Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Radiother Oncol ; 170: 213-223, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35227733

RESUMO

INTRODUCTION: Radiation therapy (RT) is a major modality for the treatment of prostate cancer (PCa), especially castration-resistant PCa (CRPC). However, hypoxia, often seen in PCa tumors, leads to radiation-resistance. This work investigates the effect of a novel oxygen-generating polymer-lipid manganese dioxide nanoparticle (PLMDs) on improving RT outcomes in CRPC xenograft models by modulating the tumor microenvironment (TME) both before and after RT. MATERIALS AND METHODS: Human PC3 and DU145 PCa cells were used to investigate clonogenic inhibition and DNA repair pathways in vitro. Tumor hypoxia and post-RT angiogenesis were evaluated in a PC3-bearing SCID mouse model. PC3 and DU145 xenografts were used to study the efficacy of PLMD in combination with single or fractionated RT. RESULTS: PLMD plus RT significantly inhibited clonogenic potential, increased DNA double-strand breaks, and reduced DNA damage repair in hypoxic PC3 and DU145 cells as compared to RT alone. PLMD significantly reduced hypoxia-positive areas, hypoxia induced factor 1α (HIF-1α) expression, and protein carbonyl levels (a measure of oxidative stress). Application of PLMD with RT decreased RT-induced angiogenic biomarkers by up to 3-fold. Treatment of the human CRPC xenografts with PLMD plus RT (single or fractionated doses) significantly prolonged median survival of the host compared to RT alone resulting in up to a 40% curative rate. CONCLUSION: PLMD treatment modulated TME and sensitized hypoxic human CRPC cells to RT thus enhancing the efficacy of RT. These results confirmed the potential of PLMD as an adjuvant to RT for the treatment of hypoxic CRPC.


Assuntos
Nanopartículas , Neoplasias de Próstata Resistentes à Castração , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Hipóxia , Masculino , Camundongos , Camundongos SCID , Oxirredução , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/radioterapia , Microambiente Tumoral
2.
Acta Pharmacol Sin ; 38(6): 835-847, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28216624

RESUMO

Lung metastasis is the major cause of death in patients with triple negative breast cancer (TNBC), an aggressive subtype of breast cancer with no effective therapy at present. It has been proposed that dual-targeted therapy, ie, targeting chemotherapeutic agents to both tumor vasculature and cancer cells, may offer some advantages. The present work was aimed to develop a dual-targeted synergistic drug combination nanomedicine for the treatment of lung metastases of TNBC. Thus, Arg-Gly-Asp peptide (RGD)-conjugated, doxorubicin (DOX) and mitomycin C (MMC) co-loaded polymer-lipid hybrid nanoparticles (RGD-DMPLN) were prepared and characterized. The synergism between DOX and MMC and the effect of RGD-DMPLN on cell morphology and cell viability were evaluated in human MDA-MB-231 cells in vitro. The optimal RGD density on nanoparticles (NPs) was identified based on the biodistribution and tumor accumulation of the NPs in a murine lung metastatic model of MDA-MB-231 cells. The microscopic distribution of RGD-conjugated NPs in lung metastases was examined using confocal microscopy. The anticancer efficacy of RGD-DMPLN was investigated in the lung metastatic model. A synergistic ratio of DOX and MMC was found in the MDA-MB-231 human TNBC cells. RGD-DMPLN induced morphological changes and enhanced cytotoxicity in vitro. NPs with a median RGD density showed the highest accumulation in lung metastases by targeting both tumor vasculature and cancer cells. Compared to free drugs, RGD-DMPLN exhibited significantly low toxicity to the host, liver and heart. Compared to non-targeted DMPLN or free drugs, administration of RGD-DMPLN (10 mg/kg, iv) resulted in a 4.7-fold and 31-fold reduction in the burden of lung metastases measured by bioluminescence imaging, a 2.4-fold and 4.0-fold reduction in the lung metastasis area index, and a 35% and 57% longer median survival time, respectively. Dual-targeted RGD-DMPLN, with optimal RGD density, significantly inhibited the progression of lung metastasis and extended host survival.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Nanopartículas/química , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos SCID , Mitomicina/administração & dosagem , Mitomicina/química , Mitomicina/farmacologia , Estrutura Molecular , Oligopeptídeos/administração & dosagem , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Relação Estrutura-Atividade
3.
J Control Release ; 246: 98-109, 2017 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-28017889

