Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Antivir Ther ; 3(Suppl 3): 113-21, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-10726061

RESUMO

Preclinical aspects of a potent anti-hepatitis B virus (HBV) L-nucleoside, 1-(2-fluoro-5-methyl-beta-L-arabino-furanosyl)uracil (L-FMAU) are described. L-FMAU was prepared from L-ribose derivatives via either L-xylose or L-arabinose. L-FMAU shows potent antiviral activity against hepatitis B virus (EC50 5.0 microM in H1 cells) with high selectivity in vitro. L-FMAU is not incorporated into mitochondrial DNA and no significant lactic acid production was observed in vitro. L-FMAU is phosphorylated by thymidine kinase as well as deoxycytidine kinase, ultimately to the triphosphate, which inhibits HBV DNA polymerase as the mechanism of antiviral action. Preliminary in vivo toxological studies suggest no apparent toxicity for 30 days at 50 mg/kg/day in mice and for 3 months in woodchucks (10 mg/kg/day). L-FMAU also has respectable bioavailability in rats. L-FMAU shows potent anti-HBV activity in vivo against woodchuck hepatitis virus in chronically infected woodchucks and there is no significant virus rebound after cessation of the drug treatment.


Assuntos
Antivirais/farmacologia , Arabinofuranosiluracila/análogos & derivados , Vírus da Hepatite B/efeitos dos fármacos , Animais , Antivirais/toxicidade , Arabinofuranosiluracila/química , Arabinofuranosiluracila/farmacologia , Arabinofuranosiluracila/toxicidade , Disponibilidade Biológica , Linhagem Celular , Desoxicitidina Quinase/metabolismo , Avaliação Pré-Clínica de Medicamentos , Patos , Infecções por Hepadnaviridae/tratamento farmacológico , Infecções por Hepadnaviridae/virologia , Vírus da Hepatite B do Pato/efeitos dos fármacos , Vírus da Hepatite B da Marmota/efeitos dos fármacos , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Dose Letal Mediana , Marmota , Camundongos , Fosforilação , Ratos , Timidina Quinase/metabolismo
2.
J Med Chem ; 39(26): 5202-7, 1996 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-8978848

RESUMO

As a part of our efforts to design prodrugs for antiviral nucleosides, 9-(beta-D-arabinofuranosyl)-6-azidopurine (6-AAP) was synthesized as a prodrug for ara-A that utilizes the azide reduction biotransformation pathway. 6-AAP was synthesized from ara-A via its 6-chloro analogue 4. The bioconversion of the prodrug was investigated in vitro and in vivo, and the pharmacokinetic parameters were determined. For in vitro studies, 6-AAP was incubated in mouse serum and liver and brain homogenates. The half-lives of 6-AAP in serum and liver and brain homogenates were 3.73, 4.90, and 7.29 h, respectively. 6-AAP was metabolized primarily in the liver homogenate microsomal fraction by the reduction of the azido moiety to the amine, yielding ara-A. However, 6-AAP was found to be stable to adenosine deaminase in a separate in vitro study. The in vivo metabolism and disposition of ara-A and 6-AAP were conducted in mice. When 6-AAP was administered by either oral or intravenous route,the half-life of ara-A was 7-14 times higher than for ara-A administered intravenously. Ara-A could not be found in the brain after the intravenous administration of ara-A. However, after 6-AAP administration (by either oral or intravenous route), significant levels of ara-A were found in the brain. The results of this study demonstrate that 6-AAP is converted to ara-A, potentially increasing the half-life and the brain delivery of ara-A. Further studies to utilize the azide reduction approach on other clinically useful agents containing an amino group are in progress in our laboratories.


Assuntos
Antivirais/farmacocinética , Pró-Fármacos/farmacocinética , Vidarabina/análogos & derivados , Vidarabina/farmacocinética , Animais , Área Sob a Curva , Azidas/química , Biotransformação , Encéfalo/metabolismo , Feminino , Camundongos , Oxirredução , Distribuição Tecidual , Vidarabina/síntese química
3.
J Med Chem ; 39(23): 4676-81, 1996 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-8917656

RESUMO

In an effort to improve the pharmacokinetic properties and tissue distribution of 2'-F-ara-ddI, two lipophilic prodrugs, 6-azido-2'-3'-dideoxy-2'-fluoro-beta-D- arabinofuranosylpurine (FAAddP, 4) and N6-methyl-2'-3'-dideoxy-2'-fluoro-beta-D-arabinofuranosyladenine (FMAddA, 5), were synthesized and their biotransformation was investigated in vitro and in vivo, in mice. Compounds 4 and 5 were synthesized via the intermediate 2. For the in vitro studies, FAAddP and FMAddA were incubated in mouse serum, liver homogenate, and brain homogenate. FAAddP was metabolized in liver homogenate by the reduction of the azido to the amino moiety followed by deamination, yielding 2'-F-ara-ddI. The conversion of FAAddP to 2'-F-ara-ddA was mediated by microsomal P-450 NADPH reductase system, as shown by the liver microsomal assay. FAAddP was also converted to 2'-F-ara-ddI at a slower rate in the brain than in the liver. FMAddA, however, was stable in brain homogenate and was slowly metabolized in the liver homogenate. Metabolic conversion of FMAddA in vitro was stimulated by the addition of adenosine deaminase. In the in vivo metabolism study, FAAddP underwent reduction to 2'-F-ara-ddA followed by deamination to 2'-F-ara-ddI. FMAddA did not result in increased brain delivery of 2'-F-ara-ddI in vivo, probably due to the slow conversion as observed in the in vitro studies. However, there was an increase in the half-life of 2'-F-ara-ddI produced from FMAddA. This report is the first example in the design of prodrugs using the azido group for adenine- and hypoxanthine-containing nucleosides. This interesting and novel approach can be extended to other antiviral and anticancer nucleosides.


