Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Appl Toxicol ; 42(10): 1570-1584, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35393688

RESUMEN

Inhibition of sodium-glucose cotransporter-2 (SGLT2) has been shown to be a safe and efficacious approach to support managing Type 2 diabetes. In the 2-year carcinogenicity study with the SGLT2 inhibitor empagliflozin in CD-1 mice, an increased incidence of renal tubular adenomas and carcinomas was identified in the male high-dose group but was not observed in female mice. An integrated review of available nonclinical data was conducted to establish a mode-of-action hypothesis for male mouse-specific tumorigenesis. Five key events were identified through systematic analysis to form the proposed mode-of-action: (1) Background kidney pathology in CD-1 mice sensitizes the strain to (2) pharmacology-related diuretic effects associated with SGLT2 inhib ition. (3) In male mice, metabolic demand increases with the formation of a sex- and species-specific empagliflozin metabolite. These features converge to (4) deplete oxidative stress handling reserve, driving (5) constitutive cellular proliferation in male CD-1 mice. The proposed mode of action requires all five key events for empagliflozin to present a carcinogenicity risk in the CD-1 mouse. Considering that empagliflozin is not genotoxic in the standard battery of genotoxicity tests, and not all five key events are present in the context of female mice, rats, or humans, nor for other osmotic diuretics or other SGLT2 inhibitors, the observed male mouse renal tumors are not considered relevant to humans.


Asunto(s)
Carcinoma de Células Renales , Diabetes Mellitus Tipo 2 , Neoplasias Renales , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Antígenos CD1/metabolismo , Compuestos de Bencidrilo/toxicidad , Carcinoma de Células Renales/complicaciones , Carcinoma de Células Renales/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Femenino , Glucósidos , Humanos , Hipoglucemiantes/toxicidad , Riñón , Neoplasias Renales/inducido químicamente , Neoplasias Renales/complicaciones , Neoplasias Renales/tratamiento farmacológico , Masculino , Ratones , Ratas , Transportador 2 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/toxicidad
2.
Int J Toxicol ; 39(3): 198-206, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32372678

RESUMEN

The Tg.rasH2 mouse was developed as an alternative model to the traditional 2-year mouse bioassay for pharmaceutical carcinogenicity testing. This model has found extensive use in support of pharmaceutical drug development over the last few decades. It has the potential to improve quality and timeliness, reduce animal usage, and in some instances allow expedient decision-making regarding the human carcinogenicity potential of a drug candidate. Despite the increased use of the Tg.rasH2 model, there has been no systematic survey of current practices in the design, interpretation of results from the bioassay, and global health authority perspectives. Therefore, the aim of this work was to poll the pharmaceutical industry on study design practices used in the dose range finding and definitive 6-month studies and on results relative to the ongoing negotiations to revise The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use S1 Guidance. Twenty-two member companies of International Consortium for Innovation and Quality in Pharmaceutical Development DruSafe Leadership Group participated in the survey, sharing experiences from studies conducted with 55 test compounds between 2010 and 2018. The survey results provide very useful insights into study design and interpretation. Importantly, the results identified several key opportunities for reducing animal use and increasing the value of testing for potential human carcinogenicity using this model. Recommended changes to study designs that would reduce animal usage include eliminating the requirement to include positive control groups in every study, use of nontransgenic wild-type littermates in the dose range finding study, and use of microsampling to reduce or eliminate satellite groups for toxicokinetics.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Animales , Bioensayo , Genes ras , Ratones Transgénicos , Proyectos de Investigación , Encuestas y Cuestionarios
3.
Regul Toxicol Pharmacol ; 87 Suppl 3: S1-S15, 2017 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-28483710

RESUMEN

The transition from nonclinical to First-in-Human (FIH) testing is one of the most challenging steps in drug development. In response to serious outcomes in a recent Phase 1 trial (sponsored by Bial), IQ Consortium/DruSafe member companies reviewed their nonclinical approach to progress small molecules safely to FIH trials. As a common practice, safety evaluation begins with target selection and continues through iterative in silico and in vitro screening to identify molecules with increased probability of acceptable in vivo safety profiles. High attrition routinely occurs during this phase. In vivo exploratory and pivotal FIH-enabling toxicity studies are then conducted to identify molecules with a favorable benefit-risk profile for humans. The recent serious incident has reemphasized the importance of nonclinical testing plans that are customized to the target, the molecule, and the intended clinical plan. Despite the challenges and inherent risks of transitioning from nonclinical to clinical testing, Phase 1 studies have a remarkably good safety record. Given the rapid scientific evolution of safety evaluation, testing paradigms and regulatory guidance must evolve with emerging science. The authors posit that the practices described herein, together with science-based risk assessment and management, support safe FIH trials while advancing development of important new medicines.