RESUMO

Brain metastasis is a fatal disease with limited treatment options and very short survival. Although systemic chemotherapy has some effect on peripheral metastases of breast cancer, it is ineffective in treating brain metastasis due largely to the blood-brain barrier (BBB). Here we developed a BBB-penetrating amphiphilic polymer-lipid nanoparticle (NP) system that efficiently delivered anti-mitotic drug docetaxel (DTX) for the treatment of brain metastasis of triple negative breast cancer (TNBC). We evaluated the biodistribution, brain accumulation, pharmacokinetics and efficacy of DTX-NP in a mouse model of brain metastasis of TNBC. Confocal fluorescence microscopy revealed extravasation of dye-loaded NPs from intact brain microvessels in healthy mice. DTX-NP also extravasated from brain microvessels and accumulated in micrometastasis lesions in the brain. Intravenously injected DTX-NPs increased the blood circulation time of DTX by 5.5-fold and the AUC0-24h in tumor-bearing brain by 5-fold compared to the clinically used DTX formulation Taxotere®. The kinetics of NPs in the brain, determined by ex vivo fluorescence imaging, showed synchronization with DTX kinetics in the brain measured by LC-MS/MS. This result confirmed successful delivery of DTX by the NPs into the brain and suggested that ex vivo fluorescence imaging of NP could be an effective and quick means for probing drug disposition in the brain. Treatment with the DTX-NP formulation delayed tumor growth by 11-fold and prolonged median survival of tumor-bearing mice by 94% compared to an equivalent dose of Taxotere®, without inducing histological changes in the major organs.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Portadores de Fármacos/metabolismo , Nanopartículas/metabolismo , Tensoativos/metabolismo , Taxoides/administração & dosagem , Neoplasias de Mama Triplo Negativas/patologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Docetaxel , Portadores de Fármacos/química , Feminino , Humanos , Camundongos SCID , Nanopartículas/química , Polímeros/metabolismo , Tensoativos/química , Taxoides/farmacocinética , Taxoides/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
4.
Drug Deliv Transl Res ; 5(1): 15-26, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25787336

RESUMO

αvß3 integrin receptors expressed on cancer cell surfaces play a crucial role in promoting tumor angiogenesis and cancer cell metastasis. Thus, cyclic arginyl-glycyl-aspartic acid (cRGD) peptides have been explored as a αvß3 integrin receptor-specific targeting moiety for the targeted delivery of nanoparticle-loaded therapeutics. However, our previous study showed that cyclic RGD could act as a double-edged sword that, on one hand, extended the retention of cRGD-modified solid lipid nanoparticles (RGD-SLNs) at αvß3 integrin receptor overexpressing breast carcinoma, and yet on the other hand, decreased the amount of tumor accumulation of RGD-SLNs attributable to the greater uptake by the mononuclear phagocyte system (MPS). Therefore, we aimed to optimize the RGD-decorated nanoparticle systems for (1) inhibiting αvß3 integrin receptor overexpressing tumor cell metastasis and (2) increasing nanoparticle accumulation to tumor site. SLNs with cRGD content ranging from 0 to 10 % mol of total polyethyleneglycol (PEG) chains were synthesized. The binding of RGD-SLNs with αvß3 integrin receptors increased with increasing cRGD concentration on the nanoparticles. RGD-SLNs were demonstrated to inhibit MDA-MB-231 cell adhesion to fibronectin and invasion through Matrigel. In vivo whole-body fluorescence imaging revealed that 1 % cRGD on the SLNs' surface had maximum tumor accumulation with extended tumor retention among all formulations tested in an orthotopic MDA-MB-231/EGFP breast tumor model. This work has laid a foundation for further development of anticancer drug-loaded optimized cRGD nanoparticle formulations for the treatment of breast cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Integrina alfaVbeta3/metabolismo , Nanopartículas/administração & dosagem , Oligopeptídeos/administração & dosagem , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Lipídeos/química , Camundongos Nus , Nanopartículas/química , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Oligopeptídeos/farmacologia , Polietilenoglicóis/química , Distribuição Tecidual
5.
ACS Nano ; 8(10): 9925-40, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25307677

