Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 52
1.
Clin Pharmacol Ther ; 102(2): 219-227, 2017 Aug.
Article En | MEDLINE | ID: mdl-28474798

Severely-debilitating or life-threatening (SDLT) diseases include conditions in which life expectancy is short or quality of life is greatly diminished despite available therapies. As such, the medical context for SDLT diseases is comparable to advanced cancer and the benefit vs. risk assessment and development of SDLT disease therapeutics should be similar to that of advanced cancer therapeutics. A streamlined development approach would allow patients with SDLT conditions earlier access to therapeutics and increase the speed of progression through development. In addition, this will likely increase the SDLT disease therapeutic pipeline, directly benefiting patients and reducing the economic and societal burden of SDLT conditions. Using advanced-stage heart failure (HF) as an example that illustrates the concepts applicable to other SDLT indications, this article proposes a streamlined development paradigm for SDLT disease therapeutics and recommends development of aligned global regulatory guidance.


Disease Progression , Drug Discovery/methods , Heart Failure/diagnosis , Heart Failure/drug therapy , Severity of Illness Index , Animals , Communicable Diseases/diagnosis , Communicable Diseases/drug therapy , Communicable Diseases/epidemiology , Drug Discovery/trends , Drug Evaluation/methods , Drug Evaluation/trends , Heart Failure/epidemiology , Humans , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/epidemiology
2.
Toxicol Sci ; 85(1): 460-7, 2005 May.
Article En | MEDLINE | ID: mdl-15659566

Vinyl acetate has been shown to induce nasal lesions in rodents in inhalation bioassays. A physiologically based pharmacokinetic (PBPK) model for vinyl acetate has been used in human risk assessment, but previous in vivo validation was conducted only in rats. Controlled human exposures to vinyl acetate were conducted to provide validation data for the application of the model in humans. Five volunteers were exposed to 1, 5, and 10 ppm 13C1,13C2 vinyl acetate via inhalation. A probe inserted into the nasopharyngeal region sampled both 13C1,13C2 vinyl acetate and the major metabolite 13C1,13C2 acetaldehyde during rest and light exercise. Nasopharyngeal air concentrations were analyzed in real time by ion trap mass spectrometry (MS/MS). Experimental concentrations of both vinyl acetate and acetaldehyde were then compared to predicted concentrations calculated from the previously published human model. Model predictions of vinyl acetate nasal extraction compared favorably with measured values of vinyl acetate, as did predictions of nasopharyngeal acetaldehyde when compared to measured acetaldehyde. The results showed that the current PBPK model structure and parameterization are appropriate for vinyl acetate. These analyses were conducted from 1 to 10 ppm vinyl acetate, a range relevant to workplace exposure standards but which would not be expected to saturate vinyl acetate metabolism. Risk assessment based on this model further concluded that 24 h per day exposures up to 1 ppm do not present concern regarding cancer or non-cancer toxicity. Validation of the vinyl acetate human PBPK model provides support for these conclusions.


Models, Biological , Nasal Cavity , Vinyl Compounds/pharmacokinetics , Adolescent , Adult , Female , Humans , Inhalation Exposure , Male , Middle Aged , Nasal Cavity/drug effects , Nasal Cavity/metabolism , Nasal Cavity/physiology , Olfactory Mucosa/drug effects , Olfactory Mucosa/metabolism , Olfactory Mucosa/physiology , Risk Assessment , Species Specificity , Vinyl Compounds/toxicity
3.
Annu Rev Pharmacol Toxicol ; 43: 485-520, 2003.
Article En | MEDLINE | ID: mdl-12415124

Although many questions remain unanswered, the general principle of the sequence of events leading to cancer after exposure to genotoxic carcinogens has become increasingly clear. This helps to understand the parameters that influence the shape of the dose-effect curve for carcinogenesis, including metabolic activation and inactivation of carcinogens, DNA repair, cell cycle control, apoptosis, and control by the immune system. A linear dose-response relationship with no observable threshold seems to be a conservative but adequate description for the carcinogenic activity of many genotoxic carcinogens, such as aflatoxin B1, the tobacco-specific nitrosoketone NNK, and probably N,N-diethylnitrosamine. However, extrapolation models connecting the high-level risk to the zero intercept have clearly resulted in overestimations of risk. Vinyl acetate is an example that is discussed extensively in this review. At extremely high and toxic doses, vinyl acetate is carcinogenic in rats and mice and causes chromosomal aberrations. In tissues of contact, vinyl acetate is converted to acetic acid and acetaldehyde. Only when threshold levels are achieved do critical steps in the mechanism ultimately leading to cancer become active, namely pH reduction in exposed cells of more than 0.15 units leading to cytotoxicity, damage to DNA, and regenerative proliferation. Consistent with the known exposure to endogenous acetic acid and acetaldehyde, tissues sustain a certain level of exposure without adverse effects. Physiological modeling shows that the conditions necessary for carcinogenesis are in place only when threshold levels of vinyl acetate are exceeded. The example of vinyl acetate underlines the importance of toxicological research that unequivocally identifies genotoxic carcinogens acting through a threshold process.