Assuntos
Fármacos Anti-HIV/farmacologia , Azidas/farmacocinética , Didanosina/análogos & derivados , Didesoxiadenosina/análogos & derivados , Pró-Fármacos/farmacocinética , Vidarabina/análogos & derivados , Animais , Azidas/química , Biotransformação , Didanosina/farmacologia , Didesoxiadenosina/farmacologia , Feminino , Espectroscopia de Ressonância Magnética , Camundongos , Microssomos Hepáticos/metabolismo , Pró-Fármacos/química , Espectrofotometria Ultravioleta , Vidarabina/química , Vidarabina/farmacocinética
4.
J Med Chem ; 33(8): 2188-92, 1990 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2374145

RESUMO

A significant number of patients with AIDS and AIDS-related complex develop neurological complications. Therefore, it is critical that anti-HIV agents penetrate the blood-brain barrier and suppress viral replication in the brain. In an effort to increase the brain delivery of anti-HIV nucleosides, in vitro and in vivo pharmacokinetics of dihydropyridine derivatives of 3'-azido-2',3'-dideoxyuridine (AzddU, AZDU, or CS-87) and 3'-azido-3'-deoxythymidine (AZT, Zidovudine) have been studied. In vitro studies of the prodrugs (AzddU-DHP and AZT-DHP) in human serum, mouse serum, and mouse brain homogenate indicated that the rates of serum conversion from prodrugs to parent drugs are species dependent: mouse brain homogenate greater than mouse serum greater than human serum. Half-lives in human serum, mouse serum, and mouse brain homogenate are 4.33, 0.56, 0.17 h, respectively, for AzddU and 7.70, 1.40, and 0.18 h, respectively, for AZT. In vivo studies of AzddU-DHP and AZT-DHP showed that the prodrugs have areas under the serum concentration-time curves (AUC) similar to those of the parent drugs. The AUC in serum for AzddU following prodrug administration is 25.79 micrograms h/mL, which is similar to the value of 25.83 micrograms h/mL when AzddU was administered. Analogously, the serum AUCs for AZT when AZT-DHP and AZT were administered are 25.38 and 26.64 micrograms h/mL, respectively. However, the brain AUCs for both AzddU and AZT derived from prodrugs, being 11.43 and 11.28 micrograms h/mL, respectively, are greater than the brain AUCs for AzddU (2.09 micrograms h/mL) and AZT (1.21 micrograms h/mL) when the parent drugs were administered. Thus, the relative brain exposure (re) for AzddU (5.47) and AZT (9.32) indicate a significant increase in exposure to the anti-HIV nucleosides following prodrug administrations. The results of extended half-lives of the synthesized prodrugs in human serum along with the higher re values in vivo warrant studies in larger animals to determine the potential usefulness of the prodrugs in humans.


Assuntos
Antivirais/farmacocinética , Encéfalo/metabolismo , Di-Hidropiridinas/farmacocinética , HIV/efeitos dos fármacos , Pró-Fármacos/farmacocinética , Zidovudina/análogos & derivados , Animais , Fenômenos Químicos , Química , Di-Hidropiridinas/síntese química , Meia-Vida , Humanos , Camundongos , Estrutura Molecular , Pró-Fármacos/síntese química , Zidovudina/administração & dosagem , Zidovudina/síntese química , Zidovudina/farmacocinética
5.
Clin Pharmacokinet ; 38(3): 271-90, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10749520

RESUMO

The plasma protein binding of drugs, particularly those that are highly bound, may have significant clinical implications. Although protein binding is a major determinant of drug action, it is only one of a myriad of factors that influence drug disposition. The extent of protein binding is a function of drug and protein concentrations, the affinity constant for the drug-protein interaction and the number of protein binding sites per class of binding site. Age-related changes in protein binding are usually not clinically important in drug therapy. Albumin levels are generally decreased in the elderly, whereas alpha1-acid glycoprotein levels are not altered by age per se. Alterations in plasma protein binding that occur in the elderly are generally not attributed to age, but rather to physiological and pathophysiological changes or disease states that may occur more frequently in the elderly and most often account for altered protein binding. Age-related physiological changes, such as decreased renal function, decreased hepatic function and decreased cardiac output, generally produce more clinically significant alterations in drug disposition than that seen with alterations in drug plasma protein binding. An understanding of the inter-relationships between drug concentrations, protein binding, the physiology of aging, disease, pharmacokinetics and pharmacodynamics is necessary for effective therapeutic monitoring. Monitoring of unbound drug concentrations simplifies these relationships and provides the fundamental information needed for dosage regimen development and adjustment. Drug therapy in the elderly should be individualised taking into account all of these factors.


Assuntos
Envelhecimento/metabolismo , Proteínas Sanguíneas/metabolismo , Preparações Farmacêuticas/metabolismo , Farmacocinética , Adulto , Idoso , Feminino , Geriatria , Humanos , Masculino , Ligação Proteica
6.
Clin Pharmacokinet ; 30(6): 445-62, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8792057

RESUMO

The plasma protein binding of drugs has been shown to have significant effects on numerous aspects of clinical pharmacokinetics and pharmacodynamics. In many clinical situations, measurement of the total drug concentration does not provide the needed information concerning the unbound fraction of drug in plasma which is available for distribution, elimination, and pharmacodynamic action. Thus, accurate determination of unbound plasma drug concentrations is essential in the therapeutic monitoring of drugs. Many methodologies are available for determining the extent of plasma protein binding of drugs, however, in the clinical evaluation of drug therapy, equilibrium dialysis and ultrafiltration are the most routinely utilised methods. Both of these methods have been proven to be experimentally sound and to yield adequate protein binding data. Furthermore, the characterisation of the interactions between drug and protein molecules is essential for the assessment of the pharmacokinetic implications of drug-protein binding. Protein binding parameters which characterise the affinity of the drug-protein association, the number of classes of binding sites, the number of binding sites per class or protein and the binding capacity are useful for predicting unbound drug concentrations. Simple graphical methods have often been used to obtain protein binding parameters, but these methods have limitations and are not useful for drugs with more than 1 class of binding site. Therefore, the fitting of protein binding models which characterise the drug-protein binding interaction for experimental data is the preferred method of calculating binding parameters. Using the appropriate model, values for binding parameters are typically estimated by using nonlinear least-squares regression analysis.


Assuntos
Proteínas Sanguíneas/metabolismo , Modelos Lineares , Microdiálise/métodos , Preparações Farmacêuticas/metabolismo , Ultrafiltração/métodos , Simulação por Computador , Humanos , Ligação Proteica
7.
AIDS Res Hum Retroviruses ; 15(18): 1625-30, 1999 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-10606085

RESUMO

(-)-beta-D-Dioxolane guanine (DXG) is a nucleoside analog possessing potent activity against human immunodeficiency virus type 1 (HIV-1) and type 2 (HIV-2), and hepatitis B virus (HBV) in vitro. Owing to the limited aqueous solubility of DXG, (-)-beta-D-2,6-diaminopurine dioxolane (DAPD), a more water-soluble prodrug of DXG, is being developed for clinical use. The purpose of this study was to characterize the pharmacokinetics of DXG after administration of DXG and DAPD to rats and monkeys. After intravenous administration of DXG, plasma concentrations of the nucleoside declined in a biexponential manner, with a terminal-phase half-life of 0.44 +/- 0.14 hr (mean +/- SD) in rats and 2.3 hr in monkeys. Total clearance of DXG was 4.28 +/- 0.99 liters/hr/kg in rats and 0.72 liters/hr/kg in monkeys. Renal excretion of unchanged DXG accounted for approximately 50% of total clearance in both species. Steady state volume of distribution of DXG was 2.30 liters/kg in rats and 1.19 liters/kg in monkeys. After intravenous administration of DAPD to rats, prodrug concentrations declined with a half-life of 0.37 +/- 0.11 hr. DXG was rapidly generated from DAPD, with approximately 61% of the dose of DAPD being converted to DXG. After administration of DAPD to monkeys, only concentrations of metabolite DXG could be determined owing to rapid conversion of DAPD to DXG during sample collection. The half-lives of DAPD and DXG after intravenous administration determined from urinary excretion data were 0.8 +/- 0.4 and 1.6 +/- 0.2 hr, respectively. Oral bioavailability of DAPD was estimated to be approximately 30%.


Assuntos
Fármacos Anti-HIV/farmacocinética , Dioxolanos/farmacocinética , Guanosina/análogos & derivados , Macaca mulatta/metabolismo , Pró-Fármacos/farmacocinética , Nucleosídeos de Purina/farmacocinética , Ratos/metabolismo , Administração Oral , Animais , Antivirais/farmacocinética , Disponibilidade Biológica , Biotransformação , Dioxolanos/sangue , Dioxolanos/urina , Guanosina/sangue , Guanosina/farmacocinética , Guanosina/urina , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Estrutura Molecular , Nucleosídeos de Purina/sangue , Nucleosídeos de Purina/urina , Ratos Sprague-Dawley , Especificidade da Espécie , Distribuição Tecidual
8.
AIDS Res Hum Retroviruses ; 6(2): 219-28, 1990 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2328158

