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










Base de dados
Intervalo de ano de publicação
1.
Xenobiotica ; 40(7): 476-84, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20429841

RESUMO

1. 4-Oxo-4,5,6,7-tetrahydro-1H-indole-3-carboxylic acid (4-methylaminomethyl-phenyl)-amide (1), developed for general anxiety disorder, was discontinued from clinical development due to unsuitable oral pharmacokinetics. 2. In humans, (1) demonstrated an unacceptable high apparent oral clearance (Cl(p)/F) that also demonstrated a supraproportional dose-exposure relationship. Secondary peaks in the plasma concentration-time profile suggested possible enterohepatic recirculation of (1). A combination of in vitro mechanistic tools was applied to better understand the processes underlying these complex clinical pharmacokinetic profiles of (1). 3. In metabolism experiments, (1) was shown to be a substrate of monoamine oxidase A (MAO-A) as well as being metabolized by cytochrome P450. The former appeared to be a high K(M) process with a high capacity, while the latter showed saturation between 1 and 10 microM, consistent with the supraproportional dose-exposure relationship. 4. In a sandwich-cultured hepatocyte model, (1) was shown to be a substrate for both uptake and efflux into the canicular space, which is consistent with the observation of pharmacokinetics suggestive of enterohepatic recirculation. Finally, in human epithelial colon adenocarcinoma cell line (Caco-2) and Madin-Darby canine kidney cells transwell flux experiments, (1) was shown to have relatively low permeability and a basolateral-to-apical flux ratio consistent with the activity of P-glycoprotein. 5. In combination, a compounding of the contributions of MAO-A, hepatic uptake and efflux transporters, and P-glycoprotein to the disposition of (1) may underlie the low oral exposure, saturable clearance, and aberrant concentration versus time profiles observed for this compound in humans.


Assuntos
Anilidas/metabolismo , Anilidas/farmacocinética , Ansiolíticos/metabolismo , Ansiolíticos/farmacocinética , Agonistas de Receptores de GABA-A , Indóis/metabolismo , Indóis/farmacocinética , Anilidas/química , Animais , Ansiolíticos/química , Linhagem Celular Tumoral , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Relação Dose-Resposta a Droga , Humanos , Técnicas In Vitro , Indóis/química , Fígado/metabolismo , Estrutura Molecular , Monoaminoxidase/metabolismo
2.
Antimicrob Agents Chemother ; 45(9): 2529-35, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11502525

RESUMO

Clinafloxacin (CI-960) is a potent broad-spectrum, fluoroquinolone antibiotic that has been studied for parenteral and oral administration in patients with serious infections. The objectives of these studies were to examine the pharmacokinetics and safety of clinafloxacin following administration of single and twice-daily intravenous (i.v.) and oral doses to volunteers. Plasma and urine samples were assayed by validated liquid chromatographic methods, and pharmacokinetic parameter values were determined by noncompartmental methods. Safety was evaluated by clinical observation and laboratory tests. Absorption was rapid after oral administration, with maximum concentrations in plasma (C(max)) generally occurring within 2 h. Concentrations in plasma declined biexponentially, with an average terminal half-life of 4 to 6 h after single doses and 5 to 7 h after multiple doses. Increases in C(max) and area under the concentration-time curves (AUC) were generally proportional to the dose. The volume of distribution was much greater than total body water. Approximately 40 to 75% of the clinafloxacin doses were excreted unchanged into urine. Absolute bioavailability of orally administered clinafloxacin was approximately 90% and did not change with increasing dose. Therefore, switching patients from i.v. to oral dosing should achieve similar concentrations in plasma. The tolerability of clinafloxacin was acceptable. No serious adverse events occurred. C(max) values and minimum plasma clinafloxacin concentrations during multiple dosing exceeded MICs for a wide range of organisms.


Assuntos
Anti-Infecciosos/farmacocinética , Fluoroquinolonas , Neutropenia/metabolismo , Administração Oral , Adolescente , Adulto , Idoso , Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/efeitos adversos , Anti-Infecciosos/sangue , Disponibilidade Biológica , Feminino , Humanos , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade
3.
J Med Chem ; 44(14): 2319-32, 2001 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11428926

RESUMO

Due largely to the emergence of multi-drug-resistant HIV strains, the development of new HIV protease inhibitors remains a high priority for the pharmaceutical industry. Toward this end, we previously identified a 4-hydroxy-5,6-dihydropyrone lead compound (CI-1029, 1) which possesses excellent activity against the protease enzyme, good antiviral efficacy in cellular assays, and promising bioavailability in several animal species. The search for a suitable back-up candidate centered on the replacement of the aniline moiety at C-6 with an appropriately substituted heterocyle. In general, this series of heterocyclic inhibitors displayed good activity (in both enzymatic and cellular tests) and low cellular toxicity; furthermore, several analogues exhibited improved pharmacokinetic parameters in animal models. The compound with the best combination of high potency, low toxicity, and favorable bioavailabilty was (S)-3-(2-tert-butyl-4-hydroxymethyl-5-methyl-phenylsulfanyl)-4-hydroxy-6-isopropyl-6-(2-thiophen-3-yl-ethyl)-5,6-dihydro-pyran-2-one (13-(S)). This thiophene derivative also exhibited excellent antiviral efficacy against mutant HIV protease and resistant HIV strains. For these reasons, compound 13-(S) was chosen for further preclinical evaluation.


Assuntos
Inibidores da Protease de HIV/síntese química , Pironas/síntese química , Sulfetos/síntese química , Animais , Disponibilidade Biológica , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Cães , Avaliação Pré-Clínica de Medicamentos , Resistência Microbiana a Medicamentos , HIV/efeitos dos fármacos , Protease de HIV/química , Protease de HIV/genética , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/farmacocinética , Inibidores da Protease de HIV/farmacologia , HIV-1/enzimologia , Humanos , Linfócitos/virologia , Camundongos , Mutação , Pironas/química , Pironas/farmacocinética , Pironas/farmacologia , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Sulfetos/química , Sulfetos/farmacocinética , Sulfetos/farmacologia
4.
J Med Chem ; 43(5): 843-58, 2000 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-10715152

RESUMO

On the basis of previous SAR findings and molecular modeling studies, a series of compounds were synthesized which possessed various sulfonyl moieties substituted at the 4-position of the C-3 phenyl ring substituent of the dihydropyran-2-one ring system. The sulfonyl substituents were added in an attempt to fill the additional S(3)' pocket and thereby produce increasingly potent inhibitors of the target enzyme. Racemic and enantiomerically resolved varieties of selected compounds were synthesized. All analogues in the study displayed decent binding affinity to HIV protease, and several compounds were shown to possess very good antiviral efficacy and safety margins. X-ray crystallographic structures confirmed that the sulfonamide and sulfonate moieties were filling the S(3)' pocket of the enzyme. However, the additional substituent did not provide improved enzymatic inhibitory or antiviral activity as compared to the resolved unsubstituted aniline. The addition of the sulfonyl moiety substitution does not appear to provide favorable pharamacokinectic parameters. Selected inhibitors were tested for antiviral activity in clinical isolates and exhibited similar antiviral activity against all of the HIV-1 strains tested as they did against the wild-type HIV-1. In addition, the inhibitors exhibited good antiviral efficacies against HIV-1 strains that displayed resistance to the currently marketed protease inhibitors.


Assuntos
Sulfonatos de Arila/síntese química , Inibidores da Protease de HIV/síntese química , Piranos/síntese química , Sulfonamidas/síntese química , Animais , Sulfonatos de Arila/química , Sulfonatos de Arila/farmacocinética , Sulfonatos de Arila/farmacologia , Disponibilidade Biológica , Linhagem Celular , Cristalografia por Raios X , Inibidores das Enzimas do Citocromo P-450 , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/farmacocinética , Inibidores da Protease de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/isolamento & purificação , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/virologia , Camundongos , Modelos Moleculares , Piranos/química , Piranos/farmacocinética , Piranos/farmacologia , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia
5.
J Clin Pharmacol ; 39(11): 1143-50, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10579144