Carcinogens/toxicity , Dose-Response Relationship, Drug , Mutagens/toxicity , Risk Assessment , Vinyl Compounds/toxicity , Animals , Carcinogenicity Tests , Carcinogens/classification , DNA/drug effects , DNA Damage , DNA Repair , Humans , Mutagens/classification , Vinyl Compounds/classification
4.
Toxicol Sci ; 67(2): 190-7, 2002 Jun.
Article En | MEDLINE | ID: mdl-12011478

Chronic administration of vinyl acetate (VA) in drinking water to rats and mice has produced upper digestive tract neoplasms. These tumors were believed to arise from the intracellular metabolism of VA by carboxylesterases to cytotoxic and genotoxic compounds. We hypothesized that prolonged VA exposure at high concentrations would induce cytotoxicity and a restorative cell proliferation (CP). These endpoints were measured in F-344 rats and BDF1 mice administered drinking water containing 0, 1000, 5000, 10,000, or 24,000 ppm VA for 92 days. On test days, Days 1, 8, 29, and 92, upper digestive tract histopathology and oral cavity CP (pulsed 5-bromodeoxyuridine [BrdU] to measure S-phase DNA synthesis) were evaluated. Analysis of test solutions showed that VA spontaneously hydrolyzed, slowly releasing acetic acid and thereby lowering pH. Statistically significant, concentration-related increases in CP occurred in basal cells of the mandibular oral cavity mucosa of mice at 10,000 and 24,000 ppm but only after 92 days. CP increases were approximately 2.4- and 3.4-fold above controls and were considered to be toxicologically significant. Some statistically significant increases in CP were also measured in the oral cavity mucosa of rats; however, these changes were considered to be of equivocal biological relevance. No histopathological evidence of mucosal injury was seen in either species. The absence of cytotoxicity in the upper digestive tract mucosa suggests that the increased CP at high administered VA concentrations may be due to a mitogenic response, ostensibly from the loss of cell growth controls in oral cavity mucosa.


Carcinogens/toxicity , Cell Division/drug effects , Vinyl Compounds/toxicity , Administration, Oral , Animals , Body Weight/drug effects , Bromodeoxyuridine/metabolism , Carcinogens/administration & dosage , DNA/biosynthesis , Dose-Response Relationship, Drug , Drinking , Esophagus/drug effects , Esophagus/metabolism , Esophagus/pathology , Gastric Mucosa/metabolism , Male , Mice , Mice, Inbred Strains , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Rats , Rats, Inbred F344 , Stomach/drug effects , Stomach/pathology , Time Factors , Toxicity Tests , Vinyl Compounds/administration & dosage , Water Supply
5.
Inhal Toxicol ; 13(5): 377-96, 2001 May.
Article En | MEDLINE | ID: mdl-11295869

Vinyl acetate is used in the manufacture of many polymers. The Clean Air Act Amendments of 1990 require that an inhalation risk assessment be conducted to assess risks to human health from ambient exposures. Vinyl acetate is a nasal carcinogen in rats and induces olfactory degeneration in rats and mice. Because of the many unique aspects of the rodent nasal cavity compared to that of humans, conventional means for extrapolating dosimetry between species are not appropriate. Physiologically based pharmacokinetic (PBPK) and pharmacodynamic (PD) modeling can address many of these unique aspects. A PBPK/PD model has been developed for vinyl acetate, but the choice of appropriate dosimeter(s) to use for interspecies extrapolation depends on a hypothesis regarding mode of action. This article summarizes the key studies that formulate a mode of action hypothesis for vinyl acetate. Dose-response relationships for vinyl acetate-induced nonneoplastic and neoplastic responses are highly nonlinear, suggesting complex kinetic processes. Carboxylesterase-dependent metabolism of vinyl acetate forms acetic acid, a potent cytotoxicant, and acetaldehyde, a weak clastogen. Cell death, proposed to be the result of intracellular acidification, results in restorative cell proliferation. In conjunction with sufficient genetic damage, induced by spontaneous mutation and acetaldehyde-induced DNA-protein cross-links (DPX), olfactory degeneration progresses to a state of elevated proliferation and eventually, at high vinyl acetate concentrations, to neoplastic transformation. Thus, reduction in intracellular pH (pHi) is proposed as the dosimeter most closely linked to the earliest stages of vinyl acetate toxicity. Consequently, risk assessments that are based on protection of nasal epithelium from intracellular acidification will be protective of all subsequent pathological responses related to vinyl acetate exposure. Proposing a reasonable mode of action is an important step in any risk assessment and is critical to the choice of dosimeter(s) to be used for interspecies dosimetry extrapolation.