RESUMO

The pharmacokinetics of the anti-human immunodeficiency virus type 1 nucleosides, 3'-azido-2',3'-dideoxyuridine (AzddU) and 3'-azido-3'-deoxythymidine (AZT) were characterized in rhesus monkeys. Half-life, total clearance, and steady-state volume of distribution were similar for both compounds. The observed pharmacokinetic parameters for AZT were comparable to those previously reported in humans. Oral absorption of AzddU and AZT was virtually complete after 60 mg/kg. However, bioavailability of both nucleosides was markedly lower (less than 50%) after 200 mg/kg, possibly indicating the involvement of a saturable absorption mechanism. The nucleosides were also well absorbed after subcutaneous administration. AzddU and AZT penetrated the cerebrospinal fluid (CSF) with concentration ratios in CSF:serum ranging from 0.05 to 0.25 one hour after drug administration. The glucuronides of AZT and AzddU were readily detected in urine. Hemogram and blood chemistry values for animals receiving short-term treatment (3 doses) with either AZT or AzddU did not exhibit any significant changes when compared with untreated control monkeys. The similar pharmacokinetic characteristics of AzddU compared with AZT suggest that clinical trials of AzddU are warranted.


Assuntos
Antivirais/farmacocinética , Zidovudina/análogos & derivados , Zidovudina/farmacocinética , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Administração Oral , Animais , Didesoxinucleotídeos , Avaliação de Medicamentos , Injeções Intravenosas , Macaca mulatta , Masculino , Projetos de Pesquisa , Zidovudina/administração & dosagem , Zidovudina/sangue , Zidovudina/líquido cefalorraquidiano
9.
Exp Gerontol ; 30(5): 505-15, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8557098

RESUMO

The pharmacodynamics of naproxen were evaluated in 5- and 24-month-old male Fischer 344 rats. Plasma naproxen concentrations and thromboxane B2 (TxB2) concentrations were measured as a function of time after intravenous administration of 25 mg/kg naproxen. Age-dependent alterations in naproxen pharmacokinetics were attributed to significant reductions in free plasma clearance (CLfree) and free steady-state volume of distribution (VSSfree), suggesting a decline in metabolic activity and naproxen binding to tissue components in aged rats, respectively. The time course of TxB2 production as a function of unbound naproxen concentrations was described by a sigmoid Emax model. Age had no significant effect on the pharmacodynamic parameter Emax, the maximum percent inhibition of TxB2 formation. Age also had no statistically significant effect on EC50, the drug concentration producing 50% of the maximum effect, however, average EC50 values were 35% higher in the aged rats. The duration of TxB2 inhibition was unaffected by age, possibly owing to similar relative decreases in receptor sensitivity (increased EC50) and increases in free naproxen concentrations (decreased free clearance and volume of distribution). Alternatively, the age-related changes in pharmacokinetic parameters were not of sufficient magnitude to produce a significant change in drug response, naproxen, pharmacokinetics, pharmacodynamics, age, rats, thromboxane B2, nonsteroidal anti-inflammatory drugs.


Assuntos
Envelhecimento/fisiologia , Anti-Inflamatórios não Esteroides/farmacologia , Naproxeno/farmacologia , Análise de Variância , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Cromatografia Líquida de Alta Pressão , Injeções Intravenosas , Masculino , Naproxeno/farmacocinética , Ligação Proteica , Ratos , Ratos Endogâmicos F344 , Tromboxano B2/sangue
10.
Biochem Pharmacol ; 34(12): 2115-21, 1985 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-4004929

RESUMO

The binding of oxazepam and its glucuronide conjugates to human serum albumin (HSA), as well as the binding interactions of the drug and its metabolites, were examined by equilibrium dialysis and kinetic probe studies. Oxazepam and its S(+) glucuronide are bound to the HSA molecule with affinity constants of 3.5 X 10(5) M-1 and 5.5 X 10(4) M-1, respectively, which were independent of protein concentration over a range of 0.1 to 5.0 g/dl. The R(-) glucuronide bound weakly to albumin, with the binding parameter, N X K, increasing at lower albumin concentrations. Pre-acetylation of fatty acid free-HSA resulted in decreased binding of all three compounds, probably by altering the conformation of the binding sites. Kinetic probe studies with p-nitrophenyl acetate indicate that oxazepam and its S(+) glucuronide shared a common binding site on HSA, but that the R(-) glucuronide bound at another site. Oxazepam binding was unaffected by the presence of its glucuronide conjugates but was inhibited by fatty acids. The percentage of oxazepam bound to plasma proteins in patients with renal impairment (94%) was lower than in normal volunteers (97%). This lower binding can neither be attributed to lower albumin concentrations because of the large binding capacity of the protein and linearity of N X K nor to displacement by elevated concentrations of glucuronide conjugates, but it may be ascribed partly to increased plasma fatty acids.


Assuntos
Glucuronatos/metabolismo , Oxazepam/metabolismo , Albumina Sérica/metabolismo , Humanos , Técnicas In Vitro , Nefropatias/metabolismo , Nitrofenóis/metabolismo , Ligação Proteica , Estereoisomerismo
11.
Antiviral Res ; 27(1-2): 39-47, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7486957

RESUMO

1-(Ethoxymethyl)-6-(phenylselenenyl)-5-ethyluracil (E-EPSeU) has been shown to exhibit potent and selective activity against human immunodeficiency virus type 1 in vitro. The pharmacokinetics of E-EPSeU were characterized after intravenous administration of 5, 10 and 15 mg/kg to rats. Plasma and urine concentrations of E-EPSeU were determined by HPLC. The plasma protein binding of E-EPSeU averaged 86 +/- 4% and the blood: plasma concentration ratio was unity. E-EPSeU concentrations after the 5 mg/kg dose were too low to reliably characterize the pharmacokinetics. The pharmacokinetics of E-EPSeU were independent of dose over the range of 10-15 mg/kg. Plasma concentrations of E-EPSeU declined in a bi-exponential manner with terminal half-life of 0.45 +/- 0.12 h (mean +/- S.D.). The steady-state volume of distribution was 0.091 +/- 0.031 1/kg, suggesting the compound distributed primarily into blood. The systemic clearance (0.63 +/- 0.13 1/h/kg) was moderate and limited, in part, by protein binding. No parent compound was detected in urine. E-EPSeU-related toxicities were observed at high doses. One rat, out of 5, died 4 h after 15 mg/kg of E-EPSeU was administered and two rats administered 20 and 25 mg/kg died within 1 h. Two mice, out of 5, administered 30 mg/kg/day of E-EPSeU intraperitoneally for 6 days died during the experiment, while significant loss of body weight was observed in the surviving mice. However, body weight of the surviving mice returned to control values within 2 weeks after E-EPSeU treatment was stopped.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Antivirais/farmacocinética , Compostos Organosselênicos/farmacocinética , Uracila/análogos & derivados , Animais , Antivirais/toxicidade , Relação Dose-Resposta a Droga , Feminino , HIV-1 , Masculino , Camundongos , Compostos Organosselênicos/sangue , Compostos Organosselênicos/toxicidade , Ratos , Ratos Sprague-Dawley , Uracila/sangue , Uracila/farmacocinética , Uracila/toxicidade
12.
Antiviral Res ; 34(3): 91-9, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9191016

RESUMO

In the search for prodrugs of 2',3'-dideoxyinosine (ddI) with potential for improving the delivery of the nucleoside analogue to the lymphatic system, we synthesized dipalmitoylphosphatidyl-2',3'-dideoxyinosine (DPP-ddI) and its pharmacokinetics were investigated in mice. The disposition of ddI in plasma and lymph nodes was examined following intravenous and oral administration of parent nucleoside (100 mg/kg) and DPP-ddI (400 mg/kg, equivalent to 100 mg/kg of ddI). Concentrations of ddI were determined by HPLC. Pharmacokinetic parameters were estimated by area/moment analysis. Intravenous administration of DPP-ddI resulted in a pattern of lower peak concentrations of ddI and more sustained exposure of parent nucleoside in plasma and lymph nodes compared to administration of the parent nucleoside. Both ddI and DPP-ddI yielded similar AUC values in lymph nodes. Oral administration of the prodrug resulted in lower concentrations and AUC values of ddI in plasma and lymph nodes when compared to administration of the parent nucleoside. The bioavailability of ddI following ddI and DPP-ddI administration was 15 and 8%, respectively. The results of the present study demonstrate that DPP-ddI administered intravenously shows potential for targeting and sustaining level of ddI in the nodular lymphatic tissues.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Didanosina/análogos & derivados , Didanosina/administração & dosagem , Didanosina/farmacocinética , Sistema Linfático/metabolismo , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Administração Oral , Animais , Fármacos Anti-HIV/sangue , Didanosina/sangue , Feminino , Injeções Intravenosas , Camundongos , Distribuição Tecidual
13.
Antiviral Res ; 32(3): 141-8, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8955509

RESUMO

The cellular localization of the polyoxometalates, K12H2[P2W12O48].24H20 (JM 1591), K10[P2W18-Zn4(H2O)2O68].20H2O (JM 1596), and [Me3NH]8[Si2W18Nb6O77] (JM 2820) were examined in cultured J774 cells by inhibition of cellular uptake of acetylated low-density lipoprotein (LDL) and by electron microscopy. All three polyoxometalates inhibited the cellular uptake of acetylated LDL, suggesting that the polyoxometalates block the association of acetylated LDL with cellular scavenger receptors. Fluorescence microscopy showed increased numbers of vacuoles in the presence of polyoxometalates, suggesting their uptake by cells. Using scanning electron microscopy (SEM), no significant cell surface morphological differences were observed between treated and non-treated J774 cells, suggesting that the compounds are not toxic to J774 cells up to a concentration of 200 micrograms/ml. Transmission electron microscopy (TEM) revealed large amounts of high electron dense granules were observed in the ramifying system of tubular cavities and vacuoles. TEM-energy dispersive spectroscopy (EDS) X-ray microanalysis was unable to differentiate the dense particles, most likely because the amount of tungsten in the cells was below the limit of detection. X-ray microanalysis conducted using the SEM-wavelength dispersive spectroscopy (WDS) detected tungsten, averaging 0.45 +/- 0.16% (mean +/- S.D.), in the J774 cells treated with JM 2820, suggesting that this polyoxometalate was taken up by the macrophages or was bound to their surface. Polyoxometalates interact at the cell surface and appear to be taken up by J774 macrophages. The cellular localization of polyoxometalates may be associated with anti-HIV activity.


