Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Laryngol Otol ; 133(10): 862-866, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31587675

RESUMEN

BACKGROUND: The evidence supporting the efficacy of antibiotic therapy in the treatment of chronic rhinosinusitis is not compelling. A limited number of studies show that the changes in the nasal microbiome in patients following drug therapy are unpredictable and variable. The evidence for the impact of oral antibiotics on the gut microbiota is stronger, possibly as a result of differences in drug distribution to various sites around the body. There are few studies on sinus mucosal and mucus levels of oral antibiotics used in the treatment of chronic rhinosinusitis. The distribution dependent effects of antibiotics on the sinonasal microbiome is unclear. CONCLUSION: This review highlights that relative drug concentrations and their efficacy on microbiota at different sites is an important subject for future studies investigating chronic rhinosinusitis.

2.
J Dent Res ; 98(9): 985-993, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31226001

RESUMEN

Chronic periodontitis (ChP) is a prevalent inflammatory disease affecting 46% of the US population. ChP produces a profound local inflammatory response to dysbiotic oral microbiota that leads to destruction of alveolar bone and tooth loss. ChP is also associated with systemic illnesses, including cardiovascular diseases, malignancies, and adverse pregnancy outcomes. However, the mechanisms underlying these adverse health outcomes are poorly understood. In this prospective cohort study, we used a highly multiplex mass cytometry immunoassay to perform an in-depth analysis of the systemic consequences of ChP in patients before (n = 28) and after (n = 16) periodontal treatment. A high-dimensional analysis of intracellular signaling networks revealed immune system-wide dysfunctions differentiating patients with ChP from healthy controls. Notably, we observed exaggerated proinflammatory responses to Porphyromonas gingivalis-derived lipopolysaccharide in circulating neutrophils and monocytes from patients with ChP. Simultaneously, natural killer cell responses to inflammatory cytokines were attenuated. Importantly, the immune alterations associated with ChP were no longer detectable 3 wk after periodontal treatment. Our findings demarcate systemic and cell-specific immune dysfunctions in patients with ChP, which can be temporarily reversed by the local treatment of ChP. Future studies in larger cohorts are needed to test the boundaries of generalizability of our results.


Asunto(s)
Periodontitis Crónica/inmunología , Células Asesinas Naturales/inmunología , Monocitos/inmunología , Neutrófilos/inmunología , Adulto , Citocinas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Porphyromonas gingivalis , Estudios Prospectivos
3.
Acta Trop ; 127(3): 165-73, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23643517

RESUMEN

The main objective of this investigation was to establish the pharmacokinetics profile and in vivo chemosuppressive activities of cryptolepine hydrochloride-loaded gelatine nanoparticles (CHN) designed for parenteral administration for the treatment of malaria in comparison to the drug free in solution (CHS). Single-dose pharmacokinetics was investigated in Wistar rats by administering CHN or CHS (equivalent to 10 mg/kg of drug) by IV bolus injection via the lateral tail vein. The drug concentration in plasma was monitored over a 24-h period following administration. Chemosuppressive activity was investigated in Wistar rats challenged with P berghei parasites. Animals were given a daily dose of either CHN or CHS, equivalent to 2.5-100 mg/kg by intraperitoneal injection. The level of parasitaemia was determined by light microscopy by examining Giemsa-stained thin blood smears prepared from the tail end on day four of infection. It was found that CHN attained a higher (4.5-folds) area under the curve (AUC (0-24)) compared to CHS. CHS however produced a higher volume of distribution (4-folds). Distribution and elimination rates were higher with CHS which resulted in a lower (11.7 h) elimination half-life compared to that of CHN (21.85 h). The superior pharmacokinetic profile of CHN translated into superior chemosuppressive activity at all dose levels relative to CHS. As a conclusion, loading cryptolepine hydrochloride into gelatine nanoparticles improved both pharmacokinetics and in vivo antiplasmodial activity of the compound with the highest chemosuppression (97.89 ± 3.10) produced by 100 mg/kg of CHN.


Asunto(s)
Gelatina/química , Alcaloides Indólicos/farmacología , Alcaloides Indólicos/farmacocinética , Nanoestructuras/química , Quinolinas/farmacología , Quinolinas/farmacocinética , Animales , Antimaláricos/química , Antimaláricos/farmacocinética , Antimaláricos/farmacología , Área Bajo la Curva , Química Encefálica , Vías de Administración de Medicamentos , Alcaloides Indólicos/química , Riñón/química , Hígado/química , Pulmón/química , Malaria , Masculino , Estructura Molecular , Miocardio/química , Quinolinas/química , Ratas , Ratas Wistar , Bazo/química
4.
Br J Pharmacol ; 162(1): 226-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20955364

RESUMEN

BACKGROUND AND PURPOSE: PA-824 is a 2-nitroimidazooxazine prodrug currently in Phase II clinical trial for tuberculosis therapy. It is bioactivated by a deazaflavin (F(420) )-dependent nitroreductase (Ddn) isolated from Mycobacterium tuberculosis to form a des-nitro metabolite. This releases toxic reactive nitrogen species which may be responsible for its anti-mycobacterial activity. There are no published reports of mammalian enzymes bioactivating this prodrug. We have investigated the metabolism of PA-824 following incubation with a subcellular fraction of human liver, in comparison with purified Ddn, M. tuberculosis and Mycobacterium smegmatis. EXPERIMENTAL APPROACH: PA-824 (250 µM) was incubated with the 9000 × g supernatant (S9) of human liver homogenates, purified Ddn, M. tuberculosis and M. smegmatis for metabolite identification by liquid chromatography mass spectrometry analysis. KEY RESULTS: PA-824 was metabolized to seven products by Ddn and M. tuberculosis, with the major metabolite being the des-nitro product. Six of these products, but not the des-nitro metabolite, were also detected in M. smegmatis. In contrast, only four of these metabolites were observed in human liver S9; M3, a reduction product previously proposed as an intermediate in the Ddn-catalyzed des-nitrification and radiolytic reduction of PA-824; two unidentified metabolites, M1 and M4, which were products of M3; and a haem-catalyzed product of imidazole ring hydration (M2). CONCLUSIONS AND IMPLICATIONS: PA-824 was metabolized by des-nitrification in Ddn and M. tuberculosis, but this does not occur in human liver S9 and M. smegmatis. Thus, PA-824 was selectively bioactivated in M. tuberculosis and there was no evidence for 'cross-activation' by human enzymes.


Asunto(s)
Antituberculosos/farmacocinética , Hígado/metabolismo , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nitroimidazoles/farmacocinética , Fracciones Subcelulares/metabolismo , Antituberculosos/farmacología , Secuencia de Bases , Biotransformación , Cartilla de ADN , Humanos , Espectrometría de Masas , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/metabolismo , Nitroimidazoles/farmacología
5.
Anesth Analg ; 111(5): 1301-7, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20530614

RESUMEN

BACKGROUND: Transcranial electrostimulation (TES) has been reported to produce clinically significant analgesia, but randomized and double-blind studies are lacking. We investigated the analgesic and antihyperalgesic effects of TES in validated human experimental pain models. METHODS: In 20 healthy male subjects we evaluated the analgesic and antihyperalgesic effects of TES(60Hz) and TES(100Hz) to heat and mechanical pain in experimentally induced ultraviolet B skin sunburns and in normal skin. Previous animal studies in our laboratory predicted that TES(60Hz) would provide significant analgesia, and TES(100Hz) was a suitable active control. The study was conducted in a double-blind, randomized, 2-way cross-over fashion. TES was administered for 35 minutes. Quantitative sensory testing evaluating heat and mechanical pain thresholds was conducted before TES, during TES, and 45 minutes after TES. RESULTS: TES (TES(60Hz) > TES(100Hz)) evoked rapidly developing, significant thermal and mechanical antihyperalgesic effects in the ultraviolet B lesion, and attenuated thermal pain in unimpaired skin. No long-lasting analgesic and antihyperalgesic effects of a single TES treatment were demonstrated in this study. CONCLUSIONS: TES produces significant, frequency-dependent antihyperalgesic and analgesic effects in humans. The characteristics of the TES effects indicate a high likelihood of its ability to modulate both peripheral sensitization of nociceptors and central hyperexcitability.


Asunto(s)
Analgesia/métodos , Encéfalo/fisiopatología , Hiperalgesia/prevención & control , Umbral del Dolor , Dolor/prevención & control , Estimulación Eléctrica Transcutánea del Nervio , Adulto , California , Estudios Cruzados , Método Doble Ciego , Calor , Humanos , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Masculino , Persona de Mediana Edad , Vías Nerviosas/fisiopatología , Dolor/etiología , Dolor/fisiopatología , Dimensión del Dolor , Presión , Quemadura Solar/complicaciones , Quemadura Solar/fisiopatología , Adulto Joven
6.
Forensic Sci Int ; 186(1-3): 63-7, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19261399

RESUMEN

There have been many reports of benzylpiperazine (BZP) and trifluoromethylphenylpiperazine (TFMPP) being used as recreational drugs which have been widely marketed in the form of 'party pills' since the late 1990's. However, there is no information currently available describing the pharmacokinetics of these drugs in humans. Human plasma concentrations of BZP were measured in blood and urine samples taken from healthy adults (n=7) over 24h following a 200mg oral dose of BZP. Plasma concentrations of BZP were found to peak at 262 ng/mL (C(max)) and 75min (T(max)). Plasma concentrations of the major metabolites of BZP, 4-OH BZP and 3-OH BZP, were found to peak at 7 ng/mL (at 60 min) and 13 ng/mL (at 75 min) respectively. The elimination half-life (t(1/2)) for BZP was found to be 5.5h. Clearance (Cl/F) was found to be 99L/h. The results of this study indicate that BZP may be detectable in plasma for up to 30 h following an oral dose. Additionally, several urinary metabolites can be detected.


Asunto(s)
Piperazinas/farmacocinética , Adulto , Disponibilidad Biológica , Toxicología Forense , Semivida , Humanos , Masculino , Estructura Molecular , Piperazinas/sangre , Piperazinas/orina , Espectrometría de Masa por Ionización de Electrospray
7.
Pain ; 139(1): 15-27, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18396374

RESUMEN

Animal studies have documented a critical role for cytokines in cell signaling events underlying inflammation and pain associated with tissue injury. While clinical reports indicate an important role of cytokines in inflammatory pain, methodological limitations have made systematic human studies difficult. This study examined the utility of a human in vivo bioassay combining microdialysis with multiplex immunoassay techniques for measuring cytokine arrays in tissue. The first experiment measured cytokines in interstitial fluid collected from non-inflamed and experimentally inflamed skin (UVB). The effects of noxious heat on cytokine release were also assessed. The second experiment examined whether anti-hyperalgesic effects of the COX-inhibitor ibuprofen were associated with decreased tissue levels of the pro-inflammatory cytokines IL-1 beta and IL-6. In the first experiment, inflammation significantly increased IL-1 beta, IL-6, IL-8, IL-10, G-CSF, and MIP-1 beta. Noxious heat but not experimental inflammation significantly increased IL-7 and IL-13. In the second experiment, an oral dose of 400 and 800 mg ibuprofen produced similar anti-hyperalgesic effects suggesting a ceiling effect. Tissue levels of IL-1 beta and IL-6 were not affected after the 400mg dose but decreased significantly (44+/-32% and 38+/-13%) after the 800 mg dose. These results support the utility of explored method for tracking cytokines in human tissue and suggest that anti-hyperalgesic and anti-inflammatory effects of ibuprofen are at least partially dissociated. The data further suggest that high clinical doses of ibuprofen exert anti-inflammatory effects by down-regulating tissue cytokine levels. Explored human bioassay is a promising tool for studying the pathology and pharmacology of inflammatory and chronic pain conditions.


Asunto(s)
Inhibidores de la Ciclooxigenasa/administración & dosificación , Citocinas/biosíntesis , Calor/efectos adversos , Mediadores de Inflamación/fisiología , Microdiálisis/métodos , Piel/química , Adulto , Biomarcadores/metabolismo , Estudios Cruzados , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Método Doble Ciego , Femenino , Humanos , Masculino , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Piel/efectos de los fármacos , Piel/metabolismo , Quemadura Solar/tratamiento farmacológico , Quemadura Solar/metabolismo
8.
Dis Aquat Organ ; 78(3): 225-33, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18380221

RESUMEN

Investigations into the metabolism of drugs used in aquatic animal therapy are useful for understanding the mechanisms of xenobiotic transformation systems and can aid the development of dosing regimens. This study investigated the metabolism of the synthetic anthelmintic praziquantel, which has application in helminthiasis treatment for several fish species including kingfish Seriola lalandi, a commercial aquaculture finfish species. At least 7 mono- or dihydroxylated derivatives of the parent compound were identified in kingfish after administration of a 150 mg kg(-1) oral praziquantel dose, paralleling findings in mammals. The structure of one representative mono-hydroxylated species that was prominent in the skin, muscle, liver, kidney and plasma of kingfish was investigated using fragmentation experiments; this revealed that hydroxylation of the parent molecule occurred in the tetrahydroisoquinoline region of praziquantel, analogous with mammalian metabolites, but different to that of the active mammalian metabolite (trans-4-OH-praziquantel). The implications of these findings with regard to biotransformation systems for this drug in mammals and fish are discussed.


Asunto(s)
Antihelmínticos/farmacocinética , Perciformes/metabolismo , Praziquantel/farmacocinética , Administración Oral , Animales , Antihelmínticos/administración & dosificación , Antihelmínticos/química , Praziquantel/administración & dosificación , Praziquantel/química , Factores de Tiempo , Distribución Tisular
9.
Dis Aquat Organ ; 69(2-3): 233-8, 2006 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16724567

RESUMEN

Oral praziquantel (PZQ) preparations have recently been investigated for the treatment of monogeneans that infect the skin and gills of kingfish Seriola lalandi cultured in sea-cages. To evaluate an oral PZQ dosing strategy, the pharmacokinetics of a dissolved and in feed oral PZQ preparation (40 mg kg(-1) body weight) were compared with an intravenous bolus in kingfish plasma and skin using HPLC. Compared with intravenous administration, PZQ bioavailability (area under curve, AUC0-24h) was slightly improved when the drug was administered with food in both kingfish plasma (56.8% in feed vs. 50.8% in solution) and skin (55.5% in feed vs. 50.3% in solution). After oral dosing, maximum drug concentrations in skin were approximately one-third of those achieved in plasma and higher when the drug was administered in solution (5.26 microg ml(-1)) than in feed (3.96 microg ml(-1)); additionally, the time to achieve maximum PZQ concentration was similar in plasma and skin, although markedly reduced when the drug was administered in solution (1 h) than in feed (6 h). However, clearance of the drug was delayed in skin; administered as an oral formulation, PZQ concentrations in the systemic circulation fell below the limit of quantification after 24 h, but remained quantifiable (0.3 microg g(-1)) in skin at this time. These initial studies indicate that a daily treatment interval will lead to the exposure of parasites to highly variable anthelmintic concentrations, which may be sub-optimal for the treatment of monogeneans in this finfish species.


Asunto(s)
Antihelmínticos/farmacocinética , Enfermedades de los Peces/tratamiento farmacológico , Helmintiasis Animal/tratamiento farmacológico , Perciformes/metabolismo , Praziquantel/farmacocinética , Animales , Antihelmínticos/administración & dosificación , Antihelmínticos/análisis , Área Bajo la Curva , Disponibilidad Biológica , Vías de Administración de Medicamentos/veterinaria , Enfermedades de los Peces/parasitología , Perciformes/parasitología , Plasma/química , Praziquantel/administración & dosificación , Praziquantel/análisis , Piel/química , Piel/metabolismo , Factores de Tiempo
10.
Environ Toxicol Pharmacol ; 21(2): 184-90, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21783656

RESUMEN

Animals provide a physiologically relevant system for evaluation of drug metabolism, but marked inter-species differences limit extrapolation to humans. Liver microsomes are used extensively as an in vitro human drug metabolising system, and with appropriate selection of parameters, such as substrate and enzyme concentrations, may predict both routes and rate of metabolism. However, variable enzyme expression between donors and overlapping substrate specificity influence reproducibility, hence recombinant human CYP enzymes expressed in human, yeast or insect cells have been developed. For complex metabolic profiles involving sequential or competing pathways, isolated hepatocytes and liver slices are of value. Altered enzyme activity and restricted availability constrain their use. Cryopreservation or culture increase availability, but changes in enzyme activity remain a constraint. To date, human in vitro systems do not predict all aspects of drug metabolism, thus a combination of in vivo animal and in vitro human studies will be required for the foreseeable future.

11.
Xenobiotica ; 32(2): 87-107, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11868972

RESUMEN

1. Mouse studies have indicated that the antitumour effects of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) are dramatically potentiated in combination with other drugs, and it has been proposed that optimization of the therapeutic potential of DMXAA would exploit combination therapy. The aim was to identify the most appropriate animal model for further investigations of the pharmacokinetics of possible DMXAA-drug combinations and their extrapolation to patients. 2. Qualitatively, the metabolic profile for DMXAA in liver microsomes was similar in mouse, rat, rabbit and humans, with glucuronidation and 6-methylhydroxylation the two major metabolic pathways. In all species, the intrinsic clearance by glucuronidation was at least 2-fold that due to hydroxylation. There was significant variability in the in vitro kinetic parameters (Km, Vmax), with the mouse being the least efficient DMXAA metabolizer compared with the other species. 3. Mouse, rat and rabbit renal microsomes exhibited DMXAA glucuronidation activity, but only the rabbit showed 6-methylhydroxylation. For the total in vitro CL(int) (Vmax/Km) by glucuronidation and 6-methylhydroxylation, the ratio of kidney:liver was 0.67, 0.03 and 0.34 in the mouse, rat and rabbit respectively. However, taking into account the liver and kidney weight difference, it is apparent that the in vivo renal metabolism would not be a major contributor to the overall elimination of DMXAA. 4. The inhibitory profile for liver DMXAA glucuronidation was similar across species, but there was remarkable interspecies variability in the inhibition of liver DMXAA 6methylhydroxylation. 5. Extrapolation of in vitro intrinsic clearance to in vivo gave a significant underestimation of plasma clearance for all species. However, there was a significant allometric relationship for plasma clearance and volume of distribution, but not for maximum tolerated dose across species. 6. The results indicate that animal models may have a limited role in the extrapolation to patients of drug interactions with agents such as DMXAA that have immunomodulating activity that may vary widely between species.