RESUMO

The pharmacokinetics of R-clinafloxacin and S-clinafloxacin enantiomers of the broad-spectrum fluoroquinolone antibiotic, clinafloxacin, were characterized in selected volunteer subjects and patients after the administration of oral and intravenous doses of racemic drug. The absorption of each enantiomer was rapid and nearly complete after a single, oral 400 mg racemic dose. The mean (+/- SD) bioavailability of R-clinafloxacin was 87.5% +/- 4.8% compared to 86.2% +/- 5.8% for S-clinafloxacin. The mean Cmax of each enantiomer was 1.19 micrograms/mL, with plasma concentrations of each enantiomer remaining above 0.1 microgram/mL for at least 12 hours. No notable differences in the disposition of R-clinafloxacin and S-clinafloxacin were observed. After a single 400 mg intravenous dose of racemic drug, mean (+/- SD) t1/2 was 5.6 +/- 0.3 hours and 5.7 +/- 0.4 hours, plasma Cl was 329 +/- 49 mL/min and 314 +/- 45 mL/min, and Vdss was 138 +/- 18 L and 134 +/- 16 L for R- and S-clinafloxacin, respectively. Two healthy volunteers each received a single 400 mg oral dose of racemic clinafloxacin (alone) and with oral administration of 1 gm probenecid separated by a 1-week washout period between treatments. With probenecid coadministration, the increase in AUC0-infinity was 75% and 83% for R-clinafloxacin and was 71% and 75% for S-clinafloxacin in each subject, respectively. Probenecid increased the total exposure (AUC) of both R-clinafloxacin and S-clinafloxacin, although it had no stereo-selective effects on the disposition of either enantiomer. The antimicrobial potency of the isomers was also evaluated. In vitro susceptibility testing showed that the two compounds were comparable in their inhibitory activities, as all MICs were within twofold for each organism tested. These results demonstrate that in addition to their similar antimicrobial potency, R- and S-clinafloxacin have nearly identical disposition characteristics and are eliminated by similar mechanisms that display no apparent enantioselectivity in man.


Assuntos
Anti-Infecciosos/farmacocinética , Fluoroquinolonas , Adulto , Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/sangue , Bactérias/efeitos dos fármacos , Disponibilidade Biológica , Interações Medicamentosas , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Probenecid/farmacologia , Estereoisomerismo , Fatores de Tempo
6.
Bioorg Med Chem Lett ; 9(11): 1481-6, 1999 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-10386921

RESUMO

Dihydropyran-2-ones possessing amino and carboxamide functionalities on 3-SPh (2-tert-butyl, 5-methyl) ring were synthesized and evaluated for their antiviral activities. Both the enantiomers of inhibitor 15 were synthesized. The in vitro resistance profile, inhibitory activities against cytochrome P450 isozymes and pharmacokinetic properties of inhibitor 15S will be discussed.


Assuntos
Inibidores da Protease de HIV/síntese química , Inibidores da Protease de HIV/farmacocinética , Piranos/síntese química , Piranos/farmacocinética , Animais , Fármacos Anti-HIV/farmacologia , Cristalografia por Raios X , Cães , Concentração Inibidora 50 , Cinética , Camundongos , Modelos Moleculares , Relação Estrutura-Atividade
7.
J Chromatogr B Biomed Sci Appl ; 709(2): 265-72, 1998 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-9657223

RESUMO

Achiral and chiral HPLC methods were developed for clinafloxacin, a quinolone antimicrobial agent. For achiral assay, analytes were isolated from plasma by precipitating plasma proteins. Separation was achieved on a C18 column using an isocratic eluent of ion pairing solution-acetonitrile (80:20, v/v) at 1.0 ml/min with UV detection at 340 nm. The ion pairing solution was 0.05 M citric acid, 1.15 mM tetrabutylammonium hydroxide and 0.1% ammonium perchlorate. Inter-assay accuracy was within 4.9% with an inter-assay precision of 3.7% over a quantitation range of 0.025 to 10.0 microg/ml. For chiral assay, analytes were isolated from plasma by solid-phase extraction. Separation was achieved on a Crownpak CR(+) column using an isocratic eluent of water-methanol (88:12, v/v) containing 0.1 mM decylamine at 1.0 ml/min with UV detection at 340 nm. Perchloric acid was added to adjust pH to 2. Inter-assay accuracy was within 3.5% with a inter-assay precision of 5.4% over a quantitation range of 0.040 to 2.5 microg/ml.