Assuntos
Antivirais/farmacocinética , Macrófagos/metabolismo , Compostos Organometálicos/farmacocinética , Compostos de Tungstênio/farmacocinética , Animais , Linhagem Celular , Microanálise por Sonda Eletrônica , Lipoproteínas LDL/farmacocinética , Macrófagos/citologia , Macrófagos/ultraestrutura , Camundongos , Microscopia Eletrônica , Microscopia Eletrônica de Varredura
14.
Antiviral Res ; 35(3): 187-93, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9298758

RESUMO

9-(beta-D-1,3-Dioxolan-4-yl)guanine (DXG) exhibits potent antiviral activity against human immunodeficiency virus type 1 (HIV-1) and hepatitis B virus (HBV) in vitro. However, since DXG possesses limited aqueous solubility, a more water soluble prodrug of DXG, 9-(beta-D-1,3-dioxolan-4-yl)-2-aminopurine (APD), was synthesized. The purpose of this study was to characterize the pharmacokinetics of APD and its antiviral metabolite DXG in mice. Female NIH-Swiss mice were administered 100 mg/kg APD intravenously or orally. Serum, brain and liver were collected at selected times following prodrug administration and concentrations of APD and DXG were determined by HPLC. APD was efficiently converted to parent nucleoside DXG following both intravenous and oral administration. Biotransformation of APD to DXG likely occurs in the liver and is mediated by xanthine oxidase. Similar pharmacokinetic profiles for DXG were observed following either route of administration in serum, liver and brain. These results demonstrate that APD appears to be a promising prodrug for the delivery of DXG.


Assuntos
2-Aminopurina/análogos & derivados , Dioxolanos/metabolismo , Dioxolanos/farmacologia , Dioxolanos/farmacocinética , Guanina/análogos & derivados , Guanosina/análogos & derivados , Pró-Fármacos/farmacocinética , Purinas/farmacocinética , 2-Aminopurina/metabolismo , 2-Aminopurina/farmacologia , Administração Oral , Alopurinol/farmacologia , Animais , Biotransformação , Química Encefálica , Dioxolanos/administração & dosagem , Dioxolanos/sangue , Dioxolanos/química , Interações Medicamentosas , Feminino , Guanina/química , Guanina/metabolismo , Guanina/farmacologia , Guanosina/administração & dosagem , Guanosina/sangue , Guanosina/farmacocinética , Injeções Intravenosas , Fígado/química , Fígado/efeitos dos fármacos , Camundongos , Purinas/administração & dosagem , Purinas/sangue , Xantina Oxidase/efeitos dos fármacos , Xantina Oxidase/metabolismo
15.
Ann N Y Acad Sci ; 616: 385-97, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-1706574

RESUMO

3'-Azido-2',3'-dideoxy-5-methylcytidine (CS-92, AzddMeC) is an antiviral nucleoside analogue structurally related to 3'-azido-3'-deoxythymidine (AZT). CS-92 is a potent and selective inhibitor of HIV-1 reverse transcriptase and HIV-1 replication in human lymphocytes and macrophages. The EC50 for CS-92 in HIV-1-infected human PBM cells was 0.09 microM. In HIV-1-infected human macrophages, the EC50 was 0.006 microM. This compound was also effective against human immunodeficiency virus type 2 in lymphocytes. The replication of Friend murine virus was only weakly inhibited, and no effect was observed against herpes simplex virus type 1 and type 2 and coxsackievirus B4. CS-92 was not toxic to PBM or Vero cells when tested up to 200 microM and was, furthermore, at least 40 times less toxic to granulocyte-macrophage and erythroid precursor cells in vitro than was AZT. The interaction of the 5'-triphosphate of CS-92 with HIV-1 reverse transcriptase indicated competitive inhibition (the inhibition constant, Kis, was 0.0093 microM) with a 30-fold greater affinity for CS-92-TP than for ddCTP. CS-92-TP inhibited HIV-1 reverse transcriptase by 50% at a concentration 6,000-fold lower than that which was required for a similar inhibition of DNA polymerase alpha. Pharmacokinetic studies showed that CS-92 was not deaminated to AZT in rats, but this compound was found to have a half-life of 2.7 hours. In rhesus monkeys, however, a compound with a retention time and ultraviolet spectra characteristics similar to AZT was detected. The mean half-life in rhesus monkeys for CS-92 was 1.52 and 1.74 h after intravenous and oral administration, respectively, and the oral bioavailability was about 21 percent. Additional preclinical studies with CS-92 will determine the ultimate utility of this antiviral agent for the treatment of HIV-1 infections.