Asunto(s)
Antineoplásicos/metabolismo , Xantenos/metabolismo , Xantonas , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Humanos , Técnicas In Vitro , Riñón/metabolismo , Hígado/metabolismo , Ratones , Microsomas/metabolismo , Microsomas Hepáticos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Conejos , Ratas , Especificidad de la Especie , Xantenos/farmacocinética , Xantenos/farmacología
12.
Br J Clin Pharmacol ; 52(2): 129-36, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11488768

RESUMEN

AIMS: To investigate the effects of various anticancer drugs on the major metabolic pathways (glucuronidation and 6-methylhydroxylation) of DMXAA in human liver microsomes. METHODS: The effects of various anticancer drugs at 100 and 500 microM on the formation of DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) in human liver microsomes were determined by high performance liquid chromatography (h.p.l.c.). For those anticancer drugs showing significant inhibition of DMXAA metabolism, the inhibition constants (Ki) were determined. The resulting in vitro data were extrapolated to predict in vivo changes in DMXAA pharmacokinetics. RESULTS: Vinblastine, vincristine and amsacrine at 500 microM significantly (P < 0.05) inhibited DMXAA glucuronidation (Ki = 319, 350 and 230 microM, respectively), but not 6-methylhydroxylation in human liver microsomes. Daunorubicin and N-[2-(dimethylamino)-ethyl]acridine-4-carboxamide (DACA) at 100 and 500 microM showed significant (P < 0.05) inhibition of DMXAA 6-methylhydroxylation (Ki = 131 and 0.59 microM, respectively), but not glucuronidation. Other drugs such as 5-fluoroucacil, paclitaxel, tirapazamine and methotrexate exhibited little or negligible inhibition of the metabolism of DMXAA. Pre-incubation of microsomes with the anticancer drugs (100 and 500 microM) did not enhance their inhibitory effects on DMXAA metabolism. Prediction of DMXAA-drug interactions in vivo based on these in vitro data indicated that all the anticancer drugs investigated except DACA appear unlikely to alter the pharmacokinetics of DMXAA, whereas DACA may increase the plasma AUC of DMXAA by 6%. CONCLUSIONS: These results indicate that alteration of the pharmacokinetics of DMXAA appears unlikely when used in combination with other common anticancer drugs. However, this does not rule out the possibility of pharmacokinetic interactions with other drugs used concurrently with this combination of anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Microsomas Hepáticos/efectos de los fármacos , Xantenos/metabolismo , Xantonas , Antineoplásicos/antagonistas & inhibidores , Antineoplásicos/metabolismo , Cromatografía Líquida de Alta Presión , Interacciones Farmacológicas , Glucurónidos/metabolismo , Humanos , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Xantenos/antagonistas & inhibidores , Xantenos/química , Xantenos/farmacocinética
13.
Xenobiotica ; 31(5): 277-93, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11491389

RESUMEN

1. The novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is extensively metabolized by glucuronidation and 6-methylhydroxylation, resulting in DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA). 2. The major human urinary metabolite of DMXAA was isolated and purified by a solid-phase extraction (SPE) method. The isolated metabolite was hydrolysed to free DMXAA by strong base, and by beta-glucuronidase. Liquid chromatography-mass spectrometry (LC-MS) and spectral data indicated the presence of a molecular ion [M + 1]+ at m/z 459, which was consistent with the molecular weight of protonated DMXAA-G. 3. The glucuronide was unstable in buffer at physiological pH, plasma and blood with species variability in half-life. Hydrolysis and intramolecular migration were major degradation pathways. 4. In vitro and in vivo formation of DMXAA-protein adducts was observed. The formation of DMXAA-protein adducts in cancer patients receiving DMXAA was significantly correlated with plasma DMXAA-G concentration and maximum plasma DMXAA concentration. 5. At least five metabolites of DMXAA were observed in patient urine, with up to 60% of the total dose excreted as DMXAA-G, 5.5% as 6-OH-MXAA and 4.5% as the glucuronide of 6-OH-MXAA. 6. These data suggest that the major metabolite in patients' urine is DMXAA beta-1-glucuronide, which may undergo hydrolysis, molecular rearrangement and covalent binding to plasma protein. The reactive properties of DMXAA-G may have important implications for the pharmacokinetics, pharmacodynamics and toxicity of DMXAA.


Asunto(s)
Antineoplásicos/farmacocinética , Glucurónidos/aislamiento & purificación , Glucurónidos/orina , Xantenos/farmacocinética , Xantonas , Animales , Antiinflamatorios no Esteroideos/farmacología , Anticoagulantes/farmacología , Antineoplásicos/orina , Cromatografía Líquida de Alta Presión , Diazepam/farmacología , Relación Dosis-Respuesta a Droga , Moduladores del GABA/farmacología , Cromatografía de Gases y Espectrometría de Masas , Glucuronidasa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Químicos , Fenilbutazona/farmacología , Unión Proteica , Conejos , Ratas , Ratas Wistar , Espectrometría de Masa por Ionización de Electrospray , Factores de Tiempo , Warfarina/farmacología , Xantenos/orina
14.
Cancer Chemother Pharmacol ; 47(6): 541-4, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11459209

