National Academies Press: OpenBook

Using 21st Century Science to Improve Risk-Related Evaluations (2017)

Chapter: 3 Advances in Toxicology

« Previous: 2 Advances in Exposure Science
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

3

Advances in Toxicology

The decade since the publication of Toxicity Testing in the 21st Century: A Vision and a Strategy (NRC 2007) has seen continued advances in an array of technological and biological tools used to understand human function and disease at the molecular level. Some advances were initially catalyzed by the Human Genome Project, which of necessity required technological innovations and large-scale collaborations to reach the ultimate goal of mapping the sequence of DNA. Other developments came from advances made by the pharmaceutical industry to screen for chemicals that have specific biological functionality but minimal off-target effects. As a result of those advances, an era of big-data development and of public access and data-sharing has arrived with ever-increasing data-storage capacity, computational speed, and open-access software. Research has also become more multidisciplinary; project teams today often include geneticists, toxicologists, computer scientists, engineers, and statisticians.

A number of advanced tools can now be used in toxicological and epidemiological research; some examples are listed below.

  • Large banks of immortalized cells that are derived from lymphocytes and collected from different populations worldwide are available for toxicological research.
  • Genetically diverse mouse strains have been created by a multi-institution collaboration (the Complex Trait Consortium; Threadgill and Churchill 2012) and are available for medical and toxicological research. They have been fully genotyped because of the relatively low cost of sequencing today, and the sequence information is publicly available.
  • Microarrays and next-generation RNA sequencing can reveal postexposure changes in the simultaneous expression of large numbers of genes (the transcriptome). Technologies are also now available to profile the epigenome (epigenetic changes, such as methylation and histone modifications), the proteome (proteins present in the cell), and metabolome (small molecules).
  • Large compilations of a wide variety of biological data are publicly available, as is software for data access, interpretation, and prediction. Text-mining tools applied to scientific-literature databases provide approaches for developing hypotheses on relationships between chemicals, genes, and diseases.
  • Automated systems that use multiwell plates provide a high-throughput platform for measuring a wide array of effects in cells and cellular components in response to chemical exposures. Automated, multiwell testing can also be applied for rapid testing of zebrafish, vertebrates that are relatively genetically homologous with humans.
  • Computational advances have enabled the development of chemical structure–based methods for predicting toxicity and systems-biology models for evaluating the effects of perturbing various biological pathways.

Some of the advanced tools could be used to address issues in toxicology and ultimately risk assessment (see Chapter 1, Box 1-3). Some of the general risk-assessment questions to which the tools could be applied are the following:

  • Planning and scoping: Which chemicals should undergo comprehensive toxicological evaluation first (that is, how should priorities be set among chemicals for testing)?
  • Hazard identification: What adverse effects might a chemical have? For example, could it pose a carcinogenic risk or affect kidney or reproductive function? If a data-sparse chemical has a structure or biological activity that is similar to that of a well-studied chemical, can the same types of toxicity be assumed and, if so, at similar exposures? Are cellular-assay responses adaptive (or inconsequential) or harbingers of adverse effects in humans? Does the chemical operate through the same pathways or processes that are associated with cancer, reproductive toxicity, or other adverse human effects?
  • Dose–response assessment: How does response change with exposure? At what exposures are risks of
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

    harm inconsequential? Is there a threshold exposure at the population level below which there is no adverse effect?

  • Mixtures: What are the hazards and dose–response characteristics of a complex mixture? How does the addition of a chemical to existing exposure contribute to risk?
  • Differential susceptibility and vulnerability: Are some populations more at risk than others after exposure to a specific drug or environmental chemical? For example, are some more susceptible because of co-exposures, pre-existing disease, or genetic susceptibility? Are exposures of the young or elderly of greater concern?

Those risk-assessment questions provide the backdrop for considering the recent advances in toxicological tools. Information obtained with the new tools can advance our understanding of the potential health effects of chemical exposures at various points along the exposure-to-outcome continuum, shown in Figure 3-1 below. The starting point along the continuum is the transformation of external exposure to internal exposure, which was discussed in Chapter 2 of this report (see Figure 2-1). The ultimate goal is prediction of the response of the organism or population to exposure, and different tools can be used to probe or inform different places along that continuum. As noted in Chapter 2, although the continuum is depicted as a linear path, the committee recognizes that multiple interconnecting paths are typically involved in the continuum.

This chapter describes a variety of new assays and computational tools that are available for addressing risk-based questions, but it is not meant to be comprehensive. The chapter organization follows the progression along the exposure-to-outcome continuum; the discussion begins with assays and computational tools that are relevant for probing interactions of chemicals with cellular components and ends with ones that are relevant for predicting population-level responses. Understanding of pharmacokinetic relationships is critically important in toxicological evaluations for many reasons—for example, to evaluate whether exposures in in vitro cultures and in vivo assays are similar in magnitude and duration to exposures that result internally in exposed humans; to extrapolate from high to low dose, from one exposure route to another, and between species; and to characterize variability in internal human dose associated with a given exposure. Advances in pharmacokinetic analyses and models were discussed in Chapter 2 and are not elaborated on further

Image
FIGURE 3-1 Computational models and biological assays are shown with the exposure-to-outcome continuum to illustrate where the models and assays might be used to provide information at various points in the pathway. The clear portion of the bar for read-across and SAR models reflects the fact that connections are typically made between analogous chemicals for either the initial biological effect or the outcome. However, biological tools can also probe the response at the cell or tissue level and provide support for read-across and SAR analyses. If sufficient data are available, read-across and SAR analyses can be performed at various points along the exposure-to-outcome continuum.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

here. The chapter concludes with a discussion of challenges and offers recommendations that should help to address the challenges.

The committee emphasizes that most Tox21 assays or systems were not developed with risk-assessment applications as an objective. Therefore, understanding on how best to apply them and interpret data in a toxicology context is evolving. For example, assay systems that were designed to detect agents that have high affinity for or potency against a particular biological target might not be optimized to detect agents that have moderate or low potency or that cause more than one effect. Some risk questions are being addressed as data from high-throughput systems become more available. However, the usefulness or applicability of various assays will need to be determined by continued data generation and critical analysis, and some assays that are highly effective for some purposes, such as pharmaceutical development, might not be as useful for risk assessment of commodity chemicals or environmental pollutants.

PREDICTING AND PROBING INTERACTIONS OF CHEMICALS WITH CELLULAR COMPONENTS

Chemical interactions with specific receptors, enzymes, or other discrete proteins and nucleic acids and promiscuous interactions, such as those between an electrophile and a protein or DNA, have long been known to have adverse effects on biological systems (NRC 2000, 2007; Bowes et al. 2012). Accordingly, the development of in vitro assays that probe molecular-level interactions of chemicals with cellular components has been rapid, driven partly by the need to reduce high attrition rates in the drug-development process. Although various new assays have been developed, only a single assay—one that evaluates the human potassium channel (hERG channel1 —has been integrated into new drug applications. Figure 3-2 illustrates some typical interactions with cellular components, and the following sections describe how the interactions are being investigated.

Predicting Interaction by Using Chemical Structure

In recent years, predicting chemical interactions with protein targets on the basis of chemical structure has become much more feasible, particularly with the development and availability of open-access data sources (Bento et al. 2014; Papadatos et al. 2015). There are many published examples of computational models that have been developed to predict the interaction of a molecule with a single protein, most notably models for predicting hERG activity (Braga et al. 2014) and interaction with the estrogen receptor (Ng et al. 2015), but prediction of multiple interactions in parallel is now possible given available computational power. For example, Bender et al. (2007) used chemical similarity to predict the protein–chemical interactions associated with a novel chemical structure with a reported average accuracy of over 92% with some proteins and high selectivity; that is, only small numbers of active predictions were later shown to be negative in vitro. Although most of the activities were predicted correctly, it was at the expense of a high false-positive rate (that is, large numbers of inactive chemicals were predicted to be active). Most of the models have been built by using pharmaceutical candidates that have a high affinity for the particular protein, but there are examples in the literature in which the same approaches have been applied to identify chemicals that bind a receptor with low affinity (see, for example, Hornung et al. 2014).

Research to improve the prediction of protein–chemical interactions continues apace. Lounkine et al. (2012) used the similarity-ensemble approach—a method first published by Keiser et al. (2007)—and predicted the activity of 656 marketed drugs with 73 protein targets that were thought to be associated with clinical adverse events. The authors reported that about 50% of the predictions of activity were later confirmed experimentally with binding affinities for the protein targets of 1 nM to 30 μM.

Image
FIGURE 3-2 Exposure-to-outcome continuum with examples of types of interactions between biological molecules and chemicals.

___________________

1 The blockade of hERG channel has been directly implicated in prolongation of the QT interval, which is thought to play a role in the potentially fatal cardiac arrhythmia torsades de pointes.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Cheng et al. (2012) evaluated chemical–protein interaction sets that were extracted from the ChEMBL database2 by using a computational method, multitarget quantitative structure–activity relationship (QSAR), that evaluates G-protein coupled receptors (GPCRs) and kinase protein targets. Sensitivities were reported to range from 48% to 100% (average, 84.4%), and specificity for the GPCR models (about 99.9%) and the kinases was high.

Assessing Interactions with Cell-Free Assays

Cell-free or biochemical assays have long been used to probe the interactions of chemicals with biological molecules, such as enzymes and hormone receptors, and their activity with these specific targets (Bhogal et al. 2005). The assays can provide reliable and valid results with high agreement between laboratories and can be applied in low-, medium-, or high-throughput formats (Zhang et al. 2012a).

The US Environmental Protection Agency (EPA) is exploring the use of the commercially available cell-free assays, run in high-throughput format, that were originally developed for preclinical drug evaluation to assess environmental chemicals (Sipes et al. 2013). The panel selected by EPA measures various activities, including binding to GPCRs, steroid-hormone and other nuclear receptors, ion channels, and transporters. The panel also covers activation of kinases, phosphatases, proteases, cytochrome P450, and histone deacetylases (Sipes et al. 2013). Roughly 70% of the assays are derived from human cells, 20% from rat cells, and the remainder from other species.

A wide variety of cell-free assays that evaluate other targets have been developed and are being used in pharmaceutical, biomedical, and academic laboratories (Xia et al. 2011; Mehta et al. 2012; Landry et al. 2015; McKinstry-Wu et al. 2015). They are being used to probe a wide array of protein types and functions, such as nod-like receptors, which are involved in immune and inflammatory responses (Harris et al. 2015), methyltransferases (Dong et al. 2015), and various membrane proteins (Wilcox et al. 2015).

The potency of the chemical’s interaction in vitro—measured, for example, as an IC50 or KI3 —provides information on the likelihood of an in vivo concentration high enough to permit observation of the phenotypic response. The degree of inhibition or activation of the protein function that is required for a phenotypic response to be observed can vary widely and will depend partly on the nature and function of the protein or enzyme. For inhibitors of GPCRs, the anticipated pharmacological response has been observed in vivo at plasma concentrations less than or equal to three times the measured IC50 of the chemical in question when corrected for plasma-protein binding (McGinnity et al. 2007). As a rule of thumb for pharmaceuticals, a 100-fold margin between the measured IC50 or KI in a cell-free assay and the circulating plasma unbound Cmax has been considered adequate to represent minimal risk of toxicity (N. Greene, AstraZeneca, personal commun., December 14, 2015). However, for environmental chemicals, which are not tested in clinical trials or followed up through medical surveillance, a different rule of thumb might be appropriate. And it is important to remember that toxicity is influenced by many factors, including the required degree of receptor occupancy, the ability of the chemical to reach the site of action (for example, to penetrate the blood–brain barrier), the nature of the modulatory effects (for example, inhibitor, agonist, or allosteric modulator), the kinetics of the binding of the interaction with the receptor, and exposure duration.

CELL RESPONSE

Cell-based in vitro assays have existed for nearly a century; the first publication of a dissociated cell culture was in 1916 (Rous and Jones 1916). Cell-culture technology has evolved to the point where many cell lines are available and more can be produced with current techniques. Cell cultures provide easy measurement of gene and protein expression and a variety of potentially adverse responses (see Figure 3-3) and can be scaled to a high-throughput format (Astashkina et al. 2012). Additionally, cell-based assays derived from genetically different populations can allow rapid assessment of some aspects of variability in response to chemical exposures that depend on genetic differences (Abdo et al. 2015).

Cell-based assays are being used to inform hazard identification and dose–response assessments, mostly as a complement to data from whole-animal or epidemiological studies to address questions of biological plausibility and mechanisms of toxicity. For example, in evaluations of chemical carcinogenicity, the International Agency for Research on Cancer (IARC) gives weight to functional changes at the cellular level (IARC 2006) and considers the relevance of the mechanistic evidence with regard to key characteristics of cancinogens (Smith et al. 2016). Cell-based assays have been critical in the IARC assessments (IARC 2015a,b). Human-derived and animal-derived cell cultures have also been used to discern dose–response relationships and toxicogenomic profiles, for example, for ethylene oxide responses (Godderis et al. 2012). The assays have potential use in addressing many

___________________

2 ChEMBL is a chemical database of biologically active molecules that is maintained by the European Bioinformatics Institute of the European Molecular Biology Laboratory.

3 IC50 is the concentration required to cause 50% of the maximal inhibitory effect in the assay, and KI is the inhibition constant for a chemical and represents the equilibrium constant of the dissociation of the inhibitor-bound enzyme complex.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

of the risk-based questions raised at the beginning of this chapter and as illustrated in Chapter 5.

Cell cultures can be grown in a variety of architectures, including monolayer and 3-D cultures of cell lines, and can be used as indicators of possible tissue, organ, and sometimes organism-level signs of possible toxicity, particularly in integrated systems that consider effects and signaling among cell types (Zhang et al. 2012a). They can be used to evaluate a number of cellular processes and responses, including receptor binding, gene activation, cell proliferation, mitochondrial dysfunction, morphological or phenotypic changes, cellular stress, genotoxicity, and cytotoxicity. Various techniques and measurements—such as impedance, gene transcription, direct staining, reporter-gene output, and fluorescence or bioluminescence resonance energy transfer—can be used to measure cellular responses and processes (An and Tolliday 2010; Song et al. 2011; Asphahani et al. 2012; Smith et al. 2012). Furthermore, simultaneous measurements of multiple toxic phenotypes are possible with high-content imaging and other novel techniques. This section describes some of the recent developments in using cell-based assays to evaluate cellular response and emphasizes advances that can improve toxicology and risk assessment.

The committee notes that cell-based assays have some limitations; one key concern involves metabolic capabilities. Specifically, do the assays capture how exogenous substances are metabolized in the body? That particular limitation might not be a concern for assays that are performed with low-throughput methods in which it might be possible to determine a priori whether metabolism is important for toxicity and, if so, to find ways to test the metabolites in addition to the parent chemicals. However, little or no metabolic capacity is a particular concern for high-throughput systems that are used for priority-setting. Parent chemicals and metabolites can differ substantially in toxicity and potency. If the in vitro assays do not sufficiently capture critical metabolites that form in humans, they might not give valid results for assessment because they are not testing the chemicals that potentially give rise to toxicity. Furthermore, although some assay systems might capture metabolism in the liver, extrahepatic metabolism might be the driver of some chemical toxicity, so the spectrum of relevant in vivo metabolic activation is an important consideration in understanding the validity of in vitro studies and interpreting the results from both in vitro and in vivo studies. EPA, the National Institute of Environmental Health Sciences, and the National Center for Advancing Translational Sciences are awarding research grants to make progress on the issue. For example, a multiagency collaborative announced in 2016 a $1 million competition in the Transform Tox Testing Challenge: Innovating for Metabolism; the challenge called on innovators to identify ways to incorporate metabolism into high-throughput screening assays (EPA/NIH/NCATS/NTP 2016). EPA is also attempting to develop a system that encapsulates microsomal fractions of human liver homogenate in a matrix, such as an alginate, that will allow diffusion of low-molecular-weight chemicals but retain the toxic lipid peroxides. As an alternative approach, EPA is attempting a method that would transfect cells with mRNAs of enzyme-encoding genes to increase metabolic transformation intracellularly. The committee views those initiatives as steps in the right direction and emphasizes the importance of addressing the issue of metabolic capacity.

Primary Cells

Primary cells are isolated directly from fresh animal or human tissue. They can be obtained from a wide variety of tissues, such as liver, brain, skin, and kidney; and they are amenable to high-content screening and analysis (Xu et al. 2008; Zhang et al. 2011; Thon et al. 2012; Raoux et al. 2013; Tse et al. 2013; Valdivia et al. 2014; Feliu et al. 2015). Although primary cells are more reflective of in vivo cellular and tissue-specific characteristics than are immortalized cells (Bhogal et al. 2005), they can

Image
FIGURE 3-3 The exposure-to-outcome continuum with examples of cell responses.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

be short-lived in culture and suffer from rapid dedifferentiation within hours to days.

Several approaches to adapt primary cell culture to a high-throughput format for chemical-toxicity testing have been made (Sharma et al. 2012; Berg et al. 2015). For example, EPA profiled over 1,000 chemicals (Houck et al. 2009; Kleinstreuer et al. 2014) to identify activity in eight primary cell systems, including ones that used fibroblasts, keratinocytes, and endothelial, peripheral blood mono-nuclear, bronchial epithelial and coronary artery smooth muscle cells. With proprietary software, chemicals were clustered by bioactivity profiles, and some possible mechanisms of chemical toxicity were identified. The lack of publicly available datasets with which to compare the results and the complexity of the resulting data precluded sensitivity and specificity calculations (Kleinstreuer et al. 2014). The standard by which to judge construct validity—that is, whether an assay system as a whole adequately represents the target biological effect—still poses a challenge for these and other assays described in this chapter (see “Challenges and Recommendations for Advancing Toxicology” later in this chapter).

A major advance in primary cell culture over the last decade is the development of 3-D cultures of cell lines.4 3-D cell cultures have better behavior and function than the monolayer cultures (van Vliet 2011) and are of increasing interest in the development of cancer drugs because they recapitulate the tumor microenvironment to a much greater extent than do conventional monolayer assays that use a flat layer of cells (Edmondson et al. 2014; Lovitt et al. 2014). A number of assays that use 3-D cultures of primary cells from various tumors have been developed. Several studies (Arai et al. 2013; Chen et al. 2014) have shown some degree of drug resistance to well-characterized cancer drugs, depending on assay type; 3-D assays show greater drug resistance.

Similarly, primary isolated hepatocytes are the most widely used for in vitro testing, and 3-D culture systems with added cofactors are being developed to overcome limitations of conventional monolayer systems (Soldatow et al. 2013), which notably include lack of sensitivity for detection of hepatotoxic drugs. The 3-D cultures that are used, for example, for enzyme induction or inhibition studies, maintain function for a relatively long period (1–3 days) and can be used to re-establish cellular polarity that is lost in monolayer cultures. Advances in liver-culture techniques and technology have led to improvements and greater complexity in 3-D liver-cell culture for use in toxicological evaluations, and the next step is development of a bioartificial liver, commonly referred to as an organ-on-a-chip, discussed in greater detail in “Tissue-Level and Organ-Level Response.”

The examples of tumor-cell and liver-cell cultures discussed in this section highlight the movement from monolayer cultures to improved 3-D cultures of greater complexity and ultimately toward organotypic models for various tissues and organs (Huh et al. 2011; Bulysheva et al. 2013; Guiro et al. 2015).

Immortalized Cell Lines

Immortalized cell lines can be derived from isolated human cancer cells or from primary cells that have been genetically altered for enhanced longevity and resilience in tissue culture. Immortalized cell lines do not need to be isolated and harvested for each use, are relatively easy to maintain and propagate, are stable when replated multiple times, and can be easily frozen and shared between laboratories and grown in large quantities. Cloning immortalized cells enables testing in genetically identical cells, and immortalized cell lines that are derived from diverse populations allow inquiry into the variability of chemical toxicity among populations (Abdo et al. 2015). However, more than the conventional monolayer cultures of primary cells, immortalized cell lines can lose native in vivo properties and functionality. They can have altered cellular polarity (Prozialeck et al. 2003; Soldatow et al. 2013), non-native genetic content (Yamasaki et al. 2007), and decreased amounts of key cellular features (such as ligands, transporters, and mucin production); and they can be contaminated with other cell lines, such as HeLa and HepG2. Alterations in cellular phenotype can result in insensitivity to and mischaracterization of test chemicals. For example, when testing the difference between mitochondrial toxicity observed in renal proximal tubule cells (primary cells) and that observed in immortalized human renal cells, researchers found that primary cells were capable of identifying more possible toxicants than were immortalized cell lines (Wills et al. 2015).

Many of the assays in the federal government’s ToxCast and Tox21 programs use immortalized carcinoma-derived cell lines (T47D breast, HepG2 liver, and HEK293T kidney). The assays have shown potential for identifying chemical carcinogens found in rodents (Kleinstreuer et al. 2014) and for exhibiting some predictive ability in the preliminary classification of hepatotoxic chemicals in guideline and guideline-like animal studies (Liu et al. 2015). However, the assays have also been shown to be unable to predict some well-recognized hazards observed in humans or animals (Silva et al. 2015; Pham et al. 2016).

ToxCast data have been proposed for use in predicting in vivo outcomes of regulatory importance (see Rotroff et al. 2013; Sipes et al. 2013; Browne et al. 2015), such as estrogenic properties of chemicals predicted by the uterotrophic assay, but their use as replacement assays has been the subject of research and discussion. For

___________________

43-D culture is a generic term that is used to describe culture systems that are grown on some sort of support or scaffold, such as a hydrogel matrix. 3-D cultures often have two or more cell types.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

example, EPA’s Federal Insecticide, Fungicide, and Rodenticide Act (FIFRA) Science Advisory Panel recommended that the agency not replace the uterotrophic assay with a computational model of estrogen receptor agonist and antagonist activity derived from ToxCast data (EPA 2014a). Although the panel noted a number of strengths of the model, it had concerns about diminished performance of the model for nonreference chemicals and the inability of the model to assess chemicals that had modified toxicity because of pharmacokinetic factors or that had toxicity pathways different from those evaluated in the assays. Thus, the panel found that further research was needed. More recently, EPA reconsidered the results of a high-throughput battery of estrogenicity assays, concluded that the test battery is a satisfactory replacement of the uterotrophic assay for tier 1 endocrine-disrupter screening, and intends to use the results of the test battery to evaluate and screen chemicals in the future (Browne et al. 2015; EPA 2015).

Because immortalized cell lines are limited in the degree to which they can represent cells in intact tissues, alternative approaches of cell immortalization have been developed and are now being made commercially available. “Conditionally immortalized” cell lines that can undergo differentiation are increasingly available for use in biomedical research with potential applications in toxicology (Liu et al. 2015).

Stem Cells

Advances in stem-cell research have allowed the generation of a wide array of cell types, some of which have metabolic competence, which makes them useful for studying the effects of chemicals on various tissues (Scott et al. 2013; Gieseck et al. 2015). Fit-for-purpose stem-cell–based tests are becoming commercially available (Anson et al. 2011; Kolaja 2014), and research is under way to develop stem cells for application in toxicology (Sjogren et al. 2014; Romero et al. 2015). For example, an in vitro murine neural embryonic stem-cell test has been advanced as an alternative for a neurodevelopmental toxicity test (Theunissen et al. 2012; Tonk et al. 2013). The ability to grow rapidly, manipulate, and characterize an array of cell types makes stem cells potentially useful for chemical-toxicity evaluations. Furthermore, assays that use stem cells harvested from genetically diverse populations show considerable promise for providing information that can help in addressing hazard and risk-assessment questions.

Stem cells of potential use in toxicology research are of three primary types: embryonic, adult, and induced pluripotent stem cells. Embryonic stem cells are harvested from embryos that are less than 5 days old and have unlimited differentiation ability. Adult stem cells are isolated from adult bone marrow, skin, cord blood, heart tissue, and brain tissue. Induced pluripotent stem cells (iPSCs) are produced from adult somatic cells that are genetically transformed into a pluripotent state (Takahashi et al. 2007). iPSCs are similar to embryonic stem cells (pseudoembryonic) and can be grown in monolayer and 3-D structures for multiple generations. They can take on a variety of cell types, including neuronal cells (Efthymiou et al. 2014; Malik et al. 2014; Sirenko et al. 2014a; Wheeler et al. 2015), hepatocytes (Gieseck et al. 2014; Sirenko et al. 2014b; Mann 2015), and cardiomyocytes (Sinnecker et al. 2014; Karakikes et al. 2015). The ability to be derived from adult cells and the capacity to differentiate into multiple cell types also make iPSCs particularly promising for exploring human diversity. Cells can be created from specific individuals to produce personalized biomarkers, and iPSCs derived from large patient populations (Hossini et al. 2015; Mattis et al. 2015) could help to identify pathways involved in disease and susceptibility (Astashkina et al. 2012). Because iPSCs are relatively cost-effective to produce on a large scale (Beers et al. 2015), they have the potential to improve cell-based toxicity testing substantially.

There are some challenges to overcome in using stem cells. They can have different expression profiles, which indicate that they might have altered cellular processes, pathways, and functions. Stem cells generally can be difficult to culture and transfect, and the difficulties could limit their application in high-throughput formats. The lack of systematic approaches for characterizing and standardizing culture practices (such as characterizing cell types, sex origin, and cell function) also presents an obstacle for using stem cells in toxicology applications. Although stem cells (and other cells) have inherent limitations, they are still useful windows into biological processes at the cellular and molecular levels and remain useful for assessing chemical toxicity. A careful evaluation of cell phenotype and properties would help to determine the extent to which human biology is recapitulated in the cellular model.

Modeling Cellular Response

Over the last decade, numerous mathematical models and systems-biology tools have been advanced to describe various aspects of cell function and response. Considerable progress has been made in describing feedback processes that control cell function. The development of cell-based modeling has benefited greatly from coordinated contributions from the fields of cell biology, molecular biology, biomedical engineering, and synthetic biology.

A few simple structural units that have specific functions and appear repeatedly in different species are referred to as network motifs (Milo et al. 2002; Alon 2007). Molecular circuits are built up from network motifs and carry out specific cellular functions, such as controlling

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

cell-cycle progression, xenobiotic metabolism, hormone function, and the activation of stress pathways—the major pathways by which cells attempt to maintain homeostasis in response to chemical and other stressors, such as oxidative stress, DNA damage, hypoxia, and inflammation. Computational models are used to examine those circuits, the consequences of their activation, and their dose–response characteristics.

Toxicity pathways defined in NRC (2007) as cellular-response pathways can be thought of as molecular circuits that, when sufficiently perturbed, lead to adverse effects or toxicity. The circuits can be modeled with computational systems-biology approaches. The tools for describing the circuits and function are developing rapidly (Tyson and Novak 2010; Zhang et al. 2010) and should enable study of the dose–response characteristics of the perturbation of toxicity pathways (Simmons et al. 2009; Zhang et al. 2014, 2015). Quantitative descriptions of the pathways hold the promise of characterizing differences in individual susceptibility to chemicals at the cellular level but will require identification of components of signaling pathways that differ among individuals; sensitivity and other analyses can be applied to determine components that most affect human variability in adverse response. Confidence in the models will increase as they are applied to a more diverse suite of signaling pathways. Model refinement coupled with careful collection of data on detailed biological responses to chemical exposure will test model structures, refine experimental strategies, and help to chart new approaches to understanding of the biological basis of cellular dose–response behaviors at low doses.

TISSUE-LEVEL AND ORGAN-LEVEL RESPONSE

The last decade has seen advances in engineered 3-D models of tissue and computational models for simulating response at the tissue level (see Figure 3-4). This section describes organotypic models, organ-on-a-chip models, and virtual-tissue models that might be particularly applicable for toxicology research.

Organotypic Models

An organotypic model is a specific type of 3-D culture in which two or more cell types are put together in an arrangement intended to mimic, at least in part, an in vivo tissue and that therefore recapitulates at least some of the physiological responses that the tissue or organ exhibits in vivo. Organotypic models of skin, which contain keratinocytes and fibroblasts, have been developed and validated for use as alternative models for testing skin irritation (Varani et al. 2007), and data from these models are now accepted in Europe for classification and labeling of topically applied products (Zuang et al. 2010). The skin model is being evaluated to improve the specificity of in vitro genotoxicity testing. Organotypic skin cultures appear to have reasonably good concordance with in vivo genotoxicity results (Pfuhler et al. 2014) probably because they retain the ability to metabolize and detoxify chemicals and because the rate of delivery of chemicals to the basal layer is more comparable with the kinetics of dermal absorption in vivo. Other organotypic models include eye, lung epithelium, liver and nervous system tissue (see NASEM 2015). The effects of environmental chemicals have been explored in mouse organoids by using proteomic tools (Williams et al. 2016).

Organ-on-a-Chip Models

An emerging scientific development is the organ-on-a-chip model (see Figure 3-5), which is a 3-D culture grown in a multichannel microfluidic device (Esch et al. 2015). The models are meant to have the same functionality as organotypic cultures but with the ability to manipulate physiological and pharmacokinetic processes (that is, the rate at which a chemical is introduced via the flow-through channels). Several organ-on-a-chip models have been engineered, including ones for liver, heart, lung, intestine, and kidney. The models allow the study of how chemicals can disrupt an integrated biological system and how the disruption might be influenced by the mechanical

Image
FIGURE 3-4 Exposure-to-outcome continuum with examples of tissue and organ effects.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

forces at play in the intact organ, such as the stretching of the alveolar-capillary barrier in lungs due to the act of breathing.

Attempts have been made to design platforms that have different organ mimics arranged in series or parallel that as a system can recapitulate aspects of tissue interactions and in vivo pharmacokinetics (Sung and Shuler 2010). A long-term goal is to introduce a parent chemical into the system and have it move through a liver compartment where it would be metabolized, flow to compartments that contain responsive cell types or to other compartments that contain hydrophobic materials that represent fat, and finally flow through a kidney compartment where it could be eliminated. To date, microfluidic platforms that have that much complexity have not yet been introduced in practice and have not achieved a realistic metabolite distribution through the various tissues in the system (Andersen et al. 2014).

Researchers face challenges in developing such experimental platforms, for example, with the synthetic materials used in the manufacture of the cell-culture substrates. They often are not good mimics of the extracellular matrix and can even absorb small hydrophobic molecules (Wang et al. 2012); that absorption might exert an undue influence on the physiological system or alter chemical concentrations. Large-scale manufacture and high-throughput operation of organ chips also present challenges to the adoption of the technology. Similarly, access to sustainable sources of human cells presents a substantial hurdle for reproducibility and interpretation of the data produced.

Microsystems that are composed of multiple synthetic organ compartments are in the early stages of development, and a number of initiatives are going on to validate model correlations with in vivo observations. For example, the National Center for Advancing Translational Sciences has a number of efforts in this field (NCATS 2016), and the European Union–funded initiative Mechanism Based Integrated Systems for the Prediction of Drug Induced Liver Injury (EU 2015) has also been exploring the use of liver-chip models to predict adverse effects of drugs. Organ-on-a-chip models are promising, but they are not yet ready for inclusion in risk assessments.

Virtual Tissues

As discussed earlier, computational systems biology might be used to describe pathway perturbations that are caused by chemical exposures and the resulting cell responses. Such modeling can be applied to multiple processes that operate in sequence or parallel and used to link cellular responses to tissue-level responses. Modeling feedforward and feedback controls through sequential dose-dependent steps also enable the examination of responses to toxicant exposure that require multiple cell types, such as Kupffer cell–hepatocyte interactions

Image
FIGURE 3-5 Generalized components of an organ-on-a-chip model. Source: Birnbaum 2011.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

involved in hepatocyte proliferation. Feedback and feedforward control might also contribute to intercellular patterns of response that require input from earlier pathway or cellular functions to activate or inhibit integrated multicellular responses. The cellular responses alter tissue function; the quantitative modeling then focuses on the interface between the cellular-level computation models and virtual-tissue models.

EPA’s Computational Toxicology Program has developed mathematical models called virtual tissues for the embryo and the liver (Shah and Wambaugh 2010; Wambaugh and Shah 2010). EPA also has developed a model of blood-vessel development. Virtual-tissue models can use “agent-based” modeling of different cells in the tissue, which relies on and mathematically describes key aspects of cellular behavior or other tissue components to derive the properties of the tissue or organ of interest (Swat and Glazier 2013). The EPA models evaluate chemical exposures that alter growth and phenotypic characteristics of the agents in the models, which in this case are the cells. The models can describe cell growth or pattern formation of different structures in the virtual embryo or regional distribution of cell response in the virtual liver.

As with any model, a critical consideration in developing response models is fidelity of biology between the modeled outcome (virtual-tissue responses) and the apical and other responses observed experimentally. Assumptions and predictions of the models can be tested by using information from human cells and co-cultures with different human cell types. Short-term targeted animal studies that use toxicogenomic tools and other approaches can be used to evaluate the model more broadly. Virtual-tissue models have the potential to help in conceptualizing and integrating current knowledge about the factors that affect key pathways and the degree to which pathways must be perturbed to activate early and intermediates responses in human tissues and, when more fully developed, to support risk assessments based on studies of key events and how the key events combine to cause adverse responses at the organism level.

