Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Oct 23.
Published in final edited form as: Environ Mol Mutagen. 2014 Dec 6;56(3):277–285. doi: 10.1002/em.21923

Opportunities to Integrate New Approaches in Genetic Toxicology: An ILSI-HESI Workshop Report

Errol Zeiger 1,*, Bhaskar Gollapudi 2, Marilyn J Aardema 3, Scott Auerbach 4, Darrell Boverhof 2, Laura Custer 5, Peter Dedon 6, Masamitsu Honma 7, Seiichi Ishida 8, Andrea L Kasinski 9, James H Kim 10, Mugimane G Manjanatha 11, Jennifer Marlowe 12, Stefan Pfuhler 13, Igor Pogribny 11, William Slikker 11, Leon F Stankowski Jr 14, Jennifer Y Tanir 10, Raymond Tice 4, Jan van Benthem 15, Paul White 16, Kristine L Witt 4, Véronique Thybaud 17
PMCID: PMC4615680  NIHMSID: NIHMS724698  PMID: 25482136

Abstract

Genetic toxicity tests currently used to identify and characterize potential human mutagens and carcinogens rely on measurements of primary DNA damage, gene mutation, and chromosome damage in vitro and in rodents. The International Life Sciences Institute Health and Environmental Sciences Institute (ILSI-HESI) Committee on the Relevance and Follow-up of Positive Results in In Vitro Genetic Toxicity Testing held an April 2012 Workshop in Washington, DC, to consider the impact of new understanding of biology and new technologies on the identification and characterization of genotoxic substances, and to identify new approaches to inform more accurate human risk assessment for genetic and carcinogenic effects. Workshop organizers and speakers were from industry, academe, and government. The Workshop focused on biological effects and technologies that would potentially yield the most useful information for evaluating human risk of genetic damage. Also addressed was the impact that improved understanding of biology and availability of new techniques might have on genetic toxicology practices. Workshop topics included (1) alternative experimental models to improve genetic toxicity testing, (2) Biomarkers of epigenetic changes and their applicability to genetic toxicology, and (3) new technologies and approaches. The ability of these new tests and technologies to be developed into tests to identify and characterize genotoxic agents; to serve as a bridge between in vitro and in vivo rodent, or preferably human, data; or to be used to provide dose response information for quantitative risk assessment was also addressed. A summary of the workshop and links to the scientific presentations are provided.

Keywords: genetic toxicity, mutation, epigenetics, genomics

INTRODUCTION

Genetic toxicology testing of chemicals for regulatory agency approval relies on in vitro and in vivo tests that measure primary DNA damage, gene mutation, and chromosome damage for identification of potential human mutagens and carcinogens. The majority of these tests have been in use for more than 30 years. During that time, our understanding of molecular biology has increased exponentially, particularly in areas such as epigenetics, noncoding RNA, genetic structure, and mechanisms responsible for the maintenance of DNA integrity. In addition, technological advances have been made within and outside the field of genetic toxicology (e.g., 3-D tissue and organ cultures; high throughput methods; flow cytometric analyses; gene expression analysis; imaging) that could permit the measurement of multiple parameters for different effects on the genome that lead to mutation or cancer. It is recognized that many of these new procedures could be of use in developing or interpreting genetic toxicity test results, enabling a better understanding of the mechanism of action of chemicals, and improving extrapolation to potential effects in humans.

Despite these advances, the regulatory testing battery has remained relatively unchanged. Most countries require new chemicals, including drugs, pesticides, industrial chemicals, and food additives, to be tested in (1) an in vitro test for gene mutation in bacteria, (2) an in vitro cytogenetic or micronucleus assay with mammalian cells or an in vitro mammalian cell gene mutation assay, and in some cases, (3) an in vivo test for chromosomal damage using rodent hematopoietic cells. The measurement of genetic damage is frequently assessed as one of the key events in the progression of cancer, although toxicologists recognize that a wide spectrum of additional human health effects, including neurological disorders, birth defects, and mitochondrial diseases, may also result. These tests are generally not used in a quantitative manner (e.g., to provide information on relative potency) but only to provide binary, yes/no, information on the genetic hazard of the test substance. It is clear that for genetic toxicity testing to progress beyond this yes/no level, new test procedures or techniques will be needed that could bridge the gaps between in vitro data, in vivo rodent data, and human data for hazard characterization and quantitative risk assessment.

