Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Appl Toxicol ; 42(10): 1570-1584, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35393688

RESUMO

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.


Assuntos
Carcinoma de Células Renais , Diabetes Mellitus Tipo 2 , Neoplasias Renais , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Antígenos CD1/metabolismo , Compostos Benzidrílicos/toxicidade , Carcinoma de Células Renais/complicações , Carcinoma de Células Renais/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Feminino , Glucosídeos , Humanos , Hipoglicemiantes/toxicidade , Rim , Neoplasias Renais/induzido quimicamente , Neoplasias Renais/complicações , Neoplasias Renais/tratamento farmacológico , Masculino , Camundongos , Ratos , Transportador 2 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/toxicidade
2.
Int J Toxicol ; 39(3): 198-206, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32372678

RESUMO

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.


Assuntos
Testes de Carcinogenicidade/métodos , Carcinógenos/toxicidade , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Animais , Bioensaio , Genes ras , Camundongos Transgênicos , Projetos de Pesquisa , Inquéritos e Questionários
3.
Toxicol Sci ; 73(2): 209-15, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12700425

RESUMO

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.


Assuntos
Genômica/educação , Projeto Genoma Humano , Medição de Risco/métodos , Toxicologia/tendências , Animais , Humanos
4.
Regul Toxicol Pharmacol ; 36(3): 234-45, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12473408

RESUMO

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.


Assuntos
Exposição por Inalação , Metilmetacrilato/farmacocinética , Modelos Teóricos , Cavidade Nasal/fisiologia , Vasodilatadores/farmacocinética , Compostos de Vinila/farmacocinética , Movimentos do Ar , Humanos , Metilmetacrilato/administração & dosagem , Cavidade Nasal/anatomia & histologia , Nariz/patologia , Medição de Risco , Relação Estrutura-Atividade , Vasodilatadores/administração & dosagem , Compostos de Vinila/administração & dosagem
5.
Toxicol Sci ; 66(2): 320-6, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11896299

RESUMO

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.


Assuntos
Hepatócitos/efeitos dos fármacos , Compostos de Vinila/toxicidade , Animais , Carboxilesterase , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Masculino , Modelos Biológicos , Nitrofenóis/farmacologia , Ratos , Ratos Endogâmicos , Medição de Risco , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA