Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Expert Opin Investig Drugs ; 33(1): 63-72, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38224050

RESUMEN

BACKGROUND: Iruplinalkib is a novel anaplastic lymphoma kinase (ALK) inhibitor for the treatment of ALK-positive crizotinib-resistant NSCLC. RESEARCH DESIGN AND METHODS: A single oral dose of 120 mg/3.7 MBq [14C]iruplinalkib was administered to healthy subjects. Blood, urine and fecal samples were collected and analyzed for iruplinalkib and its metabolites. The safety of iruplinalkib was also assessed. RESULTS: Iruplinalkib was absorbed quickly and eliminated slowly from plasma, with a Tmax of 1.5 h and t1/2 of 28.6 h. About 88.85% of iruplinalkib was excreted at 312 h, including 20.23% in urine and 68.63% in feces. Seventeen metabolites of iruplinalkib were identified, and M3b (demethylation), M7 (cysteine conjugation), M11 (oxidative dehydrogenation and cysteine conjugation of M3b) and M12 (oxidative dehydrogenation and cysteine conjugation) were considered the prominent metabolites in humans. Iruplinalkib-related compounds were found to be covalently bound to proteins, accounting for 7.70% in plasma and 17.96% in feces, which suggested chemically reactive metabolites were formed. There were no serious adverse events observed in the study. CONCLUSIONS: Iruplinalkib was widely metabolized and excreted mainly through feces in humans. Unchanged iruplinalkib, cysteine conjugates and covalent protein binding products were the main drug-related compounds in circulation. Iruplinalkib was well tolerated at the study dose. TRIAL REGISTRATION: The trial is registered at ClinicalTrials.gov (Identifier: Anonymized).


Asunto(s)
Cisteína , Inhibidores de Proteínas Quinasas , Humanos , Administración Oral , Cisteína/uso terapéutico , Voluntarios Sanos , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras
2.
Eur J Pharm Sci ; 192: 106658, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38048851

RESUMEN

Early-stage clinical evaluation of tinengotinib (TT-00420) demonstrated encouraging preliminary efficacies in multiple types of refractory cancers, including fibroblast growth factor receptors (FGFR) inhibitors relapsed cholangiocarcinoma (CCA), castrate-resistant prostate cancer (CRPC), and HR+/HER2- breast cancer and triple negative breast cancer (TNBC). To further evaluate drug-like properties of the drug candidate, it is imperative to understand its metabolism and pharmacokinetic properties. This manuscript presented the investigation results of in vitro permeability, plasma protein binding, metabolic stability, metabolite identification, and drug-drug interaction of tinengotinib. Preclinical ADME (absorption, distribution, excretion, and metabolism) studies in rats and dogs was also conducted using a radioactive labeled tinengotinib, [14C]tinengotinib. Tinengotinib was found to have high permeability and high plasma protein binding and equally distributed between blood and plasma. There were no unique metabolites in human liver microsomes and tinengotinib showed moderate hepatic clearance. Tinengotinib is neither a potential inhibitor nor an inducer of P450 enzymes at clinically relevant concentrations, and unlikely to cause drug-drug interactions when used in combination with other drugs mediated by a key transporter, either as victim or perpetrator. Taken together, tinengotinib demonstrated a minimal risk of clinically relevant drug-drug interactions. Tinengotinib showed good oral bioavailability and dose-dependent exposures in both rat and dog after oral administration. The total radioactivity was largely distributed in the gastrointestinal system and liver, and tinengotinib could not easily pass through the blood-brain barrier. The major drug-related component in rat and dog plasma was unchanged drug (>89 %) with primary route of elimination via feces (>93 % of the dose) and minor via renal excretion (<4 % of the dose). Tinengotinib metabolism is mediated largely by CYP3A4, with minor contributions from CYP2D6 and CYP2C8. Major metabolic pathways include oxidation, oxidative cleavage of the morpholine ring, glucuronide and glutathione conjugations. The overall preclinical pharmacokinetics profile supported the selection and development of tinengotinib as a clinical candidate.


