Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Drug Des Devel Ther ; 15: 557-576, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33603345

RESUMO

PURPOSE: The aim of the present study was to develop an optimized Genkwanin (GKA)-loaded self-nanoemulsifying drug delivery system (SNEDDS) formulation to enhance the solubility, intestinal permeability, oral bioavailability and anti-colitis-associated colorectal cancer (CAC) activity of GKA. METHODS: We designed a SNEDDS comprised oil phase, surfactants and co-surfactants for oral administration of GKA, the best of which were selected by investigating the saturation solubility, constructing pseudo-ternary phase diagrams, followed by optimizing thermodynamic stability, emulsification efficacy, self-nanoemulsification time, droplet size, transmission electron microscopy (TEM), drug release and intestinal permeability. In addition, the physicochemical properties and pharmacokinetics of GKA-SNEDDS were characterized, and its anti-colitis-associated colorectal cancer (CAC) activity and potential mechanisms were evaluated in AOM/DSS-induced C57BL/6J mice model. RESULTS: The optimized nanoemulsion formula (OF) consists of Maisine CC, Labrasol ALF and Transcutol HP in a weight ratio of 20:60:20 (w/w/w), in which ratio the OF shows multiple improvements, specifically small mean droplet size, excellent stability, fast release properties as well as enhanced solubility and permeability. Pharmacokinetic studies demonstrated that compared with GKA suspension, the relative bioavailability of GKA-SNEDDS was increased by 353.28%. Moreover, GKA-SNEDDS not only significantly prevents weight loss and improves disease activity index (DAI) but also reduces the histological scores of inflammatory cytokine levels as well as inhibiting the formation of colon tumors via inducing tumor cell apoptosis in the AOM/DSS-induced CAC mice model. CONCLUSION: Our results show that the developed GKA-SNEDDS exhibited enhanced oral bioavailability and excellent anti-CAC efficacy. In summary, GKA-SNEDDS, using lipid nanoparticles as the drug delivery carrier, can be applied as a potential drug delivery system for improving the clinical application of GKA.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Colite/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Flavonas/farmacologia , Nanopartículas/química , Administração Oral , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Daphne/química , Relação Dose-Resposta a Droga , Composição de Medicamentos , Emulsões , Flavonas/administração & dosagem , Flavonas/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Nanopartículas/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Ratos , Ratos Sprague-Dawley , Solubilidade , Relação Estrutura-Atividade
2.
Int J Nanomedicine ; 7: 3929-38, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22915844

RESUMO

OBJECTIVE: Previous studies on various enzymosomes (functional lipid vesicles encapsulating an enzyme) have been mostly carried out in vitro and have focused on preserving catalytic activity and improving the stability of the enzyme. Until now, few studies have focused on their in vivo fate. Similarly, although we have previously reported the increased in vitro uricolytic activity (about 2.2 times higher than that of free uricase, or three times higher than that of PEGylated uricase, Puricase(®), under physiological pH and temperature) and improved stability of the novel alkaline enzymosomes (functional lipid vesicles encapsulating uricase from Candida utilis: uricase-containing lipid vesicles, UOXLVs), it is still necessary to study the biological properties and hypouricemic effects of UOXLVs in vivo. METHODS: The enzyme kinetics, pharmacokinetics, pharmacodynamics, immunogenicity, and preliminary safety of UOXLVs were evaluated. RESULTS: The Michaelis constant (K(m)) value of the UOXLVs was slightly lower than that of the free enzyme. The enzyme release from the UOXLVs lasted over 12 hours and their circulation half-life was about sevenfold longer than that of the free uricase. Meanwhile, the UOXLVs had a 22-fold increase in the area under the curve compared with the free uricase. Furthermore, it took less than 3 hours for the UOXLVs to lower the plasma uric acid concentration from a high to a normal level, compared with 6 hours for the free uricase. In addition, the UOXLVs had much less immunogenicity than free uricase and were well tolerated by all animals throughout the observation period. CONCLUSION: The UOXLVs markedly improved the biological properties and enhanced the hypouricemic effects of uricase in vivo.