RESUMEN

PURPOSE: Coadministration of thalidomide, cyproheptadine or diclofenac has been shown to increase the area under the plasma concentration-time curve (AUC) of the novel antitumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in mice. The aim of this study was to further investigate these pharmacokinetic DMXAA-drug interactions in the rat model. METHODS: The effects of coadministration of L-thalidomide, cyproheptadine or diclofenac on the pharmacokinetics of DMXAA were investigated in male Wistar Kyoto rats. The effects of L-thalidomide, cyproheptadine and diclofenac on microsomal metabolism and plasma protein binding of DMXAA were also investigated. RESULTS: No significant alteration in the plasma concentration profile for DMXAA was observed following L-thalidomide pretreatment in rats. In contrast, when combined with diclofenac or cyproheptadine, the plasma AUC of DMXAA was significantly (P<0.05) increased by 48% and 88% and the T1/2 by 36% and 107%, respectively, compared to controls. Both diclofenac and cyproheptadine at 500 microM caused a significant inhibition of DMXAA metabolism in rat liver microsomes. In contrast, L-thalidomide had no or little inhibitory effect on DMXAA metabolism in rat liver microsomes except for causing a 32% decrease in 6methylhydroxylation at 500 microM. None of the drugs had a significant effect on the plasma protein binding of DMXAA in the rat. CONCLUSION: These studies showed that coadministration of L-thalidomide did not alter the plasma DMXAA AUC in rats, in contrast to previous studies in mice, whereas diclofenac and cyproheptadine significantly reduced the plasma clearance of DMXAA in rats in a similar manner to their effect in mice. The cause of the species difference in the pharmacokinetic response to thalidomide by DMXAA is unknown, and indicates difficulties in predicting the outcome of such a combination in patients.


Asunto(s)
Antineoplásicos/farmacocinética , Talidomida/farmacología , Xantenos/farmacocinética , Xantonas , Animales , Antineoplásicos/sangre , Ciproheptadina/farmacología , Diclofenaco/farmacología , Interacciones Farmacológicas , Glucurónidos/metabolismo , Hidroxilación , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Ratas , Ratas Wistar , Talidomida/administración & dosificación , Xantenos/sangre
15.
J Chromatogr B Biomed Sci Appl ; 757(2): 343-8, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11417880

RESUMEN

The reversed-phase HPLC methods were developed to determinate the covalently bound protein adducts of the novel anti-cancer drug 5,6-dimethylxanthenone-4-acetic acid (DMXAA) via its glucuronides after releasing aglycone by alkaline hydrolysis in human plasma and human serum albumin (HSA). An aliquot of 75 microl of the mixture was injected onto a Spherex C18 column (150x4.6 mm; 5 microm) at a flow-rate of 2.5 ml/min. The mobile phase comprising of acetonitrile:10 mM ammonium acetate buffer (24:76, v/v, pH 5.8) was used in an isocratic condition, and DMXAA was detected by fluorescence. The method was validated with respect to recovery, selectivity, linearity, precision, and accuracy. Calibration curves for DMXAA were constructed in the concentration range of 0.5-40 microM in washed blank human plasma or HSA prior to alkaline hydrolysis. The difference between the theoretical and calculated concentration and the relative standard deviation were less than 10% at all quality control (QC) concentrations. The limit of detection for the covalent adduct in human plasma or HSA is 0.20 microM. The methods presented good accuracy, precision and sensitivity for use in the preclinical and clinical studies.


Asunto(s)
Antineoplásicos/sangre , Cromatografía Líquida de Alta Presión/métodos , Xantenos/sangre , Xantonas , Antineoplásicos/análisis , Calibración , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Albúmina Sérica/química , Xantenos/análisis
16.
J Chromatogr B Biomed Sci Appl ; 757(2): 359-63, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11417882

RESUMEN

The novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is a highly protein bound drug with narrow therapeutic window. We report a simple HPLC method with fluorimetric detection for the determination of free DMXAA concentration in human plasma. Sample preparation involves the ultrafiltration of plasma by a Centrisart device for 30 min at 2000 g and extraction with acetonitrile: methanol mixture. The method was validated with respect to recovery, selectivity, linearity, precision, and accuracy. Calibration curves for DMXAA were constructed at the concentration range of 0.5-40 microM in blank plasma and phosphate buffer. The difference between the theoretical and calculated concentration and the relative standard deviation were less than 10% at all quality control (QC) concentrations. The HPLC method has been used for the analysis of preclinical studies.


Asunto(s)
Antineoplásicos/sangre , Cromatografía Líquida de Alta Presión/métodos , Ultrafiltración/métodos , Xantenos/sangre , Xantonas , Calibración , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Fluorescencia
17.
Cancer Chemother Pharmacol ; 47(4): 319-26, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11345648