Assuntos
Antivirais/farmacologia , Azidas/farmacologia , DNA Polimerase Dirigida por DNA/metabolismo , Didesoxinucleosídeos/farmacologia , HIV-1/efeitos dos fármacos , DNA Polimerase Dirigida por RNA/metabolismo , Zalcitabina/análogos & derivados , Animais , Antivirais/farmacocinética , HIV-1/enzimologia , Humanos , Técnicas In Vitro , Cinética , Macaca mulatta , Masculino , Camundongos , Ratos
16.
Cancer Chemother Pharmacol ; 39(6): 532-6, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9118465

RESUMO

PURPOSE: beta-L-Dioxolane-cytidine (OddC), a novel L-nucleoside analog with potent cytotoxicity in vitro, appears to be a promising candidate for anticancer therapy. In this study, a high performance liquid chromatography (HPLC) analytical method was developed and the preclinical pharmacokinetics of OddC were characterized in rats. METHODS: Adult male Sprague Dawley rats were given 10, 25, or 50 mg/kg of OddC both intravenously and orally with a 6-day washout period between doses. Each rat received one dosage level of OddC and the route of administration was assessed by a randomized crossover design. Plasma and urine concentrations were determined by HPLC. Pharmacokinetic parameters were generated by area-moment analysis. RESULTS: Following intravenous administration, the plasma concentrations of OddC declined rapidly in a biexponential manner with a terminal phase half-life of 1.65 +/- 1.12 h (mean +/- SD). Mean total, renal, and nonrenal clearances were 1.38 +/- 0.62, 0.30 +/- 0.14, and 1.08 +/- 0.59 1/h per kg. Approximately 22% of the administered dose was excreted unchanged in the urine. Thus, nonrenal clearance was the predominant route of elimination of OddC. The steady-state volume of distribution averaged 1.42 +/- 0.66 1/kg, indicating intracellular distribution of OddC. The nucleoside analog was slowly absorbed after oral administration and bioavailability varied greatly between individual rats, averaging 41 +/- 27% when calculated from urinary excretion data and 37 +/- 25% when calculated from plasma OddC concentration data. CONCLUSION: The pharmacokinetics of OddC in rats were linear over the dose range studied.


Assuntos
Antineoplásicos/farmacocinética , Citosina/análogos & derivados , Dioxolanos/farmacocinética , Animais , Antineoplásicos/sangue , Área Sob a Curva , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Citosina/sangue , Citosina/farmacocinética , Dioxolanos/sangue , Masculino , Ratos , Ratos Sprague-Dawley
17.
Antivir Chem Chemother ; 11(3): 203-11, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10901291

RESUMO

The pharmacokinetics of a bispivaloylthioethyl prodrug of zidovudine monophosphate (AZTMP), bis(t-butyl-SATE)-AZTMP, and intracellular conversion of the prodrug to AZTMP were characterized following intravenous (i.v.) and oral (p.o.) administration of the prodrug to mice. Concentrations of bis(t-butyl-SATE)-AZTMP, AZTMP and zidovudine (AZT) in blood, red blood cells, plasma, brain and lymph nodes were determined by HPLC. Following i.v. administration of bis(t-butyl-SATE)-AZTMP, concentrations of the prodrug declined rapidly with low levels of the prodrug detected until 4 h. Both bis(t-butyl-SATE)-AZTMP and AZTMP were detected in brain 3 min after dosing. AZTMP was found in both plasma and peripheral red blood cells, peaking at approximately 30 min and remaining detectable until 2 h. No AZTMP was detected in lymph nodes. Compared to the pharmacokinetics of AZT following its i.v. administration, i.v. administration of bis(t-butyl-SATE)-AZTMP produced lower peak concentrations of AZT in plasma, peripheral red blood cells, brain and lymph nodes. However, terminal half-lives of AZT were significantly prolonged following administration of the prodrug. Following p.o. administration of bis(t-butyl-SATE)-AZTMP, neither the prodrug nor AZTMP were detectable in whole blood. The conversion of AZT from bis(t-butyl-SATE)-AZTMP in plasma and peripheral red blood cells following p.o administration was 12.1% of that following i.v. administration of the prodrug. Bis(t-butyl-SATE)-AZTMP demonstrated promising potential for intracellular delivery of AZTMP. The prodrug also prolonged the retention of AZT in mice, and particularly increased delivery of AZT to the lymphatic and central nervous systems.


Assuntos
Fármacos Anti-HIV/farmacocinética , Encéfalo/metabolismo , Linfonodos/metabolismo , Pró-Fármacos/farmacocinética , Zidovudina/farmacocinética , Administração Oral , Animais , Didesoxinucleotídeos , Estabilidade de Medicamentos , Feminino , Meia-Vida , Humanos , Injeções Intravenosas , Camundongos , Zidovudina/análogos & derivados
18.
J Pharm Sci ; 80(1): 36-8, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1849573