Asunto(s)
Colangiocarcinoma , Drogas en Investigación , Masculino , Ratas , Humanos , Animales , Perros , Drogas en Investigación/metabolismo , Interacciones Farmacológicas , Preparaciones Farmacéuticas/metabolismo , Disponibilidad Biológica , Inhibidores de Proteínas Quinasas/farmacocinética , Administración Oral , Microsomas Hepáticos/metabolismo , Colangiocarcinoma/metabolismo
3.
Xenobiotica ; 52(1): 38-45, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34743655

RESUMEN

The metabolism and excretion of cetagliptin were investigated in healthy male subjects after a single oral dose of 100mg/50µCi [14C] cetagliptin.The mean concentration-time profile of cetagliptin was similar to that of total radioactivity in plasma after oral administration of [14C] cetagliptin in healthy male subjects. Cetagliptin was rapidly absorbed after oral administration. Unchanged cetagliptin was the most abundant radioactive component in all matrices investigated. Approximately 53.13% of plasma AUC of total radioactivity was accounted for by cetagliptin. Each metabolite plasma AUC was not higher than 2.93% of plasma AUC of total radioactivity. By 336 h after administration, 91.68% of the administered radioactivity was excreted, and the cumulative excretion in the urine and faeces was 72.88% and 18.81%, respectively. The primary route of excretion of radioactivity was via the kidneys.Four metabolites were detected at trace levels, and it involved hydroxylated (M436-1 and M436-3), N- sulphate (M500), and N-carbamoyl glucuronic acid conjugates (M640B) of cetagliptin. These metabolites were detected also in plasma, urine, and faeces at low levels, except that metabolite M640B was not detected in faeces. All metabolites were observed with <10% of parent compound systemic exposure after oral administration.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV , Administración Oral , Radioisótopos de Carbono , Heces , Voluntarios Sanos , Humanos , Hipoglucemiantes , Masculino
4.
Front Neurosci ; 16: 1079357, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36620452

RESUMEN

Spiking neural networks (SNNs), as one of the algorithmic models in neuromorphic computing, have gained a great deal of research attention owing to temporal information processing capability, low power consumption, and high biological plausibility. The potential to efficiently extract spatio-temporal features makes it suitable for processing event streams. However, existing synaptic structures in SNNs are almost full-connections or spatial 2D convolution, neither of which can extract temporal dependencies adequately. In this work, we take inspiration from biological synapses and propose a Spatio-Temporal Synaptic Connection SNN (STSC-SNN) model to enhance the spatio-temporal receptive fields of synaptic connections, thereby establishing temporal dependencies across layers. Specifically, we incorporate temporal convolution and attention mechanisms to implement synaptic filtering and gating functions. We show that endowing synaptic models with temporal dependencies can improve the performance of SNNs on classification tasks. In addition, we investigate the impact of performance via varied spatial-temporal receptive fields and reevaluate the temporal modules in SNNs. Our approach is tested on neuromorphic datasets, including DVS128 Gesture (gesture recognition), N-MNIST, CIFAR10-DVS (image classification), and SHD (speech digit recognition). The results show that the proposed model outperforms the state-of-the-art accuracy on nearly all datasets.

5.
Br J Clin Pharmacol ; 87(1): 93-105, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32415708

RESUMEN

AIMS: This trial (NCT03751956) investigated the mass balance, pharmacokinetics and pharmacodynamics of HSK3486, a novel anaesthetic, in healthy subjects. METHODS: A single dose of 0.4 mg/kg [14 C]HSK3486 was administered to six healthy subjects. Blood, urine and faecal samples were collected, analysed for radioactivity, unchanged HSK3486 and profiled for metabolites. The Modified Observer's Assessment of Alertness/Sedation (MOAA/S) scale and vital signs were closely monitored during the study. RESULTS: The mean recovery of total radioactivity in excreta was 87.3% in 240 h, including 84.6% in urine and 2.65% in faeces. The exposure (AUC0-t ) of total radioactivity was much higher than that of unchanged HSK3486 in plasma, indicating there were circulating metabolites in plasma. The glucuronide conjugate of HSK3486 (M4) was found as the only major circulating metabolite in plasma (79.3%), while unchanged HSK3486 accounted for only 3.97% of the total radiation exposure. M4 also resulted in a longer estimated elimination half-life (t1/2 ) of total radioactivity than that of unchanged HSK3486 in plasma. Fortunately, the metabolite was detected to be not specific to red blood cells and was suggested to be nonhypnotic and nontoxic. All the subjects were quickly anaesthetized (2 min) after drug administration and woke up smoothly after a short time (5.5-14.1 min) with few residual effects. The only adverse event in the study was mild (grade 1) and consisted of hypotension. CONCLUSION: HSK3486 is a promising anaesthetic candidate with rapid onset of action and clear absorption, distribution, metabolism, excretion (ADME) processes. HSK3486 showed favourable pharmacokinetic characteristics, pharmacodynamic responses and safety at the study dose.


Asunto(s)
Anestésicos , Administración Intravenosa , Administración Oral , Heces , Voluntarios Sanos , Humanos , Tasa de Depuración Metabólica
6.
Curr Drug Metab ; 21(5): 357-367, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407271

RESUMEN

BACKGROUND: Surufatinib is a potent small-molecule tyrosine kinase inhibitor and exhibited significant efficacy in the treatment of neuroendocrine tumors in clinical trials. OBJECTIVE: The absorption, metabolism and excretion of surufatinib were investigated in rats and human volunteers following a single oral dose of [14C] surufatinib. METHODS: The radioactivity was measured in plasma, urine, feces and bile by liquid scintillation counting, and the metabolites were characterized by liquid chromatography coupled to mass spectrometry. RESULTS: Surufatinib was orally absorbed similarly in rats and human volunteers, with the median Tmax of 4 hours post-dose. The estimated t1/2 appeared longer in humans than in rats (mean t1/2: 3.12 hour for male rats, 6.48 hours for female rats and 23.3 hours for male human volunteers). The excretion of surufatinib was almost complete in rats and human volunteers in the studies, with the total radioactivity recovery of >90% of the dose. Similarly, in rats and humans, fecal excretion predominated (approximately 87% of the dose recovered in feces and only 5% in urine). The parent drug was the major radioactive component detected in the plasma extracts of rats and humans, and no single circulating metabolite accounted for >10% of the total radioactivity. Unchanged drug was a minor radioactive component in the excreta of rats and humans. CONCLUSION: Fecal excretion was the predominant way for the elimination of surufatinib and its metabolites in rats and humans. No disproportionate circulating metabolite was observed in humans.


Asunto(s)
Antineoplásicos/farmacocinética , Indoles/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Administración Oral , Adulto , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/orina , Bilis/metabolismo , Heces/química , Femenino , Humanos , Indoles/efectos adversos , Indoles/orina , Absorción Intestinal , Masculino , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/orina , Pirimidinas/efectos adversos , Pirimidinas/orina , Ratas Sprague-Dawley , Sulfonamidas/efectos adversos , Sulfonamidas/orina
7.
Inorg Chem ; 56(17): 10404-10411, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28816450

RESUMEN

A new series of mixed valence perovskites, La0.75K0.25AMnTiO6 (A = Sr and Ba) nanocubes, have been synthesized by a mild hydrothermal route. From powder X-ray diffraction (XRD) analysis, the crystal structure of La0.75K0.25AMnTiO6 was solved as the orthorhombic symmetry (space group Pbnm) with a random A-site or B-site arrangement. The phase purity of the products was confirmed by ICP, SEM, and EDS analyses, and the oxidation states of the B-site metal atoms were determined to be +4 for Ti and +3/+4 for Mn from XPS results. The soft ferromagnetic behavior is present in both samples. The Curie point TC has been detected as high as 309 K in La0.75K0.25SrMnTiO6 and 217 K in La0.75K0.25BaMnTiO6. A Griffiths phase can be observed at the high temperature region, which is related to the magnetic inhomogeneity induced by the existence of short-range ferromagnetic clusters. The resistivity measurements indicate that the semiconducting properties of the samples can be depicted better by variable range hopping (VRH) due to a diluted double-exchange interaction of Mn3+-O-Mn4+ with Ti4+ doping.

8.
Drug Metab Lett ; 10(4): 228-239, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28103788

RESUMEN

OBJECTIVE: This study describes the in vivo pharmacokinetics and metabolism of [14C]labeled XQ-1H in male rats. METHODS: XQ-1H is a methanesulfonate of XQ, 10-O-(N,N-dimethylaminoethyl)-ginkgolide B, a derivative of ginkgolide B (GB) with enhanced water solubility. Since it is very difficult to synthesize radiolabeled GB, the results obtained in this study may provide helpful insight to further ADME investigation of GB and its analogue compounds. After an i.v. administration of [14C]XQ-1H to male rats, XQ (the freebase form of XQ-1H) was extensively hydrolyzed, moderately metabolized, and mainly excreted in feces (71.5% of the dose) via the biliary route. RESULTS: The main enzyme mediated metabolic pathways were mono- and di-demthylation. Using the radiolabel form of XQ-1H, the temporal binding of XQ to red blood cells was observed. CONCLUSION: Binding of XQ to RBCs may lower the blood's viscosity and thus provide symptomatic improvement of ischemic stroke patients.