The International Life Sciences Institute/Health and Environmental Sciences Institute (ILSI-HESI) Project Committee on the Relevance and Follow-up of Positive Results in In Vitro Genetic Toxicity (IVGT) Testing1 was formed in 2008 to assess the current state of genetic toxicity testing, address issues related to the performance of the assays in use at that time, and evaluate new testing techniques and technologies that hold promise for improving on the predictive performance of the tests. Additionally, efforts are underway to develop new approaches for quantitative analysis of the test data [Gollapudi et al., 2013; Johnson et al., 2014] and improve hazard and risk assessment for humans.

An initial workshop was convened in May, 2008 to review and assess methods that could be used in place of, or in addition to, this test battery for identification and characterization of genetically active substances [Lynch et al., 2011]. In that workshop, a number of new and emerging in vitro and in vivo test methods were evaluated for their potential utility for incorporation into existing testing schemes.

The subsequent April 2012 Workshop2, reported herein, was developed to consider the impact that our improved understanding of biology and new technologies might have on the identification and characterization of genotoxic substances, to define ways of bridging genetic toxicology to other disciplines, and to identify potential synergies that would result in new approaches to inform more accurate human risk assessment for genetic and carcinogenic effects. The workshop organizers identified tests and test endpoints that are being investigated by a number of laboratories and which may have promise for improving the identification of potential genetic (and epigenetic) effects, and which may be used in the future (and in most cases, upon further development) to replace or supplement the test procedures currently in use. To accomplish these goals, the Workshop brought together expertise from both within and outside the discipline of genetic toxicology, and experts in risk assessment from regulatory agencies were invited to participate and offer perspectives on how the new procedures and technologies, including some still in the research phase, might be applied to address mutagenic modes-of-action in carcinogenesis and to inform quantitative assessment of mutagenic risk.

The genetic and epigenetic effects and the technologies that were selected as topics for the Workshop were those that the Organizing Committee believed would potentially yield the most useful information relevant to human hazard and risk assessment for genetic damage. The participants were also asked to consider the impact that the improved understanding of biology and the availability the new techniques might have on genetic toxicology practices. Among the general issues discussed were the ability of these new tests/technologies to be developed into tests to identify and characterize genotoxic agents; whether the test could provide information on in vivo genetic damage or serve as a bridge between in vitro and in vivo rodent, or preferably human, data, or be used to provide dose response information for quantitative risk assessment. To help better put the new technologies into perspective, the presenters were each asked to prepare a SWOT (Strengths; Weaknesses; Opportunities; Threats) analysis as part of their presentations, which would then be discussed in detail by the Workshop participants (see Tables (IIII)). To maximize the interaction among the Workshop participants, time for discussion was allotted after each presentation, and extended periods of discussion were provided at the end of each session.

TABLE I.

Session 1 SWOT Analyses: Alternative Experimental Models to Improve Genetic Toxicity Testing

Test/system Strengths Weaknesses Opportunities Threats
Three-
Dimensional
Tissue
Constructs
Closer (than 2-D) to “in
vivo” conditions.
Technology is well-
developed and 3-D models
are commercially available.
Cells are of human origin
and function more organ-
like in terms of cell viability,
proliferation, differentiation,
morphology,
gene and protein expression.
Large interest in assay, used
by increasing number of
laboratories in various
locations.
(Pre)validation exercise
ongoing for 3-D skin.
More difficult to handle than
2-D.
High throughput possible
only for low-complexity
models.
Various levels of supporting
data available, minimal to
moderate.
Used in limited number of
labortories.
Some models in early stage
of development.
Validation is needed for
most models.
Cost.
3-D constructs have the
potential to serve as
follow-up assays based on
results in the traditional 2-
D tests, especially when
2-D models cannot be
used.
Collaborative efforts recommended
for relevant tissues
for which such
efforts are not currently
ongoing (liver, lung).
May be useful to bridge gap
between in vitro and in
vivo for quantitative risk
assessment.
Validation of methods is
very resource intense.
Limited availability of tissue
constructs.
Potential issues with patents.
Stem cell-derived
hepatocytes
Theoretically unlimited supply
of human hepatocytes.
Use of hepatocytes with different genetic back-
grounds.
Use of hepatocytes from
patients suffering specific
diseases.
Simultaneous analysis of
multiple drug metabolism
cascade in one cell.
Complexity of differentiation
process of stem cells into
hepatocytes.
Low reproducibility of differentiation process.
Interlaboratory reproducibility
needs to be defined.
Early stage of development.
Cost.
Replacement of human primary
hepatocytes.
Replacement/reduction of
animal toxicity testing.
Improvement of hepatotoxicity
prediction.
Evaluation of hepatotoxicity,
prediction of metabolites,
of induction of drug
metabolism-related
enzymes, and of drug-
drug interaction.
Supply of metabolites for
other testing.
Complexity of differentiation
process.
Requirements of higher maturation
of hepatocyte
activities.
Establishment of standards
for hepatocyte qualification.
Hepatocyte progenitor cell
line: HepaRG.
Ethical and regulatory issues
if from human origin.
Humanized animal
models
Allows for better characterization
of human hazard
and risk potential.
Relevant to humans.
Provides data on mode/
mechanism of action.
Allows comparison of rodent
vs. human metabolism and
toxicity pathways.
Involves animal use.
Expensive to create and
maintain.
Low-throughput.
Models human gene product
in mouse environment.
Refinement of hazard and
risk assessments:
decreased uncertainty
increased human relevance
New technologies are
decreasing the cost and
expanding the model species.
Can be used to further define
“toxicity pathways”
thereby facilitating development
of in vitro assays
based on MoA.
Can be used to validate in
vitro hypotheses.
Models not widely available.
Models using different
technologies may generate
different results.
Lack of acceptance of this
technology for advancing
human health risk
assessments.