RESUMEN

BACKGROUND: Previous studies have demonstrated that coadministration of L-thalidomide with the novel antitumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) results in an increased area under the plasma concentration-time curve (AUC) of DMXAA, suggesting an explanation for the observed increase in the antitumour activity. The aims of this study were to investigate the effects of L-thalidomide on the in vitro metabolism of DMXAA in mouse and human liver microsomes using diclofenac as positive control, to examine the effects of L-thalidomide and diclofenac on the plasma protein binding of DMXAA in vitro, and to investigate whether the in vivo interactions can be predicted from in vitro data, particularly in humans. METHODS: Mouse and human liver microsomes were used to investigate the effects of L-thalidomide and diclofenac on DMXAA metabolism. The resulting in vitro data were extrapolated to predict in vivo changes in DMXAA, which were then compared with the results of in vivo mouse pharmacokinetic interaction studies. The protein binding of DMXAA in mouse and human plasma was determined using ultrafiltration followed by HPLC. RESULTS: Diclofenac at 100 microM caused significant inhibition of glucuronidation (> 70%) and 6-methylhydroxylation (> 54%) of DMXAA in mouse and human liver microsomes. In vivo diclofenac (100 mg/kg i.p.) resulted in a 24% and 31% increase in the plasma DMXAA AUC, and a threefold increase in T1/2 (P < 0.05) in male and female mice, respectively. In contrast, L-thalidomide at 100 microM had no inhibitory effect on DMXAA metabolism in vitro in either species, except for a decrease of about 25% in 6-methylhydroxylation in mice. L-Thalidomide at 500 microM resulted in further significant decreases in 6-methylhydroxylation in mice (30-60%) and human (30%) microsomes. Coadministration of L-thalidomide in male mice resulted in a 23% increase in DMXAA AUC and a twofold increase in T1/2 (P < 0.05). Neither L-thalidomide nor diclofenac at 50 or 500 microM had any significant effect on the in vitro plasma protein binding of DMXAA (500 microM) in mouse or human plasma. Based on our in vitro inhibition studies, we predicted a 20% increase in DMXAA AUC in mice with concomitant diclofenac, but little or no effect (< 5%) with L-thalidomide. CONCLUSION: Both L-thalidomide and diclofenac increased the plasma DMXAA AUC in mice. In the case of diclofenac, this appeared to be due to direct competitive inhibition of DMXAA metabolism, but this mechanism does not appear to be appropriate for L-thalidomide. From the in vitro human inhibition studies, it appears unlikely that concurrent diclofenac will cause an increase in the plasma AUC of DMXAA in patients. However, the effect of L-thalidomide on DMXAA could not be readily predicted from the in vitro data. Our study demonstrated that a predictive model based on direct inhibition of metabolism is appropriate for diclofenac-DMXAA interactions, but is inappropriate for the prediction of L-thalidomide-DMXAA interactions in mice and humans in vivo.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Diclofenaco/farmacología , Diclofenaco/farmacocinética , Teratógenos/farmacología , Teratógenos/farmacocinética , Talidomida/farmacología , Talidomida/farmacocinética , Xantenos/farmacología , Xantenos/farmacocinética , Xantonas , Animales , Proteínas Sanguíneas/metabolismo , Cromatografía Líquida de Alta Presión , Interacciones Farmacológicas , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Unión Proteica/efectos de los fármacos
18.
J Pharm Pharmacol ; 53(4): 463-71, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11341362

RESUMEN

The plasma protein binding and distribution in blood cells of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) has been investigated in-vitro using filtration and an HPLC method to measure DMXAA. DMXAA (500 microM) was extensively bound in plasma from all species with an unbound fraction (fu) of 4.61+/-1.10 (mouse), 2.59+/-0.32 (rat), 2.02+/-0.48 (rabbit) and 2.07+/-0.23% (human). The binding was concentration dependent with DMXAA concentrations > or = 1,000 microM markedly increasing the fu in the plasma from all species. The estimated number of binding sites in plasma were 2.4+/-0.2 (mouse), 1.7+/-0.2 (rat), 0.8+/-0.1 (rabbit) and 2.1+/- 0.2 (human). The major binding protein in human plasma was albumin, with negligible binding to gamma-globulin and alpha1-acid glycoprotein. There was a significant linear relationship between the bound:free DMXAA concentration ratio (Cb/Cu) and albumin concentration in human serum albumin solution (r = 0.955; P < 0.05) and in healthy human plasma (r = 0.998; P< 0.05), but not in plasma from cancer patients (n = 5), nor across species. In cancer patients (n = 5) DMXAA had a significantly higher (P < 0.05) fu (4.60+/- 0.42%) compared with healthy human plasma (2.07+/-0.23%). In human plasma, the fu of DMXAA (500 microM) was significantly reduced by 500 microM diazepam (P < 0.05), but not by warfarin, phenylbutazone, salicylic acid, ibuprofen or clofibric acid at that concentration. DMXAA significantly reduced the binding of dansylsarcosine (a Site-II binder) to HSA, but significantly increased the binding of dansylamide (a Site-I binder). Within species, the blood:plasma concentration ratio (CBL/CP) of DMXAA was relatively constant (mouse, 0.581+/-0.005; rat, 0.667+/-0.025; rabbit, 0.637+/-0.019; human, 0.673+/-0.103) over the range 50-1000 microM, but increased significantly at DMXAA concentrations > 1000 microM in all species except the rabbit. These results indicate that significant alterations in DMXAA plasma binding and distribution into blood cells occur with increasing concentrations of DMXAA in all species, and also that significant interspecies differences exist. It would be more appropriate to compare plasma unbound concentrations when assessing DMXAA exposure in cancer patients or when extrapolating across species.


Asunto(s)
Antineoplásicos/metabolismo , Xantenos/metabolismo , Xantonas , Animales , Antineoplásicos/farmacocinética , Células Sanguíneas/química , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Neoplasias/tratamiento farmacológico , Unión Proteica , Conejos , Ratas , Ratas Wistar , Xantenos/farmacocinética
19.
Eur J Nutr ; 40(1): 39-47, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11315504

RESUMEN

BACKGROUND: 2-Amino-3-methyl-imidazo[4,5-f]quinoline (IQ) is a mutagen produced in cooked food. It is commonly present in the human diet and often used as a (pro)carcinogen for chemoprevention studies. Many foodstuffs act as chemopreventers by altering xenobiotic metabolising enzyme expression in favour of detoxication over bioactivation pathways. However, IQ itself can also affect enzyme expression, which may be a confounding factor in chemoprevention studies. AIM OF STUDY: Chronic low dose IQ exposure is intuitively closest to the human dietary situation. The aim was to investigate the effects of chronic dietary exposure to IQ on the expression of enzymes involved in the bioactivation and detoxification of xenobiotics and to compare this with acute exposure, often used in chemoprevention studies. METHODS: Male Fischer rats received IQ (300 ppm) in the diet (AIN-76) for 52 weeks or were given IQ (20 mg.kg-1) orally for 3 days. Animals were killed, livers removed and subcellular fractions prepared. A range of enzymes was selected to allow investigation of several cellular mechanisms. Enzyme expression and activity were determined by Western blotting and the use of selective probe substrates as appropriate. RESULTS: Chronic exposure to IQ led to an increase in phase II detoxifying enzymes. Both the activity and expression of glutathione S-transferase (GST-A1/2) were increased, as were NADPH: Quinone oxidoreductase (NQO), UDP-glucuronosyl transferase (UGT) and beta-glucuronidase activities. There were no statistically significant changes in the potential for bioactivation by three cytochrome P450s. In contrast, acute IQ exposure significantly increased the expression and activity of some cytochrome P450 (CYP1A1 and CYP1A2), UGT and beta-glucuronidase, but significantly decreased glutathione S-transferase expression and activity. There was a non-significant decrease in NQO but no change in CYP3A2 and CYP2E1 activities. CONCLUSIONS: The changes after acute exposure suggest an interaction through the Ah receptor and xenobiotic response element, modified by the glucocorticoid response element. In contrast, the pattern of effects after chronic exposure suggests activation of the antioxidant response element (ARE). Although the acute model is more practically convenient for short-term chemoprevention screening, the data suggest that an entirely new mechanism is being invoked that completely masks effects of the ARE that occur during chronic exposure. There is a danger that chemopreventive strategies developed using acute models may be misleading, since the mechanism is unlikely to occur during human dietary exposures.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Glutatión Transferasa/metabolismo , Hígado/enzimología , Mutágenos/administración & dosificación , Quinolinas/administración & dosificación , Xenobióticos/metabolismo , Animales , Western Blotting , Quimioprevención , Relación Dosis-Respuesta a Droga , Hígado/efectos de los fármacos , Masculino , Mutágenos/metabolismo , NADP , Quinolinas/metabolismo , Ratas , Ratas Endogámicas F344 , Factores de Tiempo
20.
Drug Metab Dispos ; 28(12): 1449-56, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11095582

RESUMEN

In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). The metabolite 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) was determined by HPLC with fluorescence detection. The metabolite formed in human liver microsomes and by cDNA-expressed CYP isoform was identified by liquid chromatography mass spectrometry as 6-OH-MXAA. In human liver microsomes (n = 14), 6-methylhydroxylation of DMXAA followed monophasic Michaelis-Menten kinetics, with a mean apparent K(m) of 21 +/- 5 microM and V(max) of 0.043 +/- 0.019 nmol/min/mg. An approximate 10-fold interindividual variation in the intrinsic clearance (V(max)/K(m)) of DMXAA 6-methylhydroxylation in human liver microsomes was observed. The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K'(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation.


Asunto(s)
Antineoplásicos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Hígado/enzimología , Xantenos/metabolismo , Xantonas , Adulto , Anciano , Western Blotting , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/análisis , ADN Complementario/biosíntesis , Femenino , Humanos , Hidroxilación , Técnicas In Vitro , Isoenzimas/análisis , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Masculino , Metilación , Microsomas Hepáticos/enzimología , Persona de Mediana Edad , Proteínas/metabolismo , Fracciones Subcelulares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...