQTL mapping in homozygous populations. Bioinformatics 25, 482–489. Sen, B., Wang, A., Hester, S.D., Robertson, J.L., Wolf, D.C., 2005. Gene expression profiling of
Gatti, D.M., Sypa, M., Rusyn, I., Wright, F.A., Barry, W.T., 2009b. SAFEGUI: resampling- responses to dimethylarsinic acid in female F344 rat urothelium. Toxicology 215,
based tests of categorical significance in gene expression data made easy. 214–226.
Bioinformatics 25, 541–542. Sen, B., Wolf, D.C., Turpaz, Y., Bugrim, A., Retief, J., Hester, S.D., 2007. Identification of
Genter, M.B., Burman, D.M., Bolon, B., 2002. Progression of alachlor-induced olfactory interspecies concordance of mechanisms of arsenic-induced bladder cancer. Toxicol. In
mucosal tumours. Int. J. Exp. Pathol. 83, 303–308. Vitro 21, 1513–1529.
Ginsberg, G., Smolenski, S., Hattis, D., Sonawane, B., 2002. Population distribution of Shi, L., Reid, L.H., Jones, W.D., Shippy, R., Warrington, J.A., Baker, S.C., Collins, P.J., de Longueville,
aldehyde dehydrogenase-2 genetic polymorphism: implications for risk assess- F., Kawasaki, E.S., Lee, K.Y., Luo, Y., Sun, Y.A., Willey, J.C., Setterquist, R.A., Fischer, G.M.,
ment. Regul. Toxicol. Pharmacol. 36, 297–309. Tong, W., Dragan, Y.P., Dix, D.J., Frueh, F.W., Goodsaid, F.M., Herman, D., Jensen, R.V.,
Ginsberg, G., Neafsey, P., Hattis, D., Guyton, K.Z., Johns, D.O., Sonawane, B., 2009a. Johnson, C.D., Lobenhofer, E.K., Puri, R.K., Schrf, U., Thierry-Mieg, J., Wang, C., Wilson, M.,
Genetic polymorphism in paraoxonase 1 (PON1): population distribution of PON1 Wolber, P.K., Zhang, L., Amur, S., Bao, W., Barbacioru, C.C., Lucas, A.B., Bertholet, V., Boysen,
activity. J. Toxicol. Environ. Health B Crit. Rev. 12, 473–507. C., Bromley, B., Brown, D., Brunner, A., Canales, R., Cao, X.M., Cebula, T.A., Chen, J.J., Cheng,
Ginsberg, G., Smolenski, S., Hattis, D., Guyton, K.Z., Johns, D.O., Sonawane, B., 2009b. J., Chu, T.M., Chudin, E., Corson, J., Corton, J.C., Croner, L.J., Davies, C., Davison, T.S.,
Genetic polymorphism in glutathione transferases (GST): population distribution Delenstarr, G., Deng, X., Dorris, D., Eklund, A.C., Fan, X.H., Fang, H., Fulmer-Smentek, S.,
of GSTM1, T1, and P1 conjugating activity. J. Toxicol. Environ. Health B Crit. Rev. 12, Fuscoe, J.C., Gallagher, K., Ge, W., Guo, L., Guo, X., Hager, J., Haje, P.K., Han, J., Han, T.,
389–439. Harbottle, H.C., Harris, S.C., Hatchwell, E., Hauser, C.A., Hester, S., Hong, H., Hurban, P.,
Ginsberg, G., Smolensk, i.S., Neafsey, P., Hattis, D., Walker, K., Guyton, K., Johns, D., Jackson, S.A., Ji, H., Knight, C.R., Kuo, W.P., LeClerc, J.E., Levy, S., Li, Q.Z., Liu, C., Liu, Y.,
Sonawane, B., 2009c. The influence of genetic polymorphisms on population Lombardi, M.J., Ma, Y., Magnuson, S.R., Maqsodi, B., McDaniel, T., Mei, N., Myklebost, O.,
variability in six xenobiotic-metabolizing enzymes. J. Toxicol. Environ. Health B Ning, B., Novoradovskaya, N., Orr, M.S., Osborn, T.W., Papallo, A., Patterson, T.A., Perkins, R.