Assuntos
Candida/enzimologia , Portadores de Fármacos/química , Urato Oxidase/química , Ácido Úrico/metabolismo , Alantoína/sangue , Alantoína/metabolismo , Animais , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Estabilidade Enzimática , Hemólise/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Cinética , Masculino , Ratos , Ratos Sprague-Dawley , Urato Oxidase/metabolismo , Urato Oxidase/farmacologia , Ácido Úrico/sangue
3.
Arch Pharm Res ; 35(3): 499-508, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22477197

RESUMO

A novel pyridostigmine bromide (PB)-phospholipid nanocomplex (PBPLC) was prepared to increase the bioavailability of PB. A central composite design approach was employed for process optimization. The physicochemical properties of PBPLC were investigated by means of differential scanning calorimetry, ultraviolet spectroscopy, Fourier transformed infrared spectroscopy and the n-octano/water partition coefficient. The intestinal permeability of PBPLC was observed via a single pass intestinal perfusion in rats. After oral administration of PBPLC, the concentrations of PB at predetermined time points were determined by HPLC, and the pharmacokinetic parameters were computed by DAS 2.1.1 software. Multiple linear regression analysis for process optimization revealed that the optimal PBPLC was obtained when the values of X(1), X(2), and X(3) were 8, 40°C, and 4 mg/mL, respectively. The average particle size and zeta potential of PBPLC with the optimized formulation were 204.60 nm and -25.12 mV, respectively. Non-covalent interactions between PB and phospholipids were found in the PBPLC. The n-octanol/water partition coefficient of PBPLC was substantially increased. PBPLC had better intestinal permeability in comparison with free PB. Mean plasma drug concentration-time curves of PBPLC and free PB after oral administration were both in accordance with the two-compartment open model. The values of pharmacokinetic parameters of PBPLC and free PB were the peak time (T(max)) 2 h vs 2 h, the maximum concentration (C(max)) 22.79 µg/mL vs 6.00 µg/mL, and the value of the area under the concentration vs time curve (AUC(0-∞)) 7128.21 µg·min/mL vs 1772.36 µg·min/mL, respectively. In conclusion, compared with free PB, PBPLC remarkably improves the oral bioavailability of PB, which is likely due to its higher lipophilicity and permeability.


Assuntos
Inibidores da Colinesterase/administração & dosagem , Inibidores da Colinesterase/farmacocinética , Nanopartículas , Fosfolipídeos/química , Brometo de Piridostigmina/administração & dosagem , Brometo de Piridostigmina/farmacocinética , 1-Octanol/química , Administração Oral , Animais , Disponibilidade Biológica , Varredura Diferencial de Calorimetria , Química Farmacêutica , Inibidores da Colinesterase/sangue , Inibidores da Colinesterase/química , Composição de Medicamentos , Absorção Intestinal , Mucosa Intestinal/metabolismo , Modelos Lineares , Masculino , Modelos Biológicos , Nanotecnologia , Permeabilidade , Brometo de Piridostigmina/sangue , Brometo de Piridostigmina/química , Ratos , Ratos Sprague-Dawley , Solubilidade , Solventes/química , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Tecnologia Farmacêutica/métodos , Água/química
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 43(6): 873-6, 2012 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-23387217

RESUMO

OBJECTIVE: To determine the pharmacokinetics characteristics of mestinon-phospholipid complex (PBPLC) in rats. METHODS: This study adopted a single-dose, randomized, open-label, two-period crossover trial design. Twelve healthy rats were randomly divided into two groups. One group was orally administered with mestinon-phospholipid complex, and the other group was orally administered with reference mestinon solution (1.5 mg/kg of mestinon). The plasma concentrations of the drugs in ophthalmic vein bloods were determined using HPLC. The pharmacokinetic parameters were calculated with the aid of DAS2.1.1 software. RESULTS: Pharmacokinetic parameters of mestinon-phospholipid complex were Tmax 2 h, Cmax 22.79 microg x min/mL and AUC(0-infinity) 7128.21 microg x min/mL, which were different from those of free mestinon--Tmax, 2 h, Cmax 6.00 microg/mL and AUC(0-infinity) 1772.36 microg x min/mL. The relative bioavailability of mestinon-phospholipid complex was 410.98% of free mestinon. CONCLUSION: The oral bioavailability of mestinon increases remarkably when administered as mestinon-phospholipid complex.


Assuntos
Inibidores da Colinesterase/farmacocinética , Fosfolipídeos/farmacocinética , Brometo de Piridostigmina/farmacocinética , Animais , Disponibilidade Biológica , Combinação de Medicamentos , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...