ORGANISM-LEVEL AND POPULATION-LEVEL RESPONSE

The Tox21 report (NRC 2007) emphasized a future in which routine toxicity testing would rely on in vitro assays with human cells or assays that probe molecular responses of human toxicity pathways and pathway components. But, the report also noted that in some cases testing in whole animals might be necessary, depending on the nature of the risk-assessment questions, although whole-animal studies were not intended to provide routine information for assessing risks. The need for different types of information related to the nature of the question posed was also emphasized in EPA’s report on next-generation risk assessment (EPA 2014b; Krewski et al. 2014; Cote et al. 2016). That report considered three types of assessments: screening and priority-setting assessments, limited-scope assessments, and in-depth assessments. The last one would likely involve a wide array of toxicity-testing approaches, including whole-animal studies. Approaches for assessing variability could also benefit from rodent panels that capture population variability and panels of human cells derived from a group of diverse people. As is true of toxicity-testing tools at the molecular and cellular levels, there has been continuing development of new methods for examining responses in whole animals that are likely to provide important information for the limited-scope and especially for the in-depth assessments. The approaches for assessments on different levels emphasize a fit-for-purpose orientation of designing the testing assays or batteries that depend on the risk-assessment question. This section discusses novel animal models that provide opportunities for enhancing the utility and power of whole-animal testing. It also describes recent advances in structure-based computational models and read-across approaches that provide opportunities for predicting response of data-poor chemicals at the organism level. Figure 3-6 highlights some organism-level and population-level responses.

Image
FIGURE 3-6 Exposure-to-outcome continuum with examples of organism and population-level responses.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Novel Whole-Animal Models

Advances in genetics, genomics, and model-organism development have led to genetically well-characterized whole-animal models, including transgenic rodent lines, isogenic mouse strains, and alternative species, such as zebrafish and Caenorhabditis elegans, which can be studied in a high-throughput format. Those models coupled with toxicogenomics and novel imaging offer improvements over the traditional in-life rodent studies in that they offer new ways to explore chemical interactions at tissue and cellular levels. Isogenic strains also offer new opportunities to identify determinants of human susceptibility, especially when coupled with new interrogation tools, and to define new mechanisms of toxicity. Targeted testing, which is typically hypothesis-driven and more focused than historical testing strategies, can help to develop and enhance the value of the new animal models, as well as traditional ones. It can be used to explore the mechanisms by which a chemical causes toxicity, how outcomes might differ by age and sex, and how susceptibility might vary in the population. It can help to address specific knowledge gaps in risk assessment and can link in vitro observations to molecular, cellular, or physiological effects in the whole animal. Targeted testing will be critically important in evaluating and validating the robustness and reliability of new computational models, in vitro assays, and testing batteries (Andersen and Krewski 2009; Krewski et al. 2009). As this section shows, the new animal models and outcome-interrogation tools might provide broader assessment of hazards in whole organisms.

Transgenic Rodents

The development of transgenic mouse lines (such as knockin, knockout, conditional knockout, reporter, and humanized lines) advanced biomedical research; a few transgenic rat lines are also available now. Novel gene-editing technologies, such as CRISPR/Cas9, have the potential to generate inducible gene editing in adult animals and the creation of transgenic lines in nontraditional mammalian models (Dow et al. 2015). Gene editing permits the creation of experimental approaches that are more specifically suited for various tasks, including targeted testing of susceptible strains and exploration of gene–environment interactions.

Although transgenic animals have been available for decades (Lovik 1997; Boverhof et al. 2011), testing in transgenic animals and incorporation of data from transgenic models into risk assessment has been limited, partly because of questions about applicability for risk assessment and concerns about the cost to develop the models and evaluate a chemical in multiple strains. The National Toxicology Program (NTP) continues to evaluate and develop such models. For example, NTP is using transgenic mice in the testing of the artificial sweetener aspartame, which generally tested negative in standard assays but showed a slight increase in brain tumors in a more sensitive transgenic-mouse strain. The transgenic p16 model was used because it was thought to be susceptible to brain glial-cell tumors. NTP is also testing aspartame in transgenic strains with knocked-out tumor-suppressor genes and activated oncogenes to improve characterization of susceptibility and risk related to gene–environment interactions. Transgenic-rodent mutation data have been used by EPA to understand carcinogenic mechanisms of several agents, such as acrylamide (EPA 2010), but beyond those applications their incorporation into risk assessment has been limited. They have been somewhat more widely used to test specific hypotheses about mechanism, such as the mechanism of liver-cancer induction by phthalates (Guyton et al. 2009), and to evaluate the depth of biological understanding to apply fully organotypic, computational systems-biology, physiologically based pharmacokinetic (PBPK), or other tools.

Genetically Diverse Rodents

Historically, toxicity testing has used only a few rodent species and strains. Although there are advantages in using a well-characterized strain of mice or rats to test chemical toxicity, there are many shortcomings, including concerns about inadequately accounting for profound strain differences in chemical sensitivity and metabolism (Kacew and Festing 1996; Pohjanvirta et al. 1999; De Vooght et al. 2010) and inadequate genetic and phenotypic diversity. High rates of spontaneous disease in some strains (outbred and inbred) can sometimes complicate the interpretation of results. For example, the incidence of background cardiomyopathy in the Sprague Dawley rat can be as high as 100% (Chanut et al. 2013), some strains are completely resistant to some toxicants (Shirai et al. 1990; Pohjanvirta et al. 1999), and it is unclear a priori whether the standard strain has sensitivity that is adequate or too high for identifying a potential human hazard.

Assessment in multiple strains that have known genetic backgrounds is one approach to address variable sensitivity among relatively homogeneous test strains and to address questions related to interindividual sensitivity to toxicants. Initiated in 2005, the Collaborative Cross (CC) is a large panel of novel recombinant mouse strains created from an eight-way cross of founder strains that include three wild-derived strains. The CC has a level of genetic variation akin to that of humans and captures nearly 90% of the known variation in laboratory mice (Churchill et al. 2004). Outbred progeny that have completely reproducible genomes can be produced through the generation of recombinant inbred intercrosses (RIX) (Zou et al. 2005). Because the CC strains and, by extension, the RIX lines have a population structure that randomizes exist-

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

ing genetic variation, these models provide the increased power that is required to explore the genetic underpinnings of interindividual susceptibility. For example, the CC mouse replicated human susceptibility, immunity, and outcome of West Nile virus infection more comprehensively than the standard inbred model (C57BL/6J) (Graham et al. 2015).

There are several examples of the value of the CC in toxicological evaluation. Trichloroethylene (TCE) metabolism, for example, varies considerably among people and among mouse strains, and the metabolites differ in their mechanisms, toxicity, and organ-specific effects (NRC 2006). That variability has been a critical barrier to understanding of the risk that TCE poses to humans. To address the challenge in TCE-toxicity testing, a battery of mouse lines was used to assess interindividual variability in TCE metabolism and toxicity in the liver and kidney (Bradford et al. 2011; Yoo et al. 2015a,b). Significant differences in toxicity and metabolism were observed in the different strains. Population PBPK modeling was applied to the study results to illustrate how data on diverse mouse strains can provide insight into pharmacokinetic variability in the human population (Chiu et al. 2013).

Multistrain approaches have also revealed fundamental mechanisms of hepatotoxicity of acetaminophen and biomarkers of this potentially fatal effect. Harrill et al. (2009) used a panel of 36 inbred mouse strains and found that liver injury induced by acetaminophen was associated with polymorphisms in four genes, but susceptibility to hepatotoxicity was associated with yet another, CD44. Follow-up study of two healthy human cohorts showed that variation in the human CD44 gene conferred susceptibility to acetaminophen liver toxicity. This powerful example shows how a diverse animal population (in this case, mice) can be used to characterize and identify potential susceptibility in humans.

The Diversity Outbred (DO) population is a heterogeneous stock seeded in 2009 from 144 independent lineages from the CC breeding colony. Each DO mouse is unique and has a high level of allelic heterozygosity (Churchill et al. 2012). Because they were derived from the same eight strains as the CC mice, their genome can be reconstructed with a high degree of precision—a feature that facilitates genome-wide association studies and other similar approaches. A 2015 NTP proof-of-concept study that used DO mice to capture variation in benzene susceptibility successfully identified two sulfotransferases that modify and eliminate benzene metabolites that confer resistance to benzene toxicity (French et al. 2015).

One caveat in using genetically diverse rodent models is that their use potentially can increase animal use. The most effective use of such models in toxicology requires acceptance of novel computational approaches, experimental designs, and statistical approaches that are specifically suited for the models and capable of handling the unprecedented amount of data that these studies generate (Festing 2010). For example, factorial designs can maximize genetic diversity and reduce the risk of false negatives without necessarily requiring more animals than traditional rodent studies to address the central question. Additionally, using DO mice requires accepting that each individual is unique and that there is no way to incorporate “biological replicates” in the traditional sense. Researchers and risk assessors need to be aware of and comfortable with the suite of data that results from these studies and to understand how to integrate the data with information from other sources, including more traditional animal models (see Chapter 7). Computational tools uniquely suited for these emerging animal models are available and readily adaptable to toxicological testing (Zhang et al. 2012b; Morgan and Welsh 2015). Tools for data analysis, visualization, and dissemination are also available (Morgan and Welsh 2015). As with any model system, these rodent models should be used only for questions that they are best suited to address. NTP and other groups are developing frameworks and use cases to highlight when it is advantageous to use such models, and the committee supports further discussion on this issue.

Other Whole-Animal Systems

Advances in genomics, imaging, and instrumentation have made some alternative species—such as Caenorhabditis elegans (a nematode), Drosophila melanogaster (a fruit fly), and Danio rerio (the zebrafish)—useful animal models for hazard identification and pathway discovery. Many technical advantages are shared among the three dominant nonmammalian species, but zebrafish have several useful characteristics not shared by the others. The genomes of zebrafish and humans display remarkable homology with an overall conservation of over 70%. Furthermore, 80% of the genes known to be involved in human disease are expressed in zebrafish (Howe et al. 2013b). The signal-transduction mechanisms, anatomy, and physiology of zebrafish are homologous to those of humans (Dooley and Zon 2000), and zebrafish have all the classical sensory pathways, which are generally homologous to those of humans (Moorman 2001; Colley et al. 2007).

Another important attribute that might make zebrafish particularly well suited for translational research is the capacity to generate transgenic reporter lines that express fluorescent genes in specific cells, tissues and organs. The large collection of transgenic fish lines are curated by the Zebrafish Model Organism Database and maintained by the Zebrafish International Information Network (Howe et al. 2013a). There is also a rich diversity of zebrafish-disease models and drug screens to help to understand, prevent, and develop therapies for human diseases, including various cancers (Feitsma and Cuppen

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

2008; Nguyen et al. 2012; Gallardo et al. 2015; Gordon et al. 2015), diabetes and obesity (Gut et al. 2013; Dalgin and Prince 2015; Schlegel and Gut 2015), psychiatric conditions (Panula et al. 2010; Norton 2013; Jones and Norton 2015), heart disease (Arnaout et al. 2007; Chico et al. 2008; Arnaout et al. 2014; Asnani and Peterson 2014; Walcott and Peterson 2014), neurodegenerative syndromes (Bretaud et al. 2004; Chapman et al. 2013; Mahmood et al. 2013; Da Costa et al. 2014; Martin-Jimenez et al. 2015; Preston and Macklin 2015), autism (Tropepe and Sive 2003), immunodeficiencies (Meeker and Trede 2008; Cui et al. 2011), and blood disorders (Ablain and Zon 2013). Zebrafish have been used to investigate neurotoxicants (Levin et al. 2007; Egan et al. 2009; Irons et al. 2010), and Box 3-1 provides an example of using zebrafish for behavioral assessments.

The Zebrafish Mutation Project hosted by the Sanger Institute is yet another major effort that will facilitate cross-species studies. The project aims to develop a knockout allele in every protein-coding gene in the zebrafish genome and characterize its morphological phenotype (Kettleborough et al. 2013). Mining of zebrafish gene or phenotype databases should provide powerful opportunities to identify genes involved in chemical-induced phenotypes.

An additional advantage of zebrafish is that the zebrafish genome is fully annotated, so transcriptomic and all other -omics approaches are possible. Repression of gene expression by antisense morpholinos, siRNA, and such gene-editing techniques as CRISPR/Cas9 is routinely used to assess gene functions in the intact fish, and zebrafish embryos and larvae are nearly transparent, so noninvasive observation is possible. Because larvae measure less than a few millimeters, they can be accommodated in multiwell plates, such as 384-well formats (Rennekamp and Peterson 2015). Only small quantities of test chemicals are needed, so exposure–response relationships can be evaluated over a broad concentration range and testing can be replicated to increase data confidence.

Although substantial research is going on with adult zebrafish for translational research (Phillips and Westerfield 2014; Pickart and Klee 2014), early zebrafish life stages are particularly well suited for rapid screening. During the first 5 days of life, nearly all gene products and signal-transduction pathways are expressed (Pauli et al. 2012); thus, as in other vertebrates, development is a period of heightened sensitivity to chemical exposure. Early–life-stage zebrafish also express a full battery of phase I and phase II metabolism systems, whose activities are highly similar to those of humans (Goldstone et al. 2010).

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Despite the advantages of incorporating the use of early–life-stage zebrafish as part of a strategy for making risk-based decisions, there are some noteworthy limitations. First, test chemicals typically are added directly to the aqueous media, not unlike cells in culture. However, the routes of exposure over the course of development, which can affect chemical uptake and metabolism, can be quite different. During the first 2 days of embryonic development, the primary route of exposure is passive dermal adsorption. Later in development, the gills and oral routes become available, and circulation plays a major role in chemical distribution. For the varied routes of exposure, there is little understanding of tissue concentrations, and this contributes to the challenges in comparing concentration–response results in zebrafish with dose–response studies in other systems directly.

A related potential limitation is that despite metabolic similarities to other vertebrates, subtle differences in metabolic activity could lead to inaccurate toxicity predictions, particularly if metabolic activation or inactivation is mechanistically important for specific test chemicals. Because the developing embryo constitutes a comprehensive integrated system, all potential molecular initiating events are operational during testing. Thus, zebrafish are uniquely sensitive to chemical contaminants present in test solutions in that a contaminant could act on biological targets and disrupt critical molecular events. Finally, as with any animal model, the primary sequences of individual pathway components are not necessarily highly conserved. For example, the zebrafish cyclin-dependent kinase 20 (cdc20) protein is 75% identical with the human protein at the amino acid level, and the zebrafish and human aryl hydrocarbon receptors are only 40% identical. In both cases, the homologous proteins are functionally conserved. Although variable conservation of the genomes is a source for potential discordance between zebrafish and humans, the challenge is not unique to zebrafish inasmuch as individual allelic variations between humans can also result in marked differences in chemical susceptibility.

Computational Structure-Based Models for Predicting Organism-Level Response

It has long been recognized that chemicals that have similar chemical structures can elicit the same or similar toxicological effects and that, paradoxically, almost identical chemicals can cause dissimilar biological responses. The extent to which similar chemicals or their metabolites interact with critical biological molecules, such as target proteins, and operate by similar mechanisms is a critical element in determining structure–activity relationships. The last decade has seen advances in the development of structure-based computational methods to predict human health effects. Some are computational expert systems that consider structural alerts and underlying mechanisms, others are QSAR models that rely on statistical correlations with molecular fragments, and still others are hybrids of these. Many advances have been supported by large curated databases and increased computational power. Health effects addressed include carcinogenicity (Contrera et al. 2005; Valerio et al. 2007), hepatotoxicity (Greene et al. 2010; Hewitt et al. 2013), reproductive and developmental effects (Matthews et al. 2007; Wu et al. 2013), and skin sensitization (Roberts et al. 2007a,b; Alves et al. 2015).

The structure-based computational models that are probably the most advanced in model performance and regulatory acceptance are QSAR models for genotoxicity or more specifically for mutagenicity as measured in the Ames assay, a reverse-mutation bacterial assay that is commonly used to evaluate the potential of chemicals to induce point mutations. The development of those models has benefited from the quantity and structural diversity of data available in the public domain on chemicals that have been tested in the Ames assay. As a result of performance, computational models are being accepted as surrogates for actual testing and have recently been incorporated into international guidelines for assessing mutagenic impurities in pharmaceuticals to limit potential carcinogenic risk (ICH 2014). Computational approaches for other human health effects are being considered for use in a regulatory setting (Kruhlak et al. 2012), and the Organisation for Economic Co-operation and Development has published guidance that outlines the needed components of a QSAR model in regulatory settings (OECD 2004). They include “a defined end point; an unambiguous algorithm; a defined domain of applicability; appropriate measures of goodness of fit, robustness, and predictivity; and, if possible, a mechanistic interpretation” (Gavaghan 2007).