TABLE III.

Session 3 SWOT Analyses: New Technologies and Approaches

Test/system Strengths Weaknesses Opportunities Threats
Imaging for
safety
assessment
Longitudinal study design
and minimal invasiveness.
Acceptance of imaging by
the medical community
enhances its utility as a
translatable pre-clinical
biomarker.
Can be highly quantitative.
Minimally to noninvasive.
The test animal or human
subject can serve as its
own control.
No standardized approach.
Deficits in quantization.
A powerful technology looking
for an application for
genetic toxicology and
carcinogenesis.
Broad applications due to
recent advances in resolution
and standardization of
approaches.
Identification of early toxic
or precarcinogenic
changes in situ.
Expensive technology.
Not universally available.
Tox21 strategy
for detecting
genotoxicants
Capability to test thousands
of chemicals in human, or
other species, cells.
Very small amounts of
compound needed for screening.
Rapid generation of test
data.
Eliminates or greatly reduces
use of animals.
Large number of cell lines
can be used.
Provides MOA information.
High throughput screens
currently lack means to provide
bioactivation.
Limited number of suitable
genotoxicity assays currently
available.
Tested compounds limited to
those that are DMSO-
soluble, nonvolatile, stable
in solution for a period of
time, etc.
No or limited, evaluation of
metabolites.
Expensive.
Data processing complexity
heavily software
dependent.
Ability to develop
approaches to assess differential
susceptibility on
a wide-spread scale.
Ability to identify susceptible
subpopulations using
genomic assays.
Ability to develop
comprehensive profiles of
chemical activity.
Ability to develop SAR and
chemical clustering models
based on chemical
activity.
Potential to reduce the number
of animals needed for
toxicity testing.
Challenges in anchoring
chemical profiles for
genotoxicity-associated
activity generated with
these technologies to
results from traditional
tests for genotoxicity with
sufficient accuracy to be
acceptable to regulatory
agencies.
Biological relevance and
reliability of the selected
assays for risk assessment.
Reproducibility across cell
lines.
Genomic
Signatures of
Genotoxicity
Query large swaths of biological
space in one assay.
Provides pathway/mechanistic
information.
In vivo genomics dependent
on animals.
Ability to interpret complex
data.
Variability (time-dependent,
platforms, dose).
Lack of mechanistic anchoring
(need more correlative
data).
High dimensionality allows
for querying of large
amount of biological
space including genotoxicity
and beyond (i.e., one
assay to query all).
Potential to rapidly identify
a no-effect dose.
Potential to reduce the numbers
of animals needed to
characterize toxicities.
Metrics (i.e., genes and path-
ways) are different from
traditional endpoints.
If it is not possible to
change metrics (i.e.
pathology to pathways), it
will significantly hinder
development in the regulatory
arena.
Reproducibility of results
across testing platforms
and between laboratories.

There were 108 Workshop participants from regulatory agencies, government research laboratories, industry, and academe, including presenters. A condensed summary of the workshop along with links to key scientific presentations is presented in this report. In addition, this report also documents the attempts of the genetic toxicology community to advance the field by embracing/integrating the advances in various allied scientific disciplines over the past 30 years.

WORKSHOP PRESENTATIONS

Overview (V. Thybaud, Sanofi; B. Gollapudi, The Dow Chemical Co. [Cochairs])

The introductory comments to the Workshop participants provided an overview of the history of the HESI initiative, the structure of the HESI committee, including its various subcommittees and leaders, and the organizing committee, as well as the objectives of the Workshop, are available at: http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/2-IVGT-NAWkshpThybaud.pdf.

Session 1: Alternative Experimental Models to Improve Genetic Toxicity Testing (M. J. Aardema, BioReliance Corp.; S. Pfuhler, The Procter and Gamble Co.)

This session was designed to present and discuss recently developed biological models and test systems. Issues addressed included the adaptation of these models as tools for genetic toxicity testing and the best use of these models, whether as supplements or potential replacements to the genetic toxicity test systems currently in use. The relevance of these new models to animal and human exposure scenarios, metabolic capacity, etc., and their ability to inform and improve hazard identification and risk assessment decisions was evaluated.

Stefan Pfuhler (The Procter & Gamble Co.) described and discussed the use of 3-dimensional (3-D) tissue constructs for genotoxicity testing. The in vitro genotoxicity tests currently used are based on two-dimensional mammalian cell cultures. 3-D tissue constructs are logical follow-up tools for genotoxicity testing because they allow for more natural cell-cell and cell-matrix interactions, and show “in vivo-like” behavior for key parameters such as cell viability differentiation, morphology, gene and protein expression, and function. These constructs are available at various levels of complexity from simple (e.g., epidermal skin models with only one cell type) to highly complex (e.g., vascularized human liver model). 3-D skin models have been successfully established for testing for genotoxic properties of dermally applied compounds using micronucleus and Comet assays. Several liver culture systems were also presented and their potential use in genotoxicity testing was discussed. Details of this presentation can be viewed at http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/4%Pfuhler.pdf).

Seiichi Ishida (National Institute of Health Sciences) discussed the development of in vitro toxicity tests using hepatocytes differentiated from human induced pluripotent stem cells (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/5-ISHIDAshorten.pdf). Pluripotent stem cells, including those from humans, have the potential to differentiate into tissue-specific primary cells, which could be used for genetic toxicity studies. Several kinds of stem cells are considered suitable. However, their use had been limited because of their availability and accessibility, and because of ethical issues regarding their source. The situation has changed since the establishment of the human iPS (induced pluripotent stem) cell line. Differentiation of iPS cells into hepatocytes could make possible an unlimited and uniform supply of a variety of cells with different genetic backgrounds.

Darrell Boverhof (The Dow Chemical Co.) described the construction and use of humanized animal models (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/6-Humanized_Models-DRB-FINAL.pdf). Humanized models can be defined as animal models that carry functioning human genes, cells, tissues, or organs, and include transgenic animals that express human genes or immunodeficient animals that have been engrafted with human cells or tissues. The models have been applied in research areas such as hematopoiesis, innate and adaptive immunity, autoimmunity, infectious disease, cancer biology, regenerative medicine, pharmacology, and toxicology. Two applications are humanized metabolizing enzyme models and nuclear receptor models. These, and other, models have provided valuable insights into human-specific responses and modes of action that can be used to decrease uncertainties in human hazard and risk assessments. Although these models have traditionally been limited to the mouse, new technologies are facilitating the development of models in other species.

Session 2: Biomarkers of Epigenetic Changes and Their Applicability to Genetic Toxicology (B. Gollapudi, The Dow Chemical Co.; V. Thybaud, Sanofi)

It is generally recognized that epigenetic changes to the DNA, while not causing a change in the nucleotide composition or sequence, can lead to heritable phenotypic changes, including heritable changes in the newborn. This session was organized to discuss the potential interplay between chemically induced alterations to the genome (genetic toxicity) and changes to the epigenome leading to or exacerbating heritable effects. This session considered screening systems that can be designed to identify “epimutations” and to evaluate the potential for their transgenerational inheritance, and whether new, less invasive and hopefully transspecies biomarkers of exposure, susceptibility, and effect can be identified in order to contribute to the risk assessment process.