Asunto(s)
Ensayos Clínicos Fase I como Asunto , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/efectos adversos , Humanos , Medición de Riesgo/métodos , Seguridad
4.
Int J Toxicol ; 33(6): 436-49, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25260362

RESUMEN

Empagliflozin, a selective inhibitor of the renal tubular sodium-glucose cotransporter 2, was developed for treatment of type 2 diabetes mellitus. Nonclinical safety of empagliflozin was studied in a battery of tests to support global market authorization. Safety pharmacology studies indicated no effect of empagliflozin on measures of respiratory or central nervous system function in rats or cardiovascular safety in telemeterized dogs. In CD-1 mouse, Wistar Han rat, or beagle dogs up to 13, 26, or 52 weeks of treatment, respectively, empagliflozin exhibited a toxicity profile consistent with secondary supratherapeutic pharmacology related to glucose loss and included decreased body weight and body fat, increased food consumption, diarrhea, dehydration, decreased serum glucose and increases in other serum parameters reflective of increased protein catabolism, gluconeogenesis, and electrolyte imbalances, and urinary changes such as polyuria and glucosuria. Microscopic changes were consistently observed in kidney and included tubular nephropathy and interstitial nephritis (dog), renal mineralization (rat) and tubular epithelial cell karyomegaly, single cell necrosis, cystic hyperplasia, and hypertrophy (mouse). Empagliflozin was not genotoxic. Empagliflozin was not carcinogenic in female mice or female rats. Renal adenoma and carcinoma were induced in male mice only at exposures 45 times the maximum clinical dose. These tumors were associated with a spectrum of nonneoplastic changes suggestive of a nongenotoxic, cytotoxic, and cellular proliferation-driven mechanism. In male rats, testicular interstitial cell tumors and hemangiomas of the mesenteric lymph node were observed; both tumors are common in rats and are unlikely to be relevant to humans. These studies demonstrate the nonclinical safety of empagliflozin.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Glucósidos/toxicidad , Hipoglucemiantes/toxicidad , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Perros , Evaluación Preclínica de Medicamentos , Femenino , Riñón/efectos de los fármacos , Riñón/patología , Neoplasias Renales/inducido químicamente , Neoplasias Renales/patología , Masculino , Ratones , Pruebas de Mutagenicidad , Ratas Wistar , Pruebas de Toxicidad Crónica , Pruebas de Toxicidad Subcrónica
5.
Inhal Toxicol ; 20(4): 375-90, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18302046

RESUMEN

Acetaldehyde is an important intermediate in the chemical synthesis and normal oxidative metabolism of several industrially important compounds, including ethanol, ethyl acetate, and vinyl acetate. Chronic inhalation of acetaldehyde leads to degeneration of the olfactory and respiratory epithelium in rats at concentrations > 50 ppm (90 day exposure) and respiratory and olfactory nasal tumors at concentrations > or = 750 ppm, the lowest concentration tested in the 2-yr chronic bioassay. Differences in the anatomy and biochemistry of the rodent and human nose, including polymorphisms in human high-affinity acetaldehyde dehydrogenase (ALDH2), are important considerations for interspecies extrapolations in the risk assessment of acetaldehyde. A physiologically based pharmacokinetic model of rat and human nasal tissues was constructed for acetaldehyde to support a dosimetry-based risk assessment for acetaldehyde (Dorman et al., 2008). The rodent model was developed using published metabolic constants and calibrated using upper-respiratory-tract acetaldehyde extraction data. The human nasal model incorporates previously published tissue volumes, blood flows, and acetaldehyde metabolic constants. ALDH2 polymorphisms were represented in the human model as reduced rates of acetaldehyde metabolism. Steady-state dorsal olfactory epithelial tissue acetaldehyde concentrations in the rat were predicted to be 409, 6287, and 12,634 microM at noncytotoxic (50 ppm), and cytotoxic/tumorigenic exposure concentrations (750 and 1500 ppm), respectively. The human equivalent concentration (HEC) of the rat no-observed-adverse-effect level (NOAEL) of 50 ppm, based on steady-state acetaldehyde concentrations from continual exposures, was 67 ppm. Respiratory and olfactory epithelial tissue acetaldehyde and H(+) (pH) concentrations were largely linear functions of exposure in both species. The impact of presumed ALDH2 polymorphisms on human olfactory tissue concentrations was negligible; the high-affinity, low-capacity ALDH2 does not contribute significantly to acetaldehyde metabolism in the nasal tissues. The human equivalent acetaldehyde concentration for homozygous low activity was 66 ppm, 1.5% lower than for the homozygous full activity phenotype. The rat and human acetaldehyde PBPK models developed here can also be used as a bridge between acetaldehyde dose-response and mode-of-action data as well as between similar databases for other acetaldehyde-producing nasal toxicants.