Asunto(s)
Viscosidad Sanguínea/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Eritrocitos/metabolismo , Ginkgólidos/farmacología , Lactonas/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/sangre , Radioisótopos de Carbono/química , Desmetilación , Heces/química , Ginkgo biloba/química , Ginkgólidos/química , Ginkgólidos/metabolismo , Ginkgólidos/uso terapéutico , Eliminación Hepatobiliar , Inyecciones Intravenosas , Lactonas/química , Lactonas/metabolismo , Lactonas/uso terapéutico , Masculino , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/sangre , Distribución Tisular
9.
Sci Rep ; 5: 11181, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26153673

RESUMEN

In human being's history, both the Iron Age and Silicon Age thrived after a matured massive processing technology was developed. Graphene is the most recent superior material which could potentially initialize another new material Age. However, while being exploited to its full extent, conventional processing methods fail to provide a link to today's personalization tide. New technology should be ushered in. Three-dimensional (3D) printing fills the missing linkage between graphene materials and the digital mainstream. Their alliance could generate additional stream to push the graphene revolution into a new phase. Here we demonstrate for the first time, a graphene composite, with a graphene loading up to 5.6 wt%, can be 3D printable into computer-designed models. The composite's linear thermal coefficient is below 75 ppm·°C(-1) from room temperature to its glass transition temperature (Tg), which is crucial to build minute thermal stress during the printing process.

10.
Drug Metab Dispos ; 42(5): 839-43, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24595680

RESUMEN

It has been proposed that in humans 4ß-hydroxycholesterol is formed mainly by CYP3A-catalyzed metabolism of cholesterol and thus may serve as an endogenous marker for CYP3A activity. The cynomolgus monkey is widely used as one of the nonrodent preclinical safety species in pharmaceutical research. In the current study, the potential application of 4ß-hydroxycholesterol as an endogenous biomarker of CYP3A in response to drug treatment was evaluated in cynomolgus monkeys. Following multiple oral administration of rifampicin (a known CYP3A inducer) at 15 mg/kg/d in cynomolgus monkeys, the mean serum 4ß-hydroxycholesterol levels increased 4-fold from the baseline of 55.3 ± 21.7 to 221 ± 53.4 ng/ml. The mean concentration ratios of 4ß-hydroxycholesterol to cholesterol increased 5-fold. The data suggest that 4ß-hydroxycholesterol formation from cholesterol metabolism was induced by rifampicin treatment in monkeys. This observation correlated with the metabolism of midazolam (a probe substrate of CYP3A activity) monitored in the same study. The serum exposure (area under the curve) of midazolam was markedly decreased by ∼95%, confirming the induction of CYP3A catalytic activity by rifampicin treatment in monkeys. The formation of 4ß-hydroxycholesterol from cholesterol was specifically mediated by recombinant cynomolgus CYP3A8 and CYP3A5. The Km values of CYP3A8 and CYP3A5 for 4ß-hydroxycholesterol formation from cholesterol were 204 and 104 µM, respectively, and Vmax values were 0.600 and 0.310 pg/pmol/min, respectively. The results suggest that 4ß-hydroxycholesterol can be used as an endogenous biomarker to identify strong CYP3A inducers in cynomolgus monkeys, which may help to evaluate drug-drug interaction potential of drug candidates in preclinical settings.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Hidroxicolesteroles/sangre , Administración Oral , Animales , Biomarcadores/sangre , Biotransformación , Colesterol/metabolismo , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP3A/biosíntesis , Citocromo P-450 CYP3A/genética , Evaluación Preclínica de Medicamentos , Inducción Enzimática , Femenino , Macaca fascicularis , Masculino , Midazolam/sangre , Midazolam/farmacocinética , Rifampin/farmacología , Especificidad por Sustrato , Espectrometría de Masas en Tándem
11.
Xenobiotica ; 41(12): 1063-75, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21859393

RESUMEN

Apremilast is a novel, orally available small molecule that specifically inhibits PDE4 and thus modulates multiple pro- and anti-inflammatory mediators, and is currently under clinical development for the treatment of psoriasis and psoriatic arthritis. The pharmacokinetics and disposition of [(14)C]apremilast was investigated following a single oral dose (20 mg, 100 µCi) to healthy male subjects. Approximately 58% of the radioactive dose was excreted in urine, while faeces contained 39%. Mean C(max), AUC(0-∞) and t(max) values for apremilast in plasma were 333 ng/mL, 1970 ng*h/mL and 1.5 h. Apremilast was extensively metabolized via multiple pathways, with unchanged drug representing 45% of the circulating radioactivity and <7% of the excreted radioactivity. The predominant metabolite was O-desmethyl apremilast glucuronide, representing 39% of plasma radioactivity and 34% of excreted radioactivity. The only other radioactive components that represented >4% of the excreted radioactivity were O-demethylated apremilast and its hydrolysis product. Additional minor circulating and excreted compounds were formed via O-demethylation, O-deethylation, N-deacetylation, hydroxylation, glucuronidation and/or hydrolysis. The major metabolites were at least 50-fold less pharmacologically active than apremilast. Metabolic clearance of apremilast was the major route of elimination, while non-enzymatic hydrolysis and excretion of unchanged drug were involved to a lesser extent.