Peter Dedon (Massachusetts Institute of Technology) described studies on the dynamic reprogramming of tRNA modifications and their effects on the stress response (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/8-DedonHESIGentoxWorkshop.pdf). Complex interactions between tRNA, mRNA, and ribosomes control the rate and fidelity of translation. Contributing to this are the large numbers of genes encoding tRNAs, rRNA and proteins, in addition to different ribonucleoside modifications. The modified ribonucleosides in tRNA behave as a system, and are reprogrammed in response to different types of cell stress. When cells are exposed to toxicants there are dynamic shifts in the population of RNA modifications as part a step-wise mechanism of cell responses to the chemical stresses and damage, with signature changes for each agent and for different doses of each agent. These responses involve reprogramming of tRNA modifications that leads to reprogramming of ribosome structure in the translational control of cellular stress responses.

Jennifer Marlowe (Novartis) described studies of the potential use of epigenetics in toxicology (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/9-IVGT%20meeting_24Apr2012_JMarlowe_Final.pdf). Some of the earliest events preceding the development of overt pathologies, including those arising from exposure to environmental and pharmaceutical agents, involve perturbations of the epigenome. The application of epigenomic profiling technologies to drug safety sciences has potential for providing novel insights into the molecular basis of long-lasting cellular perturbations. Genome-wide epigenomic and transcriptomic profiling, combined with molecular histopathology, are valuable components towards investigation of temporal sequences of events, and therefore mechanistic understanding of the toxicity response. Integrated molecular profiling that can be performed includes analyses of mRNA, microRNA, DNA methylation, and proteins. The mapping of epigenomes from humans and other species will enhance our ability to interpret the biological significance of xenobiotic-induced epigenetic perturbations, and may provide novel biomarkers for susceptibility to adverse events.

Igor Pogribny (US Food and Drug Administration) addressed epigenetic traits as biomarkers of carcinogenesis (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/10-Pogribny_ILSI_2012.pdf). Numerous epigenetic responses that may be used as potential biomarkers for the molecular diagnosis of cancer and prognosis for survival or treatment outcomes have been identified. Epigenetic alterations may be used as early indicators in the evaluation of the carcinogenic potential of both genotoxic and nongenotoxic substances. Incorporation of epigenetic biomarkers into cancer risk assessment holds a number of advantages over traditionally used methods, such as evaluation of DNA damage, DNA adduct formation, or bacterial mutagenicity. These advantages include their earlier appearance, stability, target tissue-specificity, and applicability to both genotoxic and nongenotoxic agents. In addition, a greater number of detectable epigenetic changes than detectable genetic alterations are present after exposure to the insult.

Andrea L. Kasinski (Yale University) described studies on the use of microRNAs (mRNAs) for treatment of lung cancers (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/11–042412Kasinski.pdf). Because they are endogenous substances, toxicities associated with mRNA use are expected to be minimal. Different delivery agents for these regulatory molecules are being investigated, and studies are ongoing to better understand their therapeutic potential, notably in the context of cancer. Targeted therapies have shown encouraging results for subsets of lung cancer patients. Likewise, gene therapies using retrovirus and replication-impaired adenoviruses to reintroduce the tumor suppressive protein p53 have had compelling results, including tumor regression and stabilization.

Session 3: New Technologies and Approaches (M. Manjanatha, US Food and Drug Administration; K. L. Witt, National Institute of Environmental Health Sciences)

This session was organized to present technology-driven approaches to assessing genetic toxicity, including the potential mode-of-action leading to the effect. There are a number of new initiatives and advances in the field of toxicology, ranging from high resolution, high content imaging assays to high throughput approaches that allow the profiling of thousands of chemicals in multiple, diverse assays. It is important from both scientific and strategic points of view that genetic toxicologists understand the implications and impact of these new technologies and embrace those approaches that hold the greatest promise for improving genetic toxicity hazard and risk assessment.

William Slikker (US Food and Drug Administration) addressed new imaging techniques that could be used in safety assessment of chemicals (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/14-SlikkerImagingGenetoxIVGTconference041512.pdf). Although the use of imaging in the clinical setting is well established, the use of imaging for preclinical assessments is infrequent. Positron Emission Tomography (MicroPET) can be used to assess any target organ and deliver quantitative information in a minimally invasive manner and in parallel with other endpoint requirements. One advantage to this approach is that the animal can be used as its own control. A number of dose-response studies were performed of CNS responses to anesthetic drugs. The data from these studies support the concept that imaging is a viable approach for assessing nervous system status by serving as a minimally invasive and translatable bio-marker. Similar imaging techniques may find use in the identification of precarcinogenic changes in organs of animals treated with carcinogenic chemicals or subject to cancer-inducing stress conditions.

Raymond Tice (National Institute of Environmental Health Sciences) described the Tox21 interagency (NIEHS, NHGRI, EPA, and FDA) studies on the responses of ~100 quantitative high throughput screens (qHTS), predominantly reporter gene assays using a 1536-well format on an automated platform (Phase I) (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/15-Tice-ILSI-HESI_GeneTox.pdf). Among the qHTS assays were several that related to the detection of activities associated with some aspect of genotoxicity. Phase II of the program screened a 10,000 compound library against a set of nuclear receptor and stress response pathway assays, to detect potentially genotoxic compounds. Confirmation of genotoxicity of a small set of selected compounds will be performed using standard in vivo tests. The lack of xenobiotic metabolism in the qHTS assays is a major limitation, so protocols that use metabolically competent cells are being developed to address this limitation. Data from these assays will be compared against existing in vitro genotoxicity data. All the data generated through Tox21 are available through a variety of publicly accessible databases.

Scott Auerbach (National Institute of Environmental Health Sciences) discussed the effect of dose level and exposure duration on genomic signatures in response to chemical exposure (http://www.hesiglobal.org/files/public/Committee%20Presentations/IVGT/16-Auerbach-IVGT-2012-3.pdf). One potential approach to the problem of setting protective exposure limits for toxic responses is the use of in vivo toxicity testing coupled with toxicogenomic dose-response analysis. Typically, the most important piece of information acquired from these studies is the benchmark dose (BMD). A systematic assessment of no-observed-effect levels indicates that they change minimally going from subacute to chronic exposures. The application of genomics allows for a broad-based query of biological perturbations that is in line with the goals of safety testing, as it is an agnostic query of nearly all the possible changes at the level of the transcriptome. Results from studies of specific agents suggest that, depending upon the mode of action, genomic signatures can identify less sensitive, in the case of genotoxic agents, or more sensitive, in the case of nongenotoxic agents, BMD values when compared with the values from tumor response data.

DISCUSSION

The disciplines of genetic toxicology and genetic toxicity testing were originally designed and developed for hazard identification. This was based on the presumption that identification of genotoxic substances would allow society to avoid or minimize exposure to agents that induce mutations in somatic and germ cells, and thereby contribute to the efforts to prevent cancer formation and other adverse effects in exposed and future generations. These uses of genetic toxicity data have not changed significantly for more than 30 years. The identification of substances posing a genetic hazard would also contribute to the risk assessment and risk management decisions.

For the field of genetic toxicity testing to move beyond the traditional test endpoints and binning of test responses into “positive” and “negative” it will be necessary to either improve the existing assays or apply new approaches used in other biology domains, and to “think outside of the box.” These new approaches would employ new experimental models and technologies and incorporate our growing understanding of the relevant biological mechanisms responsible for the development of a mutated cell and its contribution to adverse health effects (e.g., cancer).

This Workshop addressed how the new scientific discoveries and methodologies could allow for more effective and relevant screening of chemicals and other environmental substances for mutagenic risk and resulting health issues. There are a large number of test methods, testing approaches, and technologies that can potentially address these issues. However, because of time and other constraints, only a few could be addressed at the Workshop, and the lack of inclusion of other biological end-points and techniques was not meant to imply that they would be less useful. Included among the methods discussed in the Workshop were those that measured the traditional genetic endpoints (e.g., micronuclei, DNA and chromosome breaks, and gene mutation), but more efficiently or in cell, tissue, or animal constructs that are deemed to be more relevant to human exposure than the traditional test systems. The integration of new, nontraditional methods concentrated on the relevance and use of epigenetic changes as biomarkers of heritable genetic damage or cancer without the underlying change in DNA sequence. In addition, new detection techniques amenable to high-throughput screening, and noninvasive in vivo approaches, were presented. It was recognized that not all of the models addressed at the workshop appeared to be directly applicable to genetic toxicology or cancer initiation. However, the structure of the workshop and the presentations served as much to introduce the researchers and developers to the issues and needs of genetic toxicology, as they did to introduce the genetic toxicologists to the newly emerging models.

Although there were a number of discussions and suggestions of how the various test procedures might be used as adjuncts or alternatives to the current test guidelines, it would have been premature to make such recommendations, given the experimental nature or lack of validation of some of these procedures. As a result, no specific recommendations were made with regard to the future use or role of these procedures or whether they should, or could, be integrated into the existing testing schemes or replace currently used methods. It was clear that, in the future, genetic toxicology scientists will have to use more complex, often patented, commercially available models and technologies.

The Workshop participants identified issues and knowledge gaps that will require more work and resources, and that would best profit from collaborations between genetic toxicologists and the new technology researchers. The conclusions of the presenters and the discussions during and at the end of the Workshop are presented in the SWOT analyses (see Tables (IIII)) developed for the individual test approaches and assays and which were agreed to be a useful approach for evaluating the potential suitability of a process or test procedure.

Remaining issues and knowledge gaps identified by the Workshop included:

  • The relevance of surrogate models, and the ability of these models to mimic human cells and whole body complexity, with respect to DNA repair, metabolic activation, and genetic stability;

  • The relevance of the various identified end-points for human risk assessment and the ability to extrapolate the results from these models to humans based on protective mechanisms and genetic polymorphisms, including the identification of appropriate safety and uncertainty factors;

  • The need to consider all key events, including epigenetic factors, that might contribute to DNA damage or modification and affect the organism’s response to these DNA effects and DNA integrity;

  • Whether and how multiparametric and more integrated test methods can help improve our understanding of genotoxic modes of action.

The presentations and discussions at this Workshop have helped to stimulate ongoing efforts to develop and refine additional methods of identifying genotoxicants and will provide valuable information for the future paradigm of genetic toxicology testing. An ongoing activity of the ILSI/HESI IVGT (now named the Genetic Toxicology Technical Committee) is to start from a “clean sheet” to determine what a new and improved testing strategy for the identification and evaluation of genotoxicants might look like, based on both the current practices, and on new and developing approaches. This new testing strategy would be developed to cover all aspects of genomic damage, incorporate the advances in systems biology, be relevant to human risk assessment and resource efficient, and move away from a standard battery approach to a more flexible testing strategy.

TABLE II.

Session 2 SWOT Analyses: Biomarkers of Epigenetic Changes and their Applicability to Genetic Toxicology

Test/System Strengths Weaknesses Opportunities Threats
Epigenetic control
of cell
phenotype
Assay is predictive of chemical
exposures, including
genotoxins.
Analysis of RNA modification
spectra provides
insights into mechanisms
of toxicity.
Possible complementation to
transcriptional and proteomic
data for identifying
genotoxic and nongenotoxic
carcinogens.
Relatively straightforward
method for practitioners
of analytical chemistry.
Analysis of RNA modification
spectra in very early
stages of development.
Very limited data for toxicants.
Relatively specialized
method at this point.
Probably not entirely
predictive of carcinogenicity of
a chemical or drug candi-
date.
Interlaboratory validation
will be needed.
Possible coordination of RNA
modification analysis with
other ‘omic data sets.
Application to in vitro screening
for hazard assessment.
Provides insights into mechanisms
of action of toxicants.
Possible biomarkers that are
specific for non-genotoxic
carcinogens.
Allow distinction between
genotoxic and nongenotoxic
carcinogens.
Analysis of RNA modification
reprogramming is in
the earliest stages of scientific
acceptance and
application.
Translation to a high-
throughput method.
Relating RNA modification
patterns to specific
toxicities.
Epigenetics Provides comprehensive
view of modifications or
changes that may precede
events that lead to phenotypic
change or toxicity.
Rich biomarker resource.
Potential for assessment of
species-specific responses
in vivo and in vitro.
Large and complex datasets;
need for customized
bioinformatic tools.
Huge number of changes
observed many possible.
mechanisms/outcomes
High probability for
nonspecific or irrelevant
responses.
Distinguishing stress
responses from chemical-
specific responses.
Biomarkers for use in acute and
subchronic toxicity studies.
Early prediction of genotoxic
risk; inform or replace 2-year
bioassay.
Drug-induced vs. non-drug-
induced tumors.
Provide or support carcinogen
mode-of-action determinations.
May allow identification of pre-
neoplastic changes.
Tremendous complexity with
possibility of compound-
specific effects rather than
class-effects amenable to
generalized biomarkers
generation.
Translation of findings from
preclinical models/results
to humans.
Epigenetic traits as
biomarkers of
carcinogenesis
Early appearance of epigenetic
carcinogen-alterations.
Target tissue specificity.
Specificity for genotoxic and
nongenotoxic carcinogens.
Cellular epigenome individual
and dynamic variability not
completely
characterized.
Models for safety assessment
not yet developed.
Endpoints and techniques for
use in safety assessment
not established.
Interlaboratory reproducibility
not determined.
Early prediction and identification
of hazardous compounds
before their dissemination
into environment.
Provide or support carcinogen
mode-of-action
determinations.
Complexity of cellular epi-
genomic landscape and
epigenetic regulatory
mechanisms.
Correlating epigenomic
changes between test animal
species and humans.
MicroRNA analysis with
lentivirus platform
This analysis provides data
to progress towards more
clinically relevant therapeutic
applications.
The lentivirus system used is
commercially available
and serves as a proof-of-
concept to move forward.
Lentivirus platform explored
may not be the best plat-
form for human patient
delivery.
Provides background for
developing and evaluating better
and more clinically relevant
miRNA delivery mechanisms.
Lack of approval for use of
lentivirus in humans for
ethical reasons.
Advancement of better,
clinically relevant delivery
agents needed.
Multiple companies on board
with the development
phase.
Patent protection and
confidentiality agreements limit
access.

ACKNOWLEDGMENTS

The organizing committee is grateful to the researchers who agreed to participate in this Workshop. In addition, the support of the ILSI/HESI staff, particularly Eric Moore, was invaluable for the organization, logistics, and day-to-day running of the Workshop. The authors also thank the ILSI/HESI IVGT committee for its financial and intellectual support.

Footnotes

1

Steering committee for this Workshop: M. J. Aardema, L. Custer, B. Gollapudi, M. Honma, M. Manjanatha, S. Pfuhler, L. Stankowski, Jr., J. van Benthem, P. White, K.L. Witt, V. Thybaud, E. Zeiger.

2

Genetic Toxicology: Opportunities to Integrate New Approaches. April 24–25, 2012. Crowne Plaza Hotel, Silver Spring, Maryland. http://www.hesiglobal.org/i4a/pages/index.cfm?pageid=53596

AUTHOR CONTRIBUTIONS

The workshop organizing committee members (M.J.A., L.C., B.G., M.H., M.M., S.P., L.F.S. Jr., J.vB., P.W., K.L.W., and V.T.) were responsible for the development of the workshop, selection of the speakers, chairing the various sessions, and writing the section descriptions. The speakers (S.A., D.B., S.I., A.L.K., J.M., S.P., W.S., and R.T.) contributed expanded abstracts of their talks. ILSI/ HESI staff members (J.K., and J.Y.T.) were responsible for the organization of the workshop and coordination of the writing assignments. All authors contributed to the final wording of the manuscript and in addressing the internal reviewers’ comments.

Disclaimer: This document represents the consensus of the participants’ views expressed as individual scientists and does not necessarily represent the policies and procedures of their respective institutions.

Conflicts of Interest: MJ.A. and L.F.S., Jr. are employed by BioReliance Corporation, which offers genetic toxicology testing services on a contract basis.

REFERENCES

  1. Gollapudi BB, Dearfield KL, Hernandez LG, Jeffrey AM, Johnson GE, Julien E, Kim JH, Lovell DP, MacGregor JT, Moore MM, et al. Quantitative approaches for assessing dose-response in genetic toxicology studies. Environ Mol Mutagen. 2013;54:8–18. doi: 10.1002/em.21727. [DOI] [PubMed] [Google Scholar]
  2. Johnson GE, Soeteman-Hernández LG, Gollapudi BB, Bodger OG, Dearfield KL, Heflich RH, Hixon JG, Lovell DP, Macgregor JT, Pottenger LH, et al. Derivation of point of departure (PoD) estimates in genetic toxicology studies and their potential applications in risk assessment. Environ Mol Mutagen. 2014;55:609–623. doi: 10.1002/em.21870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Lynch AM, Sasaki JC, Elespuru R, Jacobson-Kram D, Thybaud V, De Boeck M, Aardema MJ, Aubrecht J, Benz RD, Dertinger SD, et al. New and emerging technologies for genetic toxicity testing. Environ Mol Mutagen. 2011;52:205–223. doi: 10.1002/em.20614. [DOI] [PubMed] [Google Scholar]

RESOURCES