Crit. Rev. 12, 307–333. G., Peters, E.H., Peterson, R., et al., 2006. The MicroArray Quality Control (MAQC) project
Ginsberg, G., Guyton, K., Johns, D., Schimek, J., Angle, K., Sonawane, B., 2010. Genetic shows inter- and intraplatform reproducibility of gene expression measurements. Nat.
polymorphism in metabolism and host defense enzymes: implications for human Biotechnol. 24, 1151–1161.
health risk assessment. Crit. Rev. Toxicol. 40, 575–619. Shi, L., Campbell, G., Jones, W.D., Campagne, F., Wen, Z., Walker, S.J., Su, Z., Chu, T.M.,
Guo, L., Lobenhofer, E.K., Wang, C., Shippy, R., Harris, S.C., Zhang, L., Mei, N., Chen, T., Goodsaid, F.M., Pusztai, L., Shaughnessy Jr., J.D., Oberthuer, A., Thomas, R.S., Paules,
Herman, D., Goodsaid, F.M., Hurban, P., Phillips, K.L., Xu, J., Deng, X., Sun, Y.A., Tong, R.S., Fielden, M., Barlogie, B., Chen, W., Du, P., Fischer, M., Furlanello, C., Gallas, B.D.,
W., Dragan, Y.P., Shi, L., 2006. Rat toxicogenomic study reveals analytical Ge, X., Megherbi, D.B., Symmans, W.F., Wang, M.D., Zhang, J., Bitter, H., Brors, B.,
consistency across microarray platforms. Nat. Biotechnol. 24, 1162–1169. Bushel, P.R., Bylesjo, M., Chen, M., Cheng, J., Chou, J., Davison, T.S., Delorenzi, M.,
Hackett, J.L., Lesko, L.J., 2003. Microarray data — the USFDA, industry and academia. Nat. Deng, Y., Devanarayan, V., Dix, D.J., Dopazo, J., Dorff, K.C., Elloumi, F., Fan, J., Fan, S.,
Biotechnol. 21, 742–743. Fan, X., Fang, H., Gonzaludo, N., Hess, K.R., Hong, H., Huan, J., Irizarry, R.A., Judson, R.,
Hamadeh, H.K., Bushel, P.R., Jayadev, S., DiSorbo, O., Bennett, L., Li, L., Tennant, R., Stoll, R., Juraeva, D., Lababidi, S., Lambert, C.G., Li, L., Li, Y., Li, Z., Lin, S.M., Liu, G., Lobenhofer,
Barrett, J.C., Paules, R.S., Blanchard, K., Afshari, C.A., 2002a. Prediction of compound E.K., Luo, J., Luo, W., McCall, M.N., Nikolsky, Y., Pennello, G.A., Perkins, R.G., Philip, R.,
signature using high density gene expression profiling. Toxicol. Sci. 67, 232–240. Popovici, V., Price, N.D., Qian, F., Scherer, A., Shi, T., Shi, W., Sung, J., Thierry-Mieg, D.,

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017
10 V.S. Wilson et al. / Toxicology and Applied Pharmacology xxx (2011) xxx–xxx

Thierry-Mieg, J., Thodima, V., Trygg, J., Vishnuvajjala, L., Wang, S.J., Wu, J., Wu, Y., U.S. EPA, 2004b. Potential Implications of Genomics for Regulatory and Risk Assessment
Xie, Q., Yousef, W.A., Zhang, L., Zhang, X., Zhong, S., Zhou, Y., Zhu, S., Arasappan, D., Applications at EPA. eGenomics Tasks Force Workgroup, Science Policy Council. US
Bao, W., Lucas, A.B., Berthold, F., Brennan, R.J., Buness, A., Catalano, J.G., Chang, C., EPAhttp://www.epa.gov/osa/genomics-external-review-draft.pdf.
Chen, R., Cheng, Y., Cui, J., et al., 2010. The MicroArray Quality Control (MAQC)-II U.S. EPA, 2006. Revised Reregistration Eligibility Decision Document (RED) for MSMA; DSMA,
study of common practices for the development and validation of microarray- and CAMA and Cacodylic AcidWashington, DC http://www.epa.gov/oppsrrd1/REDs/
based predictive models. Nat. Biotechnol. 28, 827–838. organic_arsenicals_red.pdf2006.
Shippy, R., Fulmer-Smentek, S., Jensen, R.V., Jones, W.D., Wolber, P.K., Johnson, C.D., Pine, P.S., U.S. EPA, 2009. Proceedings of the Computational Toxicology Centers STAR Progress Review
Boysen, C., Guo, X., Chudin, E., Sun, Y.A., Willey, J.C., Thierry-Mieg, J., Thierry-Mieg, D., Workshop. http://www.epa.gov/ncerqa/publications/workshop/10_01_09/comptox-
Setterquist, R.A., Wilson, M., Lucas, A.B., Novoradovskaya, N., Papallo, A., Turpaz, Y., Baker, proceedings100109.pdf2009.