RESUMO

The pharmacokinetics of 2',3'-dideoxycytidine (DDC) was characterized after iv administration of a high dose (500 mg/kg) of DDC to rats. The high dose was administered to optimally characterize plasma DDC concentration and urinary excretion rate versus time profiles. Drug concentrations in plasma and urine were determined by HPLC. Plasma DDC concentrations and DDC urinary excretion rates as a function of time were fitted simultaneously to a two-compartment model. Drug concentrations in plasma and urinary excretion rates declined in parallel with a terminal half-life of 1.29 +/- 0.07 h (mean +/- SD). Total, renal, and nonrenal clearances were 1.48 +/- 0.15, 0.73 +/- 0.38, and 0.75 +/- 0.36 L/h/kg, respectively. Renal clearance exceeds glomerular filtration rate in the rat, indicating that DDC undergoes active renal tubular secretion. The unbound secretory intrinsic clearance for DDC renal excretion was moderate, with a value of 0.4 L/h. The steady-state volume of distribution of DDC was 1.25 +/- 0.13 L/kg. Pharmacokinetic parameters after iv administration of 500 mg/kg of DDC were virtually identical to those reported previously after administration of 10-200 mg/kg of the nucleoside to rats. Thus, the disposition of DDC in the rat is independent of dose over a range of 10 to 500 mg/kg. High doses of DDC can be administered to rats to allow for complete characterization of the disposition pattern of the drug without complexities due to any nonlinearity.


Assuntos
Túbulos Renais/metabolismo , Zalcitabina/farmacocinética , Animais , Masculino , Matemática , Taxa de Depuração Metabólica/fisiologia , Modelos Biológicos , Ratos , Ratos Endogâmicos , Zalcitabina/administração & dosagem , Zalcitabina/urina
19.
J Pharm Sci ; 73(6): 774-80, 1984 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6204037

RESUMO

The effects of drug stability, radioactive tracer purity, buffer composition, protein concentration, and fluid shifts on the nonlinear plasma protein binding of prednisolone were examined by equilibrium dialysis. Prednisolone exhibits a concentration-dependent degradation; however, the limited extent of this does not affect protein binding. Impure tritiated prednisolone used as a tracer produces incorrect, low fractional binding values with the binding parameters generated for transcortin affected more than those for albumin. Isotonic sodium phosphate and Krebs original Ringer phosphate buffers yield similar fractional binding of prednisolone and identical protein binding parameters. Fractional binding of the steroid decreases with total plasma protein concentration, but the association constants remain constant over a twofold dilution of plasma proteins. Further dilution increases these parameters. A time-dependent colloidal osmotic fluid shift during dialysis causes dilution of plasma protein concentrations and diminished drug binding. Theoretical simulations show that the osmotic fluid shifts produce the largest changes in fractional binding for compounds that are bound by low-capacity proteins with low association constants (K less than 10(6) M-1). A mathematical equation was developed to correct bound drug concentrations and fraction bound for protein dilution caused by this effect. The fluid shifts can be prevented by the addition of dextran (mol. wt. 70,000) to the dialysis buffer in a concentration of 55% of the total protein concentration. Multiple factors can diminish the nonlinear prednisolone binding as artifacts during equilibrium dialysis, but the changes are relatively modest.


Assuntos
Prednisolona/sangue , Proteínas Sanguíneas/metabolismo , Líquidos Corporais/metabolismo , Soluções Tampão , Fenômenos Químicos , Química , Dextranos , Diálise , Estabilidade de Medicamentos , Humanos , Marcação por Isótopo , Membranas Artificiais , Ligação Proteica
20.
J Pharm Sci ; 85(9): 965-70, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8877888

RESUMO

The disposition of zidovudine (AZT) was investigated in near-term (day 20) pregnant rats after intravenous bolus administration of AZT at 50 mg/kg. A compartmental pharmacokinetic model was developed to describe AZT concentrations in maternal plasma (1), placenta (2), fetus (3), amniotic fluid (4), and the maternal tissue compartment (5). Model equations were fitted simultaneously to all concentration data by NONLIN least-squares regression. The model that best described the AZT concentration data (F test, AIC, sum of weighted squared residuals) incorporated bidirectional transfer between maternal plasma reversible placenta, placenta reversible fetus, placenta reversible amniotic fluid, and maternal plasma reversible tissue compartment. Transfer rate constants (1/h) were as follows: k12, 0.58 +/- 0.41; k21, 47.64 +/- 46.61; k23, 67.50 +/- 42.03; k32, 13.09 +/- 8.80; k24, 0.62 +/- 0.03; k42, 0.32 +/- 0.06; k15, 5.75 +/- 7.00; k51, 4.12 +/- 1.01; and k10, 1.51 +/- 0.80. AZT rapidly distributed into tissue and placenta compartments. However, AZT accumulated more slowly into amniotic fluid. Intercompartmental distributional clearances suggest that the mechanism of maternal-placental, placental-fetal, and fetal-amniotic fluid transfer of AZT was by passive diffusion. This maternal-fetal model for AZT may offer a useful approach for describing the placental transfer kinetics of other antiviral nucleosides as well.


Assuntos
Fármacos Anti-HIV/farmacocinética , Feto/metabolismo , Prenhez/metabolismo , Zidovudina/farmacocinética , Líquido Amniótico/metabolismo , Animais , Feminino , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA