scispace - formally typeset
Search or ask a question

Showing papers in "Critical Reviews in Toxicology in 2005"


Journal ArticleDOI
TL;DR: The human health risks from clinical and animal research studies are reviewed, including aldehydes as haptens in allergenic hypersensitivity diseases, respiratory allergies, and idiosyncratic drug toxicity; the potential carcinogenic risks of the carbonyl body burden.
Abstract: Aldehydes are organic compounds that are widespread in nature. They can be formed endogenously by lipid peroxidation (LPO), carbohydrate or metabolism ascorbate autoxidation, amine oxidases, cytochrome P-450s, or myeloperoxidase-catalyzed metabolic activation. This review compares the reactivity of many aldehydes towards biomolecules particularly macromolecules. Furthermore, it includes not only aldehydes of environmental or occupational concerns but also dietary aldehydes and aldehydes formed endogenously by intermediary metabolism. Drugs that are aldehydes or form reactive aldehyde metabolites that cause side-effect toxicity are also included. The effects of these aldehydes on biological function, their contribution to human diseases, and the role of nucleic acid and protein carbonylation/oxidation in mutagenicity and cytotoxicity mechanisms, respectively, as well as carbonyl signal transduction and gene expression, are reviewed. Aldehyde metabolic activation and detoxication by metabolizing enzymes are also reviewed, as well as the toxicological and anticancer therapeutic effects of metabolizing enzyme inhibitors. The human health risks from clinical and animal research studies are reviewed, including aldehydes as haptens in allergenic hypersensitivity diseases, respiratory allergies, and idiosyncratic drug toxicity; the potential carcinogenic risks of the carbonyl body burden; and the toxic effects of aldehydes in liver disease, embryo toxicity/teratogenicity, diabetes/hypertension, sclerosing peritonitis, cerebral ischemia/neurodegenerative diseases, and other aging-associated diseases.

608 citations


Journal ArticleDOI
TL;DR: The intention of this review is to collate and discuss the currently available data on OTA-mediated toxicity with particular focus on their relevance for the in vivo situation, and to suggest possible future strategies for unlocking the secrets of ochratoxin A.
Abstract: The mycotoxin ochratoxin A (OTA) has been linked to the genesis of several disease states in both animals and humans It has been described as nephrotoxic, carcinogenic, teratogenic, immunotoxic, and hepatotoxic in laboratory and domestic animals, as well as being thought to be the probable causal agent in the development of nephropathies (Balkan Endemic Nephropathy, BEN and Chronic Interstitial Nephropathy, CIN) and urothelial tumors in humans As a result, several international agencies are currently attempting to define safe legal limits for OTA concentration in foodstuffs (eg, grain, meat, wine, and coffee), in processed foods, and in animal fodder In order to achieve this goal, an accurate risk assessment of OTA toxicity including mechanistic and epidemiological studies must be carried out Ochratoxin has been suggested by various researchers to mediate its toxic effects via induction of apoptosis, disruption of mitochondrial respiration and/or the cytoskeleton, or, indeed, via the generation of DNA adducts Thus, it is still unclear if the predominant mechanism is of a genotoxic or an epigenetic nature One aspect that is clear, however, is that the toxicity of OTA is subject to and characterized by large species- and sex-specific differences, as well as an apparently strict structure-activity relationship These considerations could be crucial in the investigation of OTA-mediated toxicity Furthermore, the use of appropriate in vivo and in vitro model systems appears to be vital in the generation of relevant experimental data The intention of this review is to collate and discuss the currently available data on OTA-mediated toxicity with particular focus on their relevance for the in vivo situation, and also to suggest possible future strategies for unlocking the secrets of ochratoxin A

378 citations


Journal ArticleDOI
TL;DR: It is biologically implausible that parabens could increase the risk of any estrogen-mediated endpoint, including effects on the male reproductive tract or breast cancer, and a comparative approach involving both dose and potency is used to assess whether in utero or adult exposure to paraben might be associated with adverse effects mediated via an estrogen-modulating mode of action.
Abstract: Parabens are a group of the alkyl esters of p-hydroxybenzoic acid and typically include methylparaben, ethylparaben, propylparaben, butylparaben, isobutylparaben, isopropylparaben, and benzylparabe...

371 citations


Journal ArticleDOI
TL;DR: A review on the essentiality and toxicity of manganese and its transport across the blood-brain barrier, and its distribution within the central nervous system (CNS) is presented in this article.
Abstract: Manganese (Mn) is an essential mineral that is found at low levels in food, water, and the air. Under certain high-dose exposure conditions, elevations in tissue manganese levels can occur. Excessive manganese accumulation can result in adverse neurological, reproductive, and respiratory effects in both laboratory animals and humans. In humans, manganese-induced neurotoxicity (manganism) is the overriding concern since affected individuals develop a motor dysfunction syndrome that is recognized as a form of parkinsonism. This review primarily focuses on the essentiality and toxicity of manganese and considers contemporary studies evaluating manganese dosimetry and its transport across the blood-brain barrier, and its distribution within the central nervous system (CNS). These studies have dramatically improved our understanding of the health risks posed by manganese by determining exposure conditions that lead to increased concentrations of this metal within the CNS and other target organs. Most individuals are exposed to manganese by the oral and inhalation routes of exposure; however, parenteral injection and other routes of exposure are important. Interactions between manganese and iron and other divalent elements occur and impact the toxicokinetics of manganese, especially following oral exposure. The oxidation state and solubility of manganese also influence the absorption, distribution, metabolism, and elimination of manganese. Manganese disposition is influenced by the route of exposure. Rodent inhalation studies have shown that manganese deposited within the nose can undergo direct transport to the brain along the olfactory nerve. Species differences in manganese toxicokinetics and response are recognized with nonhuman primates replicating CNS effects observed in humans while rodents do not. Potentially susceptible populations, such as fetuses, neonates, individuals with compromised hepatic function, individuals with suboptimal manganese or iron intake, and those with other medical states (e.g., pre-parkinsonian state, aging), may have altered manganese metabolism and could be at greater risk for manganese toxicity.

310 citations


Journal ArticleDOI
TL;DR: Evidence for metabolic activation of a drug to a reactive intermediate is a necessary, yet insufficient, step in the generation of an idiosyncratic reaction is reviewed and the potential role that high dosages may play in these adverse reactions is considered.
Abstract: Rare adverse reactions to drugs that are of unknown etiology, or idiosyncratic reactions, can produce severe medical complications or even death in patients. Current hypotheses suggest that metabolic activation of a drug to a reactive intermediate is a necessary, yet insufficient, step in the generation of an idiosyncratic reaction. We review evidence for this hypothesis with drugs that are associated with hepatotoxicity, one of the most common types of idiosyncratic reactions in humans. We identified 21 drugs that have either been withdrawn from the U.S. market due to hepatotoxicity or have a black box warning for hepatotoxicity. Evidence for the formation of reactive metabolites was found for 5 out of 6 drugs that were withdrawn, and 8 out of 15 drugs that have black box warnings. For the other drugs, either evidence was not available or suitable studies have not been carried out. We also review evidence for reactive intermediate formation from a number of additional drugs that have been associated with idiosyncratic hepatotoxicity but do not have black box warnings. Finally, we consider the potential role that high dosages may play in these adverse reactions.

291 citations


Journal ArticleDOI
TL;DR: The findings of this article support the conclusion that hormetic dose responses are broadly generalizable, being independent of biological model, endpoint measured, and stressor agent, and represent a basic feature of biological responsiveness to chemical and physical stressors.
Abstract: This article assesses the nature of the dose-response relationship of human tumor cell lines with a wide range of agents including antineoplastics, toxic substances (i.e., environmental pollutants), nonneoplastic drugs, endogenous agonists, and phyto-compounds. Hormetic-like biphasic dose responses were commonly reported and demonstrated in 136 tumor cell lines from over 30 tissue types for over 120 different agents. Quantitative features of these hormetic dose responses were similar, regardless of tumor cell line or agent tested. That is, the magnitude of the responses was generally modest, with maximum stimulatory responses typically not greater than twice the control, while the width of the stimulatory concentration range was usually less than 100-fold. Particular attention was directed to possible molecular mechanisms of the biphasic nature of the dose response, as well as clinical implications in which a low concentration of chemotherapeutic agent may stimulate tumor cell proliferation. Finally, these findings further support the conclusion that hormetic dose responses are broadly generalizable, being independent of biological model, endpoint measured, and stressor agent, and represent a basic feature of biological responsiveness to chemical and physical stressors.

186 citations


Journal ArticleDOI
TL;DR: The HRF features a “concordance” analysis of MOA information from both animal and human sources, with a focus on determining the appropriate role for each MOA data set in human risk assessment.
Abstract: A complete mode of action human relevance analysis—as distinct from mode of action (MOA) analysis alone—depends on robust information on the animal MOA, as well as systematic comparison of the animal data with corresponding information from humans. In November 2003, the International Life Sciences Institute's Risk Science Institute (ILSI RSI) published a 2-year study using animal and human MOA information to generate a four-part Human Relevance Framework (HRF) for systematic and transparent analysis of MOA data and information. Based mainly on non-DNA-reactive carcinogens, the HRF features a “concordance” analysis of MOA information from both animal and human sources, with a focus on determining the appropriate role for each MOA data set in human risk assessment. With MOA information increasingly available for risk assessment purposes, this article illustrates the further applicability of the HRF for reproductive, developmental, neurologic, and renal endpoints, as well as cancer. Based on qualitative and ...

178 citations


Journal ArticleDOI
TL;DR: A set of key events whereby DNA reactivity initiates the process of carcinogenicity that leads to the subsequent mutation induction and enhanced cell proliferation that ultimately results in tumor development is presented.
Abstract: It has been known for decades that mutagenicity plays an important role in the activity of most carcinogens. This mutagenicity can result from direct damage to DNA through a chemical being DNA reactive or from indirect effects, such as through the production of oxygen radicals that then react with DNA. This article presents a set of key events whereby DNA reactivity initiates the process of carcinogenicity that leads to the subsequent mutation induction and enhanced cell proliferation that ultimately results in tumor development. This set of key events for DNA-reactive chemicals was applied to two case studies (aflatoxin B1 and dichloromethane) with the aim of assessing the utility of the Human Relevance Framework (HRF) for this class of chemicals. The conclusions were that the HRF was a viable approach for the use of mechanistic data for DNA-reactive chemicals obtained from both laboratory animals and human cells in vivo and in vitro for predicting human carcinogenicity. In the case of aflatoxin B1, the HRF could be used to predict that carcinogenicity in humans was a likely outcome. In contrast, the HRF predicted that the human carcinogenic potential of dichloromethane was at best less likely than in rodents; this conclusion was supported by the available epidemiological data.

140 citations


Journal ArticleDOI
TL;DR: It is demonstrated that immune-system-related hormetic-like biphasic dose-response relationships are common and highly generalizable according to model, endpoint, and chemical class.
Abstract: This article provides an assessment of the occurrence of immune-system-related hormetic-like biphasic dose-response relationships. Such dose-response relationships are extensive, with over 90 different immune response-related endpoints reported, induced by over 70 endogenous agonists, over 100 drugs, and over 40 environmental contaminants. Such hormetic responses were reported in over 30 animal models, over a dozen mammalian and human cell lines. These findings demonstrate that immune-system-related hormetic-like biphasic dose-response relationships are common and highly generalizable according to model, endpoint, and chemical class. The quantitative features of the dose response are generally consistent with previously published examples of hormetic dose responses for other biological endpoints. These findings were generally recognized and explicitly discussed by the original authors, often with consideration given to possible mechanistic foundations as well as numerous clinical implications. Despite the recognition by individual authors of the hormetic nature of these observed responses, the overall widespread nature of immune-related hormetic responses has been only little appreciated, with a general lack of insight into the highly generalizable nature of this phenomenon as well as the complex regulatory networks affecting biological switching mechanisms that result in the hormetic responses.

128 citations


Journal ArticleDOI
TL;DR: While the analysis predicts that PTU doses that produce thyroid hormone insufficiency in humans would adversely affect the developing brain, careful monitoring of PTU administration in pregnant and lactating humans keeps infant serum thyroid hormone levels within the normal range.
Abstract: Because thyroid hormone is essential for normal brain development before and after birth, environmental chemicals that interfere with thyroid hormone signaling can adversely affect brain development. Adverse consequences of thyroid hormone insufficiency depend both on severity and developmental timing, indicating that environmental antithyroid factors may produce different effects at different developmental windows of exposure. Mechanistic studies can provide important insight into the potential impact of chemicals on human thyroid function, but relevance to humans must be systematically evaluated. This kind of analysis depends on data sets that include information about animals and humans. The drug 6-n-propyl-2-thiouracil (PTU) is used in animals to experimentally manipulate serum thyroid hormone levels, and in humans to treat patients, including pregnant women, with Graves' disease. A systematic analysis of the mode of action (MOA) of PTU in rats and in humans discloses similar modes of action. While the analysis predicts that PTU doses that produce thyroid hormone insufficiency in humans would adversely affect the developing brain, careful monitoring of PTU administration in pregnant and lactating humans keeps infant serum thyroid hormone levels within the normal range.

98 citations


Journal ArticleDOI
TL;DR: Funds of the hepatocarcinogenicity of vinyl chloride appear now well established, providing a solid scientific basis for regulatory activities and supporting a linear nonthreshold extrapolation for low-dose assessment of carcinogenic risks due to vinyl chloride.
Abstract: The carcinogenicity of vinyl chloride in humans was recognized in 1974 based on observations of hepatic angiosarcomas in highly exposed workers. A multiplicity of endpoints has been demonstrated. The primary target organ, the liver, displays differential susceptibilities of hepatocytes and sinusoidal cells, which are modified by factors of age and dose. There is consistency in organotropism between experimental animals and humans. Vinyl chloride is a pluripotent carcinogen, predominantly directed toward hepatic endothelial (sinusoidal) cells, and second toward the parenchymal cells of the liver. The similarity of results between experimental animals and humans is a solid basis of an amalgamation of experimental and epidemiological risk estimates. Vinyl chloride requires metabolic activation for carcinogenicity and mutagenicity, and toxicokinetics are a key to interpret the dose response. Practically the entire initial metabolism of vinyl chloride is oxidative. At higher exposure concentrations this is non...

Journal ArticleDOI
TL;DR: Developmental exposure to nicotine in rats results in neurobehavioral effects such as reduced locomotor and cognitive function and the question as to the relative importance of smoking per se versus nicotine alone may be determined.
Abstract: Developmental exposure to nicotine in rats results in neurobehavioral effects such as reduced locomotor and cognitive function. Key events in the animal mode of action (MOA) include binding to the nicotinic cholinergic receptor during prenatal and/or early postnatal development. This leads to premature onset of cell differentiation at the expense of cell replication, which leads to brain cell death or structural alterations in regional brain areas. Other events include an initial increase followed by a decrease in adenyl cyclase activity, as well as effects on the noradrenergic, dopaminergic, and serotonergic neurotransmitter systems. Because the nicotine receptor is also present in the developing human brain and the underlying biology for DNA synthesis and cell signaling is comparable, this MOA is likely to be relevant for humans. Although the effects of nicotine exposure in developing humans is not well documented, nicotine exposure as a result of cigarette smoking during pregnancy is associated with several physiological and behavioral outcomes that are reminiscent of the effects of nicotine alone in animal models. As data become available with the advent of the use of the nicotine patch in pregnant humans, the question as to the relative importance of smoking per se versus nicotine alone may be determined.

Journal ArticleDOI
TL;DR: The correspondence of results on signaling and of effects on development in animals and humans supports the view that alteration of Wnt signaling is a relevant MOA in humans.
Abstract: Valproic acid (VPA) has long been known to cause spina bifida, a neural tube defect, and other effects in fetuses of women treated with this drug. Toxicological tests in laboratory mice and rats at human therapeutic doses also show neural tube and other defects. Studies show that VPA alters Wnt signaling in human and animal cells, inducing Wnt-dependent gene expression at doses that cause developmental effects. Structural analogues of VPA that do not have this effect on Wnt signaling do not cause developmental effects. Similarly, Trichostatin A, a compound that mimics VPA in its effects on Wnt gene expression, also causes similar developmental effects. Alteration of Wnt signaling is empirically well supported as the postulated mode of action (MOA) for VPA's developmental effects in animals. VPA causes alteration of Wnt signaling in both human and animal cells systems at the same dose levels. The correspondence of effects on signaling and of effects on development in animals and humans supports the view that alteration of Wnt signaling is a relevant MOA in humans.

Journal ArticleDOI
TL;DR: Because humans may be exposed to multiple phthalate esters producing adverse effects on reproductive development, these compounds may be useful in testing the utility of the Human Relevance Framework approach for evaluating cumulative and aggregate risk.
Abstract: Certain phthalate esters (di-2-ethylhexyl; di-n-butyl and butyl benzyl) have profound effects on the developing male reproductive system when administered orally to pregnant experimental animals during a critical window of development. These esters produce a syndrome of adverse effects that are characteristic of a disturbance in androgen-mediated development and include a variety of reproductive tract malformations and effects on developmental phenotypic markers. A testicular dysgenesis syndrome has been proposed to explain the secular increases in a number of human male reproductive deficits, including decreased semen parameters, increased incidence of cryptorchidism and hypospadias (two of the most common human birth defects), and increased incidence of testicular (germ cell derived) cancer. The rodent phthalate data lend support to the hypothesis. This example illustrates a number of points in the use of the Human Relevance Framework. First, chemical agents may have more than one mode of action (MOA): for example, phthalate-induced peroxisome proliferation leading to hepatocarcinogensis, compared with the induction of developmental effects via effects on androgen signaling. Second, the case demonstrates the life-stage sensitivity of the response to these compounds. Third, because humans may be exposed to multiple phthalate esters producing adverse effects on reproductive development, these compounds may be useful in testing the utility of the Human Relevance Framework (HRF) approach for evaluating cumulative and aggregate risk.

Journal ArticleDOI
TL;DR: It is concluded that there is no relationship between the damage to the chromosomes at high, toxic doses of aniline and its major metabolites p-aminophenol/p-hydroxyacetanilide and theAniline-induced spleen tumors in the rat.
Abstract: Aniline (in the form of its hydrochloride) has been shown to induce a rather rare spectrum of tumors in the spleen of Fischer 344 rats. The dose levels necessary for this carcinogenic activity were in a range where also massive effects on the blood and non-neoplastic splenotoxicity as a consequence of methemoglobinemia were to be observed. This review aimed at clarifying if aniline itself or one of its metabolites has a genotoxic potential which would explain the occurrence of the spleen tumors in rats as a result of a primary genetic activity. The database for aniline and its metabolites is extremely heterogeneous. With validated assays it ranges from a few limited Ames tests (o- and m-hydroxyacetanilide, phenylhydroxylamine, nitrosobenzene) to a broad range of studies covering all genetic endpoints partly with several studies of the same or different test systems (aniline, p-aminophenol, p-hydroxyacetanilide). This makes a direct comparison rather difficult. In addition, a varying number of results with as yet not validated systems are available for aniline and its metabolites. Most results, especially those with validated and well performed/documented studies, did not indicate a potential of aniline to induce gene mutations. In five different mouse lymphoma tests, where colony sizing was performed only in one test, aniline was positive. If this indicates a peculiar feature of a point mutagenic potential or does represent a part of the clastogenic activity for which there is evidence in vitro as well as in vivo remains to be investigated. There is little evidence for a DNA damaging potential of aniline. The clastogenic activity in vivo is confined to dose levels, which are close to lethality essentially due to hematotoxic effects. The quantitatively most important metabolites for experimental animals as well as for humans (p-aminophenol, p-hydroxyacetanilide) seem to have a potential for inducing chromosomal damage in vitro and, at relatively high dose levels, also in vivo. This could be the explanation for the clastogenic effects that have been observed after high doses/concentrations with aniline. They do not induce gene mutations and there is little evidence for a DNA damaging potential. None of these metabolites revealed a splenotoxic potential comparable to that of aniline in studies with repeated or long-term administration to rats. The genotoxicity database on those metabolites with a demonstrated and marked splenotoxic potential, i.e. phenylhydroxylamine, nitrosobenzene, is unfortunately very limited and does not allow to exclude with certainty primary genotoxic events in the development of spleen tumors. But quite a number of considerations by analogy from other investigations support the conclusion that the effects in the spleen do not develop on a primary genotoxic basis. The weight of evidences suggests that the carcinogenic effects in the spleen of rats are the endstage of a chronic high-dose damage of the blood leading to a massive overload of the spleen with iron, which causes chronic oxidative stress. This conclusion, based essentially on pathomorphological observations, and analogy considerations thereof by previous authors, is herewith reconfirmed under consideration of the more recently reported studies on the genotoxicity of aniline and its metabolites, on biochemical measurements indicating oxidative stress, and on the metabolism of aniline. It is concluded that there is no relationship between the damage to the chromosomes at high, toxic doses of aniline and its major metabolites p-aminophenol/p-hydroxyacetanilide and the aniline-induced spleen tumors in the rat.

Journal ArticleDOI
TL;DR: Confidence is high in the animal MOA and medium confidence for the interspecies concordance for the key events in the MOA, but there is a question of whether ambient human exposures are large enough to cause human fetal hypothyroxinemia to the degree needed to cause hearing loss.
Abstract: An increasing incorporation of mode of action (MOA) information into risk assessments has led to examination of animal MOAs to determine relevance to humans. We examined a specific MOA for developmental neurotoxicity using the MOA/Human Relevance Framework (Meek et al., 2003). The postulated MOA of ototoxicity in rats involves early postnatal exposure to polychlorinated biphenyls (PCBs) via lactation, an upregulation of hepatic uridine diphosphoglucuronyltransferases (UGTs), and subsequent hypothyroxinemia during a critical period of cochlear development, with the ultimate neurotoxic consequence of hearing loss. This review concludes with high confidence in the animal MOA and medium confidence for the interspecies concordance for the key events in the MOA. Possible interspecies differences in toxicodynamic factors moderate confidence in some key events. In addition, there is a question of whether ambient human exposures are large enough to cause human fetal hypothyroxinemia to the degree needed to cause hearing loss. Data gaps identified by this analysis include a need to characterize the induciblity of human fetal UGTs and the comparative sensitivity of UGT induction by xenobiotics in rats and humans. Research on these areas of uncertainty will increase confidence that this MOA for PCBs is not likely to not occur in humans, assuming normal conditions of limited ambient exposure.

Journal ArticleDOI
TL;DR: Confidence is high that the animal mode of action (MOA) for vinclozolin-induced malformation of the male reproductive tract is highly plausible in humans.
Abstract: Vinclozolin is a fungicide that has been shown to cause Leydig cell tumors and atrophy of the accessory sex glands in adult rodents. In addition, exposure of rats during pregnancy causes a pattern of malformations in the male urogenital tract. A wealth of standard toxicological studies and targeted research efforts is available related to this adverse effect, and these were used to evaluate the Human Relevance Framework (HRF) for noncancer health effects. Vinclozolin and two of its metabolites, designated M1 and M2, have been shown to bind and inhibit the function of the rat and human androgen receptor. Other means of interfering with androgen receptor function (e.g., by exposure to the pharmaceutical agent flutamide) lead to similar adverse health outcomes. There is direct in vivo evidence in the rat prostate that androgen-dependent gene expression changes occur after exposure to vinclozolin. There are no proposed alternatives to the androgen receptor-mediated mode of action. Based on what is known about kinetic and dynamic factors, confidence is high that the animal mode of action (MOA) for vinclozolin-induced malformation of the male reproductive tract is highly plausible in humans.

Journal ArticleDOI
TL;DR: Overall, peroxisome proliferators produce changes in oxidative stress, but whether these changes are important in the carcinogenic process is not clear at this time.
Abstract: In this review, the evidence about the role of oxidative stress in the induction of hepatocellular carcinomas by peroxisome proliferators is examined. The activation of PPAR-alpha by peroxisome proliferators in rats and mice may produce oxidative stress, due to the induction of enzymes like fatty acyl coenzyme A (CoA) oxidase (AOX) and cytochrome P-450 4A1. The effect of peroxisome proliferators on the antioxidant defense system is reviewed, as is the effect on endpoints resulting from oxidative stress that may be important in carcinogenesis, such as lipid peroxidation, oxidative DNA damage, and transcription factor activation. Peroxisome proliferators clearly inhibit several enzymes in the antioxidant defense system, but studies examining effects on lipid peroxidation and oxidative DNA damage are conflicting. There is a profound species difference in the induction of hepatocellular carcinomas by peroxisome proliferators, with rats and mice being sensitive, whereas species such as nonhuman primates and guinea pigs are not susceptible to the effects of peroxisome proliferators. The possible role of oxidative stress in these species differences is also reviewed. Overall, peroxisome proliferators produce changes in oxidative stress, but whether these changes are important in the carcinogenic process is not clear at this time.

Journal ArticleDOI
TL;DR: This review highlights the discrepancies in molecular events triggered by trichostatin A, the reference compound of hydroxamic acid-containing HDAC inhibitors, in hepatoma cells and primary hepatocytes (which are key targets for drug-induced toxicity).
Abstract: Histone deacetylase (HDAC) inhibitors target key steps of tumor development: They inhibit proliferation, induce differentiation and/or apoptosis, and exhibit potent antimetastatic and antiangiogenic properties in transformed cells in vitro and in vivo. Preliminary studies in animal models have revealed a relatively high tumor selectivity of HDAC inhibitors, strenghtening their promising potential in cancer chemotherapy. Until now, preclinical in vitro research has almost exclusively been performed in cancer cell lines and oncogene-transformed cells. However, as cell proliferation and apoptosis are essential for normal tissue and organ homeostasis, it is important to investigate how HDAC inhibitors influence the regulation of and interplay between proliferation, differentiation, and apoptosis in primary cells as well. This review highlights the discrepancies in molecular events triggered by trichostatin A, the reference compound of hydroxamic acid-containing HDAC inhibitors, in hepatoma cells and primary hepatocytes (which are key targets for drug-induced toxicity). The implications of these differential outcomes in both cell types are discussed with respect to both toxicology and drug development. In view of the future use of HDAC inhibitors as cytostatic drugs, it is highly recommended to include both tumor cells and their healthy counterparts in preclinical developmental studies. Screening the toxicological properties of compounds early in their development process, using a battery of different cell types, will enable researchers to discard those compounds bearing undesirable adverse activity before entering into expensive clinical trials. This will not only reduce the risk for harmful exposure of patients but also save time and money.

Journal ArticleDOI
TL;DR: This critical review is offered to facilitate an updated assessment of the carcinogenicity of diesel exhaust and to provide a rationale for future animal research of new diesel technology to evaluate the health effects of the new technology.
Abstract: Diesel engines play a vital role in world economy, especially in transportation. Exhaust from traditional diesel engines using high-sulfur fuel contains high concentrations of respirable carbonaceous particles with absorbed organic compounds. Recognition that some of these compounds are mutagenic has raised concern for the cancer-causing potential of diesel exhaust exposure. Extensive research addressing this issue has been conducted during the last three decades. This critical review is offered to facilitate an updated assessment of the carcinogenicity of diesel exhaust and to provide a rationale for future animal research of new diesel technology. Life-span bioassays in rats, mice, and Syrian hamsters demonstrated that chronic inhalation of high concentrations of diesel exhaust caused lung tumors in rats but not in mice or Syrian hamsters. In 1989, the International Agency for Research on Cancer (IARC) characterized the rat findings as “sufficient evidence of animal carcinogenicity,” and, with “limited” evidence from epidemiological studies, classified diesel exhaust Category 2A, a “probable human carcinogen.” Subsequent research has shown that similar chronic high concentration exposure to particulate matter generally considered innocuous (such as carbon black and titanium dioxide) also caused lung tumors in rats. Thus, in 2002, the U.S. Environmental Protection Agency (EPA) concluded that the findings in the rats should not be used to characterize the cancer hazard or quantify the cancer risk of diesel exhaust. Concurrent with the conduct of the health effects studies, progressively more stringent standards have been promulgated for diesel exhaust particles and NOx. Engine manufacturers have responded with new technology diesel (improved engines, fuel injection, fuels, lubricants, and exhaust treatments) to meet the standards. This review concludes with an outline of research to evaluate the health effects of the new technology, research that is consistent with recommendations included in the U.S. EPA 2002 health assessment document. When this research has been completed, it will be appropriate for IARC to evaluate the potential cancer hazard of the new technology diesel.

Journal ArticleDOI
TL;DR: This case study with PCE demonstrates the danger of relying on parent chemical kinetic data to validate a model that will be used for the prediction of metabolism, and the closest predictions of the urinary excretion observed in these low-concentration exposures.
Abstract: One of the more problematic aspects of the application of physiologically based pharmacokinetic (PBPK) models in risk assessment is the question of whether the model has been adequately validated to provide confidence in the dose metrics calculated with it. A number of PBPK models have been developed for perchloroethylene (PCE), differing primarily in the parameters estimated for metabolism. All of the models provide reasonably accurate simulations of selected kinetic data for PCE in mice and humans and could thus be considered to be "validated" to some extent. However, quantitative estimates of PCE cancer risk are critically dependent on the prediction of the rate of metabolism at low environmental exposures. Recent data on the urinary excretion of trichloroacetic acid (TCA), the major metabolite of PCE, for human subjects exposed to lower concentrations than those used in previous studies, make it possible to compare the high- to low-dose extrapolation capability of the various published human models. The model of Gearhart et al., which is the only model to include a description of TCA kinetics, provided the closest predictions of the urinary excretion observed in these low-concentration exposures. Other models overestimated metabolite excretion in this study by 5- to 15-fold. A systematic discrepancy between model predictions and experimental data for the time course of the urinary excretion of TCA suggested a contribution from TCA formed by metabolism of PCE in the kidney and excreted directly into the urine. A modification of the model of Gearhart et al. to include metabolism of PCE to TCA in the kidney at 10% of the capacity of the liver, with direct excretion of the TCA formed in the kidney into the urine, markedly improved agreement with the experimental time-course data, without altering predictions of liver metabolism. This case study with PCE demonstrates the danger of relying on parent chemical kinetic data to validate a model that will be used for the prediction of metabolism.

Journal ArticleDOI
TL;DR: The relevance to humans of ethylene glycol-induced developmental toxicity in animals is unknown and the mechanism by which glycolic acid perturbs development has not been investigated sufficiently to develop a plausible hypothesis of mode of action.
Abstract: Ethylene glycol can cause both renal and developmental toxicity, with metabolism playing a key role in the mode of action (MOA) for each form of toxicity. Renal toxicity is ascribed to the terminal metabolite oxalic acid, which precipitates in the kidney in the form of calcium oxalate crystals and is believed to cause physical damage to the renal tubules. The human relevance of the renal toxicity of ethylene glycol is indicated by the similarity between animals and humans of metabolic pathways, the observation of renal oxalate crystals in toxicity studies in experimental animals and human poisonings, and cases of human kidney and bladder stones related to dietary oxalates and oxalate precursors. High-dose gavage exposures to ethylene glycol also cause axial skeletal defects in rodents (but not rabbits), with the intermediary metabolite, glycolic acid, identified as the causative agent. However, the mechanism by which glycolic acid perturbs development has not been investigated sufficiently to develop a plausible hypothesis of mode of action, nor have any cases of ethylene glycol-induced developmental effects been reported in humans. Given this, and the variations in sensitivity between animal species in response, the relevance to humans of ethylene glycol-induced developmental toxicity in animals is unknown at this time.

Journal ArticleDOI
TL;DR: It is concluded that hormesis is an important phenomenon, but that its consequences for risk assessment and the possibilities for in vitro to in vivo extrapolation may remain limited without additional mechanistic insight.
Abstract: This article comments on some of the basic questions put forward in state-of-the-art discussions on hormesis. There seems to be a need for a better definition of the concept itself and reconsideration of whether all biphasic dose-response curves should be considered representative for hormesis. Hormesis may be restricted to phenomena that proceed by mechanisms that are broadly generalizable and represent possibly beneficial overcompensation in response to an adverse stimulus. Using the concept that hormesis is defined as such, the biphasic effect of quercetin on cell proliferation, but also several other receptor-mediated biphasic dose-response phenomena, should not be related to hormesis. Taking into account hormesis in the procedures for risk assessment on compounds characterised by a threshold for the adverse effect is another matter for considerable debate. In our opinion, this would require the reduction of safety factors, providing the possibility for beneficial hormesis-type effects for some people, at the cost of increased chances on adverse effects for other parts of the population. Whether this is a proper way forward remains to be discussed. Improvement of risk assessment strategies may include taking into account biphasic dose-response curves, but should rather start with the consideration of proper physiologically based pharmacokinetic (PBPK) models for better extrapolation of differences in toxicokinetics going from high- to low-dose exposure, as well as taking into account kinetics for gene repair systems. Without considering in vivo toxicokinetics in the in vitro models, extrapolation from in vitro biphasic dose-response curves on cell proliferation to in vivo cell proliferation is difficult to do. Altogether, it is concluded that hormesis is an important phenomenon, especially from the scientific point of view, but that its consequences for risk assessment and the possibilities for in vitro to in vivo extrapolation may remain limited without additional mechanistic insight.

Journal ArticleDOI
TL;DR: Differences in the timing of development of critical target organ systems, particularly the renal system and RAS, explain the absence of definitive structural abnormalities in test animals.
Abstract: Relative to species tested in laboratory studies, the human fetus displays higher vulnerability to enalapril and other angiotensin-converting enzyme inhibitors (ACEI) exhibiting a malformative syndrome that does not appear to have a similar counterpart in experimental animals. An important reason for this higher vulnerability is the earlier intrauterine development of the kidney and the renin-angiotensin-aldosterone (RAS) system in humans, organ systems that are specific targets of ACEI's pharmacological effect. In humans, these systems begin developing prior to the onset of skeletal ossification at the end of the first trimester, with continuing vulnerability throughout the pregnancy. In most animal species tested, these target systems develop close to term, when the fetus is relatively more mature and less vulnerable to the effects of developmental toxicants. For this reason, animal studies that follow standard protocols and evaluate developmental toxicity only for exposures during embryogenesis will miss developmental effects arising secondary to disruption of target systems that develop after the period of major organogenesis. Thus, although the animal mode of action (MOA) for enalapril and other ACEI is plausible in humans, differences in the timing of development of critical target organ systems, particularly the renal system and RAS, explain the absence of definitive structural abnormalities in test animals.

Journal ArticleDOI
TL;DR: Hemoglobin-based oxygen carriers (HBOC), being developed as blood substitutes, are not expected to cause abnormal development in humans or other mammals that do not develop an invYSP, in view of the existing species-specific placental differences and HBOC's demonstrated, exclusive interference withInvYSP histiotrophic nutrition.
Abstract: Rodents form an early inverted yolk sac placenta (invYSP) by apposing the yolk sac to the uterine wall. The invYSP supplies nutrients via histiotrophic nutrition involving pinocytosis of materials from uterine gland secretions, lysosomal degradation, and transfer of the products to the embryo. Interference with histiotrophic trafficking through the invYSP by high-molecular-weight molecules (such as trypan blue) causes malformations and resorptions. Later in gestation, rodents form a definitive chorioallantoic placenta (CAP). By contrast, humans and dogs never develop an invYSP, relying exclusively on the CAP. Given their large size (∼ 250 kD), hemoglobin-based oxygen carriers (HBOC), being developed as blood substitutes, could be expected to interfere with histiotrophic trafficking through the invYSP. During initial toxicity testing, intravenous infusions of HBOC caused pronounced developmental toxicity in rats exposed during the pre-CAP period. Assuming that HBOC interfered with invYSP function, we hypot...

Journal ArticleDOI
TL;DR: The observation that certain drugs with biphasic effects on cancer cell growth in vitro may also produce an “up–down” effect on cancer growth in humans supports Calabrese's suggestion that the concept of the hormetic dose response must be taken seriously by toxicologists and regulators.
Abstract: Evidence is presented that the ability of hormones and antihormones to cause biphasic (hormetic) proliferative responses in cancer cells in vitro correlates with a similar effect of these substances in humans with cancer. Certain antidepressants also produce biphasic growth responses of cancer cells in vitro and stimulate cancer growth in rodents, correlating with an increased risk of breast and other cancers in some, but not all, epidemiological studies assessing early and/or late cancer incidence in patients on antidepressant drugs. The observation that certain drugs with biphasic effects on cancer cell growth in vitro may also produce an "up-down" effect on cancer growth in humans supports Calabrese's suggestion that the concept of the hormetic dose response must be taken seriously by toxicologists and regulators.

Journal ArticleDOI
TL;DR: Comparison of the dose and concentration ranges confirms that hormesis could be an issue in vivo for therapeutic drugs, phytochemicals, and environmental toxicants, and for several examples where in vivo titers are known.
Abstract: Large numbers of chemicals of a variety of types exhibit apparent hormetic effects on cultured human cancer cells, causing stimulation of cell growth or related changes at low doses, followed by inhibition at higher doses. Many of the studies listed are not fully convincing, due to lack of appropriate controls or sufficient number of doses. However, the proposed hormetic response seems firmly established in a subset of these experiments. Significance with regard to in vivo cancer growth has not been pursued and must be a priority for the future. For several examples where in vivo titers are known, such as, resveratrol, suramin, and tamoxifen, comparison of the dose and concentration ranges confirms that hormesis could be an issue in vivo. Further investigations are warranted, especially for therapeutic drugs, phytochemicals, and environmental toxicants.

Journal ArticleDOI
TL;DR: The weight of evidence is insufficient to define the mode of action for reproductive toxicity in the male rat and, based on traditional risk assessment principles, any toxic effects are assumed to be relevant for human risk assessment.
Abstract: Molinate is a preemergent herbicide that has been demonstrated to affect reproduction in the rat via alterations in sperm production. A wealth of standard toxicological studies and targeted research efforts relating to this adverse effect is available, and these were used to evaluate the utility of the Human Relevance Framework (HRF) for noncancer health effects. The hypothesized mode of action involved inhibition of the hydrolysis of cholesterol from high-density lipoprotein in the rat testes, followed by an androgen withdrawal syndrome on spermatogenesis. Some evidence is available that a similar mode of action would not be operable in humans. Despite the wealth of studies conducted in the rat, the weight of evidence is insufficient to define the mode of action for reproductive toxicity in the male rat. A principal deficiency in the database was discordance between the exposure levels observed to cause biochemical disturbances in the testes related to the hypothesized mode of action and the dose levels observed to induce the adverse outcome on spermatogenesis. For this reason, a complete MOA/human relevance analysis is not possible and, based on traditional risk assessment principles, any toxic effects are assumed to be relevant for human risk assessment.

Journal ArticleDOI
TL;DR: This review of the Calabrese review suggests that toxicologists in general have long recognized the existence of nonlinear dose-response curves, but may differently interpret their meaning or importance.
Abstract: Linear-threshold and nonthreshold dose-response relationships have been of fundamental importance in toxicology risk assessment and regulation. However, many examples exist in the literature where immunologic and other data display inverted U-shaped (hormetic) dose-response curves. These data are characterized by stimulation of response at low doses but inhibition at higher doses (Calabrese and Baldwin, 2003). As an example, the nonsteroidal estrogenic compound diethylstilbestrol (DES) at physiologic doses stimulates mitogen-induced lymphocyte proliferation, while pharmacologic exposures inhibit this same response. Similar biphasic dose-response observations have been made for other bioassays that form the foundation of immunotoxicity testing—for example, antibody or cytokine production, macrophage phagocytosis, chemotaxis, or chemiluminescence; surface antigen expression; and ultimately, disease resistance. Dr. Calabrese effectively demonstrates that nonlinear doseresponse relationships are common in the immunology literature. He has reviewed an extraordinary number of references to support this idea. His abstract suggests that such biphasic immune responses have been little appreciated, which may not accurately represent researchers of the discipline. The several hundred graphs shown to support immunologic hormesis represent the work of immunologists, who then often discussed possible operating mechanisms behind the responses as well as potential clinical relevances. The authors of this review of the Calabrese review are among those who have shown nonlinear immunologic responses to immune modulating agents (Calemine et al., 2002), without describing these results as atypical or unexpected. Thus, at least from our perspective, the validity of many of these nonlinear immunologic dose responses is well accepted. We suggest that toxicologists in general have long recognized the existence of nonlinear dose-response curves, but may differently interpret their meaning or importance. David Eaton and

Journal ArticleDOI
Shu-Zheng Liu1
TL;DR: The dose-effect curves of most parameters in cancer cell lines in response to x-irradiation or toxic agents are usually nonlinear, but the shape of the nonlinear dose- effect curves was found to be different for lymphocytes and macrophages.
Abstract: The dose-effect curves of most parameters in cancer cell lines in response to x-irradiation or toxic agents are usually nonlinear. Such a nature is demonstrated in this article with cell lines of the immune system. However, the shape of the nonlinear dose-effect curves was found to be different for lymphocytes (EL-4) and macrophages (J774A.1). The mechanisms and implications are briefly discussed.