Asunto(s)
Acetaldehído/farmacocinética , Contaminantes Ocupacionales del Aire/farmacocinética , Aldehído Deshidrogenasa/genética , Proteínas Mitocondriales/genética , Modelos Biológicos , Cavidad Nasal/metabolismo , Polimorfismo Genético , Aldehído Deshidrogenasa/metabolismo , Aldehído Deshidrogenasa Mitocondrial , Animales , Simulación por Computador , Relación Dosis-Respuesta a Droga , Humanos , Exposición por Inhalación , Proteínas Mitocondriales/metabolismo , Cavidad Nasal/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Ratas
6.
Toxicol Sci ; 97(2): 336-47, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17341482

RESUMEN

Chronic exposure to propylene does not result in any increased incidence of tumors, yet does increase N7-hydroxypropylguanine (N7-HPGua) adducts in tissue DNA. To investigate any potential for genotoxicity (mutagenicity or clastogenicity), male F344 rats were exposed via inhalation to up to 10,000 ppm propylene for 1, 3, or 20 days (6 h/day, 5 days/week). The endpoints examined included gene (Hprt, splenocytes) and chromosomal (bone marrow micronucleus [MN]) mutations, hemoglobin (hydroxypropylvaline, HPVal) adducts in systemic blood, and DNA adducts (N7-HPGua) in several tissues. Similarly exposed female and male F344 rats, implanted with bromodeoxyuridine (BrdU) minipumps, were evaluated for nasal effects (irritation via histopathology and cell proliferation via BrdU). Internal dose measures provided clear evidence for propylene exposure, with HPVal increased for all exposures; N7-HPGua was increased in all tissues from rats exposed for more than 1 day (except lymphocytes). Saturation of propylene conversion to propylene oxide was apparent from the adduct dose-response curves. There were no biologically significant genotoxic effects demonstrated at any exposure level, with no increase in Hprt mutant frequency or in bone marrow MN formation. In addition, no histopathological changes were noted in rodent nasal tissues nor any induction of cell proliferation in nasal tissues. These results demonstrate that repeated exposure of rats to high concentrations of propylene (< or = 10,000 ppm) does not produce evidence of local nasal cavity toxicity or evidence of systemic genotoxicity to hematopoietic tissue, despite the formation of N7-HPGua adducts. In addition, these data indicate that formation of N7-HPGua does not correlate with any measure of genotoxic effect, neither mutagenic nor clastogenic.


Asunto(s)
Alquenos/toxicidad , Mutágenos , Alquenos/administración & dosificación , Animales , Antimetabolitos , Bromodesoxiuridina , Proliferación Celular/efectos de los fármacos , ADN/genética , ADN/aislamiento & purificación , Aductos de ADN/efectos de los fármacos , Determinación de Punto Final , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Femenino , Cromatografía de Gases y Espectrometría de Masas , Globinas/aislamiento & purificación , Globinas/metabolismo , Hemoglobinas/efectos de los fármacos , Hemoglobinas/metabolismo , Hidrólisis , Hipoxantina Fosforribosiltransferasa/metabolismo , Exposición por Inhalación , Masculino , Pruebas de Micronúcleos , Mucosa Nasal/patología , Ratas , Ratas Endogámicas F344 , Espectrometría de Masa por Ionización de Electrospray
7.
Toxicol Lett ; 158(2): 116-21, 2005 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-16039400

RESUMEN

Vinyl acetate exposure in drinking water has been associated with tumor formation in the upper gastrointestinal tract of rats and mice. One potential mechanism for inducing carcinogenesis involves acidification of the intracellular environment due to the metabolism of vinyl acetate to acetic acid. Prolonged intracellular acidification is thought to produce cytotoxic and/or mitogenic responses that are the sentinel pharmacodynamic steps toward cancer. To determine whether exposure to vinyl acetate affects the intracellular pH of intact oral cavity tissue, isolated mouse oral buccal epithelium was loaded with the pH-sensitive dye BCECF, and then exposed to vinyl acetate concentrations ranging from 10 to 1000 microM for up to 4 min. Extracellular vinyl acetate exposure induced a progressive intracellular acidification that was reversible upon removal of the vinyl acetate. The rate of the acidification was concentration-dependent and increased exponentially within the concentration range tested. The magnitude of the vinyl acetate-induced acidification was inhibited by pretreatment with the carboxylesterase inhibitor bis(p-nitrophenyl)phosphate. These results are consistent with the hypothesis that vinyl acetate contributes to the generation and progression of oral cavity tumors via a process of intracellular acidification. Such a process has been proposed to have practical dose-response thresholds below which the intracellular environment can be maintained within homeostatic bounds and the contribution of exposure to carcinogenic risk is negligible.


Asunto(s)
Ácidos , Carcinógenos/toxicidad , Células Epiteliales/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Mucosa Bucal/efectos de los fármacos , Compuestos de Vinilo/toxicidad , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Células Epiteliales/patología , Concentración de Iones de Hidrógeno , Espacio Intracelular/metabolismo , Ratones , Mucosa Bucal/metabolismo , Mucosa Bucal/patología
8.
Toxicol Appl Pharmacol ; 201(3): 203-25, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15582645

RESUMEN

Scientists and decision makers from all sectors agree that risk assessments should be based on the best available science. Several years ago, the Health and Environmental Sciences Institute (HESI), a global branch of the International Life Sciences Institute (ILSI), identified the need for better scientific understanding of dose-dependent transitions in mechanisms of toxicity as one avenue by which the best and latest science can be integrated into the decision making process. In July 2001, the HESI Project Committee on Dose-Dependent Transitions in Mechanisms of Toxicity established a group of academic, government, and industry scientists to engage in active technical discourse on the issue of dose-dependent transitions in mechanisms of toxicity. Over the next 18 months, case studies were examined. These case studies included acetaminophen, butadiene, ethylene glycol, formaldehyde, manganese, methylene chloride, the peroxisome proliferator-activated receptor, progesterone/hydroxyflutamide, propylene oxide, vinyl acetate, vinyl chloride, vinylidene chloride, and zinc (Slikker, W., Jr., Andersen, M.E., Bogdanffy, M.S., Bus, J.S., Cohen, S.D., Conolly, R.B., David, R.M., Doerrer, N.G., Dorman, D.C., Gaylor, D.W., Hattis, D., Rogers, J.M., Setzer, R.W., Swenberg, J.A., Wallace, K., 2004. Dose-dependent transitions in mechanisms of toxicity: case studies. Toxicol. Appl. Pharmacol. 201(3), 226-294 (this issue)). The HESI Project Committee sponsored two technical workshops in 2003. The first of these workshops took place on February 12-13, 2003, and was co-sponsored by the Agency for Toxic Substances and Disease Registry, the American Chemistry Council, the National Institute of Environmental Health Sciences, the Society of Toxicology, and the U.S. Environmental Protection Agency. Additional support was provided by Health Canada. Invited experts from government, academia, and industry provided scientific perspectives and recommendations at the workshop. The purpose of the workshop was to examine approaches to dose-response analysis, learn from the case study examples, and gather feedback from invited participants on the impact of dose-dependent transitions on the risk assessment process. The second forum consisted of a workshop in March 2003 at the Society of Toxicology Annual Meeting in Salt Lake City, UT. This paper addresses the issues discussed at both workshops, and presents the consensus conclusions drawn by expert participants.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Animales , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Inactivación Metabólica , Proyectos de Investigación , Medición de Riesgo
9.
Toxicol Appl Pharmacol ; 201(3): 226-94, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15582646

RESUMEN

Experience with dose response and mechanisms of toxicity has shown that multiple mechanisms may exist for a single agent along the continuum of the full dose-response curve. It is highly likely that critical, limiting steps in any given mechanistic pathway may become overwhelmed with increasing exposures, signaling the emergence of new modalities of toxic tissue injury at these higher doses. Therefore, dose-dependent transitions in principal mechanisms of toxicity may occur, and could have significant impact on the interpretation of reference data sets for risk assessment. To illustrate the existence of dose-dependent transitions in mechanisms of toxicity, a group of academic, government, and industry scientists, formed under the leadership of the ILSI Health and Environmental Sciences Institute (HESI), developed a series of case studies. These case studies included acetaminophen, butadiene, ethylene glycol, formaldehyde, manganese, methylene chloride, peroxisome proliferator-activated receptor (PPAR), progesterone/hydroxyflutamide, propylene oxide, vinyl acetate, vinyl chloride, vinylidene chloride, and zinc. The case studies formed the basis for technical discourse at two scientific workshops in 2003.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Flutamida/análogos & derivados , Acetaminofén/administración & dosificación , Acetaminofén/farmacocinética , Acetaminofén/toxicidad , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/farmacocinética , Analgésicos no Narcóticos/toxicidad , Antagonistas de Andrógenos/administración & dosificación , Antagonistas de Andrógenos/farmacocinética , Antagonistas de Andrógenos/toxicidad , Animales , Butadienos/administración & dosificación , Butadienos/farmacocinética , Butadienos/toxicidad , Dicloroetilenos/administración & dosificación , Dicloroetilenos/farmacocinética , Dicloroetilenos/toxicidad , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/farmacocinética , Compuestos Epoxi/toxicidad , Glicol de Etileno/administración & dosificación , Glicol de Etileno/farmacocinética , Glicol de Etileno/toxicidad , Flutamida/administración & dosificación , Flutamida/farmacocinética , Flutamida/toxicidad , Formaldehído/administración & dosificación , Formaldehído/farmacocinética , Formaldehído/toxicidad , Humanos , Manganeso/administración & dosificación , Manganeso/farmacocinética , Intoxicación por Manganeso/metabolismo , Cloruro de Metileno/administración & dosificación , Cloruro de Metileno/farmacocinética , Cloruro de Metileno/toxicidad , Oxidación-Reducción , Receptores Activados del Proliferador del Peroxisoma/fisiología , Progesterona/administración & dosificación , Progesterona/farmacocinética , Progesterona/toxicidad , Compuestos de Vinilo/administración & dosificación , Compuestos de Vinilo/farmacocinética , Compuestos de Vinilo/toxicidad , Zinc/administración & dosificación , Zinc/farmacocinética , Zinc/toxicidad
10.
Inhal Toxicol ; 16(9): 581-92, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16036751