Vinyl Compounds/toxicity , Animals , Humans , Inhalation Exposure , Risk Assessment , Species Specificity , Vinyl Compounds/chemistry
6.
Toxicol Sci ; 61(1): 18-31, 2001 May.
Article En | MEDLINE | ID: mdl-11294970

Significant advancements have been made toward the use of all relevant scientific information in health risk assessments. This principle has been set forth in risk-assessment guidance documents of international agencies including those of the World Health Organization's International Programme on Chemical Safety, the U.S. Environmental Protection Agency, and Health Canada. Improving the scientific basis of risk assessment is a leading strategic goal of the Society of Toxicology. In recent years, there has been a plethora of mechanistic research on modes of chemical toxicity that establishes mechanistic links between noncancer responses to toxic agents and subsequent overt manifestations of toxicity such as cancer. The research suggests that differences in approaches to assessing risk of cancer and noncancer toxicity need to be resolved and a common broad paradigm for dose-response assessments developed for all toxicity endpoints. In November 1999, a workshop entitled "Harmonization of Cancer and Noncancer Risk Assessment" was held to discuss the most critical issues involved in developing a more consistent and unified approach to risk assessment for all endpoints. Invited participants from government, industry, and academia discussed focus questions in the areas of mode of action as the basis for harmonization, common levels of adverse effect across toxicities for use in dose-response assessments, and scaling and uncertainty factors. This report summarizes the results of those discussions. There was broad agreement, albeit not unanimous, that current science supports the development of a harmonized set of principles that guide risk assessments for all toxic endpoints. There was an acceptance among the participants that understanding the mode of action of a chemical is ultimately critical for nondefault risk assessments, that common modes of action for different toxicities can be defined, and that our approach to assessing toxicity should be biologically consistent.


Decision Making , Hazardous Substances/pharmacology , Hazardous Substances/toxicity , Neoplasms/chemically induced , Practice Guidelines as Topic/standards , Risk Assessment , United States Environmental Protection Agency/standards , Animals , Dose-Response Relationship, Drug , Forecasting , Humans , Risk Factors , Species Specificity , United States , World Health Organization
7.
J Toxicol Environ Health A ; 60(8): 567-86, 2000 Aug 25.
Article En | MEDLINE | ID: mdl-10983523

Cyanazine is a member of the chloro-s-triazine class of herbicides. Other triazine herbicides have been shown to induce mammary-gland tumors in rats, although the response is unique to the Sprague-Dawley strain. Cyanazine is nongenotoxic. The present study was conducted to evaluate the chronic toxicity and oncogenic potential of cyanazine. Groups of 62 male and female rats were fed diets containing cyanazine at concentrations of 1, 5, 25, or 50 ppm for up to 2 yr. Mean body weight and body weight gain of male and female rats of the 25- and 50-ppm groups were significantly reduced over the course of the study. Food consumption and food efficiency were also reduced in these groups. Survival was not adversely affected in the treatment groups compared to controls. A significant increase in the incidence of masses of the inguinal region was noted among female rats of the 50-ppm group. These masses were correlated with a significant increase in the incidence of female rats with mammary-gland adenocarcinomas and carcinosarcomas. The incidence of rats with malignant mammary-gland tumors was elevated in the 5-, 25-, and 50-ppm groups, although the incidence within the 5-ppm group was within historical controls. There were no other toxicologically significant observations with respect to ophthalmological, clinical laboratory, or pathological evaluations. Under the conditions of this study, the no-observed-adverse-effect level was 5 ppm. Research into the mechanism of action suggests these mammary tumors are mediated through a prolactin mechanism that is thought to be of low relevance to humans.


Herbicides/toxicity , Mammary Neoplasms, Animal/chemically induced , Triazines/toxicity , Adenocarcinoma/chemically induced , Animal Feed/analysis , Animals , Biological Assay , Body Weight/drug effects , Carcinosarcoma/chemically induced , Eating/drug effects , Female , Food Contamination , Herbicides/administration & dosage , Herbicides/blood , Herbicides/urine , Male , Rats , Rats, Sprague-Dawley , Triazines/administration & dosage , Triazines/blood , Triazines/urine
8.
Mutat Res ; 460(2): 105-15, 2000 Jul 25.
Article En | MEDLINE | ID: mdl-10882851

The ability to repair promutagenic damage resulting from exposure to carcinogens is a critical factor in determining quantitative relationships in carcinogenesis, including the target cell for neoplasia. One major pathway for the repair of alkylating agent-induced DNA damage involves removal of alkylated bases by N-methylpurine-DNA-glycosylase (MPG), the first enzyme in base excision repair. We have measured the expression level of MPG mRNA in liver, lung, and kidney of Sprague-Dawley rats as a function of age. A quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR) method was used to measure cellular MPG mRNA. MPG mRNA was readily detectable in each tissue analyzed and the age-dependent and tissue specific expressions were not statistically different. The lowest amount of mRNA was measured in preweanling liver and the highest amounts were found in preweanling lung and kidney. Since MPG is reported to be responsible for excision of 1,N(6)-ethenoadenine and N(2),3-ethenoguanine, two promutagenic DNA adducts of vinyl chloride (VC) and vinyl fluoride (VF), we examined the regulation of this enzyme after carcinogen exposure. Expression of MPG was induced in rat liver by these carcinogens. In order to determine the repair capacity in different cell populations of liver, we measured MPG gene expression in isolated hepatocytes and nonparenchymal cells (NPC). The amount of MPG mRNA was 4.5-5 times higher in hepatocytes than in NPC of control rats. Induction of MPG expression was observed in hepatocytes of VF exposed-rats but not in NPC. The expression of MPG in NPC was only 15% of that of the hepatocytes from exposed rats. Western blots of MPG protein confirmed the cell type differences, but did not show increased protein in exposed vs. control liver and hepatocytes. Since metabolism of VC and VF requires CYP2E1, an enzyme exhibiting much greater activity in hepatocytes, formation of etheno adducts preferentially occurs in hepatocytes. These data suggest that cellular differences in the repair of N-alkylpurines may be a critical mechanism in the development of cell specificity in VC carcinogenesis.


DNA Glycosylases , N-Glycosyl Hydrolases/deficiency , N-Glycosyl Hydrolases/genetics , Vinyl Chloride/pharmacology , Vinyl Compounds/pharmacology , Aging/physiology , Alkylating Agents/chemistry , Alkylating Agents/pharmacology , Animals , Blotting, Western , Carcinogens/chemistry , Carcinogens/pharmacology , Enzyme Induction/drug effects , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Lung/drug effects , Lung/enzymology , Male , Mutagens/chemistry , Mutagens/pharmacology , N-Glycosyl Hydrolases/biosynthesis , N-Glycosyl Hydrolases/metabolism , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Vinyl Chloride/chemistry , Vinyl Compounds/chemistry
9.
Int J Cancer ; 85(2): 223-7, 2000 Jan 15.
Article En | MEDLINE | ID: mdl-10629081

Previous studies have shown that a high proportion (5/6) of human liver angiosarcomas (ASL) associated with exposure to vinyl chloride (VC) contains a GC-->AT mutation at the Ki-ras codon 13. This mutation, however, has not been found in 5 ASL or 2 hepatocellular carcinomas (HCC) induced in rats by VC. These 2 HCC did contain a mutation at codon 61 of the Ha-ras gene. In order to extend this study and further explore the mechanisms of tumour induction, an additional 6 ASL and 6 HCC induced in rats by VC were analysed for ras gene point mutations, as well as 10 rat and 10 murine ASL induced by vinyl fluoride (VF), and 5 ASL, 6 Kupffer cell sarcomas, 4 HCC and 2 cholangiocellular carcinomas induced by Thorotrast in rats. Tumour DNA was analysed by PCR-SSCP and direct sequencing. None of the rodent ASL contained a mutation at codon 13 of the Ki-ras gene showing that the ras gene mutational pattern is species-specific. The CAA-->CTA mutation, previously found at codon 61 of the Ha-ras gene in rat HCC, was observed in 5 further VC-induced HCC but was not detected in the Thorotrast-induced HCC, suggesting carcinogen-specificity. This mutation was also absent in VC-induced ASL, which supports the cell-specificity of the ras mutational pattern in chemically induced tumours. No predominant mutation was detected in VF- and Thorotrast-induced tumours. Thus, a given mutation in a tumour may be carcinogen-specific but also depend on the species and the cell type.


Carcinogens/toxicity , Carcinoma, Hepatocellular/genetics , Genes, ras , Hemangiosarcoma/genetics , Liver Neoplasms/genetics , Mutagens/toxicity , Vinyl Chloride/toxicity , Vinyl Compounds/toxicity , Animals , Carcinoma, Hepatocellular/chemically induced , DNA Mutational Analysis , Exons , Female , Hemangiosarcoma/chemically induced , Liver Neoplasms/chemically induced , Male , Mice , Mutagenesis , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Rats , Rats, Sprague-Dawley , Thorium Dioxide/toxicity
10.
Inhal Toxicol ; 11(10): 927-41, 1999 Oct.
Article En | MEDLINE | ID: mdl-10509027

Vinyl acetate induces nasal tumors in rats, but not mice. Species differences in airflow patterns, physiology, and biochemistry complicate extrapolation of nasal dosimetry from rats to humans. Physiologically based pharmacokinetic modeling of vinyl acetate dosimetry in rats suggested the presence of a saturable metabolic removal pathway in rat nasal mucus. We explored the possibility that this pathway is either a cytochrome P-450 2E1 (CYP2E1) or high-affinity carboxylesterase. Nasal extraction of vinyl acetate vapor (150 ppm) was measured in the surgically isolated nasal cavity of anesthetized rats. Vinyl acetate (150 ppm) was extracted with 73% efficiency in controls. Pretreatment of rats with the CYP2E1 inhibitor diallyl sulfide (DAS) had no effect on extraction, despite significantly reducing CYP2E1 activity. Pretreatment with bis(p-nitrophenyl) phosphate (BNPP), a carboxylesterase inhibitor, reduced extraction to approximately 41%. Acetaldehyde production was similarly unaffected by DAS but was reduced to 55% of control by BNPP. Rat nasal mucus carboxylesterase activity had a K(m) value (32 microM) similar, within a factor of 2, to the value predicted by the physiologically based model, although V(max) was significantly lower than the model prediction. Histochemical observations support the inference that the high-affinity carboxylesterase is bound to the luminal plasma membrane of nasal tissue and is not readily released by nasal lavage, providing an explanation for the low V(max) of the lavage enzyme. This high-affinity isoenzyme could be important in the removal of odorants from the sensory cell-rich nasal olfactory epithelium.


Carboxylic Ester Hydrolases/metabolism , Inhalation Exposure , Nasal Mucosa/enzymology , Nasal Mucosa/metabolism , Vinyl Compounds/pharmacokinetics , Algorithms , Animals , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cytochrome P-450 CYP2E1/metabolism , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Models, Biological , Nasal Lavage Fluid/chemistry , Nasal Lavage Fluid/cytology , Nasal Mucosa/ultrastructure , Nitrophenols/pharmacology , Rats , Rats, Inbred Strains , Vinyl Compounds/administration & dosage
11.
Toxicol Sci ; 51(1): 19-35, 1999 Sep.
Article En | MEDLINE | ID: mdl-10496674

The 1990 Clean Air Act Amendments require that health risk from exposure to vinyl acetate be assessed. Vinyl acetate is a nasal carcinogen in rats, but not mice, and induces olfactory degeneration in both species. A biologically based approach to extrapolating risks of inhalation exposure from rats to humans was developed, which incorporates critical determinants of interspecies dosimetry. A physiologically based pharmacokinetic (PBPK) model describing uptake and metabolism of vinyl acetate in rat nose was validated against nasal deposition data collected at three airflow rates. The model was also validated against observations of metabolically derived acetaldehyde. Modifying the rat nose model to reflect human anatomy created a PBPK model of the human nose. Metabolic constants from both rats and humans specific for vinyl acetate and acetaldehyde metabolism enabled predictions of various olfactory tissue dosimeters related to the mode of action. Model predictions of these dosimeters in rats corresponded well with observations of vinyl acetate toxicity. Intracellular pH (pHi) of olfactory epithelial cells was predicted to drop significantly at airborne exposure concentrations above the NOAEL of 50 ppm. Benchmark dose methods were used to estimate the ED10 and LED10 for olfactory degeneration, the precursor lesion thought to drive cellular proliferation and eventually tumor development at excess cellular acetaldehyde levels. A concentration x time adjustment was applied to the benchmark dose values. Human-equivalent concentrations were calculated by using the human PBPK model to predict concentrations that yield similar cellular levels of acetic acid, acetaldehyde, and pHi. After the application of appropriate uncertainty factors, an ambient air value of 0.4 to 1.0 ppm was derived. The biologically based approach supports a workplace standard of 10 ppm.


Nasal Cavity/drug effects , Nose Neoplasms/chemically induced , Vinyl Compounds/toxicity , Acetaldehyde/pharmacokinetics , Acetic Acid/pharmacokinetics , Administration, Inhalation , Animals , Epithelium/drug effects , Epithelium/metabolism , Humans , Hydrogen-Ion Concentration , Male , Models, Biological , Nasal Cavity/metabolism , No-Observed-Adverse-Effect Level , Nose Neoplasms/metabolism , Nose Neoplasms/pathology , Olfactory Mucosa/drug effects , Olfactory Mucosa/metabolism , Olfactory Mucosa/pathology , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Rats , Risk Assessment , Species Specificity , Vinyl Compounds/pharmacokinetics
13.
Regul Toxicol Pharmacol ; 29(1): 23-36, 1999 Feb.
Article En | MEDLINE | ID: mdl-10051416

Experimental observations and theoretical considerations indicate a dose threshold for most chemically induced noncancer toxic effects below which the increased risk of toxicity is zero. Thus, the historical approach for minimizing risk from toxic chemicals has been to experimentally determine a no-observed-adverse-effect-level (NOAEL) and then to apply safety or uncertainty factors to estimate a dose not expected to produce that toxic effect in humans. In contrast, for radiation and chemically induced cancer, it has been assumed that all agents operate by a genotoxic mode of action and that some risk can be assigned to even vanishingly small doses. Accordingly, risk assessments for carcinogens have commonly been based on the assumption that the tumor dose-response curve at low doses is linear and passes through the origin. Mode of action is defined as a fundamental obligatory step in the induction of toxicity or cancer. It is now clear that tumor induction can arise in a variety of ways including not only a DNA-reactive genotoxic mode of action, but also non-DNA-reactive nongenotoxic-cytotoxic and nongenotoxic-mitogenic modes of action. Initial risk assessment approaches that recognized this distinction identified a chemical carcinogen as either genotoxic or nongenotoxic, with no middle ground. The realization that there is a continuum whereby different chemicals can act by a combination of modes of action and the recent explosion of research into molecular mechanisms of carcinogenesis indicate that all relevant information should be integrated into the risk assessment process on a case by case basis. A comprehensive approach to risk assessment demands that default assumptions be replaced with an integrated understanding of the rate-limiting steps in the induction of toxicity or cancer along with quantitative measures of the shapes of those dose-response curves. The examples of more contemporary risk assessments are presented for chloroform and vinyl acetate.


Carcinogens/toxicity , Neoplasms/chemically induced , Animals , Humans , Neoplasms/pathology , Neoplasms, Experimental/chemically induced , Risk Assessment
14.
IARC Sci Publ ; (150): 29-43, 1999.
Article En | MEDLINE | ID: mdl-10626206

Vinyl chloride is a known human and animal carcinogen that induces angiosarcomas of the liver. We review here studies on the formation and repair of DNA adducts associated with vinyl chloride and vinyl fluoride in exposed and control rodents and unexposed humans. These vinyl halides induce etheno (epsilon) adducts that are identical to those formed after lipid peroxidation. Of these adducts, N2,3-ethenoguanine (epsilon G) is present in greatest amounts in tissues of exposed animals. After exposure to vinyl chloride for four weeks, epsilon G levels attain steady-state concentrations, such that the amount of newly formed adducts equals the number of adducts that are lost each day. We report the first dosimetry of epsilon G in rats exposed to 0, 10, 100 or 1100 ppm vinyl chloride for five days or four weeks. The number of adducts increased in a supralinear manner. Exposure to 10 ppm vinyl chloride for five days caused a two- to threefold increase in epsilon G over that of the controls, while four weeks' exposure resulted in a fivefold increase. This was confirmed with [13C2]vinyl chloride and by measuring exogenous and endogenous adducts in the same animals. Exposure to 100 ppm vinyl chloride for four weeks caused a 25-fold increase in epsilon G levels over that found in control rats, while exposure to 1100 ppm resulted in a 42-fold increase. The amount of endogenous epsilon G was similar in liver DNA from rats and humans. A comparable response to exposure was seen in rats and mice exposed to 0, 25, 250 or 2500 ppm vinyl fluoride for 12 months. There was a very high correlation between epsilon G levels in rat and mouse liver at 12 months and the incidence of haemangiosarcoma at two years. We were able to demonstrate that the target cell population for angiosarcoma, the nonparenchymal cells, contained more epsilon G than hepatocytes, even though nonparenchymal cells are exposed by diffusion of vinyl halide metabolites formed in hepatocytes. The expression of N-methylpurine-DNA glycosylase mRNA was induced in rat liver after exposure to either 25 or 2500 ppm vinyl fluoride. When this induction was investigated in hepatocytes and nonparenchymal cells, it was found that the latter had only 20% of the N-methylpurine-DNA glycosylase mRNA of hepatocytes, and that only the hepatocytes had induction of this expression after exposure to vinyl fluoride. Thus, the target cells for vinyl halide carcinogenesis have much lower expression of this DNA repair enzyme, which has been associated with etheno adduct repair.


Carcinogens/toxicity , DNA Adducts/biosynthesis , DNA Repair , Liver Neoplasms, Experimental/chemically induced , Vinyl Chloride/toxicity , Vinyl Compounds/toxicity , Animals , DNA Adducts/analysis , Dose-Response Relationship, Drug , Female , Gas Chromatography-Mass Spectrometry , Humans , Liver/drug effects , Male , Mice , Rats , Time Factors
15.
Toxicol Sci ; 46(2): 235-46, 1998 Dec.
Article En | MEDLINE | ID: mdl-10048126

Physiologically based pharmacokinetic (PBPK) models require estimates of catalytic rate constants controlling the metabolism of xenobiotics. Usually, these constants are derived from whole tissue homogenates wherein cellular architecture and enzyme compartmentation are destroyed. Since the nasal cavity epithelium is composed of a heterogeneous cell population measurement of xenobiotic metabolizing enzymes using homogenates could yield artifactual results. In this article a method for measuring rates of metabolism of vinyl acetate, a metabolism-dependent carcinogen, is presented that uses whole-tissue samples and PBPK modeling techniques to estimate metabolic kinetic parameters in tissue compartments. The kinetic parameter estimates were compared to those derived from homogenate experiments using two methods of tissue normalization. When the in vitro gas uptake constants were compared to homogenate-derived values, using a normalization procedure that does not account for tissue architecture, there was poor agreement. Homogenate-derived values from rat nasal tissue were 3- to 23-fold higher than those derived using the in vitro gas uptake method. When the normalization procedure for the rat homogenate-derived values took into account tissue architecture, a good agreement was observed. Carboxylesterase activity in homogenates of human nasal tissues was undetectable. Using the in vitro gas uptake technique, however, carboxylesterase activity was detected. Rat respiratory carboxylesterase and aldehyde dehydrogenase activities were about three and two times higher than those of humans, respectively. Activities of the rat olfactory enzymes were about equivalent to those of humans. K(m) values did not differ between species. The results suggest that the in vitro gas uptake technique is useful for deriving enzyme kinetic constants where effects of tissue architecture are preserved. Furthermore, the results suggest that caution should be exercised when scaling homogenate-derived values to whole-organ estimates, especially in organs of cellular heterogeneity.


Aldehyde Dehydrogenase/metabolism , Carboxylic Ester Hydrolases/metabolism , Gases/pharmacokinetics , Nasal Mucosa/metabolism , Vinyl Compounds/metabolism , Aged , Animals , Carboxylesterase , Chromatography, High Pressure Liquid , Female , Humans , In Vitro Techniques , Male , Middle Aged , Models, Biological , Rats , Species Specificity , Statistics as Topic
17.
Mutat Res ; 380(1-2): 77-96, 1997 Oct 31.
Article En | MEDLINE | ID: mdl-9385391

The purpose of this review is to provide a compilation of work examining DNA damage in the nasal cavity. There are numerous methods to identify and quantify damage to DNA and the diversity of methods and toxicologic endpoints is illustrated by the range of studies presented here. There are a large number of independent studies measuring endpoints in the upper respiratory tract; however, with regard to toxicant induced DNA damage in the nasal passageway, the effects of two compounds, 4-(N-Methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and formaldehyde (HCHO), appear to have been extensively characterized. The body of work on NNK and formaldehyde have provided insights into molecular mechanisms of DNA damage and repair and induced cell replication and its relationship to nasal cancer. With new technologies and molecular techniques, the sensitivity to enable evaluations of the minute quantities of nasal tissue available in test species and human biopsy impact the study of the nasal-toxicant interactions. As methods used to characterize DNA damage increase in sensitivity, the importance of both exogenous and endogenous sources of DNA damage, steady-state levels of cellular damage, repair, and resulting mutations, low-dose exposure assessments and inter-species extrapolation will become increasingly complex. Additional studies of DNA damage in the nasal passage will undoubtedly challenge future estimations of risk and impact what are perceived to be acceptable levels of exposure to known and predicted carcinogens. The aim of this paper is to provide to the interested scientist literature relevant to the effects of agents on nasal DNA, so that areas of insufficient information can be identified and used to further develop and expand the knowledge base for nasal DNA toxicant interactions.


Carcinogens/toxicity , DNA Damage , Nasal Cavity/drug effects , Animals , Cricetinae , Female , Haplorhini , Humans , Male , Rats
18.
Mutat Res ; 380(1-2): 125-41, 1997 Oct 31.
Article En | MEDLINE | ID: mdl-9385394

Anatomical, physiological, biochemical and molecular factors that contribute to chemical-induced nasal carcinogenesis are either largely divergent between test species and humans, or we know very little of them. These factors, let alone the uncertainty associated with our knowledge gap, present a risk assessor with the formidable task of making judgments about risks to human health from exposure to chemicals that have been identified in rodent studies to be nasal carcinogens. This paper summarizes some of the critical attributes of the hazard identification and dose-response aspects of risk assessments for nasal carcinogens that must be accounted for by risk assessors in order to make informed decisions. Data on two example compounds, dimethyl sulfate and hexamethylphosphoramide, are discussed to illustrate the diversity of information that can be used to develop informed hypotheses about mode of action and decisions on appropriate dosimeters for interspecies extrapolation. Default approaches to interspecies dosimetry extrapolation are described briefly and are followed by a discussion of a generalized physiologically based pharmacokinetic model that, unlike default approaches, is flexible and capable of incorporating many of the critical species-specific factors. Recent advancements in interspecies nasal dosimetry modeling are remarkable. However, it is concluded that without the development of research programs aimed at understanding carcinogenic susceptibility factors in human and rodent nasal tissues, development of plausible modes of action will lag behind the advancements made in dosimetry modeling.


Carcinogens/toxicity , Nasal Cavity/drug effects , Animals , Dose-Response Relationship, Drug , Haplorhini , Hempa/toxicity , Humans , Pulmonary Ventilation , Rats , Risk Assessment , Species Specificity , Sulfuric Acid Esters/toxicity
19.
Mutat Res ; 380(1-2): 155-65, 1997 Oct 31.
Article En | MEDLINE | ID: mdl-9385396

Hexamethylphosphoramide (HMPA) is a rat nasal carcinogen that induces squamous cell carcinomas in the anterior portions of the nasal cavity following chronic inhalation exposures as low as 50 ppb. These tumors may arise as a result of P-450-mediated release of formaldehyde (HCHO), a known rat nasal carcinogen. The goal of this research was to investigate early responses of the nasal epithelium to inhaled HMPA. Rats were exposed nose-only to approximately 3 ppm HMPA for 6 h, and killed 18, 48, 96 or 144 h post-exposure. In a separate study, rats were exposed nose-only for 6 h for 1, 2, 3, or 5 consecutive days and killed 18 or 96 h post-exposure. With both single and repeated doses of HMPA, there was no evidence of cytotoxicity in the anterior nose. Olfactory degeneration and necrosis of the dorsal meatus, Bowman's glands and tips of the ethmoid turbinates increased in severity with repeated exposures to HMPA. Cell proliferation was assessed in levels of nasal tissue that included regions of squamous, respiratory, transitional and olfactory epithelium. Regional induction of cell proliferation was measured by BrdU incorporation, and reported as the number of labeled cells/mm basement membrane. At 18 h after a single exposure, there was an increase in cell proliferation in squamous epithelium, which returned to control levels within 48 h. A transitory increase in cell proliferation was observed regions of respiratory and transitional epithelium, although the response of each tissue, in terms of magnitude and peak time of response post-exposure, also differed. Along the dorsal meatus in Level 9, olfactory labeling initially decreased, returned to control levels by 96 h, but again declined at 144 h post-exposure. In repeat dose studies, the squamous epithelium response was variable 18 h post-exposure. For respiratory and transitional epithelium, increased cell proliferation 18 h post-exposure was correlated with increased dose (exposure) of HMPA. Cell proliferation responses following two or more exposures returned to near control levels within 96 h post-exposure. In conclusion, HMPA induced cell proliferation, but not cytotoxicity, in the anterior nose at approximately 3 ppm. These data suggest that HMPA induces proliferative, perhaps mitogenic, responses in the nasal epithelium, and this response may facilitate the fixation of low level genetic damage induced by liberated HCHO.


Carcinogens/toxicity , Hempa/toxicity , Nasal Cavity/drug effects , Administration, Inhalation , Animals , Cell Division/drug effects , Epithelial Cells/drug effects , Hempa/administration & dosage , Male , Rats , Time Factors
20.
Toxicol Appl Pharmacol ; 142(2): 386-400, 1997 Feb.
Article En | MEDLINE | ID: mdl-9070362

Chronic inhalation exposure to vinyl acetate (VA) causes lesions in the nasal cavity of the rat. This effect appears to be related to tissue exposure to either acetaldehyde (AAld) or acetic acid (AA) metabolites of VA or both. A physiologically based pharmacokinetic model was constructed to describe the deposition of VA in the nasal cavity of the rat and provide estimates of regional tissue exposure to VA, AAld, and AA. Since formation of AA in the nasal tissue should cause intracellular acidification, a submodel which describes free intracellular hydrogen ion concentration and intracellular pH (pHi) changes was linked to the VA model. The dosimetry model was applied to data from a series of experiments designed to measure the uptake and metabolism of VA in the isolated upper respiratory tract of the rat at exposure concentrations ranging from 73 to 2190 ppm. Extraction of VA from the nasal cavity was nonlinear with respect to exposure concentration and ranged from 36 to 94%, with the greatest deposition occurring at the lowest VA concentrations. Pretreatment with bis(p-nitrophenyl)phosphate, an inhibitor of carboxylesterases, significantly reduced fractional deposition of VA compared to naive rats exposed to similar VA concentrations. The best model fits for VA extraction and AAld appearance were achieved when a second carboxylesterase isozyme, with high-affinity characteristics, was included. Simulations of 6-h inhalation exposures to VA predicted that the order of nasal tissue exposures will be to AA > AAld > VA. In addition, based on measured tissue hydrolysis rates, sufficient acid should be formed by the metabolism of VA to cause significant changes in pHi. VA exposures of 200 and 600 ppm were predicted to result in a pHi of less than 7.2 and 6.7, respectively. This model provides nasal dosimetry estimates needed to develop mechanistically based risk assessment approaches for human exposures to VA vapor.


Nasal Cavity/metabolism , Vinyl Compounds/pharmacokinetics , Acetaldehyde/metabolism , Acetic Acid/metabolism , Animals , Computer Simulation , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Male , Models, Biological , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Nasal Cavity/drug effects , Nitrophenols/pharmacology , Rats , Solubility
...