The lack of wide use of QSAR models for end points other than mutagenicity might reflect predictive performance that falls short of that required for practical applications. Most approaches predict only whether a chemical will cause the adverse effect. The inability to predict a plasma concentration that would be expected to elicit toxicity ultimately limits utility for differentiating between closely related structures on which little or no safety information is available for comparison.

Read-Across Predictions

Read-across is a process that uses two-dimensional chemical-structure information to identify chemicals (analogues) that have been well studied toxicologically that are then used to predict the toxicity of a similar chemical that has inadequate toxicological data or to group chemicals for the purpose of evaluating their toxicity collectively. Structural similarity can be determined by atom-by-atom matching that results, for example, in a chemical-similarity score or by identifying core molecular structures or

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

functional groups that are thought to be important in conferring toxicity potential. There should also be a consideration of physicochemical similarity among analogues because significant differences in, for example, partition coefficients (such as logKOW, a measure of lipophilicity) will have important effects on pharmacokinetic and often pharmacodynamic behavior of a chemical. Read-across approaches are receiving much attention because they can help to satisfy the information requirements under European Union Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) regulations; the general concept has been accepted by the European Chemicals Agency (ECHA) and member-state authorities (Patlewicz et al. 2013). When robust toxicological data are available on one or more structurally related chemicals, they can be used to infer the activity of a chemical that has not been adequately tested. ECHA (2015) has recently published a framework by which it evaluates read-across submissions under REACH. ECHA’s framework groups the read-across into six categories according to such factors as whether the read-across is for a single analogue or an entire category, whether it is based on metabolism to a common product, and the relative potencies of members of a chemical series.

Phthalate esters provide a well-studied example of the utility of read-across for male reproductive toxicity. Phthalate esters that have chain lengths of four to six carbons (more if branched) cause testicular toxicity (Foster et al. 1980) and adverse effects on male reproductive-system development (Gray et al. 2000; NRC 2008) in rats. Studies of global gene expression in the fetal rat testis show comparable effects of all the developmentally toxic phthalates (Liu et al. 2005) and support a conclusion that these chemicals act via the same mechanism. Phthalate esters with shorter chains, such as dimethyl and diethyl phthalate, do not produce similar effects on gene expression or on testicular function or male reproductive-system development. Thus, well-studied phthalate esters in this group would serve as anchor chemicals for other phthalates that have chains of four to six carbons in a read-across approach.

Read-across can be problematic, and caution is needed before its conclusions are relied on heavily. For example, thalidomide has two stereoisomers, (S)-thalidomide and (R)-thalidomide, that are virtually identical from a structural perspective in all aspects except for the 3-D orientation of the two ring systems in relation to one another (see Figure 3-7). Their physical characteristics are also identical, so read-across analysis might conclude that the chemicals will have similar or identical safety profiles. However, (S)-thalidomide causes birth defects, embryo death or altered development, growth retardation, and functional defects, whereas (R)-thalidomide does not. Still, the enantiomers are capable of interconverting in vivo, so it is impossible to eliminate the teratogenic effects by administering only the (R)-enantiomer.

Despite the limitations, read-across remains a screening approach for assessing the safety of a molecule in the absence of data on which to base an assessment. The 2015 ECHA framework provides guidance on how protein binding, metabolism, and other data can be used in read-across analyses and potentially overcome the limitations. Furthermore, a recent European study team proposed evaluation of read-across for four basic chemical-group scenarios (Berggren et al. 2015): chemicals that do not undergo metabolism to exert toxicity, that exert their toxicity through the same or structurally similar metabolites, that have low toxicity, or that are structurally similar but have variable toxicity on the basis of their hypothesized mechanism. They have selected chemical groups for case studies in each of the four categories.

Low et al. (2013) extended the concept of similarity in read-across from chemical structure to bioactivity, specifically responses in a variety of in vitro and genomic assays. They proposed a hazard classification and visualization method that draws on both chemical structure and biological features to establish similarity among chemicals in read-across. The approach incorporates mechanistic data to increase the confidence of read-across.

In addition to serving as a screening approach, read-across can be regarded as a hypothesis-generating exercise. The hypotheses can be lumped into two broad cat-

Image
FIGURE 3-7 Molecular structures of (S)-thalidomide and (R)-thalidomide.
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

egories: the new chemical is metabolized to a chemical that has already been tested (or it and its analogue are metabolized to the same chemical), or the new chemical and its analogues are sufficiently similar in chemical structure and properties that their biological activity is the same (that is, they have the same mechanism). In the former case, there are long-standing methods for assessing chemical metabolism that can be applied to support or refute the hypothesis that the new chemical is metabolized to something that has already been tested. In the latter case, if the mechanism of the analogous chemicals is known, it is reasonably straightforward to test for effects on the initial events of the mechanism (for example, receptor occupancy or enzyme inhibition). In most cases, however, mechanisms are not known; in such cases, it is still possible to compare the responses of the chemical and its analogues in screening systems that globally assess toxicological responses. Global gene-expression analysis is likely to provide universal coverage of possible mechanisms. Gene expression in an animal model in which the target tissues (for the tested analogues) are known or in an in vitro system that represents the target tissue is a reasonable way to test the hypothesis of a comparable mechanism among analogues. It still might be possible to use gene expression in in vitro models to identify a mechanism when target tissue is not known, but it will probably require testing in more than one cell type. Lamb et al. (2006) evaluated the gene-expression changes elicited in four cell types by a large number of drugs; they clearly showed the connections between agents that have the same pharmacological action and demonstrated that this approach has high potential for toxicology. High-throughput screening batteries, such as ToxCast, might also have utility for that purpose, but it will need to be determined whether the current battery covers the universe of known toxicity mechanisms. Higher-order models, such as organ-on-a-chip or zebrafish, might also be used for testing hypotheses of biological similarity if it can be shown that these models have the biological machinery that is critical for the mechanism in question. As data streams are added more systematically to the read-across process, integrated approaches, such as Bayesian models, that provide for a more agnostic evaluation and promote consistency in output could be developed. Figure 3-8 illustrates several scenarios for read-across and how it can be used to infer hazard and dose–response relationships.

INCORPORATING DATA STREAMS

Various chemicals will have multiple data streams along the exposure-to-outcome continuum that can be used to characterize hazard or risk. For example, pharmacokinetic studies might point to tissues that have particularly high concentrations of a chemical that are potentially increased by active transport as indicated in in vitro studies. Cell-free assays might suggest a set of key receptors, with cell-response assays indicating response; the results, when considered in the context of high concentrations of a chemical in tissues, might indicate particular hazards, such as particular cancers or reproductive toxicity. Targeted studies might show early markers of effect histopathologically, and gene expression in the studies might show consistency with the findings of cell-based assays. The results might be supported by findings on similar chemicals that predict the activity through structure–activity analyses. Robust assessments will identify the more influential data streams with which to develop an integrated assessment. Some streams will be more information-rich than others. The integration of multiple data streams is discussed further in Chapter 7.

CHALLENGES AND RECOMMENDATIONS FOR ADVANCING TOXICOLOGY

This chapter shows how emerging scientific tools generate toxicological evidence on hazard and dose–response relationships of chemicals and other risk issues. It emphasizes how the tools apply to different components in the exposure-to-outcome continuum. Some tools, such as PBPK and systems-biology models, provide a basis for linking components along the continuum. Others, such as high-throughput assays or targeted testing, provide a direct readout of chemical effect within a single component or in multiple components. The tools vary in their maturity for application, their scope of applicability among chemical classes, and the questions that they can address. The committee emphasizes that the level of performance required for the various tools will depend on the question that is being addressed (context) and on agency policies.

There are specific technical and research challenges. Some have been mentioned in preceding sections of this chapter; the challenges related to molecular and cell-based assays are particularly notable. Some important challenges in advancing the tools for risk-assessment application are described below, and some recommendations are offered.

Advancing the New Testing Paradigm

Challenge: Obtaining the vision described in the Tox21 report in which traditional whole-animal testing is replaced with a broad toxicity-testing strategy that uses primarily in vitro assays, computational methods, and targeted animal testing for assessing the biological activity of chemicals is a complex and labor-intensive task that requires focus, commitment, and resources (NRC 2007). The strategy for achieving the vision involves research to understand the spectrum of perturbations that could result in human toxicity and the nature and extent of the toxicity caused by the perturbations and research to understand

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Image
FIGURE 3-8 Scenarios for conducting read-across.

how determinants of human variability (for example, underlying nutritional, genetic, or disease state or life stage) and exposure duration might affect biological responses or toxicity. The scientific community needs to recognize that the current approach to toxicity testing and data analysis is often compartmentalized, and this prevents a holistic approach in trying to determine toxicity of chemical exposure.

Recommendation: Broad consideration of research that is needed to advance the development of a suite of tests that essentially achieves the vision in the Tox21 report is beyond the present committee’s charge, but the committee notes that the research described above in the challenge statement should have high priority so that the vision can be achieved. The committee expresses its concurrence with the Tox21 committee and emphasizes that testing should not be limited to the goal of one-to-one replacement but rather should extend toward development of the most salient and predictive assays for the end point or disease being considered.

Optimizing Tools to Probe Biological Response

Challenge: Developing a comprehensive in vitro system that covers the important biological responses to chemical exposure that contribute to human adverse health effects is a considerable challenge. Most assays used in the ToxCast program were developed to meet the needs of the pharmaceutical industry and were not designed to cover the full array of biological response, given the extensive testing in whole animals and humans that is conducted for drug development. Thus, not all major forms of toxicity are captured in the current assays, and correlating tested activities with toxicity-hazard traits has been limited. For example, few or no ToxCast or Tox21 assays test for several of the key characteristics of carcinogenesis (Smith et al. 2016). There is also the question of how short-term assay exposures are related to chronic exposure or developmental exposures in vivo. Responses that depend on higher levels of biological complexity could be missed by cell-based assays. A number of issues for assay development acknowledged in NRC (2007) remain, including coverage of the necessary biological space to ensure that human sensitivity and susceptibility to toxicants are adequately captured.

Recommendation: Whole-animal testing should move beyond standard approaches, including those associated with experimental design and statistical methods, to maximize their utility. An array of whole-animal tools are now available, and their adoption could address knowledge gaps in risk assessment more comprehensively and begin to address the breadth of genetic sensitivity in response to chemical exposure and other contributors to human variability in response. Guidance for incorporating these whole-animal tools into risk assessment would likely speed their adoption and use.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Recommendation: Use of targeted rodent tests that incorporate the use of -omics technologies, such as sentinel-tissue transcriptomics, should be encouraged. The experimental design should include strategies for data interpretation and analysis, such as Bayesian approaches, that are specifically developed for these studies. Strategic whole-animal testing could help to identify the broader suite of pathways that are beyond the scope of current molecular and cell-based tests, guide the development of in vitro assays that could enhance confidence in extrapolating from in vitro tests to whole-animal responses, and provide a stronger basis of hazard identification and dose–response assessment.

Recommendation: Tools for probing genomic, epigenetic, transcriptomic, proteomic, and metabolomic changes in cells should be advanced because they provide an opportunity to assess cellular changes in a nontargeted and non–pathway-specific manner. Because virtually all toxicity is accompanied by specific changes in gene expression (and presumably changes in protein expression and metabolic profile), continued exploration of these in vivo and in vitro approaches as standalone screens or as complements to in vitro screens might be a way to cover more biological space.5

Understanding and Addressing Limitations of Cell Systems

Challenge: Substantial progress has been made in developing and adapting a wide array of assays for screening environmental chemicals, but cell cultures have several important limitations. There are challenges in incorporating metabolic capacity into the assays to ensure that assay conditions generate chemical exposures that are representative of the exposures in humans that could lead to toxicity. Cell cultures also tend to be extremely sensitive to environmental conditions; changes in microenvironments can alter cellular phenotypes and responses and result in skewed results of toxicity screens. Furthermore, conventional monolayer cultures are less sensitive than 3-D cultures, and the response obtained from an in vitro assay can depend on the cell type that is used—a liver cell versus a neuron or a primary cell versus an immortalized cell. Current in vitro assays evaluate only chemicals that have particular properties; chemicals typically must be soluble in dimethyl sulfoxide, have low volatility, meet molecular-weight cutoffs, and be available in high enough quantity and purity.

Recommendation: Formalized approaches should be developed to characterize the metabolic competence of assays, to determine for which assays metabolic competence is not an essential consideration, and to account for the toxicity of metabolites appropriately. Approaches could include the development and application of better in silico methods for predicting metabolism and elimination and the development of methods for including metabolic capability without compromising other aspects of assay performance. Federal agencies have initiated some research to address the metabolic-capacity issue, and the committee recommends that the research have high priority.

Recommendation: Research should be conducted to understand the breadth of cell types needed to capture toxicity that might occur only with specific cell lines. It is possible to identify common pathways of toxicity that exist in all cell types, but biology specific to cell types could be of great use in identifying organ-specific toxicities.

Recommendation: Cell batches—even those from established cell lines—should be characterized sufficiently before, during, and after experimentation. Genetic variability, phenotypic characteristics, and purity should be reported in published literature or on publicly accessible Web sites or interfaces.

Recommendation: Assay development should be coordinated with development of computational models of cellular responses involved in pathway perturbations to promote deeper understanding of shapes of dose–response curves at the cellular level.

Addressing the Whole Human and the Human Population

Challenge: The exposure-to-outcome continuum in reality can be complex. Chemicals can perturb multiple pathways and lead to various forms of toxicity. Furthermore, toxicity can be influenced by genetics, diet, lifestyle choices, social factors, sex, life stage, health status, and past and present exposures. All those factors can influence responses at different points in the exposure-to-outcome continuum and occur in the exposure milieu and context of human experience.

Recommendation: Efforts to capture human variability better in in vitro and in vivo toxicity tests should be explored. Broader testing of multiple cell lines from diverse human populations could find idiosyncratic sensitivity of some populations, as has been seen in in vivo testing of panels of isogenic mouse strains, although this approach addresses only variability due to genetic factors for a single upstream end point. Approaches for better characterization of the variety of possible responses to chemicals in food, drugs, or the environment are needed. Experi-

___________________

5 If in vitro methods are used for this purpose, it will be important to identify the minimum number of cell types necessary for full coverage. Identifying the cell types will require a combination of statistical approaches that retrospectively analyze the available transcriptomic data and prospective experimentation to determine the number of cell types that are responsive to a broad array of mechanisms. High-content imaging techniques that capture effects on multiple cellular-toxicity indicators simultaneously—including mitochondrial integrity, cell viability, lipid accumulation, cytoskeletal integrity, and formation of reactive oxygen species (Grimm et al. 2015)—can also be used for nontargeted screening and offer the potential to integrate multiple aspects of cell function.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

mental approaches could be coupled with computational approaches for better characterization.

Recommendation: Relatively low-cost, rapid molecular and cellular assays should be used to investigate the toxicity of chemical mixtures. Furthermore, humans are not exposed to single chemicals in isolation but instead are constantly exposed to myriad chemicals in their environment, endogenous chemicals produced in the body or modulated as a consequence of social and behavioral factors, and complex chemical mixtures. Cell-based assays can be used to explore at the molecular and pathway level how the addition of a chemical exposure to existing exogenous and endogenous exposures might contribute to risk.

REFERENCES

Abdo, N., M. Xia, C.C. Brown, O. Kosyk, R. Huang, S. Sakamuru, Y.H. Zhou, J.R. Jack, P. Gallins, K. Xia, Y. Li, W.A. Chiu, A.A. Motsinger-Reif, C.P. Austin, R.R. Tice, I. Rusyn, and F.A. Wright. 2015. Population-based in vitro hazard and concentration-response assessment of chemicals: The 1000 genomes high-throughput screening study. Environ. Health Perspect. 123(5):458-466.

Ablain, J., and L.I. Zon. 2013. Of fish and men: Using zebrafish to fight human diseases. Trends Cell. Biol. 23(12):584-586.

Al-Imari, L., and R. Gerlai. 2008. Sight of conspecifics as reward in associative learning in zebrafish (Danio rerio). Behav. Brain. Res. 189(1):216-219.

Alon, U. 2007. Network motifs: Theory and experimental approaches. Nat. Rev. Genet. 8(6):450-461.

Alves, V.M., E. Murastov, D. Fourches, J. Strickland, N. Kleinstreuer, C.H. Andrade, and A. Tropsha. 2015. Predicting chemically-induced skin reactions. Part I: QSAR models of skin sensitization and their application to identify potentially hazardous compounds. Toxicol. Appl. Pharmacol. 284(2):262-272.

An, W.F., and N. Tolliday. 2010. Cell-based assays for high-throughput screening. Mol. Biotechnol. 45(2):180-186.

Andersen, M.E., and D. Krewski. 2009. Toxicity testing in the 21st century: Bringing the vision to life. Toxicol. Sci. 107(2):324-330.

Andersen, M.E., K. Betts, Y. Dragan, S. Fitzpatrick, J.L. Goodman, T. Hartung, J. Himmelfarb, D.E. Ingber, A. Jacobs, R. Kavlock, K. Kolaja, J.L. Stevens, D. Tagle, D. Lansing Taylor, and D. Throckmorton. 2014. Developing microphysiological systems for use as regulatory tools—challenges and opportunities. ALTEX 31(3):364-367.

Anson, B.D., K.L. Kolaja, and T.J. Kamp. 2011. Opportunities for use of human iPS cells in predictive toxicology. Clin. Pharmacol. Ther. 89(5):754-758.

Arai, K., R. Sakamoto, D. Kubota, and T. Kondo. 2013. Proteomic approach toward molecular backgrounds of drug resistance of osteosarcoma cells in spheroid culture system. Proteomics 13(15):2351-2360.

Arnaout, R., T. Ferrer, J. Huisken, K. Spitzer, D.Y.R. Stainier, M. Tristani-Firouzi, and N.C. Chi. 2007. Zebrafish model for human long QT syndrome. Proc. Natl. Acad. Sci. US 104(27):11316-11321.

Arnaout, R., S., Reischauer, and D.Y. Stainier. 2014. Recovery of adult zebrafish hearts for high-throughput applications. J. Vis. Exp. 94:e52248.

Asnani, A., and R.T. Peterson. 2014. The zebrafish as a tool to identify novel therapies for human cardiovascular disease. Dis. Model Mech. 7(7):763-767.

Asphahani, F., M. Thein, K. Wang, D. Wood, S.S. Wong, J. Xu, and M. Zhang. 2012. Real-time characterization of cytotoxicity using single-cell impedance monitoring. Analyst. 137(13):3011-3019.

Astashkina, A., B. Mann, and D.W. Grainger. 2012. A critical evaluation of in vitro cell culture models for high-throughput drug screening and toxicity. Pharmacol. Ther. 134(1):82-106.

Beers, J., K.L. Linask, J.A. Chen, L.I. Siniscalchi, Y. Lin, W. Zheng, M. Rao, and G. Chen. 2015. A cost-effective and efficient reprogramming platform for large-scale production of integration-free human induced pluripotent stem cells in chemically defined culture. Sci. Rep. 5:11319.

Bender, A., J. Scheiber, M. Glick, J.W. Davies, K. Azzaoui, J. Hamon, L. Urban, S. Whitebread, and J.L. Jenkins. 2007. Analysis of pharmacology data and the prediction of adverse drug reactions and off-target effects from chemical structure. Chem. Med. Chem. 2(6):861-873.

Bento, A.P., A. Gaulton. A. Hersey, L.J. Bellis, J. Chambers, M. Davies, F.A. Krüger, Y. Light, L. Mark, S. McGlinchey, M. Nowotka, G. Papadatos, R. Santos, and J.P. Overington. 2014. The ChEMBL bioactivity database: An update. Nucleic Acids Res. 42:D1083-D1090.

Berg, E.L., M.A. Polokoff, A. O’Mahony, D. Nguyen, and X. Li. 2015. Elucidating mechanisms of toxicity using phenotypic data from primary human cell systems--a chemical biology approach for thrombosis-related side effects. Int. J. Mol. Sci. 16(1):1008-1029.

Berggren, E., P. Amcoff, R. Benigni, K. Blackburn, E. Carney, M. Cronin, H. Deluyker, F. Gautier, R.S. Judson, G.E. Kass, D. Keller, D. Knight, W. Lilienblum, C. Mahony, I. Rusyn, T. Schultz, M. Schwarz, G. Schüürmann, A. White, J. Burton, A.M. Lostia, S. Munn, and A. Worth. 2015. Chemical safety assessment using read-across: Assessing the use of novel testing methods to strengthen the evidence base for decision making. Environ. Health Perspect. 123(12):1232-1240.

Best, J.D., S. Berghmans, J.J. Hunt, S.C. Clarke, A. Fleming, P. Goldsmith, and A.G. Roach. 2008. Non-associative learning in larval zebrafish. Neuropsychopharmacology 33(5):1206-1215.

Bhogal, N., C. Grindon, R. Combes, and M. Balls. 2005. Toxicity testing: Creating a revolution based on new technologies. Trends Biotechnol. 26(6):299-307.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Birnbaum, L. 2011. Presentation at NIEHS Workshop: Engineered Tissue Models for Environmental Health Science Research, June 27-28, 2011, Washington, DC.

Boverhof, D.R., M.P. Chamberlain, C.R. Elcombe, F.J. Gonzalez, R.H. Heflich, L.G. Hernandez, A.C. Jacobs, D. Jacobson-Kram, M. Luijten, A. Maggi, M.G. Manjanatha, J. Benthem, and B.B. Gollapudi. 2011. Transgenic animal models in toxicology: Historical perspectives and future outlook. Toxicol. Sci. 121(2):207-233.

Bowes, J., A.J. Brown, J. Hamon, W. Jarolimek, A. Sridhar, G. Waldron, and S. Whitebread. 2012. Reducing safety-related drug attrition: The use of in vitro pharmacological profiling. Nat. Rev. Drug Discov. 11(12):909-922.

Bradford, B.U., E.F. Lock, O. Kosyk, S. Kim, T. Uehara, D. Harbourt, M. DeSimone, D.W. Threadgill, V. Tryndyak, I.P. Pogribny, L. Bleyle, D.R. Koop, and I. Rusyn. 2011. Interstrain differences in the liver effects of trichloroethylene in a multistrain panel of inbred mice. Toxicol. Sci. 120(1):206-217.

Braga, R.C., V.M. Alves, M.F. Silva, E. Muratov, D. Fourches, A. Tropsha, and C.H. Andrade. 2014. Tuning hERG out: Antitarget QSAR models for drug development. Curr. Top Med. Chem. 14(11):1399-1415.

Bretaud, S., S. Lee, and S. Guo. 2004. Sensitivity of zebrafish to environmental toxins implicated in Parkinson’s disease. Neurotoxicol. Teratol. 26(6):857-864.

Browne, P., R.S. Judson, W.M. Casey, N.C. Kleinstreuer, and R.S. Thomas. 2015. Screening chemicals for estrogen receptor bioactivity using a computational model. Environ. Sci. Technol. 49(14):8804-8814.

Bulysheva, A.A., G.L. Bowlin, S.P. Petrova, and W.A. Yeudall. 2013. Enhanced chemoresistance of squamous carcinoma cells grown in 3D cryogenic electrospun scaffolds. Biomed. Mater. 8(5):055009.

Champagne, D.L., C.C. Hoefnagels, R.E. de Kloet, and M.K. Richardson. 2010. Translating rodent behavioral repertoire to zebrafish (Danio rerio): Relevance for stress research. Behav. Brain Res. 214(2):332-342.

Chanut, F., C. Kimbrough, R. Hailey, B. Berridge, A. Hughes-Earle, R. Davies, K. Roland, A. Stokes, A. Casartelli, M. York, H. Jordan, F. Crivellente, P. Cristofori, H. Thomas, J. Klapwijk, and R. Adler. 2013. Spontaneous cardiomyopathy in young Sprague-Dawley rats: Evaluation of biological and environmental variability. Toxicol. Pathol. 41(8):1126-1136.

Chapman, A.L., E.J. Bennett, T.M. Ramesh, K.J. De Vos, and A.J. Grierson. 2013. Axonal transport defects in a mitofusin 2 loss of function model of Charcot-Marie-Tooth disease in zebrafish. PLoS One 8(6):e67276.

Chen, J., S.R. Das, J. La Du, M.M. Corvi, C. Bai, Y. Chen, X. Liu, G. Zhu, R.L. Tanguay, Q. Dong, and C. Huang. 2013. Chronic PFOS exposures induce life stage-specific behavioral deficits in adult zebrafish and produce malformation and behavioral deficits in F1 offspring. Environ. Toxicol. Chem. 32(1):201-206.

Chen, J., J. Wang, Y. Zhang, D. Chen, C. Yang, C. Kai, X. Wang, F. Shi, and J. Dou. 2014. Observation of ovarian cancer stem cell behavior and investigation of potential mechanisms of drug resistance in three-dimensional cell culture. J. Biosci. Bioeng. 118(2):214-222.

Cheng, F., Y. Zhou, J. Li, W. Li, G. Liu, and Y. Tang. 2012. Prediction of chemical-protein interactions: MultitargetQSAR versus computational chemogenomic methods. Mol. Biosyst. 8(9):2373-2384.

Chico, T.J., P.W. Ingham, and D.C. Crossman. 2008. Modeling cardiovascular disease in the zebrafish. Trends Cardiovasc. Med. 18(4):150-155.

Chiu, W.A., J. Jinot, C.S. Scott, S.L. Makris, G.S. Cooper, R.C. Dzubow, A.S. Bale, M.V. Evans, K.Z. Guyton, N. Keshava, J.C. Lipscomb, S. Barone Jr., J.F. Fox, M.R. Gwinn, J. Schaum, and J.C. Caldwell. 2013. Human health effects of trichloroethylene: Key findings and scientific issues. Environ. Health Perspect. 121(3):303-311.

Churchill, G.A., D.C. Airey, H. Allayee, J.M. Angel, A.D. Attie, J. Beatty, W.D. Beavis, J.K. Belknap, B. Bennett, W. Berrettini, A. Bleich, M. Bogue, K.W. Broman, K.J. Buck, E. Buckler, M. Burmeister, E.J. Chesler, J.M. Cheverud, S. Clapcote, M.N. Cook, R.D. Cox, J.C. Crabbe, W.E. Crusio, A. Darvasi, C.F. Deschepper, R.W. Doerge, C.R. Farber, J. Forejt, D. Gaile, S.J. Garlow, H. Geiger, H. Gershenfeld, T. Gordon, J. Gu, W. Gu, G. de Haan, N.L. Hayes, C. Heller, H. Himmelbauer, R. Hitzemann, K. Hunter, H.C. Hsu, F.A. Iraqi, B. Ivandic, H.J. Jacob, R.C. Jansen, K.J. Jepsen, D.K. Johnson, T.E. Johnson, G. Kempermann, C. Kendziorski, M. Kotb, R.F. Kooy, B. Llamas, F. Lammert, J.M. Lassalle, P.R. Lowenstein, L. Lu, A. Lusis, K.F. Manly, R. Marcucio, D. Matthews, J.F. Medrano, D.R. Miller, G. Mittleman, B.A. Mock, J.S. Mogil, X. Montagutelli, G. Morahan, D.G. Morris, R. Mott, J.H. Nadeau, H. Nagase, R.S. Nowakowski, B.F. O’Hara, A.V. Osadchuk, G.P. Page, B. Paigen, K. Paigen, A.A. Palmer, H.J. Pan, L. Peltonen-Palotie, J. Peirce, D. Pomp, M. Pravenec, D.R. Prows, Z. Qi, R.H. Reeves, J. Roder, G.D. Rosen, E.E. Schadt, L.C. Schalkwyk, Z. Seltzer, K. Shimomura, S. Shou, M.J. Sillanpaa, L.D. Siracusa, H.W. Snoeck, J.L. Spearow, K. Svenson, L.M. Tarantino, D. Threadgill, L.A. Toth, W. Valdar, F.P. de Villena, C. Warden, S. Whatley, R.W. Williams, T. Wiltshire, N. Yi, D. Zhang, M. Zhang, F. Zou, and Complex Trait Consortium. 2004. The collaborative cross, a community resource for the genetic analysis of complex traits. Nat. Genet. 36(11):1133-1137.

Churchill, G.A., D.M. Gatti, S.C. Munger, and K.L. Svenson. 2012. The diversity outbred mouse population. Mamm. Genome 23(9-10):713-718.

Colley, H., D. James, K. Diment, and M. Tedder. 2007. Learning as becoming in vocational education and training: Class, gender and the role of vocational habitus. J. Voc. Educ. Train. 55(4):471-498.

Contrera, J.F., P. MacLaughlin, L.H. Hall, and L.B. Kier. 2005. QSAR modeling of carcinogenic risk using discrim-

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

inant analysis and topological molecular descriptors. Curr. Drug Discov. Technol. 2(2):55-67.

Cote, I., M.E. Andersen, G.T. Ankley, S. Barone, L.S. Birnbaum, K. Boekelheide, F.Y. Bois, L.D. Burgoon, W.A. Chiu, D. Crawford-Brown, K.M. Crofton, M. DeVito, R.B. Devlin, S.W. Edwards, K. Guyton, D. Hattis, R.S. Judson, D. Knight, D. Krewski, J. Lambert, E.A. Maull, D. Mendrick, G.M. Paoli, C.J. Patel, E. Perkins, G. Poje, C.J. Portier, I. Rusyn, P.A. Schulte, A. Simeonov, M.T. Smith, K. Thayer, R.S. Thomas, R. Thomas, R.R. Tice, J.J. Vandenberg, D. Villeneuve, S. Wesselkamper, M. Whelan, C. Whittaker, R. White, M. Xia, C. Yauk, L. Zeise, J. Zhao, and R. DeWoskin. 2016. The next generation of risk assessment multiyear study- highlights of findings, applications to risk assessment and future directions. Environ. Health Perspect. 121(11):1671-1682.

Crosby, E.B., J.M. Bailey, A.N. Oliveri, and E.D. Levin. 2015. Neurobehavioral impairments caused by developmental imidacloprid exposure in zebrafish. Neurotoxicol. Teratol. 49:81-90.

Cui, C., E.L. Benard, Z. Kanwal, O.W. Stockhammer, M. van der Vaart, A. Zakrzewska, H.P. Spaink, and A.H. Meijer. 2011. Infectious disease modeling and innate immune function in zebrafish embryos. Methods Cell Biol.105:273-308.

Da Costa, M.M., C.E. Allen, A. Higginbottom, T. Ramesh, P.J. Shaw, and C.J. McDermott. 2014. A new zebrafish model produced by TILLING of SOD1-related amyotrophic lateral sclerosis replicates key features of the disease and represents a tool for in vivo therapeutic screening. Dis. Model Mech. 7(1):73-81.

Dalgin, G., and V.E. Prince. 2015. Differential levels of Neurod establish zebrafish endocrine pancreas cell fates. Dev. Biol. 402(1):81-97.

De Vooght, V., J. A. Vanoirbeek, K. Luyts, S. Haenen, B. Nemery, and P.H. Hoet. 2010. Choice of mouse strain influences the outcome in a mouse model of chemical-induced asthma. PLoS One 5(9):e12581.

Dong, H., W. Xu, J.K. Pillai, C. Packianathan, and B.P. Rosen. 2015. High-throughput screening-compatible assays of As(III) S-adenosylmethionine methyltransferase activity. Anal. Biochem. 480:67-73.

Dooley, K., and L.I. Zon. 2000. Zebrafish: A model system for the study of human disease. Curr. Opin. Genet. Dev. 10(3):252-256.

Dow, L.E., J. Fisher, K.P. O’Rourke, A. Muley, E.R. Kastenhuber, G. Livshits, D.F. Tschaharganeh, N.D. Socci and S.W. Lowe. 2015. Inducible in vivo genome editing with CRISPR-Cas9. Nat. Biotechnol. 33(4):390-394.

ECHA (European Chemicals Agency). 2015. Read-Across Assessment Framework (RAAF) [online]. Available: http://echa.europa.eu/documents/10162/13628/raaf_en.pdf [accessed July 19, 2016].

Edmondson, R., J.J. Broglie, A.F. Adcock, and L. Yang. 2014. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev. Technol. 12(4):207-218.

Efthymiou, A., A. Shaltouki, J.P. Steiner, B. Jha, S.M. Heman-Ackah, A. Swistowski, X. Zeng, M.S. Rao, and N. Malik. 2014. Functional screening assays with neurons generated from pluripotent stem cell-derived neural stem cells. J. Biomol. Screen. 19(1):32-43.

Egan, R.J., C.L. Bergner, P.C. Hart, J.M. Cachat, P.R. Canavello, M.F. Elegante, S.I. Elkhayat, B.K. Bartels, A.K. Tien, D.H. Tien, S. Mohnot, E. Beeson, E. Glasgow, H. Amri, Z. Zukowska, and A.V. Kalueff. 2009. Understanding behavioral and physiological phenotypes of stress and anxiety in zebrafish. Behav. Brain Res. 205(1):38-44.