RESUMEN

Dimethyl sulfate (DMS) is an alkylating agent that is carcinogenic to the respiratory tract of rodents. DNA adducts, cell proliferation, and histopathology were assessed in rats to better understand the molecular dosimetry and tissue dynamics associated with repeated inhalation exposure to DMS. For DNA methylation, rats were exposed to DMS vapor 6 h/day for up to 10 days to 0.0, 0.1, 0.7 and 1.5 ppm. N7-Methylguanine and N3-methyladenine were detected in neutral thermal hydrolysates of DNA isolated from respiratory tract tissues by high-performance liquid chromatography (HPLC) using fluorescence and ultraviolet (UV) detection. DNA methylation was greatest in DNA isolated from nasal respiratory mucosa, less in olfactory, and little was found in lung. N7-Methylguanine levels in respiratory mucosa approached steady-state levels by day 5, and N7-methylguanine persistence following exposure for 5 consecutive days was also determined. Loss of N7-methylguanine from respiratory and olfactory mucosa appeared to follow first-order kinetics. N3-Methyladenine levels were at or below detection limits in all samples. The effect of DMS on histopathology and cell proliferation in the nasal epithelium was also investigated. Rats were exposed nose-only for 2 wk to DMS vapor at concentrations of 0, 0.1, 0.7, or 1.5 ppm. Inhalation exposure to DMS induced degenerative and inflammatory changes in nasal epithelium at >or=0.7 ppm. Cell proliferation evaluations showed a trend towards an increased response at 1.5 ppm. These experiments demonstrate that DMS can induce cytotoxic and proliferative effects and is a potent methylating agent of the nasal mucosa in vivo. These experiments will provide data for the development of dosimetry models useful for risk extrapolation.


Asunto(s)
Adenina/análogos & derivados , Alquilantes/toxicidad , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Guanina/análogos & derivados , Mutágenos/toxicidad , Ésteres del Ácido Sulfúrico/toxicidad , Adenina/metabolismo , Administración por Inhalación , Alquilantes/administración & dosificación , Alquilantes/farmacocinética , Animales , Peso Corporal/efectos de los fármacos , ADN/metabolismo , Aductos de ADN/metabolismo , Guanina/metabolismo , Masculino , Metilación , Modelos Biológicos , Mutágenos/administración & dosificación , Mutágenos/farmacocinética , Mucosa Olfatoria/patología , Purinas/metabolismo , Ratas , Riesgo , Ésteres del Ácido Sulfúrico/administración & dosificación , Ésteres del Ácido Sulfúrico/farmacocinética
11.
Inhal Toxicol ; 16(9): 593-605, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16036752

RESUMEN

Dimethyl sulfate (DMS) is a volatile sulfuric acid ester used principally as a methylating agent in a wide variety of industrial applications. DMS reacts with organic macromolecules by a SN2 mechanism. The weight of experimental evidence suggests that DMS possesses genotoxic and carcinogenic potential. Inhalation studies have shown that repeated exposure to DMS leads to tumors in the nasal cavity and lower respiratory tract in both rats and mice. Here we present a quantitative assessment for cross-species dose extrapolation for inhaled DMS using a physiologically based pharmacokinetic (PBPK) model. The model is designed to simulate N7-methylguanine (N7 mG) DNA adduct levels in the nasal mucosa following DMS exposure in rats and humans. This model was parameterized and predictions were tested by comparison against experimentally measured N7 mG DNA adduct levels in rat nasal mucosa following inhalation exposure to DMS. The model-based interspecies dose comparison, using N7 mG adduct levels in the nasal respiratory tissue as the appropriate dose metrics, predicts a dose rate seven times higher in rats compared to humans.


Asunto(s)
Alquilantes/administración & dosificación , Alquilantes/toxicidad , Guanina/análogos & derivados , Guanina/metabolismo , Mutágenos/administración & dosificación , Mutágenos/toxicidad , Cavidad Nasal/metabolismo , Ésteres del Ácido Sulfúrico/administración & dosificación , Ésteres del Ácido Sulfúrico/toxicidad , Administración por Inhalación , Algoritmos , Animales , Cámaras de Exposición Atmosférica , Aductos de ADN/metabolismo , Metilación de ADN , Relación Dosis-Respuesta a Droga , Humanos , Modelos Biológicos , Ratas , Especificidad de la Especie
12.
Toxicol Sci ; 75(2): 423-31, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12883081

RESUMEN

Vinyl acetate is a synthetic organic ester that has been shown to produce nasal tumors in rats following exposure to 600 ppm in air. The proposed mechanism of action involves the metabolism of vinyl acetate by carboxylesterases and the production of protons leading to cellular acidification. While vinyl acetate-induced decreases in intracellular pH (pHi) have been demonstrated in rat hepatocytes, comparable data from nasal epithelial cells do not exist. Using an in vitro assay system, we have determined the effects of vinyl acetate exposure on pHi in respiratory and olfactory nasal epithelial cells from rats. The respiratory and olfactory epithelial cells were isolated from dissected maxillo- and ethmoturbinates by enzyme digestion. The cells were plated; loaded with the pH-sensitive dye, carboxyseminaphthorhodafluor-1 (SNARF-1); and observed using confocal microscopy. Individual cellular analysis demonstrated that both respiratory and olfactory epithelial cells responded to vinyl acetate exposures with a dose-dependent decrease in pHi. Changes occurred at 100 microM but reached a plateau above 250 microM. Maximal decreases in pHi were 0.3 pH unit in respiratory epithelial cells. While pHi values were normally distributed for the respiratory epithelial cells, the olfactory epithelial cells demonstrated a bimodal distribution, indicating at least two populations of cells, with only one population of cells responding to vinyl acetate. Acidification in these cells did not plateau but continued to increase at 1000 microM. Bis(p-nitrophenyl)phosphate (BNPP), a carboxylesterase inhibitor, was able to attenuate the vinyl acetate-induced decrease in pHi. Data obtained from the isolated cells were validated using tissue explants. These results are consistent with the proposed mode of action for vinyl acetate and supply further data for developing appropriate risk assessments for vinyl acetate exposure.