Assuntos
Anti-Infecciosos/sangue , Fluoroquinolonas , Quinolonas/sangue , Anti-Infecciosos/administração & dosagem , Cromatografia Líquida de Alta Pressão/métodos , Estabilidade de Medicamentos , Humanos , Infusões Intravenosas , Quinolonas/administração & dosagem , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Estereoisomerismo
8.
J Chromatogr B Biomed Appl ; 668(1): 141-51, 1995 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-7550971

RESUMO

CI-980, a 1-deaza-7,8-dihydropteridine, is a novel anticancer agent that is a potent mitotic inhibitor acting as a tubulin binder similar to the vinca alkaloids. CI-980 has shown equivalent or superior anticancer activity in vitro compared to vincristine and retains full activity against vincristine resistant tumors in vitro. A high-performance liquid chromatographic (HPLC) assay was developed and validated for human plasma and urine to support Phase 1 clinical trials. CI-980 and PD 080658, internal standard, were isolated from 2-ml samples of human plasma and urine by solid-phase extraction with Bond-Elut C18 cartridges. Urine samples must be pretreated with bovine serum albumin (BSA) to minimize the binding of CI-980 to glass and some plastics. The eluate from the cartridges for both matrices was evaporated to dryness and taken up in mobile phase. Zorbax RX C18 columns, mobile phase buffer of 10 mM ammonium dihydrogen phosphate at pH 7.5 and a flow-rate of 0.75 ml/min were used for both matrices. Column dimensions, column temperature and mobile phase acetonitrile-buffer ratio were 300 mm x 4.6 mm I.D., 30 degrees C and 38:62 (v/v), respectively, for the plasma assay and 250 mm x 4.6 mm I.D., 35 degrees C and 40:60 (v/v), respectively, for the urine assay. Column effluent was monitored fluorometrically for the plasma method using excitation and emission wavelengths of 388 nm and 473 nm, respectively. Ultraviolet detection at 380 nm was used for the urine method. Peak-area ratios were proportional to CI-980 concentrations from 0.2 to 25 ng/ml and 1 to 100 ng/ml for plasma and urine, respectively. CI-980 in water will bind to glass and plastics but not PTFE or stainless steel. Urine calibration standards were frozen prior to use in order to compensate for loss of CI-980 due to freezing in this matrix. The accuracy of the assay was within 4.7%, with a precision of 5.6% for both matrices. Recoveries ranged from 93.8 to 102% and 90.7 to 92.3% for plasma and urine, respectively. CI-980 was stable in plasma and urine for at least 275 and 217 days, respectively, when stored at -70 degrees C. The assay is suitable for studying the clinical pharmacokinetics of CI-980.


Assuntos
Antineoplásicos/farmacocinética , Carbamatos/farmacocinética , Cromatografia Líquida de Alta Pressão/métodos , Pirazinas/farmacocinética , Piridinas/farmacocinética , Antineoplásicos/sangue , Antineoplásicos/urina , Carbamatos/sangue , Carbamatos/urina , Estabilidade de Medicamentos , Congelamento , Humanos , Pirazinas/sangue , Pirazinas/urina , Piridinas/sangue , Piridinas/urina , Reprodutibilidade dos Testes , Espectrometria de Fluorescência
9.
Cancer ; 67(2): 367-71, 1991 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-1985732

RESUMO

5-Fluorouracil (5-FU) was delivered in a dose-intensive schedule to 23 patients with metastatic or unresectable colorectal carcinoma. The schedule consisted of bolus single-dose 5-FU therapy 400 to 500 mg followed by 4-day infusion of 5-FU, 600 to 800 mg/m2/day, followed by a 17-day to 24-day infusion of 200 to 250 mg/m2/day. Partial remissions were seen in 22% of all eligible patients. Significant toxicity, including mucositis, diarrhea, and hand-foot syndrome, necessitated dose reductions in most patients. The authors conclude that 5-FU given in this moderately intensive schedule is associated with a moderate level of response, as easily achieved with more conventional schedules or with 5-FU-leucovorin combinations. Tumor responsiveness to dose intensive 5-FU regimens may be limited.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Fluoruracila/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/metabolismo , Esquema de Medicação , Feminino , Fluoruracila/efeitos adversos , Fluoruracila/farmacocinética , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Neoplasias Pélvicas/tratamento farmacológico , Neoplasias Pélvicas/secundário , Indução de Remissão , Neoplasias Retroperitoneais/tratamento farmacológico , Neoplasias Retroperitoneais/secundário
10.
Cancer Chemother Pharmacol ; 27(1): 27-32, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2245490

RESUMO

Both the capacity of healthy human, cancer patient, and mouse plasma proteins to bind flavone acetic acid (FAA) and the qualitative differences in the plasma protein-binding site were studied. The binding capacity of plasma proteins for FAA was saturated within the therapeutic range in both species. The binding of FAA to plasma protein was significantly greater in both healthy human and cancer patient plasma than in mouse plasma. Plasma from patients with cancer bound on the average less FAA than did healthy patient plasma. The concentration of albumin in the plasma varied between healthy humans, cancer patients, and mice, being 5.3 +/- 0.7, 4.7 +/- 0.8, and 3.9 +/- 0.3 g/100 ml, respectively. The protein binding of FAA was found to be dependent on the plasma albumin concentration, but albumin concentration alone was not adequate for the accurate prediction of the percentage of FAA protein bound. Scatchard plots indicated that healthy human plasma had a greater number of high-affinity binding sites than did mouse plasma. FAA binds at the indolebenzodiazepine binding area on albumin and can be displaced from this site by salicylic acid and clofibric acid, but only at supratherapeutic concentrations. Our results indicate that alterations in plasma albumin could contribute to a variable effect with FAA. Therefore, the influence of serum albumin concentration and the nonlinearity of FAA protein binding should be considered in assessment of the appropriateness of a dose schedule for FAA.


Assuntos
Proteínas Sanguíneas/metabolismo , Flavonoides/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Ácido Clofíbrico/farmacologia , Humanos , Técnicas In Vitro , Camundongos , Neoplasias/sangue , Ligação Proteica , Salicilatos/farmacologia , Ácido Salicílico , Albumina Sérica/análise
11.
Drug Metab Dispos ; 18(1): 20-7, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-1970773

RESUMO

Metabolism of benzene by the liver has been suggested to play an important role in the hepatotoxicity of benzene. The role of the different benzene metabolites and the causes of species differences in benzene hepatotoxicity are, however, not known. The metabolism and covalent binding of 14C-benzene by liver microsomal fractions and liver slices from rat, mouse, and human subjects have been studied. Rat microsomal fraction formed phenol at a rate of 0.32 nmol/min/mg of protein; mouse microsomal fraction formed phenol at 0.64 nmol/min/mg and hydroquinone at 0.03 nmol/min/mg; and human microsomal fraction formed phenol at 0.46 nmol/min/mg and hydroquinone at 0.07 nmol/min/mg. Covalent binding of 14C-benzene metabolites to rat, mouse, and human liver microsomal protein was 29, 113, and 169 pmol/min/mg of protein, respectively. The rates of metabolite formation from benzene by liver slices in nmol/min/g of tissue were: rat, phenol 0.15, hydroquinone 0.26, and phenylsulfate 1.22; mouse: phenol 0.13, hydroquinone 0.29, phenylsulfate 1.37, and phenylglucuronide 1.34; and human: phenol 0.16, hydroquinone 0.27, phenylsulfate 0.83, and phenylglucuronide 0.52. trans,trans-Muconic acid formation was not detected with liver slices of any species. Covalent binding of 14C-benzene metabolites to rat, mouse, and human liver slices was 8.2, 79.7, and 27.3 pmol/min/g liver, respectively. There was no correlation between ascorbic acid levels in the human liver slices and covalent binding of 14C-benzene metabolites. The results show that phenol and hydroquinone found in extrahepatic tissues, including bone marrow, of animals exposed to benzene could originate from the liver. There was no evidence for the release of highly reactive benzene metabolites such as trans,trans-muconaldehyde or p-benzoquinone from liver cells.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Benzeno/metabolismo , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Adolescente , Idoso , Animais , Ácido Ascórbico/metabolismo , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos F344 , Especificidade da Espécie , Espectrofotometria Ultravioleta , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo
12.
Cancer Chemother Pharmacol ; 24(5): 277-83, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2758557

RESUMO

The chemical breakdown of carmethizole [1-methyl-2-methylthio-4,5-bis-(hydroxymethyl)imidazole-4',5'- bis(N-methylcarbamate)hydrochloride] and its pharmacokinetics in the mouse and beagle dog were studied. Carmethizole was relatively unstable in aqueous media, having a half-life of less than or equal to 1 h in 0.9% sodium chloride, human whole blood, human plasma, and dog urine at 37 degrees C. Its major breakdown product in 0.9% sodium chloride and pH 5.0 sodium phosphate buffer was carmethizole diol. When carmethizole was added to pH 7.0 or pH 9.0 sodium phosphate buffer, the major breakdown product was carmethizole diol-4'-monophosphate. Carmethizole reacted directly with glutathione at pH 8.0, forming a glutathione adduct of carmethizole monocarbamate. Elimination of the drug from the plasma of the beagle dog following i.v. bolus doses of 22.4 and 4.3 mg/kg was biphasic. At these doses the terminal half-life was 39 and 46 min, respectively, and the respective total body clearance was 4.6 and 7.7 ml/min per kg. The 22.4 mg/kg dose was lethal to the beagle dog by day 4. Elimination of carmethizole from the plasma of mice following an i.v. bolus dose of 115 mg/kg was monoexponential, with a half-life of 11.6 min and a total body plasma clearance of 43.6 ml/min per kg. When the drug was infused at 230 mg/kg over 8 h into mice, the total body clearance was 40.8 ml/min per kg. Following the i.v. bolus administration of carmethizole to mice, 30% of the total dose was excreted in urine over 3 h as carmethizole diol, 10%, as carmethizole diol-sulfate, 3.4%, as carmethizole 4'-monocarbamate, and 2.4%, as unchanged drug.


Assuntos
Antineoplásicos/farmacologia , Imidazóis/farmacologia , Animais , Antineoplásicos/análise , Antineoplásicos/farmacocinética , Cromatografia Líquida de Alta Pressão , Cães , Interações Medicamentosas , Estabilidade de Medicamentos , Glutationa/metabolismo , Concentração de Íons de Hidrogênio , Imidazóis/análise , Imidazóis/farmacocinética , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Endogâmicos F344 , Soluções , Fatores de Tempo
13.
J Chromatogr ; 427(2): 247-55, 1988 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-3410909

RESUMO

A sensitive gas chromatographic assay with electron-capture detection has been developed for sulfamic acid diester (sulfamic acid 1,7-heptanediyl ester, NSC 329680) based on its conversion to 1,7-diiodoheptane in the presence of excess sodium iodide. The assay is linear up to 1 microgram/ml sulfamic acid diester and has a lower limit of detection of 25 ng/ml from 0.5 ml plasma. The coefficient of variation of the assay is 6.4% at 1 microgram/ml and 8.0% at 100 ng/ml. Sulfamic acid diester is relatively stable in 0.9% sodium chloride and 0.1 M sodium phosphate buffers, pH 7.0-9.0, with half-lives greater than 38 h. The major breakdown product of sulfamic acid diester is sulfamic acid 1,7-heptane-monoyl ester. When added to whole blood sulfamic acid diester shows concentration-dependent breakdown. At 50 and 100 micrograms/ml sulfamic acid diester, the half-time in whole blood is 6.9 h and 65% of the drug is sequestered by the blood cells. At 10 micrograms/ml sulfamic acid diester in blood, there is no detectable breakdown of the drug over 24 h and all of the drug is sequestered by the blood cells. Protein binding of sulfamic acid diester in human plasma is 82% at 10 micrograms/ml and 68% at 100 micrograms/ml.


Assuntos
Antineoplásicos/análise , Ácidos Sulfônicos/análise , Antineoplásicos/sangue , Antineoplásicos/urina , Proteínas Sanguíneas/análise , Cromatografia Gasosa , Eritrócitos/análise , Humanos , Espectrometria de Massas , Ácidos Sulfônicos/sangue , Ácidos Sulfônicos/urina
14.
Invest New Drugs ; 6(1): 3-9, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3410664

RESUMO

The antitumor drug pyrazine-2-diazohydroxide exhibits cytotoxicity to A204 tumor cells in vitro under acid conditions. The IC50 with a 1 hr drug exposure at pH of 7.4 was 61 micrograms/ml and at pH of 6.0 it was 31 micrograms/ml. It is suggested that the increased cytotoxicity is due to the acid catalyzed formation of a reactive pyrizinyldiazonium ion from pyrazine-2-diazohydroxide. Pyrazine-2-diazohydroxide is also more cytotoxic to A204 cells under hypoxic conditions in the presence of glucose with an IC50 at pH 7.4 of 22 micrograms/ml. The increased cytotoxicity of pyrazine-2-diazohydroxide under acid and hypoxic conditions may favor selective toxicity to solid tumors in vivo. Coincubation with rat hepatic microsomes increased the cytotoxicity of pyrazine-2-diazohydroxide to A204 cells. The effect did not require NADPH and was not due to formation of metabolites. There was an increased rate of degradation of pyrazine-2-diazohydroxide in the presence of microsomes, presumably with formation of the pyrizinyldiazonium ion. The final degradation product 2-hydroxypyrazine was not cytotoxic to A204 cells. The effect of microsomes on pyrazine-2-diazohydroxide cytotoxicity is probably of little in vivo significance.


