Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Mutat Res ; 752(2): 99-118, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23262374

RESUMO

Genetic toxicity testing is used as an early surrogate for carcinogenicity testing. Genetic toxicity testing is also required by regulatory agencies to be conducted prior to initiation of first in human clinical trials and subsequent marketing for most small molecule pharmaceutical compounds. To reduce the chances of advancing mutagenic pharmaceutical candidates through the drug discovery and development processes, companies have focused on developing testing strategies to maximize hazard identification while minimizing resource expenditure due to late stage attrition. With a large number of testing options, consensus has not been reached on the best mutagenicity platform to use or on the best time to use a specific test to aid in the selection of drug candidates for development. Most companies use a process in which compounds are initially screened for mutagenicity early in drug development using tests that require only a few milligrams of compound and then follow those studies up with a more robust mutagenicity test prior to selecting a compound for full development. This review summarizes the current applications of bacterial mutagenicity assays utilized by pharmaceutical companies in early and late discovery programs. The initial impetus for this review was derived from a workshop on bacterial mutagenicity screening in the pharmaceutical industry presented at the 40th Annual Environmental Mutagen Society Meeting held in St. Louis, MO in October, 2009. However, included in this review are succinct summaries of use and interpretation of genetic toxicity assays, several mutagenicity assays that were not presented at the meeting, and updates to testing strategies resulting in current state-of the art description of best practices. In addition, here we discuss the advantages and liabilities of many broadly used mutagenicity screening platforms and strategies used by pharmaceutical companies. The sensitivity and specificity of these early mutagenicity screening assays using proprietary compounds and their concordance (predictivity) with the regulatory bacterial mutation test are discussed.


Assuntos
Bactérias/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica , Testes de Mutagenicidade , Mutagênicos/toxicidade , Mutação/genética , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Humanos
3.
Toxicol Mech Methods ; 18(2-3): 277-95, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-20020921

RESUMO

ABSTRACT Genetic toxicity data from various sources were integrated into a rigorously designed database using the ToxML schema. The public database sources include the U.S. Food and Drug Administration (FDA) submission data from approved new drug applications, food contact notifications, generally recognized as safe food ingredients, and chemicals from the NTP and CCRIS databases. The data from public sources were then combined with data from private industry according to ToxML criteria. The resulting "integrated" database, enriched in pharmaceuticals, was used for data mining analysis. Structural features describing the database were used to differentiate the chemical spaces of drugs/candidates, food ingredients, and industrial chemicals. In general, structures for drugs/candidates and food ingredients are associated with lower frequencies of mutagenicity and clastogenicity, whereas industrial chemicals as a group contain a much higher proportion of positives. Structural features were selected to analyze endpoint outcomes of the genetic toxicity studies. Although most of the well-known genotoxic carcinogenic alerts were identified, some discrepancies from the classic Ashby-Tennant alerts were observed. Using these influential features as the independent variables, the results of four types of genotoxicity studies were correlated. High Pearson correlations were found between the results of Salmonella mutagenicity and mouse lymphoma assay testing as well as those from in vitro chromosome aberration studies. This paper demonstrates the usefulness of representing a chemical by its structural features and the use of these features to profile a battery of tests rather than relying on a single toxicity test of a given chemical. This paper presents data mining/profiling methods applied in a weight-of-evidence approach to assess potential for genetic toxicity, and to guide the development of intelligent testing strategies.