Asunto(s)
Concentración de Iones de Hidrógeno/efectos de los fármacos , Mucosa Olfatoria/efectos de los fármacos , Compuestos de Vinilo/toxicidad , Animales , Benzopiranos , Carboxilesterasa/antagonistas & inhibidores , Células Cultivadas , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Inhibidores Enzimáticos/farmacología , Masculino , Microscopía Confocal , Naftoles/metabolismo , Nitrofenoles/farmacología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/patología , Ratas , Ratas Endogámicas , Rodaminas/metabolismo
13.
Toxicol Sci ; 73(2): 209-15, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12700425

RESUMEN

The completion of the Human Genome Project has provided the foundation to analyze the expression of all genes transcribed in a specific cell, as well as a reference against which to assess genetic variability and its impact on susceptibility. Recent advances in genomic technologies have set the stage for better understanding and predicting individual adaptive and toxicological responses after toxicant exposure. Thus, it is now possible to simultaneously assess expression levels for thousands of different genes using DNA microarrays, as well as assess posttranscriptional and posttranslational events using high throughput proteomics. Similarly, the risk of toxicant exposure-induced disease used to be estimated across populations with widely varying responses. However, new high-throughput genomic technologies have the potential to greatly improve the accuracy of risk assessment, allowing identification of sensitive subpopulations at risk and ultimately leading to personalized risk profiles based on genetic composition. Recognizing the importance these technologies will have on the practice of risk assessment and the development of regulatory guidelines, the Society of Toxicology sponsored a workshop to evaluate the current status of genomic research; the ethical, legal, and social issues associated with these approaches; and, in this context, how data derived from these technologies would impact risk assessment. This article summarizes the evaluation by experts in genomic research and risk assessment in the first workshop to provide a forum for interaction between these scientific disciplines.


Asunto(s)
Genómica/educación , Proyecto Genoma Humano , Medición de Riesgo/métodos , Toxicología/tendencias , Animales , Humanos
14.
Toxicol Lett ; 140-141: 83-98, 2003 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-12676454

RESUMEN

Understanding the mode of action of carcinogens is critical to scientifically assessing exposure-related risk. Regulatory hazard classification schemes and dose-response assessment paradigms generally require basic knowledge of genotoxic potential to guide decisions on which scheme or paradigm is most appropriate. Although convention suggests that classification and dose-response assessment of genotoxic chemicals should be assessed using conservative assumptions of no threshold, several examples, such as vinyl acetate, exist that challenge this assumption. Vinyl acetate is carcinogenic at portals of entry (nasal cavity and upper gastrointestinal tract). Local metabolism of vinyl acetate produces DNA-reactive acetaldehyde but also produces acetic acid and protons, which contribute to intracellular acidification, cytotoxicity and cell proliferation. This paper reviews their relative contributions to the overall mode of action. Elevated cellular proliferation, well understood to be a risk factor for carcinogenesis, is observed at concentrations associated with tumor formation. Cytotoxicity and compensatory tissue regeneration is one pathway for stimulating cellular proliferation while intracellular acidification is a mitogenic stimulus. Both of these pathways may be operative in nasal tissues while mitogenic proliferation alone appears to be induced in the upper gastrointestinal tract. Using a physiologically-based pharmacokinetic model, quantitative relationships between critical tissue dosimeters and tissue responses are developed to assess the relative importance of genotoxicity and cell proliferation in the overall mode of action of vinyl acetate. This approach supports the concept that intracellular acidification is the sentinel response that precedes cytotoxicity and cellular proliferation. Secondarily, the carcinogenic potential of vinyl acetate is expressed only when tissue exposure to acetaldehyde is high and when cellular proliferation is simultaneously elevated. This mode of action suggests that exposure levels that do not increase intracellular acidification beyond homeostatic bounds will be adequately protective of adverse downstream responses including cancer. These mechanistic insights provide the scientific basis for a cancer classification that incorporates thresholds for cytotoxic and/or mitogenic cell proliferation secondary to intracellular acidification.


Asunto(s)
Carcinógenos/clasificación , Carcinoma de Células Escamosas/inducido químicamente , Pruebas Inmunológicas de Citotoxicidad/métodos , Mitógenos/toxicidad , Neoplasias de la Boca/inducido químicamente , Compuestos de Vinilo/toxicidad , Animales , Carcinógenos/administración & dosificación , Relación Dosis-Respuesta a Droga , Medición de Riesgo , Compuestos de Vinilo/metabolismo , Compuestos de Vinilo/farmacocinética
15.
Toxicol Lett ; 138(1-2): 103-17, 2003 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-12559695

RESUMEN

The respiratory tract is frequently identified as a site of toxicity for inhaled xenobiotic chemicals. Usually, these observations come from controlled animal studies. For these studies to be of quantitative value to human health risk assessment, species-specific factors governing dosimetry of inhaled substances must be taken into account. Toxicokinetics of vapours in the respiratory tract are defined by absorption, distribution, metabolism, and excretion, as they are in other tissues; however, these concepts take on new dimensions when considering respiratory tract toxicants, especially those that elicit portal of entry effects by directly interacting with the tissue lining the respiratory tract. Species-specific factors related to anatomy, physiology and biochemistry govern inter-species extrapolation of toxicokinetics. This article discusses critical factors of respiratory tract kinetics that should be considered when developing physiological-based toxicokinetic (PBTK) models for inhaled vapours. Important considerations such as impact of regional airflow-delivery, water solubility, reactivity, and rates of local biotransformation on respiratory tract tissue dosimetry are highlighted. These factors can be accounted for only to a limited extent when using default approaches to extrapolate dosimetry of inhaled substances across species. On the other hand, PBTK modeling has the flexibility to accommodate many of the critical determinants of respiratory tract toxicity. PBTK models can also help identify the most critical toxicokinetic data necessary to replace defaults. PBTK approaches have led to more informed estimates of human target tissue dose, and therefore human health risk, especially where these risk assessments have been based on extrapolation of animal dosimetry studies. Experience derived from the development of more intensive case studies have, in turn, enabled simplified approaches to the use of PBTK modeling for respiratory tract toxicants. Whether simplified or highly complex, PBTK modeling approaches are proven to be of great utility to risk assesors interested in applying quantitative information to informed risk assessment evaluations.


Asunto(s)
Gases/farmacocinética , Gases/toxicidad , Modelos Biológicos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Animales , Humanos , Exposición por Inhalación , Medición de Riesgo
16.
Regul Toxicol Pharmacol ; 36(3): 234-45, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12473408

RESUMEN

Mathematical models have been developed to describe nasal epithelial tissue dosimetry with two compounds, vinyl acetate (VA) and methyl methacrylate (MMA), that cause toxicity in these tissues These models couple computational fluid dynamics (CFD) calculations that map airflow patterns within the nose with physiologically based pharmacokinetic (PBPK) models that integrate diffusion, metabolism, and tissue interactions of these compounds. Dose metrics estimated in these models for MMA and VA, respectively, were rates of MMA metabolism per volume of tissue and alterations in pH in target tissues associated with VA hydrolysis and metabolism. In this article, four scientists who have contributed significantly to development of these models describe the many similarities and relatively few differences between the MMA and VA models. Some differences arise naturally because of differences in target tissues, in the calculated measures of tissue dose, and in the modes of action for highly extracted vapors (VA) compared with poorly extracted vapors (MMA). A difference in the approach used to estimate metabolic parameters from human tissues provides insights into interindividual extrapolation and identifies opportunities for studies with human nasal tissues to enhance current risk assessments. In general, the differences in model structure for these two esters were essential for describing the biology of the observed responses and in accounting for the different measures of target tissue dose. This article is intended to serve as a guide for understanding issues of optimum model structure and optimal data sources for these nasal tissue dosimetry models. We also hope that it leads to greater international acceptance of these hybrid CFD/PBPK modeling approaches for improving risk assessment for many nasal toxicants. In general, these models predict either equivalent (VA) or lower (MMA) nasal tissue doses in humans compared with tissue doses at equivalent exposure concentrations in rats.


Asunto(s)
Exposición por Inhalación , Metilmetacrilato/farmacocinética , Modelos Teóricos , Cavidad Nasal/fisiología , Vasodilatadores/farmacocinética , Compuestos de Vinilo/farmacocinética , Movimientos del Aire , Humanos , Metilmetacrilato/administración & dosificación , Cavidad Nasal/anatomía & histología , Nariz/patología , Medición de Riesgo , Relación Estructura-Actividad , Vasodilatadores/administración & dosificación , Compuestos de Vinilo/administración & dosificación
17.
Toxicology ; 180(3): 209-20, 2002 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-12393291

RESUMEN

Vinyl acetate (VA) is widely used within the chemical industry, in the manufacture of polyvinyl alcohol, and as polyvinyl acetate emulsions in latex paints, adhesives, paper and paper board coatings. Chronic oral exposure of rodents to high concentrations of VA induces tumours within the oral cavity. Carboxylesterase-dependent hydrolysis of VA is thought to be critical in the development of nasal tumours following inhalation exposure of animals to VA. Therefore, carboxylesterase activity was determined histochemically in the oral cavities of male F344 rats and BDF mice in order to explore the potential role of carboxylesterase-dependent hydrolysis of VA in the development of oral tumours. Following fixation in 10% neutral buffered formalin heads were decalcified in neutral saturated EDTA, embedded in resin, sectioned at six levels (three each for the upper and lower jaws), and carboxylesterase activity revealed in the tissue using alpha-naphthyl butyrate as substrate. The localisation of carboxylesterase activity in freshly dissected rat oral tissue was compared to that of the resin sections and found to be identical, thus validating the decalcification process. A similar pattern of carboxylesterase activity was observed for the two species. Staining was low in areas surrounding the teeth, and medium/high in the buccal mucosa, the central/posterior upper palate and those regions of the lower jaw not proximal to the teeth. In general the intensity of staining was greater in sections from the rat compared to those from the mouse. By comparison, carboxylesterase activity was considerably higher in mouse nasal olfactory epithelium than in any of the oral tissues. Thus the mucosa of the oral cavity has the potential to hydrolyse VA to its metabolites, acetic acid and acetaldehyde, and the presence of carboxylesterases at this site is consistent with, and may be an important determining factor in, the development of oral cavity tumours following exposure to VA.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Mucosa Bucal/enzimología , Animales , Mejilla , Histocitoquímica , Cinética , Ratones , Mucosa Bucal/anatomía & histología , Ratas , Ratas Endogámicas F344 , Especificidad de la Especie , Fijación del Tejido
18.
Toxicol Sci ; 66(2): 320-6, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11896299

RESUMEN

Cancerbioassays have demonstrated the carcinogenic activity of vinyl acetate in rodents. Tumors appear only at the site of contact and mechanistic data suggest that the carcinogenic mechanism involves carboxylesterase-mediated metabolism of vinyl acetate to acetic acid. It has been hypothesized that intracellular formation of acetate causes a reduction of intracellular pH (pH(i)) at noncytotoxic levels, but that prolonged exposure to reduced pH(i) is cytotoxic and/or mitogenic and drives proliferative responses. Coupled with exposure to metabolically formed acetaldehyde at high administered concentrations, nonlinear dose-response curves for epithelial tumors are produced. Freshly isolated rat hepatocytes were used as a model system to test the concept that exposure of cells to vinyl acetate causes a reduction in pH(i). Quantitative fluorescence imaging ratio microscopy showed that exposure of hepatocytes to vinyl acetate concentrations ranging from 10 to 1000 microM caused rapid and sustained reductions of approximately 0.03 to 0.65 pH units. Cellular acidification was rapidly reversed to control pH(i) upon removal of vinyl acetate. There was minimal accumulation of protons during the exposure period, as suggested by minor differences in pH(i) of cells with or without prior exposure to vinyl acetate. The effect of vinyl acetate on pH(i) was attenuated by prior exposure to the carboxylesterase inhibitor bis(p-nitrophenyl)phosphate. These results support the concept that intracellular acidification is a sentinel pharmacodynamic response of cells to vinyl acetate exposure and that pH(i) is an appropriate metric dose for use in quantitative risk assessments of cancer and noncancer human health risk assessment.


Asunto(s)
Hepatocitos/efectos de los fármacos , Compuestos de Vinilo/toxicidad , Animales , Carboxilesterasa , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Masculino , Modelos Biológicos , Nitrofenoles/farmacología , Ratas , Ratas Endogámicas , Medición de Riesgo , Factores de Tiempo
19.
Drug Metab Dispos ; 30(1): 47-54, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11744611

RESUMEN

TY029, an N-pyrrolo[1,2-c]imidazolylphenyl sulfonamide herbicide, controls economically important weeds through inhibition of protoporphyrinogen oxygenase. As partial satisfaction of regulatory requirements to establish safety and to aid in the interpretation of toxicology bioassays, a rat metabolism study of TY029 was performed to define the pharmacokinetics and tissue distribution of this compound. Animals were exposed to single 50- and 2-mg/kg doses of [hydantoin-5-(14)C]TY029 by oral gavage. The tissue distribution studies revealed that generally greater than 5% of the oral dose was found in the carcass, gastrointestinal tract, liver, and the whole blood when plasma microgram equivalents per gram of TY029 was at maximum or at half of the maximum. However, these concentrations rapidly declined to negligible levels. By 96 h after the oral administration of [hydantoin-5-(14)C]TY029, the highest value reported for any one of the collected tissues was below 0.5% of administered dose. Therefore, neither TY029 nor its metabolites was sequestered in tissues to appreciable levels. The C(max), C(max/2), and area under the curve (AUC(INF)) obtained from the plasma pharmacokinetics suggested that in general single-dosed female rats absorbed and eliminated the test compounds faster than their male counterparts. Mass spectral evaluations of the plasma from single high- and low-dose male and female rats identified the plasma constituents related to the test compound. Although the parent molecule was present in all plasma samples, the three acidic metabolites were the predominant plasma metabolites in the high-dose groups. The overall plasma profile included TY029 and six metabolites.


Asunto(s)
Herbicidas/metabolismo , Herbicidas/farmacocinética , Pirroles/metabolismo , Pirroles/farmacocinética , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Animales , Radioisótopos de Carbono , Femenino , Masculino , Ratas , Espectrometría de Masa por Ionización de Electrospray , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...