Assuntos
Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Pirazinas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Animais , Meia-Vida , Humanos , Concentração de Íons de Hidrogênio , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Endogâmicos F344 , Rabdomiossarcoma/patologia , Compostos de Sulfidrila/metabolismo , Células Tumorais Cultivadas/patologia
15.
Cancer Chemother Pharmacol ; 21(4): 269-73, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3370734

RESUMO

The pharmacokinetics and metabolism of pyrazine-2-diazohydroxide have been studied in the beagle dog and mouse. When pyrazine-2-diazohydroxide was administered to beagle dogs at a dose of 18.6 mg/kg (428 mg/m2) by i.v. bolus, the plasma half-life (t1/2) was 7.3 min, the apparent volume of distribution (Vd) 577 ml/kg, and the total body clearance (Cl) 55 ml/min per kg. In mice given pyrazine-2-diazohydroxide by i.v. bolus at 100 mg/kg (428 mg/m2), the t1/2 was 5.8 min, the Vd 250 ml/kg, and the Cl 30 ml/min per kg. When [2-14C]pyrazine-2-diazohydroxide was infused i.v. to mice at 100 mg/kg over 8 h, the Cl for parent drug was 122 ml/min per kg. The major product formed from pyrazine-2-diazohydroxide was 2-hydroxypyrazine, which accounted for 80% of the total radioactivity in the plasma after a 6-h drug infusion. There were three other metabolites in plasma, two more polar than pyrazine-2-diazohydroxide, which accounted for 7% of the radioactivity, and one less polar, which accounted for 5% of the radioactivity. Following an i.v. bolus dose of [2-14C]pyrazine-2-diazohydroxide, 79% of the radioactivity was excreted in the urine in 24 h, 3% in the feces, and 0.4% in the expired air; 18% remained in the carcass. The liver and kidney showed the highest tissue levels of radioactivity. 2-Hydroxypyrazine accounted for 45% of the urinary radioactivity, pyrazine-2-diazohydroxide for 14%, and a glucuronide or sulfate conjugate of 2-hydroxypyrazine for 17%. Twenty-four percent of the radioactivity eluted near the void volume on high-performance liquid chromatography and was not identified.


Assuntos
Antineoplásicos/farmacocinética , Pirazinas/farmacocinética , Animais , Cães , Meia-Vida , Taxa de Depuração Metabólica , Camundongos , Distribuição Tecidual
16.
Cancer Chemother Pharmacol ; 22(2): 120-5, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3409442

RESUMO

The pharmacokinetics and metabolism of sulfamic acid diester were studied in the beagle dog and mouse. Elimination of sulfamic acid diester from the plasma and whole blood following i.v. administration at a dose of 193 mg/m2 was best approximated by a three-compartment model in both species. The compound was relatively rapidly cleared from the plasma, with a plasma beta half-life of 2.3 h and 0.9 h and a gamma half-life of 16 h and 3 h in the dog and the mouse, respectively. Sulfamic acid diester was taken up by blood cells and only slowly eliminated with a whole blood gamma half-life of 42 h in the dog and 32 h in the mouse. When sulfamic acid diester was infused i.v. to mice at 15 mg/kg over 8 h, the clearance for the parent drug was 13.2 ml/min kg from the plasma and 3.3 ml/min kg from the whole blood. Urine collected from mouse and dog contained the parent drug and three metabolic/breakdown products, namely, sulfamic acid 1,7-heptanemonoyl ester, sulfamic acid 3-hydroxyl-1,7-heptanediyl ester, and an unidentified product. Excretion of unchanged drug and products in mouse urine over 8 h accounted for less than 16% of the dose of sulfamic acid diester. Sulfamic acid diester did not react with glutathione in buffer, whole blood, or 100,000 g rat liver cytosol.


Assuntos
Antineoplásicos/farmacocinética , Ácidos Sulfônicos/farmacocinética , Animais , Antineoplásicos/metabolismo , Cães , Glutationa/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Ratos , Ácidos Sulfônicos/metabolismo
17.
Biochem Pharmacol ; 36(19): 3161-7, 1987 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-3663231

RESUMO

Ascorbic acid deficiency causes qualitative and quantitative differences in the guinea pig hepatic flavin-containing monooxygenase (FMO). Kinetic studies with purified FMO indicated no significant change in the apparent Km of dimethylaniline or NADPH in ascorbate-supplemented or -deficient animals. Following purification of ascorbate-deficient guinea pig FMO by DEAE-cellulose and blue agarose chromatography, exogenous FAD was required for 15% of the FMO microsomal activity recovered. In contrast, only 5% of the total microsomal enzyme recovered from ascorbate-supplemented animals required exogenous FAD. Furthermore, there was an enhanced sensitivity to time-dependent nonlinearity with the purified ascorbate-deficient guinea pig FMO. The degree of time-dependent nonlinearity was related to the concentration of substrate. Also, purified ascorbate-supplemented guinea pig FMO was stable for 4 weeks at -20 degrees, whereas the ascorbate-deficient enzyme was inactivated. A decrease in the quantity of ascorbate-deficient guinea pig FMO compared to ascorbate-supplemented was indicated by a marked reduction in total FMO activity recovered from blue agarose chromatography and reduced protein staining intensity with SDS-PAGE at 56,000 daltons.


Assuntos
Deficiência de Ácido Ascórbico/enzimologia , Oxigenases/análise , Animais , Ácido Ascórbico/farmacologia , Flavina-Adenina Dinucleotídeo/farmacologia , Cobaias , Cinética , Peso Molecular
18.
J Nutr ; 117(2): 286-90, 1987 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2881990

RESUMO

Modulation of the flavin-containing monooxygenase (FMO) by varying the ascorbic acid and food intake was investigated. Hepatic activity of the FMO in ascorbic acid-deficient guinea pigs fed a restricted amount of diet which resulted in a 10-15% body weight loss, was 17% of that in animals fed restricted amounts of the adequate diet. FMO hepatic activity in ascorbic acid-supplemented guinea pigs on a food-restricted regimen was 176% of that found in animals fed the adequate diet ad libitum. This increase in activity was not related to stress. Alteration in the activity of this important drug-metabolizing enzyme system by a combination of ascorbic acid deficiency and reduced food intake could potentially alter the rate of metabolism of a great variety of pharmaceutical drugs and environmental chemicals.


Assuntos
Deficiência de Ácido Ascórbico/enzimologia , Privação de Alimentos/fisiologia , Fígado/enzimologia , Oxigenases/metabolismo , Animais , Ácido Ascórbico/farmacologia , Cobaias , Fígado/efeitos dos fármacos , Masculino , Tirosina Transaminase/metabolismo
20.
Biochem Pharmacol ; 35(4): 637-44, 1986 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-3947395

RESUMO

Activity of the flavin-containing monooxygenase (FMO) was reduced significantly in ascorbic acid deficient guinea pigs. Reduction in oxidation of dimethylaniline (DMA) and of thiobenzamide was associated with a decrease in the activity of the FMO. In both ascorbate supplemented and deficient guinea pig hepatic 12,000 g supernatant fractions, SKF-525A and n-octylamine did not inhibit DMA N-oxidation. Phenobarbital pretreatment did not increase the rate of N-oxidation of DMA. In addition, hepatic supernatant fractions thermally treated at 50 degree were unable to N-oxidize DMA, but 80% of the cytochrome P-450 activity was retained. Also, N-oxidation of DMA was reduced by 53% at pH 7.0, while oxidation of cytochrome P-450 specific substrates was inhibited by only 19%. Kinetic studies of DMA N-oxidation indicate no significant change in the apparent Km in ascorbate supplemented or deficient animals. The in vitro addition of ascorbic acid had no effect on the activity of the FMO. The toxicological implications of the reduction in FMO activity in ascorbic acid deficiency are discussed.


Assuntos
Deficiência de Ácido Ascórbico/enzimologia , Oxigenases/metabolismo , Aminopirina N-Desmetilase/metabolismo , Compostos de Anilina/metabolismo , Anilina Hidroxilase/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Redutases do Citocromo/metabolismo , Grupo dos Citocromos b/metabolismo , Citocromo-B(5) Redutase , Citocromos b5 , Cobaias , Temperatura Alta , Concentração de Íons de Hidrogênio , Cinética , Masculino , Oxirredução , Tioamidas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...