4.
Chem Res Toxicol ; 14(12): 1590-5, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11743741

RESUMO

The metabolic fate of 3-butene-1,2-diol (BDD), a secondary metabolite of the industrial carcinogen, 1,3-butadiene, is unclear. The current study characterizes BDD oxidation to hydroxymethylvinyl ketone (HMVK), a reactive Michael acceptor. Because of its instability in aqueous medium, HMVK was trapped by conjugation with GSH, a reaction that occurred readily at physiological conditions (pH 7.4, 37 degrees C) to yield 1-hydroxy-2-keto-4-(S-glutathionyl)butane. The results show that BDD was oxidized to HMVK by mouse, rat, and human liver microsomes and by cDNA-expressed human cytochrome P450s. Eadie-Hofstee plots demonstrated biphasic kinetics of BDD oxidation with mouse and rat liver microsomes and one of three individual human liver microsomes; BDD oxidation by the other two human liver microsomal samples was best described by monophasic kinetics. Of the human P450 enzymes examined, only P450 2E1 exhibited activity at 1 mM BDD. P450 3A4 was capable of catalyzing the reaction at a high BDD (10 mM) concentration; P450 1A1, 1A2, 1B1, 2D6-Met, and 2D6-Val produced only trace amounts of HMVK-GSH whereas P450 2A6, 2C8, 2C9, and 4A11 had no detectable activity. Detection of HMVK or the HMVK-GSH conjugate was dependent on reaction time, protein, and BDD concentrations, and the presence of NADPH. Collectively, the results provide clear evidence for BDD bioactivation to yield HMVK. Because mouse, rat, and human liver microsomes exhibited K(m) values of 50-80 microM, the results also suggest that HMVK could be formed after rodent or human exposure to BDD or its parent compound, BD.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Glicóis/metabolismo , Microssomos Hepáticos/enzimologia , Adulto , Animais , Sistema Enzimático do Citocromo P-450/genética , DNA Complementar/genética , Cromatografia Gasosa-Espectrometria de Massas , Glutationa/metabolismo , Humanos , Cetonas/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Oxirredução , Ratos
5.
Chem Biol Interact ; 135-136: 239-48, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11397394

RESUMO

Species differences in 1,3-butadiene (BD) bioactivation and detoxication have been implicated in the greater sensitivity of mice to the carcinogenic effects of BD compared to rats, but the molecular basis for species differences in BD metabolism is not well understood. Previous and recent work conducted in this laboratory has examined the relative rates of BD oxidation to epoxybutene (EB) in male and female B6C3F1 mouse tissues, characterized the major cytochrome P450 enzymes involved in BD bioactivation in these tissues, and determined the potential utility of the freshly isolated hepatocyte model to investigate species differences in metabolism of BD and related compounds. Collectively, the results suggest a role for P450s 2E1, 2A5, and 4B1 in sex and tissue differences in BD bioactivation in the mouse. When coordinated metabolism of EB was investigated in male B6C3F1 mouse and Sprague-Dawley rat hepatocytes, the hepatocytes from both species were found to catalyze EB oxidation to meso- and (+/-)-diepoxybutane (DEB), EB hydrolysis to 3-butene-1,2-diol (BDD), and EB conjugation to form GSH conjugates (GSEB). The metabolite area under the curve (AUC) exhibited dependence on the EB concentration used. However, the EB activation/detoxication ratios with the mouse hepatocytes were much higher than the ratios obtained with the rat hepatocytes. These results illustrate the potential utility of the hepatocyte model for estimating flux through competing metabolic pathways and predicting in-vivo metabolism of EB. Collectively, the results may allow a better understanding of the molecular and kinetic basis of species differences in BD metabolism and may lead to a more accurate assessment of human risk.


Assuntos
Butadienos/metabolismo , Animais , Biotransformação , Butadienos/toxicidade , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Compostos de Epóxi/metabolismo , Compostos de Epóxi/toxicidade , Feminino , Glutationa/metabolismo , Inativação Metabólica , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Distribuição Tecidual
6.
Drug Metab Dispos ; 29(6): 830-6, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11353751

RESUMO

1,3-Butadiene (BD) is a multisite carcinogen in rodents, with mice being much more susceptible than rats. This species difference in carcinogenicity has been attributed to differences in metabolism. In this study, coordinated metabolism of butadiene monoxide (BMO, 5, 25, and 250 microM), the primary reactive metabolite of BD, was investigated in freshly isolated male B6C3F1 mouse and Sprague-Dawley rat hepatocytes. The hepatocytes from both species catalyzed BMO oxidation to meso- and (+/-)-diepoxybutane (DEB), BMO hydrolysis to 3-butene-1,2-diol (BDD), and BMO conjugation with glutathione (GSH) to form GSH conjugates (GSBMO). Metabolite area under the curve (AUC) exhibited dependence on the BMO concentration and incubation time (0-45 min). However, the observed BMO activation/detoxication ratios (obtained by dividing the AUC for total DEB by the summed AUC values for BDD and GSBMO) with mouse hepatocytes were approximately 15- to 40-fold higher than the corresponding ratios observed with rat hepatocytes. At 5 microM BMO, bioactivation in the mouse exceeded detoxication by approximately 2-fold, whereas at the 250 microM concentration, activation was only about 31% of total detoxication. In rat hepatocytes, the activation-detoxication ratio was relatively independent of the initial BMO concentration, with flux through the oxidative pathway at approximately 2 to 5% of the total detoxication. These results, which are more consistent with in vivo mouse and rat toxicity data than the metabolic rates obtained with subcellular fractions, illustrate the potential utility of the isolated hepatocyte model for estimating flux through competing metabolic pathways and predicting in vivo metabolism of BMO and its parent compound, BD.


Assuntos
Compostos de Epóxi/farmacocinética , Hepatócitos/metabolismo , Animais , Cromatografia Gasosa-Espectrometria de Massas , Glutationa/metabolismo , Hidrólise , Masculino , Camundongos , Oxirredução , Ratos , Ratos Sprague-Dawley
8.
Drug Metab Dispos ; 26(9): 914-20, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9733671

RESUMO

3-Butene-1,2-diol (BDD), a metabolite of 1,3-butadiene, is rapidly metabolized by B6C3F1 mice at doses ranging from 10 to 250 mg/kg. Calculation of plasma clearance suggested that the kinetics of BDD metabolism were dose-dependent. Clearance varied 5-fold in this dose range. Urinary excretion of BDD was also dose-dependent but did not exceed 5% of the administered dose. A small fraction of the dose (<1%) was excreted as glucuronide or sulfate conjugates. Benzylimidazole, a cytochrome P450 inhibitor, decreased the clearance of BDD (25 mg/kg) by 44%, whereas 4-methylpyrazole, an alcohol dehydrogenase and cytochrome P450 inhibitor, decreased BDD clearance by 82%. BDD administration (250 mg/kg) resulted in depletion of hepatic and renal nonprotein thiols, by 48 and 22%, respectively. Pretreatment of mice with 4-methylpyrazole provided partial protection against depletion of nonprotein thiols, whereas pretreatment with benzylimidazole was ineffective. Incubation of BDD with NADPH and mouse liver microsomes resulted in time-dependent inactivation of p-nitrophenol hydroxylase (PNPH) activity, a marker for cytochrome P450. Inclusion of glutathione, with or without glutathione peroxidase, did not attenuate the inactivation of PNPH, whereas deferoxamine, superoxide dismutase, catalase, and mannitol provided modest protection. These results are consistent with suicide inhibition of PNPH by BDD, with a minor role for reactive oxygen species in the loss of PNPH. Treatment of mice with BDD (250 mg/kg) inactivated hepatic microsomal PNPH activity by 50% after 60 min. These results suggest that BDD is extensively and rapidly metabolized in mice, and they provide evidence for the formation of reactive intermediates that could play a role in the toxicity of 1, 3-butadiene.


Assuntos
Álcool Desidrogenase/metabolismo , Butadienos/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Glicóis/farmacocinética , Álcool Desidrogenase/antagonistas & inibidores , Álcool Desidrogenase/urina , Animais , Antioxidantes/farmacologia , Butadienos/urina , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/urina , Inibidores Enzimáticos/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Glicóis/urina , Masculino , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia
9.
Chem Res Toxicol ; 9(7): 1127-34, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8902267

RESUMO

3-Butene-1,2-diol (BDD) is a metabolite of the carcinogenic petrochemical 1,3-butadiene. BDD is produced by cytochrome P450-mediated oxidation of 1,3-butadiene to butadiene monoxide, followed by enzymatic hydrolysis by epoxide hydrolase. The metabolic disposition of BDD is unknown. The current work characterizes BDD oxidation by purified horse liver alcohol dehydrogenase (ADH) and by cytosolic ADH from mouse, rat, and human liver. BDD is oxidized by purified horse liver ADH in a stereoselective manner, with (S)-BDD oxidized at approximately 7 times the rate of (R)-BDD. Attempts to detect and identify metabolites of BDD using purified horse liver ADH demonstrated formation of a single stable metabolite, 1-hydroxy-2-butanone (HBO). A second possible metabolite, 1-hydroxy-3-butene-2-one (HBONE), was tentatively identified by GC/MS, but HBONE formation could not be clearly attributed to BDD oxidation, possibly due to its rapid decomposition in the incubation mixture. Formation of HBO by ADH was dependent upon reaction time, protein concentration, substrate concentration, and the presence of NAD. Inclusion of GSH or 4-methylpyrazole in the incubation mixture resulted in inhibition of HBO formation. Based on these results and other lines of evidence, a mechanism is proposed for HBO formation involving generation of several potentially reactive intermediates which could contribute to toxicity of 1,3-butadiene in exposed individuals. Comparison of kinetics of BDD oxidation in rat, mouse, and human liver cytosol did not reveal significant differences in catalytic efficiency (Vmax/K(m)) between species. These results may contribute to a better understanding of 1,3-butadiene metabolism and toxicity.


Assuntos
Álcool Desidrogenase/farmacologia , Glicóis/metabolismo , Adolescente , Adulto , Animais , Cromatografia Gasosa , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Cinética , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Oxirredução , Ratos , Ratos Sprague-Dawley
10.
Toxicol Appl Pharmacol ; 135(1): 110-8, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7482530

RESUMO

Vinyl carbamate epoxide (VCO) is believed to be the metabolite of ethyl carbamate (EC) ultimately responsible for its carcinogenic effects. This study investigates the role of glutathione (GSH) in protection against VCO-mediated adduct formation, and the involvement of glutathione S-transferases (GSTs) in detoxification of VCO. Formation of 1,N6-ethenoadenosine from VCO and adenosine in vitro was employed as a measure of VCO toxicity. GSH inhibited formation of ethenoadenosine in a concentration-dependent manner at concentrations ranging from 1 to 8 mM. This effect was significantly enhanced by addition of rat liver GST. Mouse liver cytosol was also found to inhibit formation of ethenoadenosine in a concentration-dependent manner, and the inhibition was relieved by addition of S-octylglutathione, a competitive inhibitor of GST. Pretreatment of mice with 1% dietary (2(3)-tert-butyl-4-hydroxyanisole (BHA) caused parallel increases in cytosolic GST activity and cytosolic enhancement of detoxification of VCO by GSH. Furthermore, BHA increased hepatic steady-state concentrations of GSH greater than twofold. The effect of BHA on detoxification of EC in vivo was examined using formation of 2-oxoethylvaline (OEV) adducts of hemoglobin as a biomarker. Pretreatment with BHA decreased overall formation of OEV adducts 23%. The major conclusions of this study are (1) VCO can be detoxified by spontaneous conjugation with GSH, (2) conjugation of VCO with GST can be catalyzed by GST(s), (3) pretreatment with BHA protects against binding of active EC metabolites in vitro and in vivo, and (4) the protective effect of BHA against EC is mediated by increases in GST activity and GSH concentration.


Assuntos
Carcinógenos/farmacocinética , Glutationa Transferase/farmacocinética , Uretana/análogos & derivados , Uretana/farmacocinética , Animais , Hidroxianisol Butilado/farmacologia , Catálise , Citosol/metabolismo , Relação Dose-Resposta a Droga , Glutationa/farmacologia , Glutationa Transferase/farmacologia , Hemoglobinas/efeitos dos fármacos , Hemoglobinas/metabolismo , Inativação Metabólica , Fígado/química , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos A , Uretana/toxicidade
11.
Drug Metab Dispos ; 19(2): 388-93, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1676642

RESUMO

Ethanol and a variety of other compounds previously have been shown to acutely inhibit the metabolism of ethyl carbamate (EC) when given concurrently in mice. On the other hand, ethanol pretreatment (10% in drinking water for the period 48 to 12 hr prior to EC treatment) is known to have the opposite effect and enhance the clearance of EC from blood of mice. In the present work, acetone has been shown to act similarly. Concurrent acetone treatment inhibits the metabolism of EC (11.1 mg/kg po) in male A/JAX mice in a dose-response manner. Blood clearance (Cl) of this po dose of EC from mice following concurrent acetone treatment (50 mg/kg, 0.86 mmol/kg ip) averaged 185 +/- 5.4 (SE) ml hr-1 kg-1 vs. controls of 804 +/- 24.6 ml hr-1 kg-1. Comparing doses that produce equal effects on the blood clearance values of EC, acetone is approximately 50-fold more potent as an inhibitor than ethanol. Pretreatment of mice with acetone (2 g/kg ip) 48 hr and 24 hr before EC administration po increased the clearance of EC approximately 3-fold (CI = 2623 +/- 123 ml hr-1 kg-1). 2-Propanol was found to be at least as potent as inhibitor as acetone, but with a longer duration of inhibition; this longer duration was explained by the longer persistence of acetone in blood from conversion of 2-propanol to acetone.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Acetona/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Uretana/metabolismo , 1-Propanol/sangue , Acetona/sangue , Animais , Inibidores das Enzimas do Citocromo P-450 , Etanol/sangue , Masculino , Camundongos , Camundongos Endogâmicos A , Estreptozocina/metabolismo , Estreptozocina/urina
12.
Drug Metab Dispos ; 19(1): 239-40, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1673406

RESUMO

Acute administration of ethanol, acetaldehyde, dimethyl sulfoxide and several other compounds has been reported previously by this laboratory to inhibit the metabolism of ethyl carbamate (E.C.) in mice. Since many enzyme systems that are inhibited by a compound are also induced by that chemical, the effect of chronic administration of ethanol on the metabolism of E.C. was studied in male, A/JAX mice. Ethanol was given in three pretreatment schedules: 1, 5% in drinking water for 7 days with a 24-hr washout before E.C.; 2, 10% in drinking water 48-12 hr before E.C.; 3, 5 g/kg orally as 10% in saline 48 and 24 hr before E.C. E.C. (11.125 mg/kg) in saline was administered orally and blood samples taken at frequent intervals for analysis of E.C. by a GC/MS technique developed in this laboratory. AUCs of E.C. concentration vs. time were calculated by trapezoidal estimation. From these data, E.C. blood clearance values (dose/AUC; ml hr-1kg-1) were calculated: control, 751 +/- 49.7; group 1,803 +/- 43.5; group 2, 1225 +/- 24.6; group 3, 815 +/- 75.4. Only group 2 was significantly different (p less than 0.01) from control and other groups by Newman-Keuls test. These results indicate that ethanol may be an inducer of E.C. metabolism only under certain limited conditions. The induction may be detected 12 hr after ethanol administration but is not apparent at 24 hr after ethanol pretreatment.


Assuntos
Etanol/farmacologia , Uretana/metabolismo , Animais , Biotransformação , Etanol/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Camundongos , Camundongos Endogâmicos
13.
Biomed Environ Mass Spectrom ; 19(1): 27-31, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2306547

RESUMO

Methodology is presented for convenient, reproducible and direct measurement of blood concentrations of ethyl carbamate, an experimental animal carcinogen. Extraction techniques requiring 20 microliters of blood and selected ion monitoring using ethyl (13C, 15N)carbamate as internal standard enabled quantification of ethyl carbamate concentrations ranging from 50 ng ml-1 to 100 micrograms ml-1. Coefficients of variation at several representative concentrations averaged less than 4%. The method was used to determine the time course of elimination of ethyl carbamate from mice receiving doses of 125 mumol kg-1.


Assuntos
Uretana/sangue , Animais , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Camundongos , Camundongos Endogâmicos A , Uretana/farmacocinética
14.
Am J Ophthalmol ; 73(1): 140, 1972 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-4550198
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...