Asunto(s)
Inhibidores de Fosfodiesterasa 4/administración & dosificación , Inhibidores de Fosfodiesterasa 4/farmacocinética , Talidomida/análogos & derivados , Administración Oral , Adulto , Radioisótopos de Carbono , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/metabolismo , Radiactividad , Talidomida/administración & dosificación , Talidomida/química , Talidomida/metabolismo , Talidomida/farmacocinética , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
12.
Drug Metab Dispos ; 37(8): 1738-48, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19420130

RESUMEN

The metabolism and disposition of [(14)C]apixaban, a potent, reversible, and direct inhibitor of coagulation factor Xa, were investigated in mice, rats, rabbits, dogs, and humans after a single oral administration and in incubations with hepatocytes. In plasma, the parent compound was the major circulating component in mice, rats, dogs, and humans. O-Demethyl apixaban sulfate (M1) represented approximately 25% of the parent area under the time curve in human plasma. This sulfate metabolite was present, but in lower amounts relative to the parent, in plasma from mice, rats, and dogs. Rabbits showed a plasma metabolite profile distinct from that of other species with apixaban as a minor component and M2 (O-demethyl apixaban) and M14 (O-demethyl apixaban glucuronide) as prominent components. The fecal route was a major elimination pathway, accounting for >54% of the dose in animals and >46% in humans. The urinary route accounted for <15% of the dose in animals and 25 to 28% in humans. Apixaban was the major component in feces of every species and in urine of all species except rabbit. M1 and M2 were common prominent metabolites in urine and feces of all species as well as in bile of rats and humans. In vivo metabolite profiles showed quantitative differences between species and from in vitro metabolite profiles, but all human metabolites were found in animal species. After intravenous administration of [(14)C]apixaban to bile duct-cannulated rats, the significant portion (approximately 22%) of the dose was recovered as parent drug in the feces, suggesting direct excretion of the drug from gastrointestinal tracts of rats. Overall, apixaban was effectively eliminated via multiple elimination pathways in animals and humans, including oxidative metabolism, and direct renal and intestinal excretion.


Asunto(s)
Fibrinolíticos/farmacocinética , Pirazoles/farmacocinética , Piridonas/farmacocinética , Administración Oral , Adolescente , Adulto , Animales , Área Bajo la Curva , Bilis/metabolismo , Biotransformación , Radioisótopos de Carbono , Remoción de Radical Alquila , Perros , Inhibidores del Factor Xa , Heces/química , Femenino , Fibrinolíticos/administración & dosificación , Fibrinolíticos/sangre , Fibrinolíticos/orina , Glucurónidos/metabolismo , Hepatocitos/metabolismo , Humanos , Infusiones Intravenosas , Macaca fascicularis , Masculino , Metabolómica/métodos , Ratones , Ratones Endogámicos ICR , Persona de Mediana Edad , Oxidación-Reducción , Pirazoles/administración & dosificación , Pirazoles/sangre , Pirazoles/orina , Piridonas/administración & dosificación , Piridonas/sangre , Piridonas/orina , Conejos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Adulto Joven
13.
World J Gastroenterol ; 15(12): 1518-23, 2009 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-19322928

RESUMEN

AIM: To observe the biotransformation process of a Chinese compound, aesculin, by human gut bacteria, and to identify its metabolites in rat urine. METHODS: Representative human gut bacteria were collected from 20 healthy volunteers, and then utilized in vitro to biotransform aesculin under anaerobic conditions. At 0, 2, 4, 8, 12, 16, 24, 48 and 72 h post-incubation, 10 mL of culture medium was collected. Metabolites of aesculin were extracted 3 x from rat urine with methanol and analyzed by HPLC. For in vivo metabolite analysis, aesculetin (100 mg/kg) was administered to rats via stomach gavage, rat urine was collected from 6 to 48 h post-administration, and metabolite analysis was performed by LC/ESI-MS and MS/MS in the positive and negative modes. RESULTS: Human gut bacteria could completely convert aesculin into aesculetin in vitro. The biotransformation process occurred from 8 to 24 h post-incubation, with its highest activity was seen from 8 to 12 h. The in vitro process was much slower than the in vivo process. In contrast to the in vitro model, six aesculetin metabolites were identified in rat urine, including 6-hydroxy-7-gluco-coumarin (M1), 6-hydroxy-7-sulf-coumarin (M2), 6, 7-di-gluco-coumarin (M3), 6-glc-7-gluco-coumarin (M4), 6-O-methyl-7-gluco-coumarin (M5) and 6-O-methyl-7-sulf-coumarin (M6). Of which, M2 and M6 were novel metabolites. CONCLUSION: Aesculin can be transferred into aesculetin by human gut bacteria and is further modified by the host in vivo. The diverse metabolites of aesculin may explain its pleiotropic pharmaceutical effects.


Asunto(s)
Bacterias Anaerobias/metabolismo , Esculina/metabolismo , Intestinos/microbiología , Umbeliferonas/metabolismo , Animales , Bacterias Anaerobias/crecimiento & desarrollo , Bacterias Anaerobias/aislamiento & purificación , Biotransformación , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Medios de Cultivo , Esculina/orina , Femenino , Humanos , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Ratas
14.
Invest New Drugs ; 26(6): 541-51, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18600301

RESUMEN

Combination of platinum with topoisomerase-I inhibitors are synergistic. The objectives of this study were to determine MTD range and toxicity profile of combinations of oral 9-nitrocamptothecin (9NC) and intravenous cisplatin in patients with refractory solid tumors. Each course was 28 days starting on day 1 with cisplatin, and then 9NC daily for 5 days/week for three weeks. A new two arm crossing design was created: patients in arm 1 were treated with at the single agent recommended dose of cisplatin (50 mg/m(2)), and increasing doses of 9NC and in arm 2 with the single agent recommended dose of 9NC (1.5 mg/day) and increasing dose of cisplatin. Once a dose limiting toxicity was observed, the dose of the escalated drug was decreased by one level, and the fixed-dose drug was then escalated. A 3 + 3 design was used. Eligibility criteria were standard for a phase I trial. Pharmacokinetics was performed. Eighteen patients were treated on Arm 1, 3 at the crossing level, and 33 on Arm 2. Dose limiting toxicities were gastrointestinal at the crossing dose level. After crossing, prolonged grade 3 thrombocytopenia was the DLT in arm 1, and grade 4 neutropenia in Arm 2. Only one patient with ovarian cancer had a partial remission, and 12 patients had disease stabilization (24% of clinical benefit). A Bayesian optimal dose finding was tested post-facto. The recommended doses for phase II studies by the 3 + 3 design are cisplatin 60 mg/m(2) and 9NC 1.25 mg/day and cisplatin 40 mg/m(2) and 9NC 2.0 mg/day. The Bayesian optimal dose finding suggested a different solution, closest to that of the latter dosing which may be less toxic.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias/tratamiento farmacológico , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Teorema de Bayes , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Cisplatino/administración & dosificación , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Inducción de Remisión/métodos , Adulto Joven
15.
J Pharm Biomed Anal ; 46(1): 75-81, 2008 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-17931815

RESUMEN

The lipophilicity and solubility profiles of bis(12)-hupyridone (B12H) and bis(7)-tacrine (B7T), two novel acetylcholinesterase inhibitors dimerized from huperzine A fragments and tacrine, respectively, were investigated over a broad pH range. Lipophilicity was assessed by both shake flask method with 1-octanol-water system and a reverse-phase HPLC system with methanol-water as mobile phase. The former method was used for determining the lipophilicities of the ionized forms (log D) of the dimers while the latter method was used for that of the neutral forms (log P). The log P values for B12H and B7T were found to be 5.4 and 8.2, respectively, indicating that the two dimers are highly lipophilic. The solubilities of both dimers were found to be affected by pH. The solubility of B12H was >1.41 mg/ml when the pH was <7, but <0.06 mg/ml when the pH was >8. The solubility of B7T was >0.26 mg/ml when the pH was <9, but <0.005 mg/ml when the pH was >12. The ionic strength of a solution could affect the solubilities considerably (11.16 mg/ml for B12H and 12.71 mg/ml for B7T in water; 2.07 mg/ml for B12H and 0.36 mg/ml for B7T in saline). The ionization constants (pK(a)) of the two dimers were determined by UV spectrophotometry. Both dimers were found to have two pK(a) values: 7.5+/-0.1 (pK(a1)) and 10.0+/-0.2 (pK(a2)) for B12H; and 8.7+/-0.1 (pK(a1)) and 10.7+/-0.4 (pK(a2)) for B7T. Furthermore, an in vivo pharmacological assay conducted in mice showed that a maximum AChE inhibition occurred 15 min after the single-dose and intraperitoneal administration of either dimer. This indicates that the two dimers may easily cross the blood-brain barrier. In summary, these physiochemical characteristics suggest that the two dimers may be promising candidates for the development of better drugs for Alzheimer's disease.


Asunto(s)
Química Física/métodos , Inhibidores de la Colinesterasa/química , Quinolonas/química , Sesquiterpenos/química , Tacrina/análogos & derivados , Tacrina/química , Administración Oral , Algoritmos , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Química Física/normas , Inhibidores de la Colinesterasa/administración & dosificación , Inhibidores de la Colinesterasa/farmacología , Cromatografía Líquida de Alta Presión/métodos , Dimerización , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos ICR , Estructura Molecular , Quinolonas/farmacología , Sesquiterpenos/administración & dosificación , Sesquiterpenos/farmacología , Solubilidad , Espectrofotometría Ultravioleta/métodos , Tacrina/administración & dosificación , Tacrina/farmacología
16.
J Pharm Biomed Anal ; 44(5): 1133-8, 2007 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-17628383

RESUMEN

An analytical method using on-line high performance liquid chromatography-tandem mass spectrometry with electrospray ionization was developed and applied for the quantification of bis(7)-tacrine (B7T) in rat blood. B7T and pimozide (internal standard, IS) were extracted in a single step from 100 microl of alkalized blood with ethyl acetate. Analytes were separated using an Extend C-18 column at 25 degrees C. The elution was achieved isocratically with a mobile phase composed of 0.05% aqueous formic acid and acetonitrile (60:40, v/v) at a flow rate of 0.35 ml/min. Quantification was achieved by monitoring the selected ions at m/z 247 for B7T and m/z 462-->m/z 328 for pimozide. Retention times were 1.45 and 2.23 min for B7T and IS, respectively. Calibration curves were linear in the range from 86.4 to 2160.0 ng/ml. The established method is rapid, selective and sensitive for the identification and quantification of B7T in biological samples. The assay is accurate (bias <10%) and reproducible (intra- and inter-day variation <10%), with detection and quantification limit of 3.6 and 42.3 ng/ml, respectively. Furthermore, it was successfully applied for the pharmacokinetic measurement of B7T in rat with a single intravenous administration at 0.3mg/kg.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Cromatografía Líquida de Alta Presión/métodos , Tacrina/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Animales , Área Bajo la Curva , Estabilidad de Medicamentos , Congelación , Semivida , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Tacrina/administración & dosificación , Tacrina/sangre , Tacrina/química , Tacrina/farmacocinética , Tacrina/uso terapéutico , Factores de Tiempo
17.
Am J Ther ; 14(3): 280-90, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17515705

RESUMEN

The in vitro metabolism of ciclesonide, a novel inhaled nonhalogenated glucocorticoid for the treatment of asthma, was compared in cryopreserved hepatocytes from mice, rats, rabbits, dogs, and humans. Incubations of C-ciclesonide with individual hepatocyte suspensions revealed similar metabolite profiles in all 5 in vitro systems used. Ciclesonide was rapidly converted to its active metabolite, desisobutyryl-ciclesonide (des-CIC). Des-CIC was then extensively metabolized to pharmacologically inactive metabolites through oxidation and reduction, followed by glucuronidation. A total of 12 groups of metabolites derived from des-CIC were characterized and identified by liquid chromatography/radioactivity monitor/mass spectrometry. Oxidation occurred on both the cyclohexane ring and the steroid moiety. Hippuric acid formation by cleavage of the cyclohexylmethyl moiety of ciclesonide, followed by aromatization of the cyclohexane ring through multiple steps of hydroxylation, dehydration, and conjugation with glycine, was found in rat, rabbit, and human hepatocyte incubations. The results indicated that ciclesonide and its active metabolite, des-CIC, were extensively metabolized in vitro in animal and human hepatocytes and that the metabolite profiles in mouse, rat, rabbit, and dog hepatocytes were similar to the profiles in human hepatocytes.


Asunto(s)
Antialérgicos/metabolismo , Hepatocitos/metabolismo , Pregnenodionas/metabolismo , Animales , Perros , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos ICR , Conejos , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
18.
Am J Ther ; 13(6): 490-501, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17122529

RESUMEN

The pharmacokinetics, metabolism, and excretion of ciclesonide, a novel and effective inhaled glucocorticoid for the treatment of asthma, were investigated after intravenous and oral administration of 14C-ciclesonide in the mouse, rat, rabbit, and dog. The pharmacokinetics of ciclesonide in all animal species were characterized by a low oral bioavailability (approximately 6% or less), a high clearance, and a large volume of distribution. The apparent terminal half-life of ciclesonide was short; the apparent terminal half-life of the active desisobutyryl-ciclesonide metabolite (des-CIC or M1) was longer and ranged from 2.4 to 6.9 hours in the 4 species. Metabolites derived from ciclesonide in serum (or plasma) and excreta samples from the 4 animal species were profiled and identified by LC/RAM/MS (liquid chromatography/radioactivity monitor/mass spectrometry). Ciclesonide was extensively metabolized to yield des-CIC, which was further metabolized to primarily yield hippuric acid and hydroxylated metabolites, namely, isomers of cyclohexane-monohydroxylated des-CIC and B-ring-monohydroxylated des-CIC. Greater than 90% of intravenous and oral 14C-ciclesonide doses were recovered in all species; the main elimination route was fecal/biliary. A comparison of in vitro and in vivo metabolite profiles between mice, rats, rabbits, and dogs with those from humans indicated that metabolic pathways for ciclesonide were qualitatively similar in humans and in the 4 animal species.


Asunto(s)
Antiasmáticos/farmacocinética , Pregnenodionas/farmacocinética , Administración Oral , Animales , Antiasmáticos/administración & dosificación , Antiasmáticos/sangre , Antiasmáticos/orina , Área Bajo la Curva , Disponibilidad Biológica , Biotransformación , Cromatografía Liquida , Perros , Heces/química , Femenino , Semivida , Humanos , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Ratones , Pregnenodionas/administración & dosificación , Pregnenodionas/sangre , Pregnenodionas/orina , Conejos , Ratas , Ratas Wistar , Espectrometría de Masas en Tándem
19.
Rapid Commun Mass Spectrom ; 20(19): 2969-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16952212

RESUMEN

Some compounds readily form [M+46]+ adduct ions during positive ion electrospray ionization mass spectrometry ((+)ESI-MS) analysis. These [M+46]+ ions were characterized as [M+CH3CH2NH2+H]+ by accurate mass determination. Ethylamine involved in the adduct was proposed to be the reduction product of acetonitrile and this was confirmed using deuterated acetonitrile. Other nitrile-containing compounds tested, including isobutyronitrile and benzonitrile, also formed the adduct ions of the respective amine forms under (+)ESI-MS conditions. Hydrogen/deuterium exchange experiments demonstrated that the reductive hydrogen originated from water. Reduction of nitriles (R-CN) to their respective amines (R-CH2NH2) under (+)ESI-MS conditions expands the ability to identify nitrile-containing chemical unknowns.


Asunto(s)
Aminas/química , Nitrilos/química , Espectrometría de Masa por Ionización de Electrospray/métodos , Acetonitrilos/química , Deuterio/química , Etilaminas/química , Marcaje Isotópico , Oxidación-Reducción
20.
Rapid Commun Mass Spectrom ; 20(5): 764-70, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16463359

RESUMEN

Acceleration of liquid chromatography/mass spectrometric (LC/MS) analysis for metabolite identification critically relies on effective data processing since the rate of data acquisition is much faster than the rate of data mining. The rapid and accurate identification of metabolite peaks from complex LC/MS data is a key component to speeding up the process. Current approaches routinely use selected ion chromatograms that can suffer severely from matrix effects. This paper describes a new method to automatically extract and filter metabolite-related information from LC/MS data obtained at unit mass resolution in the presence of complex biological matrices. This approach is illustrated by LC/MS analysis of the metabolites of verapamil from a rat microsome incubation spiked with biological matrix (bile). MS data were acquired in profile mode on a unit mass resolution triple-quadrupole instrument, externally calibrated using a unique procedure that corrects for both mass axis and mass spectral peak shape to facilitate metabolite identification with high mass accuracy. Through the double-filtering effects of accurate mass and isotope profile, conventional extracted ion chromatograms corresponding to the parent drug (verapamil at m/z 455), demethylated verapamil (m/z 441), and dealkylated verapamil (m/z 291), that contained substantial false-positive peaks, were simplified into chromatograms that are substantially free from matrix interferences. These filtered chromatograms approach what would have been obtained by using a radioactivity detector to detect radio-labeled metabolites of interest.


Asunto(s)
Cromatografía Líquida de Alta Presión , Espectrometría de Masa por Ionización de Electrospray/métodos , Animales , Bilis/metabolismo , Técnicas In Vitro , Masculino , Microsomas Hepáticos/metabolismo , Ratas , Reproducibilidad de los Resultados , Verapamilo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...