Engeszer, R.E., L.A. da Barbiano, M.J. Ryan, and D.M. Parichy. 2007. Timing and plasticity of shoaling behaviour in the zebrafish, Danio rerio. Anim. Behav. 74(5):1269-1275.

EPA (US Environmental Protection Agency). 2010. Toxicological Review of Acrylamide (CAS No. 79-06-1). EPA/635/R-07/009F [online]. Available: https://cfpub.epa.gov/ncea/iris/iris_documents/documents/toxreviews/0286tr.pdf [accessed July 19, 2016].

EPA (US Environmental Protection Agency). 2014a. FIFRA Scientific Advisory Panel Minutes No. 2015-01: A Set of Scientific Issues Being Considered by the Environmental Protection Agency Regarding Integrated Endocrine Bioactivity and Exposure-Based Prioritization and Screening, FIFRA Scientific Advisory Panel Meeting, December 2-4, 2014, Arlington, VA [online]. Available: https://www.epa.gov/sites/production/files/2015-06/documents/120214minutes.pdf [accessed July 19, 2016].

EPA (US Environmental Protection Agency). 2014b. Next Generation Risk Assessment: Incorporation of Recent Advances in Molecular, Computational, and Systems Biology. EPA/600/R-14/004. National Center for Environmental Assessment, Office of Research and Development, US Environmental Protection Agency, Washington, DC [online]. Available: https://cfpub.epa.gov/si/si_public_record_report.cfm?dirEntryId=286690 [accessed July 19, 2016].

EPA (US Environmental Protection Agency). 2015. Use of High Throughput Assays and Computational Tools in the Endocrine Disruptor Screening Program-Overview [online]. Available: https://www.epa.gov/endocrine-disruption/use-high-throughput-assays-and-computational-tools-endocrine-disruptor [accessed December 1, 2016].

EPA/NIH/NCATS/NTP (US Environmental Protection Agency, National Institutes of Health, National Center for Advancing Translational Sciences, and National Toxicology Program). 2016. Transform Tox Testing Challenge: Innovating for Metabolism-Challenge Overview [online]. Available: https://www.challenge.gov/wp-content/uploads/2016/09/Transform-Tox-Testing-Challenge-Brief.pdf [accessed October 14, 2016].

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Esch, E.W., A. Bahinski, and D. Huh. 2015. Organs-on-chips at the frontiers of drug discovery. Nat. Rev. Drug Discov. 14:248-260.

EU (European Union). 2015. Mechanism Based Integrated Systems for the Prediction of Drug Induced Liver Injury [online]. Available: http://www.mip-dili.eu/ [accessed July 19, 2016].

Feitsma, H., and E. Cuppen. 2008. Zebrafish as a cancer model. Mol. Cancer Res. 6(5):685-694.

Feliu, N., P. Kohonen, J. Ji, Y. Zhang, H.L. Karlsson, L. Palmberg, A. Nyström, and B. Fadeel. 2015. Next-generation sequencing reveals low-dose effects of cationic dendrimers in primary human bronchial epithelial cells. ACS Nano. 9(1):146-163.

Fernandes, Y., S. Tran, E. Abraham, and R. Gerlai. 2014. Embryonic alcohol exposure impairs associative learning performance in adult zebrafish. Behav. Brain Res. 265:181-187.

Festing, M.F. 2010. Improving toxicity screening and drug development by using genetically defined strains. Methods Mol. Biol. 602:1-21.

Foster, P.M., L.V. Thomas, M.W. Cook, and S.D. Gangolli. 1980. Study of the testicular effects and changes in zinc excretion produced by some n-alkyl phthalates in the rat. Toxicol. Appl. Pharmacol. 54(3):392-398.

French, J.E., D.M. Gatti, D.L. Morgan, G.E. Kissling, K.R. Shockley, G.A. Knudsen, K.G. Shepard, H.C. Price, D. King, K.L. Witt, L.C. Pedersen, S.C. Munger, K.L. Svenson, and G.A. Churchill. 2015. Diversity outbred mice identify population-based exposure thresholds and genetic factors that influence benzene-induced genotoxicity. Environ. Health Perspect. 123(3):237-245.

Gallardo, V.E., G.K. Varshney, M. Lee, S. Bupp, L. Xu, P. Shinn, N.P. Crawford, J. Inglese, and S.M. Burgess. 2015. Phenotype-driven chemical screening in zebrafish for compounds that inhibit collective cell migration identifies multiple pathways potentially involved in metastatic invasion. Dis. Model Mech. 8(6):565-576.

Gavaghan, C. 2007. Practical Considerations in Using QSARs in Pharmaceutical Safety Assessment [online]. Available: http://www.ukqsar.org/slides/ClaireGavaghan_2007.pdf [accessed January 3, 2017].

Gerlach, G., A. Hodgins-Davis, C. Avolio, and C. Schunter. 2008. Kin recognition in zebrafish: A 24-hour window for olfactory imprinting. Proc. Biol. Sci. 275(1647):2165-2170.

Gieseck, R.L., III, N.R. Hannan, R. Bort, N.A. Hanley, R.A. Drake, G.W. Cameron, T.A. Wynn, and L. Vallier. 2014. Maturation of induced pluripotent stem cell derived hepatocytes by 3D-culture. PLoS One 9(1):e86372.

Gieseck, R.L., III, L. Vallier, and N.R. Hannan. 2015. Generation of hepatocytes from pluripotent stem cells for drug screening and developmental modeling. Methods Mol. Biol. 1250:123-142.

Godderis, L., R. Thomas, A.E. Hubbard, A.M. Tabish, P. Hoet, L. Zhang, M.T. Smith, H. Veulemans, and C.M. McHale. 2012. Effect of chemical mutagens and carcinogens on gene expression profiles in human TK6 cells. PLOS One 7(6):e39205.

Goldstone, J.V., A.G. McArthur, A. Kubota, J. Zanette, T. Parente, M.E. Jonsson, D.R. Nelson, and J.J. Stegeman. 2010. Identification and developmental expression of the full complement of cytochrome P450 genes in zebrafish. BMC Genomics 11:643.

Gordon, M.W., F. Yan, X. Zhong, P.B. Mazumder, Z.Y. Xu-Monette, D. Zou, K.H. Young, K.S. Ramos, and Y. Li. 2015. Regulation of p53-targeting microRNAs by polycyclic aromatic hydrocarbons: Implications in the etiology of multiple myeloma. Mol. Carcinog. 54(10):1060-1069.

Graham, J.B., S. Thomas, J. Swarts, A.A. McMillan, M.T. Ferris, M.S. Suthar, P.M. Treuting, R. Ireton, M. Gale, Jr., and J.M. Lund. 2015. Genetic diversity in the collaborative cross model recapitulates human West Nile virus disease outcomes. MBio 6(3):e00493-15.

Gray, L.E., J. Ostby, J. Furr, M. Price, D.N. Veeramacheni, and L. Parks. 2000. Perinatal exposure to the phthalates DEHP, BBP, and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation of the male rat. Toxicol. Sci. 58(2):350-365.

Greene, N., L. Fisk, R.T. Naven, R.R. Note, M.L. Patel, and D.J. Pelletier. 2010. Developing structure-activity relaitonships for the preduction of hepatotoxicity. Chem. Res. Toxicol. 23(7):1215-1222.

Grimm, F.A., Y. Iwara, O. Sirenko, M. Bittner, and I. Rusyn. 2015. High-content assay multiplexing for toxicity screening in induced pluripotent stem cell-derived cardiomyocytes and hepatocytes. Assay Drug Dev. Technol. 13(9):529-546.

Guiro, K., S.A. Patel, S.J. Greco, P. Rameshwar, and T.L. Arinzeh. 2015. Investigating breast cancer cell behavior using tissue engineering scaffolds. PLoS One 10(3):e0118724.

Gut, P., B. Baeza-Raja, O. Andersson, L. Hasenkamp, J. Hsiao, D. Hesselson, K. Akassoglou, E. Verdin, M.D. Hirschey, and D.Y. Stainier. 2013. Whole-organism screening for gluconeogenesis identifies activators of fasting metabolism. Nat. Chem. Biol. 9(2):97-104.

Guyton, K.Z., W.A. Chiu, T.F. Bateson, J. Jinot, C.S. Scott, R.C. Brown, and J.C. Caldwell. 2009. A reexamination of the PPAR-alpha activation mode of action as a basis forassessing human cancer risks of environmental contaminants. Environ. Health Perspect. 117(11):1664-1672.

Harrill, A.H., P.B. Watkins, S. Su, P.K. Ross, D.E. Harbourt, I.M. Stylianou, G.A. Boorman, M.W. Russo, R.S. Sackler, S.C. Harris, P.C. Smith, R. Tennant, M. Bogue, K. Paigen, C. Harris, T. Contractor, T. Wiltshire, I. Rusyn, and D.W. Threadgill. 2009. Mouse population-guided resequencing reveals that variants in CD44 contribute to

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

acetaminophen-induced liver injury in humans. Genome Res. (9):1507-1515.

Harris, P.A., C. Duraiswami, D.T. Fisher, J. Fornwald, S.J. Hoffman, G. Hofmann, M. Jiang, R. Lehr, P.M. McCormick, L. Nickels, B. Schwartz, Z. Wu, G. Zhang, R.W. Marquis, J. Bertin, and P.J. Gough. 2015. High throughput screening identifies ATP-competitive inhibitors of the NLRP1 inflammasome. Bioorg. Med. Chem. Lett. 25(14):2739-2743.

Hewitt, M., S.J. Enoch, J.C. Madden, K.R. Przybylak, and M.T. Cronin. 2013. Hepatotoxicity: A scheme for generation chemical categories for read-across, structural alerts and insights into mechanism(s) of action. Crit. Rev. Toxicol. 43(7):537-558.

Hornung, M.W., M.A. Tapper, J.S. Denny, R.C. Kolanczyk, B.R. Sheedy, P.C. Hartig, H. Aladjov, T.R. Henry, and P.K. Schmieder. 2014. Effects-based chemical category approach for prioritization of low affinity estrogenic chemicals. SAR QSAR Environ. Res. 25(4):289-323.

Hossini, A.M., M. Megges, A. Prigione, B. Lichtner, M.R. Toliat, W. Wruck, F. Schröter, P. Nuernberg, H. Kroll, E. Makrantonaki, C.C. Zouboulis, and J. Adjaye. 2015. Induced pluripotent stem cell-derived neuronal cells from a sporadic Alzheimer’s disease donor as a model for investigating AD-associated gene regulatory networks. BMC Genomics 16:84.

Houck, K.A., D.J. Dix, R.S. Judson, R.J. Kavlock, J. Yang, and E.L. Berg. 2009. Profiling bioactivity of the ToxCast chemical library using BioMAP primary human cell systems. J. Biomol. Screen 14(9):1054-1066.

Howe, D.G., Y.M. Bradford, T. Conlin, A.E. Eagle, D. Fashena, K. Frazer, J. Knight, P. Mani, R. Martin, S.A. Moxon, H. Paddock, C. Pich, S. Ramachandran, B.J. Ruef, L. Ruzicka, K. Schaper, X. Shao, A. Singer, B. Sprunger, C.E. Van Slyke, and M. Westerfield. 2013a. ZFIN, the Zebrafish Model Organism Database: Increased support for mutants and transgenics. Nucleic Acids Res. 41:D854-D860.

Howe, K., M.D. Clark, C.F. Torroja, J. Torrance, C. Berthelot, M. Muffato, J.E. Collins, S. Humphray, K. McLaren, L. Matthews, S. McLaren, I. Sealy, M. Caccamo, C. Church-er, C. Scott, J.C. Barrett, R. Koch, G.J. Rauch, S. White, W. Chow, B. Kilian, L.T. Quintais, J.A. Guerra-Assuncao, Y. Zhou, Y. Gu, J. Yen, J.H. Vogel, T. Eyre, S. Redmond, R. Banerjee, J. Chi, B. Fu, E. Langley, S.F. Maguire, G.K. Laird, D. Lloyd, E. Kenyon, S. Donaldson, H. Sehra, J. Almeida-King, J. Loveland, S. Trevanion, M. Jones, M. Quail, D. Willey, A. Hunt, J. Burton, S. Sims, K. McLay, B. Plumb, J. Davis, C. Clee, K. Oliver, R. Clark, C. Riddle, D. Elliot, G. Threadgold, G. Harden, D. Ware, S. Begum, B. Mortimore, G. Kerry, P. Heath, B. Phillimore, A. Tracey, N. Corby, M. Dunn, C. Johnson, J. Wood, S. Clark, S. Pelan, G. Griffiths, M. Smith, R. Glithero, P. Howden, N. Barker, C. Lloyd, C. Stevens, J. Harley, K. Holt, G. Panagiotidis, J. Lovell, H. Beasley, C. Henderson, D. Gordon, K. Auger, D. Wright, J. Collins, C. Raisen, L. Dyer, K.

Leung, L. Robertson, K. Ambridge, D. Leongamornlert, S. McGuire, R. Gilderthorp, C. Griffiths, D. Manthravadi, S. Nichol, G. Barker, S. Whitehead, M. Kay, J. Brown, C. Murnane, E. Gray, M. Humphries, N. Sycamore, D. Barker, D. Saunders, J. Wallis, A. Babbage, S. Hammond, M. Mashreghi-Mohammadi, L. Barr, S. Martin, P. Wray, A. Ellington, N. Matthews, M. Ellwood, R. Woodmansey, G. Clark, J. Cooper, A. Tromans, D. Grafham, C. Skuce, R. Pandian, R. Andrews, E. Harrison, A. Kimberley, J. Garnett, N. Fosker, R. Hall, P. Garner, D. Kelly, C. Bird, S. Palmer, I. Gehring, A. Berger, C.M. Dooley, Z. Ersan-Urun, C. Eser, H. Geiger, M. Geisler, L. Karotki, A. Kirn, J. Konantz, M. Konantz, M. Oberlander, S. Rudolph-Geiger, M. Teucke, C. Lanz, G. Raddatz, K. Osoegawa, B. Zhu, A. Rapp, S. Widaa, C. Langford, F. Yang, S. C. Schuster, N.P. Carter, J. Harrow, Z. Ning, J. Herrero, S.M. Searle, A. Enright, R. Geisler, R.H. Plasterk, C. Lee, M. Westerfield, P.J. de Jong, L.I. Zon, J.H. Postlethwait, C. Nusslein-Volhard, T.J. Hubbard, H. Roest Crollius, J. Rogers, and D.L. Stemple. 2013b. The zebrafish reference genome sequence and its relationship to the human genome. Nature. 496(7446):498-503.

Huh, D., G.A. Hamilton, and D.E. Ingber. 2011. From 3D cell culture to organs-on-chips. Trends Cell Biol. 21(12):745-754.

IARC (International Agency for Research on Cancer). 2006. Preamble. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Lyon, France: IARC [online]. Available: http://monographs.iarc.fr/ENG/Preamble/CurrentPreamble.pdf [accessed July 19, 2016].

IARC (International Agency for Research on Cancer). 2015a. Diazinon in Some Organophosphate Insecticides and Herbicides. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans Vol. 112 [online]. Available: http://monographs.iarc.fr/ENG/Monographs/vol112/mono112-08.pdf [accessed May 15, 2016].

IARC (International Agency for Research on Cancer). 2015b. Malathion in Some Organophosphate Insecticides and Herbicides. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans Vol. 112 [online]. Available: http://monographs.iarc.fr/ENG/Monographs/vol112/mono112-07.pdf [accessed May 15, 2016].

ICH (International Conference on Harmonization). 2014. ICH Harmonised Tripartite Guideline: Assessment and Control of DNA Reactive (Mutagenic) Impurities in Pharmaceuticals to Limit Potential Carcinogenic Risk. M7: Current Step 4 Version, June 23, 2014. International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use [online]. Available: http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Multidisciplinary/M7/M7_Step_4.pdf [accessed July 22, 2016].

Irons, T.D., R.C. MacPhail, D.L. Hunter, and S. Padilla. 2010. Acute neuroactive drug exposures alter locomotor activity in larval zebrafish. Neurotoxicol. Teratol. 32(1):84-90.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Jones, L.J., and W.H. Norton. 2015. Using zebrafish to uncover the genetic and neural basis of aggression, a frequent comorbid symptom of psychiatric disorders. Behav. Brain Res. 276:171-180.

Kacew, S., and M.F. Festing. 1996. Role of rat strain in the differential sensitivity to pharmaceutical agents and naturally occurring substances. J. Toxicol. Environ. Health 47(1):1-30.

Karakikes, I., M. Ameen, V. Termglinchen, and J.C. Wu. 2015. Human induced pluripotent stem cell-derived cardiomyocytes: Insights into molecular, cellular, and functional phenotypes. Circ. Res. 117(1):80-88.

Keiser, M.J., B.L. Roth, B.N. Armbruster, P. Ernsberger, J.J. Irwin, and B.K. Shoichet. 2007. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 25(2):197-206.

Kettleborough, R.N., E.M. Busch-Nentwich, S.A. Harvey, C.M. Dooley, E. de Bruijn, F. van Eeden, I. Sealy, R.J. White, C. Herd, I.J. Nijman, F. Fenyes, S. Mehroke, C. Scahill, R. Gibbons, N. Wali, S. Caruthers, A. Hall, J. Yen, E. Cuppen, and D.L. Stemple. 2013. A systematic genome-wide analysis of zebrafish protein-coding gene function. Nature 496(7446):494-497.

Kleinstreuer, N.C., J. Yang, E.L. Berg, T.B. Knudsen, A.M. Richard, M.T. Martin, D.M. Reif, R.S. Judson, M. Polokoff, D.J. Dix, R.J. Kavlock, and K.A. Houck. 2014. Phenotypic screening of the ToxCast chemical library to classify toxic and therapeutic mechanisms. Nat. Biotechnol. 32(6):583-591.

Kokel, D., J. Bryan, C. Laggner, R. White, C.Y. Cheung, R. Mateus, D. Healey, S. Kim, A.A. Werdich, S.J. Haggarty, C.A. Macrae, B. Shoichet, and R.T. Peterson. 2010. Rapid behavior-based identification of neuroactive small molecules in the zebrafish. Nat. Chem. Biol. 6(3):231-237.

Kolaja, K. 2014. Stem cells and stem cell-derived tissues and their use in safety assessment. J. Biol. Chem. 289(8):4555-4561.

Krewski, D., M.E. Andersen, E. Mantus, and L. Zeise. 2009. Toxicity testing in the 21st century: Implications for human health risk assessment. Risk Anal. 29(4):474-479.

Krewski, D., M. Westphal, M.E. Andersen, G. Paoli, W. Chiu, M. Al-Zoughool, M.C. Croteau, L. Burgoon, and I. Cote. 2014. A framework for the next generation of risk science. Environ Health Perspect.122(8):796-805.

Kruhlak, N.L., R.D. Benz, H. Zhou, and T.J. Colatsky. 2012. (Q)SAR modeling and safety assessment in regulatory review. Clin. Pharmacol. Ther. 91(3):529-534.

Lamb, J., E.D. Crawford, D. Peck, J.W. Modell, I.C. Blat, M.J. Wrobel, J. Lerner, J.P. Brunet, A. Subramanian, K.N. Ross, M. Reich, H. Hieronymus, G. Wei, S.A. Armstrong, S.J. Haggerty, P.A. Clemons, R. Wei, S.A. Carr, E.S. Lander, and T.R. Golub. 2006. The connectivity map: Using gene-expression signatures to connect small molecules, genes, and disease. Science 313(5795):1929-1935.

Landry, J.P., G. Malovichko, and X.D. Zhu. 2015. High-throughput dose-response measurement using a label-free microarray-in-microplate assay platform. Anal. Chem. 87(11):5640-5648.

Lebold, K.M., C.V. Lohr, C.L. Barton, G.W. Miller, E.M. Labut, R.L.Tanguay, and M.G. Traberet. 2013. Chronic vitamin E deficiency promotes vitamin C deficiency in zebrafish leading to degenerative myopathy and impaired swimming behavior. Comp. Biochem. Phys. C Toxicol. Pharmacol. 157(4):382-389.

Levin, E.D. Z. Bencan, and D.T. Cerutti. 2007. Anxiolytic effects of nicotine in zebrafish. Physiol. Behav. 90(1):54-58.

Liu, J., K. Mansouri, R.S. Judson, M.T. Martin, H. Hong, M. Chen, X. Xu, R.S. Thomas, and I. Shah. 2015. Predicting hepatotoxicity using ToxCast in vitro bioactivity and chemical structure. Chem. Res. Toxicol. 28(4):738-751.

Liu, K., K.P. Lehmann, M. Sar, S.S. Young, and K.W.Gaido. 2005. Gene expression profiling following in utero exposure to phthalate esters reveals new gene targets in the etiology of testicular dysgenesis. Biol. Reprod. 73(1):180-192.

Lounkine, E., M.J. Keiser, S. Whitebread, D. Mikhailov, J. Hamon, J.L. Jenkins, P. Lavan, E.Weber, A.K. Doak, S. Côté, B.K. Shoichet, and L. Urban. 2012. Large-scale prediction and testing of drug activity on side-effect targets. Nature 486(7403):361-367.

Lovik, M. 1997. Mutant and transgenic mice in immunotoxicology: An introduction. Toxicology119(1):65-76.

Lovitt, C.J., T.B. Shelper, and V.M. Avery. 2014. Advanced cell culture techniques for cancer discovery. Biology 3(2):345-367.

Low, Y., A. Sedykh, D. Fourches, A. Golbraikh, M. Whelan, I. Rusyn, and A. Tropsha. 2013. Integrative chemical-biological read-across approach for chemical hazard classification. Chem. Res. Toxicol. 26(8):1199-208.

Mahmood, F., S. Fu, J. Cooke, S.W. Wilson, J.D. Cooper, and C. Russell. 2013. A zebrafish model of CLN2 disease is deficient in tripeptidyl peptidase 1 and displays progressive neurodegeneration accompanied by a reduction in proliferation. Brain 136(Pt. 5):1488-1507.

Malik, N., A.G. Efthymiou, K. Mather, N. Chester, X. Wang, A. Nath, M.S. Rao, and J.P. Steiner. 2014. Compounds with species and cell type specific toxicity identified in a 2000 compound drug screen of neural stem cells and rat mixed cortical neurons. Neurotoxicology 45:192-200.

Mann, D.A. 2015. Human induced pluripotent stem cell-derived hepatocytes for toxicology testing. Expert Opin. Drug Metab. Toxicol. 11(1):1-5.

Mann, K.D., E.R. Turnell, J. Atema, and G. Gerlach. 2003. Kin recognition in juvenile zebrafish (Danio rerio) based on olfactory cues. Biol. Bull. 205(2):224-225.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Martin-Jimenez, R., M. Campanella, and C. Russell. 2015. New zebrafish models of neurodegeneration. Curr. Neurol. Neurosci. 15(6):33.

Matthews, E.J., N.L. Kruhlak, R. Daniel Benz, J. Ivanov, G. Klopman, and J.F. Contrera. 2007. A comprehensive model for reproductive and developmental toxicity hazard identification: II. Construction of QSAR models to predict activities of untested chemicals. Regul. Toxicol. Pharmacol. 47(2):136-155.

Mattis, V.B., C. Tom, S. Akimov, J. Saeedian, M.E. Østergaard, A.L. Southwell, C.N. Doty, L. Ornelas, A. Sahabian, L. Lenaeus, B. Mandefro, D. Sareen, J. Arjomand, M.R. Hayden, C.A. Ross, and C.N. Svendesn. 2015. HD iPSC-derived neural progenitors accumulate in culture and are susceptible to BDNF withdrawal due to glutamate toxicity. Hum. Mol. Genet. 24(11):3257-3271.

McGinnity, D.F., J. Collington, R.P. Austin, and R.J. Riley. 2007. Evaluation of human pharmacokinetics, therapeutic dose and exposure predictions using marketed oral drugs. Curr. Drug Metab. 8(5):463-479.

McKinstry-Wu, A.R., W. Bu, G. Rai, W.A. Lea, B.P. Weiser, D.F. Liang, A. Simeonov, A. Jadhav, D.J. Maloney, and R.G. Eckenhoff. 2015. Discovery of a novel general anesthetic chemotype using high-throughput screening. Anesthesiology 122(2):325-333.

Meeker, N.D., and N.S. Trede. 2008. Immunology and zebrafish: Spawning new models of human disease. Dev. Comp. Immunol. 32(7):745-757.

Mehta, J., E. Rouah-Martin, B. Van Dorst, B. Maes, W. Herrebout, M.L. Scippo, F. Dardenne, R. Blust, and J. Robbens. 2012. Selection and characterization of PCB-binding DNA aptamers. Anal. Chem. 84(3):1669-1676.

Miller, N., and R. Gerlai. 2012. From schooling to shoaling: Patterns of collective motion in zebrafish (Danio rerio). PLoS One 7(11):e48865.

Milo, R., S. Shen-Orr, S. Itzkovitz, N. Kashtan, D. Chklovskii, and U. Alon. 2002. Network motifs: Simple building blocks of complex networks. Science 298(5594):824-827.

Moorman, S.J. 2001. Development of sensory systems in zebrafish (Danio rerio). ILAR J. 42(4):292-298.

Morgan, A.P., and C.E. Welsh. 2015. Informatics resources for the Collaborative Cross and related mouse populations. Mamm. Genome 26(9-10):521-539.

NASEM (National Academies of Sciences, Engineering, and Medicine). 2015. Application of Modern Toxicology Approaches for Predicting Acute Toxicity for Chemical Defense. Washington, DC: The National Academies Press.

NCATS (National Center for Advancing Translational Sciences). 2016. About Tissue Chip. Available: http://ncats.nih.gov/tissuechip/about [accessed July 20, 2016].

Ng, H.W., S.W. Doughty, H. Luo, H. Ye, W. Ge, W. Tong, and H. Hong. 2015. Development and validation of decision forest model for estrogen receptor binding prediction of chemicals using large data sets. Chem. Res. Toxicol. 28(12):2343-2351.

Nguyen, A.T., A. Emelyanov, C.H. Koh, J.M. Spitsbergen, S. Parinov, and Z. Gong. 2012. An inducible kras(V12) transgenic zebrafish model for liver tumorigenesis and chemical drug screening. Dis. Model Mech. 5(1):63-72.

Norton, W.H. 2013. Towards developmental models of psychiatric disorders in zebrafish. Front. Neural Circuits 7:79.

NRC (National Research Council). 2000. Scientific Frontiers in Developmental Toxicology and Risk Assessment. Washington, DC: National Academy Press.

NRC (National Research Council). 2006. Assessing the Human Health Risks of Trichloroethylene: Key Scientific Issues. Washington, DC: The National Academies Press.

NRC (National Research Council). 2007. Toxicity Testing in the 21st Century: A Vision and a Strategy. Washington, DC: The National Academies Press.

NRC (National Research Council). 2008. Phthalates and Cumulative Risk Assessment: The Tasks Ahead. Washington, DC: The National Academies Press.

OECD (Organisation for Economic Co-operation and Development). 2004. The Report from the Expert Group on (Quantitative) Structure- Activity Relationships [QSARs] on the Principles for the Validation of (Q)SARs. ENV/JM/MONO(2004)24. OECD Series on Testing and Assessment No. 49. Paris: OECD [online]. Available: http://www.oecd.org/officialdocuments/publicdisplaydocumentpdf/?cote=env/jm/mono(2004)24&doclanguage=en [accessed July 20, 2016].

Padilla, S., D. Corum, B. Padnos, D.L. Hunter, A. Beam, K.A. Houck, N. Sipes, N. Kleinstreuer, T. Knudsen, D.J. Dix, and D.M. Reif. 2012. Zebrafish developmental screening of the ToxCast Phase I chemical library. Reprod. Toxicol. 33(2):174-187.

Panula, P., V. Sallinen, M. Sundvik, J. Kolehmainen, V. Torkko, A. Tiittula, M. Moshnyakov, and P. Podiasz. 2006. Modulatory neurotransmitter systems and behavior: Towards zebrafish models of neurodegenerative diseases. Zebrafish 3(2):235-247.

Panula, P., C.Y. Chen, M. Priyadarshini, H. Kudo, S. Semenova, M. Sundvik, and V. Sallinen. 2010. The comparative neuroanatomy and neurochemistry of zebrafish CNS systems of relevance to human neuropsychiatric diseases. Neurobiol. Dis. 40(1):46-57.

Papadatos, G., A. Gaulton, A. Hersey, and J.P. Overington. 2015. Activity, assay and target data curation and quality in the ChEMBL database. J. Comput. Aided Mol. Des. 29(9):885-896.

Patlewicz, G., N. Ball, E.D. Booth, E. Hulzebos, E. Zvinavashe, and C. Hennes. 2013. Use of category approaches, read-across and (Q)SAR: General considerations. Regular. Toxicol. Pharmacol. 67(1):1-12.

Pauli, A., E. Valen, M.F. Lin, M. Garber, N.L. Vastenhouw, J.Z. Levin, L. Fan, A. Sandelin, J.L. Rinn, A. Regev, and A.F. Schier. 2012. Systematic identification of long non-

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

coding RNAs expressed during zebrafish embryogenesis. Genome Res. 22(3):577-591.

Pfuhler, S., R. Fautz, G. Ouedraogo, A. Latil, J. Kenny, C. Moore, W. Diembeck, N.J. Hewitt, K. Reisinger, and J. Barroso. 2014. The Cosmetics Europe strategy for animal-free genotoxicity testing: project status up-date. Toxicol. In Vitro 28(1):18-23.

Pham, N., S. Iyer, E. Hackett, B.H. Lock, M. Sandy, L. Zeise, G. Solomon, and M. Marty. 2016. Using ToxCast to explore chemical activities and hazard traits: A case study with ortho-phthalates. Toxicol. Sci. 151(2):286-301.

Phillips, J.B., and M. Westerfield. 2014. Zebrafish models in translational research: Tipping the scales toward advancements in human health. Dis. Model Mech. 7(7):739-743.

Pickart, M.A., and E.W. Klee. 2014. Zebrafish approaches enhance the translational research tackle box. Transl. Res. 163(2):65-78.

Pohjanvirta, R., M. Viluksela, J.T. Tuomisto, M. Unkila, J. Karasinska, M.A. Franc, M. Holowenko, J.V. Giannone, P.A. Harper, J. Tuomisto, and A.B. Okey. 1999. Physicochemical differences in the AH receptors of the most TCDD-susceptible and the most TCDD-resistant rat strains. Toxicol. Appl. Pharmacol. 155(1):82-95.

Preston, M.A., and W.B. Macklin. 2015. Zebrafish as a model to investigate CNS myelination. Glia 63(2):177-193.

Prozialeck, W.C., P.C. Lamar, and S.M. Lynch. 2003. Cadmium alters the localization of N-cadherin, E-cadherin, and beta-catenin in the proximal tubule epithelium. Toxicol. Appl. Pharmacol. 189(3):180-195.

Raoux, M., N. Azorin, C. Colomban, S. Rivoire, T. Merrot, P. Delmas, and M. Crest. 2013. Chemicals inducing acute irritant contact dermatitis mobilize intracellular calcium in human keratinocytes. Toxicol. In Vitro. 27(1):402-408.

Reif, D.M., L. Truong, D. Mandrell, S. Marvel, G. Zhang, and R.L. Tanguay. 2016. High-throughput characterization of chemical-associated embryonic behavioral changes predicts teratogenic outcomes. Arch. Toxicol. 90(6):1459-1470.

Rennekamp, A.J., and R.T. Peterson. 2015. 15 years of zebrafish chemical screening. Curr. Opin. Chem. Biol. 24:58-70.

Roberts, D.W., A.O. Aptula, and G. Patlewicz. 2007a. Electrophilic chemistry related to skin sensitization. Reaction mechanistic applicability domain classification for a published data set of 106 chemicals tested in the mouse local lymph node assay. Chem. Res. Toxicol. 20(1):44-60.

Roberts, D.W., G. Patlewicz, S.D. Dimitrov, L.K. Low, A.O. Aptula, P.S. Kern, G.D. Dimitrova, M.I.H. Comber, R.D. Phillips, J. Niemelä, C. Madsen, E.B. Wedebye, P.T. Bailey, and O.G. Mekenyan. 2007b. TIMES-SS-A mechanistic evaluation of an external validation study using reaction chemistry principles. Chem. Res. Toxicol. 20(9):1321-1330.

Romero, A.C., E. Del Río, E. Vilanova, and M.A. Sogorb. 2015. RNA transcripts for the quantification of differentiation allow marked improvements in the performance of embryonic stem cell test (EST). Toxicol. Lett. 238(3):60-69.

Rotroff, D.M., D.J. Dix, K.A. Houck, T.B. Knudsen, M.T. Martin, K.W. McLaurin, D.M. Reif, K.M. Crofton, A.V. Singh, M. Xia, R. Huang, and R.S. Judson. 2013. Using in vitro high throughput screening assays to identify potential endocrine-disrupting chemicals. Environ. Health Perspect. 121(1):7-14.

Rous, P., and F.S. Jones. 1916. A method for obtaining suspensions of living cells from the fixed tissues, and for the plating out of individual cells. J. Exp. Med. 23(4):549-555.

Schlegel, A., and P. Gut. 2015. Metabolic insights from zebrafish genetics, physiology, and chemical biology. Cell. Mol. Life Sci. 72(12):2249-2260.

Scott, C.W., M.F. Peters, and Y.P. Dragan. 2013. Human induced pluripotent stem cells and their use in drug discovery for toxicity testing. Toxicol. Lett. 219(1):49-58.

Shah, I., and J. Wambaugh. 2010. Virtual tissues in toxicology. J. Toxicol. Environ. Health B. Crit. Rev. 13(2-4):314-328.

Sharma, P., D.M. Ando, A. Daub, J.A. Kaye, and S. Finkbeiner. 2012. High-throughput screening in primary neurons. Methods Enzymol. 506:331-360.

Shirai, T., A. Nakamura, S. Fukushima, A. Yamamoto, M. Tada, and N. Ito. 1990. Different carcinogenic responses in a variety of organs, including the prostate, of five different rat strains given 3,2’-dimethyl-4-aminobiphenyl. Carcinogenesis 11(5):793-797.

Silva, M., N. Pham, C. Lewis, S. Iyer, E. Kwok, G. Solomon, and L. Zeise. 2015. A comparison of ToxCast test results with In vivo and other In vitro endpoints for neuro, endocrine, and developmental toxicities: A case study using endosulfan and methidathion. Birth Defects Res. B Dev. Reprod. Toxicol. 104(2):71-89.

Simmons, S.O., C.Y. Fan, and R. Ramabhadran. 2009. Cellular stress response pathway system as a sentinel ensemble in toxicological screening. Toxicol. Sci. 111(2):202-225.

Sinnecker, D., K.L. Laugwitz, and A. Moretti. 2014. Induced pluripotent stem cell-derived cardiomyocytes for drug development and toxicity testing. Pharmacol. Ther. 143(2):246-252.

Sipes, N.S., M.T. Martin, P. Kothiya, D.M. Reif, R.S. Judson, A.M. Richard, K.A. Houck, D.J. Dix, R.J. Kavlock, and T.B. Knudsen. 2013. Profiling 976 ToxCast chemicals across 331 enzymatic and receptor signaling assays. Chem. Res. Toxicol. 26(6):878-895.

Sirenko, O., J. Hesley, I. Rusyn, and E.F. Cromwell. 2014a. High-content high-throughput assays for characterizing the viability and morphology of human iPSC-derived neuronal cultures. Assay Drug Dev. Technol. 12(9-10):536-547.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Sirenko, O., J. Hesley, I. Rusyn, and E. Cromwell. 2014b. High-content assays for hepatotoxicity using induced pluripotent stem cell (iPSC)-derived cells. Assay Drug Dev. Technol. 12(1):43-54.

Sjogren, A.K., M. Liljevald, B. Glinghammar, J. Sagemark, X.Q. Li, A. Jonebring, I. Cotgreave, G. Brolén, and T.B. Andersson. 2014. Critical differences in toxicity mechanisms in induced pluripotent stem cell-derived hepatocytes, hepatic cell lines and primary hepatocytes. Arch. Toxicol. 88(7):1427-1437.

Sledge, D., J. Yen, T. Morton, L. Dishaw, A. Petro, S. Donerly, E. Linney, and E.D. Levin. 2011. Critical duration of exposure for developmental chlorpyrifos-induced neurobehavioral toxicity. Neurotoxicol. Teratol. 33(6):742-751.

Smith, A.J., M.K. Hancock, K. Bi, J. Andrews, P. Harrison, and T.J. Vaughan. 2012. Feasibility of implementing cell-based pathway reporter assays in early high-throughput screening assay cascades for antibody drug discovery. J. Biomol. Screen. 17(6):713-726.

Smith, M.T., K.Z. Guyton, C.F. Gibbons, J.M. Fritz, C.J. Portier, I. Rusyn, D.M. DeMarini, J.C. Caldwell, R.J. Kavlock, P. Lambert, S.S. Hecht, J.R. Bucher, B.W. Stewart, R. Baan, V.J. Cogliano, and K. Straif. 2016. Key characteristics of carcinogens as a basis for organizing data on mechanisms of carcinogenesis. Environ Health Perspect. 124(6):713-721.

Soldatow, V.Y., E.L. Lecluyse, L.G. Griffith, and I. Rusyn. 2013. In vitro models for liver toxicity testing. Toxicol. Res. (Camb). 2(1):23-39.

Song, Y., V. Madahar, and J. Liao. 2011. Development of FRET assay into quantitative and high-throughput screening technology platforms for protein-protein interactions. Ann. Biomed. Eng. 39(4):1224-1234.

Spence, R., and C. Smith. 2005. Male territoriality mediates density and sex ratio effects on oviposition in the zebrafish. Anim. Behav. 69(6):1317-1323.

Steenbergen, P. J., M.K. Richardson, and D.L. Champagne. 2011. The use of the zebrafish model in stress research. Prog. Neuropsychopharmacol. Biol. Psychiatry 35(6):1432-1451.

Sung, J.H., and M.L. Shuler. 2010. In vitro microscale systems for systematic drug toxicity study. Bioprocess Biosyst. Eng. 33(1):5-19.

Swat, M., and J.A. Glazier. 2013. Agent-based virtual-tissue simulations. Biomed. Comput. Rev. (Fall):28-29.

Takahashi, K., K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka, K. Tomoda, and S. Yamanaka. 2007. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131(5):861-872.

Theunissen, P.T., J.F. Robinson, J.L. Pennings, M.H. van Herwijnen, J.C. Kleinjans, and A.H. Piersma. 2012. Compound-specific effects of diverse neurodevelopmental toxicants on global gene expression in the neural embryonic stem cell test (ESTn). Toxicol. Appl. Pharmacol. 262(3):330-340.

Thon, J.N., M.T. Devine, A. Jurak Bgeonja, J. Tibbitts, and J.E. Italiano Jr. 2012. High-content live-cell imaging assay used to establish mechanism of trastuzumab emtansine (TDM1)–mediated inhibition of platelet production. Blood 120(10):1975-1984.

Threadgill, D.W., and G.A. Churchill. 2012. Ten years of the Collaborative Cross. Genetics 190(2):291-294.

Tonk, E.C., J.F. Robinson, A. Verhoef, P.T. Theunissen, J.L. Pennings, and A.H. Piersma. 2013. Valproic acid-induced gene expression responses in rat whole embryo culture and comparison across in vitro developmental and non-developmental models. Reprod. Toxicol. 41:57-66.

Tropepe, V., and H.L. Sive. 2003. Can zebrafish be used as a model to study the neurodevelopmental causes of autism? Genes Brain. Behav. 2(5):268-281.

Truong, L., K.S. Saili, J.M. Miller, J.E. Hutchison, and R.L. Tanguay. 2012. Persistent adult zebrafish behavioral deficits results from acute embryonic exposure to gold nanoparticles. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 155(2):269-274.

Truong, L., D.M. Reif, L. St Mary, M.C. Geier, H.D. Truong, and R.L. Tanguay. 2014. Multidimensional in vivo hazard assessment using zebrafish. Toxicol. Sci. 137(1):212-233.

Tse, A.C., K.Y. Lau, W. Ge, and R.S. Wu. 2013. A rapid screening test for endocrine disrupting chemicals using primary cell culture of the marine medaka. Aquat. Toxicol. 144-145:50-58.

Tyson, J.J., and B. Novák. 2010. Functional motifs in biochemical reaction networks. Annu. Rev. Phys. Chem. 61:219-240.

Valdivia, P., M. Martin, W.R. LeFew, J. Ross, K.A. Houck, and T.J. Shafer. 2014. Multi-well microelectrode array recordings detect neuroactivity of ToxCast compounds. Neurotoxicology 44:204-217.

Valerio, L.G., K.B. Arvidson, R.F. Chanderbhan, and J.F. Contrera. 2007. Prediction of rodent carcinogenic potential of naturally occurring chemicals in the human diet using high-throughput QSAR predictive modeling. Toxicol. Appl. Pharmacol. 222(1):1-16.

Van Vliet, E. 2011. Current standing and future prospects for the technologies proposed to transform toxicity testing in the 21st century. ALTEX 28(1):17-44.

Varani, J., P. Perone, D.M. Spahlinger, L.M. Singer, K.L. Diegel, W.F. Bobrowski, and R. Dunstan. 2007. Human skin in organ culture and human skin cells (keratinocytes and fibroblasts) in monolayer culture for assessment of chemically induced skin damage. Toxicol. Pathol. 35(5):693-701.

Walcott, B.P., and R.T. Peterson. 2014. Zebrafish models of cerebrovascular disease. J. Cereb. Blood Flow Metab. 34(4):571-577.

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×

Wambaugh, J., and I. Shah. 2010. Simulating microdosimetry in a virtual hepatic lobule. PLoS Comput. Biol. 6(4):e1000756.

Wang, J.D., N.J. Douville, S. Takayama, and M. El Sayed. 2012. Quantitative analysis of molecular absorption into PDMS microfluidic channels. Ann. Biomed. Eng. 40(9):1862-1873.

Wheeler, H.E., C. Wing, S.M. Delaney, M. Komatsu, and M.E. Dolan. 2015. Modeling chemotherapeutic neurotoxicity with human induced pluripotent stem cell-derived neuronal cells. PLoS One 10(2):e0118020.

Wilcox, K.C., M.R. Marunde, A. Das, P.T. Velasco, B.D. Kuhns, M.T. Marty, H. Jiang, C.H. Luan, S.G. Sligar, and W.L. Klein. 2015. Nanoscale synaptic membrane mimetic allows unbiased high throughput screen that targets binding sites for Alzheimer’s-associated Aβ oligomers. PLoS One 10(4):e0125263.

Williams, K.E., G.A. Lemieux, M.E. Hassis, A.B. Olshen, S.J. Fisher, and Z. Werb. 2016. Quantitative proteomic analyses of mammary organoids reveals distinct signatures after exposure to environmental chemicals. Proc. Natl. Acad. Sci. US 113(10):E1343-E1351.

Wills, L.P., G.C. Beeson, D.B. Hoover, R.G. Schnellmann, and C.C. Beeson. 2015. Assessment of ToxCast Phase II for mitochondrial liabilities using a high-throughput-respirometric assay. Toxicol Sci. 146(2):226-234.

Wu, S., J. Fisher, J. Naciff, M. Laufersweiler, C. Lester, G. Daston, and K. Blackburn. 2013. Framework for identifying chemicals with structural features associated with the potential act as developmental or reproductive toxicants. Chem. Res. Toxicol. 26(12):1840-1861.

Xia, W., Y.J. Wan, X. Wang, Y.Y. Li, W.J. Yang, C.X. Wang, and S.Q. Xu. 2011. Sensitive bioassay for detection of PPARα potentially hazardous ligands with gold nanoparticle probe. J. Hazard Mater. 192(3):1148-1154.

Xu, J.J., P.V. Henstock, M.C. Dunn, A.R. Smith, J.R. Chabot, and D. de Graaf. 2008. Cellular imaging predictions of clinical drug-induced liver injury. Toxicol. Sci. 105(1):97-105.

Yamasaki, K., S. Kawasaki, R.D. Young, H. Fukuoka, H. Tanioka, M. Nakatsukasa, A.J. Quantock, and S. Kinoshita. 2007. Genomic aberrations and cellular heterogeneity in SV40-immortalized human corneal epithelial cells. Invest. Ophthalmol. Vis. Sci. 50(2):604-613.

Yoo, H.S., B.U. Bradford, O. Kosyk, S. Shymonyak, T. Uehara, L.B. Collins, W.M. Bodnar, L.M. Ball, A. Gold, and I. Rusyn. 2015a. Comparative analysis of the relationship between trichloroethylene metabolism and tissue-specific toxicity among inbred mouse strains: liver effects. J. Toxicol. Environ. Health A. 78(1):15-31.

Yoo, H.S., B.U. Bradford, O. Kosyk, T. Uehara, S. Shymonyak, L.B. Collins, W.M. Bodnar, L.M. Ball, A. Gold, and I. Rusyn. 2015b. Comparative analysis of the relationship between trichloroethylene metabolism and tissue-specific toxicity among inbred mouse strains: Kidney effects. J. Toxicol. Environ. Health A. 78(1):32-49.

Zhang, Q., S. Bhattacharya, M.E. Andersen, and R.B. Conolly. 2010. Computational systems biology and dose-response modeling in relation to new directions in toxicity testing. J. Toxicol. Environ. Health B Crit. Rev. 13(2-4):253-276.

Zhang, X., S. Wiseman, H. Yu, H. Liu, J.P. Giesy, and M. Hecker. 2011. Assessing the toxicity of napthenic acids using a microbial genome wide live cell reporter array system. Environ. Sci. Technol. 45(5):1984-1991.

Zhang, Z., N. Guan, T. Li, D.E. Mais, and M. Wang. 2012a. Quality control of cell-based high-throughput drug screening. Acta. Pharma. Sin. B 2(5):429-438.

Zhang, W., R. Korstanje, J. Thaisz, F. Staedtler, N. Hartt-man, L. Xu, M. Feng, L. Yanas, H. Yang, W. Valdar, G. A. Churchill, and K. Dipetrillo. 2012b. Genome-wide association mapping of quantitative traits in outbred mice. G3 (Bethesda) 2(2):167-174.

Zhang, Q., S. Bhattacharya, R.B. Conolly, H.J. Clewell, N.E. Kaminski, and M.E. Andersen. 2014. Molecular signaling network motifs provide a mechanistic basis for cellular threshold responses. Environ. Health Perspect. 122(12):1261-1270.

Zhang, Q., S. Bhattacharya, J. Pi., R.A. Clewell, P.L. Carmichael, and M.E. Andersen. 2015. Adaptive posttranslational control in cellular stress response pathways and its relationship to toxicity testing and safety assessment. Toxicol. Sci. 147(2):302-316.

Zou, F., J.A. Gelfond, D.C. Airey, L. Lu, K.F. Manly, R.W. Williams, and D.W. Threadgill. 2005. Quantitative trait locus analysis using recombinant inbred intercrosses: Theoretical and empirical considerations. Genetics 170(3):1299-1311.

Zuang, V., J. Barroso, S. Bremer, S. Casati, M. Ceridono, S. Coecke, R. Corvi, C.Eskes, A. Kinsner, C. Pellizzer, P. Prieto, A. Worth, and J. Kreysa. 2010. ECVAM Technical Report on the Status of Alternative Methods for Cosmetics Testing (2008-2009). Luxembourg: Publications Office of the European Union [online]. Available: https://eurl-ecvam.jrc.ec.europa.eu/eurl-ecvam-status-reports/files/ecvam-report-2008-2009 [accessed July 19, 2016].

Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 51
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 52
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 53
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 54
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 55
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 56
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 57
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 58
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 59
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 60
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 61
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 62
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 63
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 64
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 65
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 66
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 67
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 68
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 69
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 70
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 71
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 72
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 73
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 74
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 75
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 76
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 77
Suggested Citation:"3 Advances in Toxicology." National Academies of Sciences, Engineering, and Medicine. 2017. Using 21st Century Science to Improve Risk-Related Evaluations. Washington, DC: The National Academies Press. doi: 10.17226/24635.
×
Page 78
Next: 4 Advances in Epidemiology »
Using 21st Century Science to Improve Risk-Related Evaluations Get This Book
×
Buy Paperback | $79.00 Buy Ebook | $64.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Over the last decade, several large-scale United States and international programs have been initiated to incorporate advances in molecular and cellular biology, -omics technologies, analytical methods, bioinformatics, and computational tools and methods into the field of toxicology. Similar efforts are being pursued in the field of exposure science with the goals of obtaining more accurate and complete exposure data on individuals and populations for thousands of chemicals over the lifespan; predicting exposures from use data and chemical-property information; and translating exposures between test systems and humans.

Using 21st Century Science to Improve Risk-Related Evaluations makes recommendations for integrating new scientific approaches into risk-based evaluations. This study considers the scientific advances that have occurred following the publication of the NRC reports Toxicity Testing in the 21st Century: A Vision and a Strategy and Exposure Science in the 21st Century: A Vision and a Strategy. Given the various ongoing lines of investigation and new data streams that have emerged, this publication proposes how best to integrate and use the emerging results in evaluating chemical risk. Using 21st Century Science to Improve Risk-Related Evaluations considers whether a new paradigm is needed for data validation, how to integrate the divergent data streams, how uncertainty might need to be characterized, and how best to communicate the new approaches so that they are understandable to various stakeholders.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!