S.C., Warrington, J.A., Shi, L., Herman, D., 2006. Using RNA sample titrations to assess Van Aggelen, G., Ankley, G.T., Baldwin, W.S., Bearden, D.W., Benson, W.H., Chipman, J.K.,
microarray platform performance and normalization techniques. Nat. Biotechnol. 24, Collette, T.W., Craft, J.A., Denslow, N.D., Embry, M.R., Falciani, F., George, S.G.,
1123–1131. Helbing, C.C., Hoekstra, P.F., Iguchi, T., Kagami, Y., Katsiadaki, I., Kille, P., Liu, L., Lord,
Staab, C.A., Ceder, R., Roberg, K., Grafstrom, R.C., Hoog, J.O., 2008. Serum-responsive P.G., McIntyre, T., O'Neill, A., Osachoff, H., Perkins, E.J., Santos, E.M., Skirrow, R.C.,
expression of carbonyl-metabolizing enzymes in normal and transformed human Snape, J.R., Tyler, C.R., Versteeg, D., Viant, M.R., Volz, D.C., Williams, T.D., Yu, L., 2010.
buccal keratinocytes. Cell. Mol. Life Sci. 65, 3653–3663. Integrating omic technologies into aquatic ecological risk assessment and
Staab, J.M., O'Connell, T.M., Gomez, S.M., 2010. Enhancing metabolomic data analysis with environmental monitoring: hurdles, achievements, and future outlook. Environ.
Progressive Consensus Alignment of NMR Spectra (PCANS). BMC Bioinform. 11, 123. Health Perspect. 118, 1–5.
Thybaud, V., Le Fevre, A.C., Boitier, E., 2007. Application of toxicogenomics to genetic Vondracek, M., Weaver, D.A., Sarang, Z., Hedberg, J.J., Willey, J.C., Warngard, L.,
toxicology risk assessment. Environ. Mol. Mutagen. 48, 369–379. Grafstrom, R.C., 2002. Transcript profiling of enzymes involved in detoxification of
Tong, W., Lucas, A.B., Shippy, R., Fan, X., Fang, H., Hong, H., Orr, M.S., Chu, T.M., Guo, X., xenobiotics and reactive oxygen in human normal and simian virus 40 T antigen-
Collins, P.J., Sun, Y.A., Wang, S.J., Bao, W., Wolfinger, R.D., Shchegrova, S., Guo, L., immortalized oral keratinocytes. Int. J. Cancer 99, 776–782.
Warrington, J.A., Shi, L., 2006. Evaluation of external RNA controls for the Walker, K., Ginsberg, G., Hattis, D., Johns, D.O., Guyton, K.Z., Sonawane, B., 2009. Genetic
assessment of microarray performance. Nat. Biotechnol. 24, 1132–1139. polymorphism in N-Acetyltransferase (NAT): population distribution of NAT1 and
U. S. EPA, 2002. Interim Policy on Genomics. Prepared by the Science Policy Council. NAT2 activity. J. Toxicol. Environ. Health B Crit. Rev. 12, 440–472.
eEnvironmental Protection Agencyhttp://www.epa.gov/osa/spc/genomics.htm. Ward, W.O., Delker, D.A., Hester, S.D., Thai, S.F., Wolf, D.C., Allen, J.W., Nesnow, S., 2006.
U. S. EPA, 2007. Interim guidance for microarray-based assays: data submission, quality, Transcriptional profiles in liver from mice treated with hepatotumorigenic and
analysis, management and training considerations. External review draft. Tasks Force nonhepatotumorigenic triazole conazole fungicides: propiconazole, triadimefon,
Workgroup, Science Policy Council, Environmental Protection Agency, Washington, DC. and myclobutanil. Toxicol. Pathol. 34, 863–878.
http://www.epa.gov/osa/spc/genomicsguidance.htm. Waters, M.D., Fostel, J.M., 2004. Toxicogenomics and systems toxicology: aims and
U. S. EPA, 2009. The U.S. Environmental Protection Agency's Strategic Plan for Evaluating the prospects. Nat. Rev. Genet. 5, 936–948.
Toxicity of Chemicals. eEnvironmental Protection Agency, Washington, DC. http://www. Wolf, D.C., Allen, J.W., George, M.H., Hester, S.D., Sun, G., Moore, T., Thai, S.F., Delker, D.,
epa.gov/spc/toxicitytesting/index.htm. Winkfield, E., Leavitt, S., Nelson, G., Roop, B.C., Jones, C., Thibodeaux, J., Nesnow, S.,
U.S. EPA, 2004a. Acetochlor Report of the Cancer Assessment Review Committee 2006. Toxicity profiles in rats treated with tumorigenic and nontumorigenic
(CARC). Fourth Evaluation. Office of Pesticide Programs, Environmental Protection triazole conazole fungicides: propiconazole, triadimefon, and myclobutanil.
Agency, Washington, DC. Toxicol. Pathol. 34, 895–902.

Please cite this article as: Wilson, V.S., et al., Utilizing toxicogenomic data to understand chemical mechanism of action in risk assessment,
Toxicol. Appl. Pharmacol. (2011), doi:10.1016/j.taap.2011.01.017

You might also like