RESUMO

Metastatic brain cancers, in particular cancers with multiple lesions, are one of the most difficult malignancies to treat owing to their location and aggressiveness. Chemotherapy for brain metastases offers some hope. However, its efficacy is severely limited as most chemotherapeutic agents are incapable of crossing the blood-brain barrier (BBB) efficiently. Thus, a multifunctional nanotheranostic system based on poly(methacrylic acid)-polysorbate 80-grafted-starch was designed herein for the delivery of BBB-impermeable imaging and therapeutic agents to brain metastases of breast cancer. In vivo magnetic resonance imaging and confocal fluorescence microscopy were used to confirm extravasation of gadolinium and dye-loaded nanoparticles from intact brain microvessels in healthy mice. The targetability of doxorubicin (Dox)-loaded nanoparticles to intracranially established brain metastases of breast cancer was evaluated using whole body and ex vivo fluorescence imaging of the brain. Coexistence of nanoparticles and Dox in brain metastatic lesions was further confirmed by histological and microscopic examination of dissected brain tissue. Immuno-histochemical staining for caspase-3 and terminal-deoxynucleotidyl transferase dUTP nick end labeling for DNA fragmentation in tumor-bearing brain sections revealed that Dox-loaded nanoparticles selectively induced cancer cell apoptosis 24 h post-injection, while sparing normal brain cells from harm. Such effects were not observed in the mice treated with free Dox. Treatment with Dox-loaded nanoparticles significantly inhibited brain tumor growth compared to free Dox at the same dose as assessed by in vivo bioluminescence imaging of the brain metastases. These findings suggest that the multifunctional nanoparticles are promising for the treatment of brain metastases.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Doxorrubicina/uso terapêutico , Animais , Antibióticos Antineoplásicos/administração & dosagem , Barreira Hematoencefálica , Doxorrubicina/administração & dosagem , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Imageamento por Ressonância Magnética , Camundongos
6.
ACS Nano ; 8(4): 3202-12, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24702320

RESUMO

Insufficient oxygenation (hypoxia), acidic pH (acidosis), and elevated levels of reactive oxygen species (ROS), such as H2O2, are characteristic abnormalities of the tumor microenvironment (TME). These abnormalities promote tumor aggressiveness, metastasis, and resistance to therapies. To date, there is no treatment available for comprehensive modulation of the TME. Approaches so far have been limited to regulating hypoxia, acidosis, or ROS individually, without accounting for their interdependent effects on tumor progression and response to treatments. Hence we have engineered multifunctional and colloidally stable bioinorganic nanoparticles composed of polyelectrolyte-albumin complex and MnO2 nanoparticles (A-MnO2 NPs) and utilized the reactivity of MnO2 toward peroxides for regulation of the TME with simultaneous oxygen generation and pH increase. In vitro studies showed that these NPs can generate oxygen by reacting with H2O2 produced by cancer cells under hypoxic conditions. A-MnO2 NPs simultaneously increased tumor oxygenation by 45% while increasing tumor pH from pH 6.7 to pH 7.2 by reacting with endogenous H2O2 produced within the tumor in a murine breast tumor model. Intratumoral treatment with NPs also led to the downregulation of two major regulators in tumor progression and aggressiveness, that is, hypoxia-inducible factor-1 alpha and vascular endothelial growth factor in the tumor. Combination treatment of the tumors with NPs and ionizing radiation significantly inhibited breast tumor growth, increased DNA double strand breaks and cancer cell death as compared to radiation therapy alone. These results suggest great potential of A-MnO2 NPs for modulation of the TME and enhancement of radiation response in the treatment of cancer.


Assuntos
Acidose/tratamento farmacológico , Compostos de Manganês/farmacologia , Nanopartículas/química , Óxidos/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Soroalbumina Bovina/química , Microambiente Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Transporte Biológico , Bovinos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Humanos , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Compostos de Manganês/química , Compostos de Manganês/metabolismo , Compostos de Manganês/uso terapêutico , Camundongos , Modelos Moleculares , Óxidos/química , Óxidos/metabolismo , Óxidos/uso terapêutico , Oxigênio/metabolismo , Conformação Proteica
7.
Eur J Pharm Biopharm ; 82(3): 587-97, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22995704

RESUMO

This work investigated the capability of a new nanoparticulate system, based on terpolymer of starch, polymethacrylic acid and polysorbate 80, to load and release doxorubicin (Dox) as a function of pH and to evaluate the anticancer activity of Dox-loaded nanoparticles (Dox-NPs) to overcome multidrug resistance (MDR) in human breast cancer cells in vitro. The Dox-NPs were characterized by Fourier transform infrared spectroscopy (FTIR), isothermal titration calorimetry (ITC), transmission electron microscopy (TEM), and dynamic light scattering (DLS). The cellular uptake and cytotoxicity of the Dox-loaded nanoparticles were investigated using fluorescence microscopy, flow cytometry, and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay. The nanoparticles were able to load up to 49.7±0.3% of Dox with a high loading efficiency of 99.9±0.1%, while maintaining good colloidal stability. The nanoparticles released Dox at a higher rate at acidic pH attributable to weaker Dox-polymer molecular interactions evidenced by ITC. The Dox-NPs were taken up by the cancer cells in vitro and significantly enhanced the cytotoxicity of Dox against human MDR1 cells with up to a 20-fold decrease in the IC50 values. The results suggest that the new terpolymeric nanoparticles are a promising vehicle for the controlled delivery of Dox for treatment of drug resistant breast cancer.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/farmacologia , Nanopartículas , Animais , Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/patologia , Coloides , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Estabilidade de Medicamentos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Polímeros/química , Ácidos Polimetacrílicos/química , Polissorbatos/química , Ratos , Amido/química
8.
ACS Nano ; 5(3): 1958-66, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21338075

RESUMO

Despite broad applications of quantum dots (QDs) in vitro, severe toxicity and dominant liver uptake have limited their clinical application. QDs that excite and emit in the ultraviolet and visible regions have limited in vivo applicability due to significant optical interference exerted by biological fluids and tissues. Hence we devised a new biocompatible hybrid fluorophore composed of near-infrared-emitting PbSe quantum dots encapsulated in solid fatty ester nanoparticles (QD-FEN) for in vivo imaging. The quantum yield and tissue penetration depth of the QD-FEN were characterized, and their biological fate was examined in a breast tumor-bearing animal model. It was found for the first time that chemical modification of the headgroup of QD-encapsulating organic fatty acids was a must as these groups quenched the photoluminescence of PbSe nanocrystals. The use of fatty esters enhanced aqueous quantum yields of PbSe QDs up to ∼45%, which was 50% higher than that of water-soluble PbSe nanocrystals in an aqueous medium. As a result, a greater than previously reported tissue penetration depth of fluorescence was recorded at 710 nm/840 nm excitation/emission wavelengths. The QD-FEN had much lower short-term cytotoxicity compared to nonencapsulated water-soluble QDs. More importantly, reduced liver uptake, increased tumor retention, lack of toxic response, and nearly complete clearance of QD-FEN from the tested animals was demonstrated. With a combination of near-infrared spectral properties, enhanced optical properties,and significantly improved biosafety profile, this novel hybrid nanoparticulate fluorophore system demonstrably provides real-time, deep-tissue fluorescent imaging of live animals, laying a foundation for further development toward clinical application.


Assuntos
Neoplasias da Mama/patologia , Ácidos Graxos/química , Microscopia de Fluorescência/métodos , Nanocápsulas , Pontos Quânticos , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Ácidos Graxos/farmacocinética , Humanos , Camundongos , Camundongos Nus , Nanocápsulas/química , Ratos
9.
Breast Cancer Res Treat ; 121(2): 323-33, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19618264

RESUMO

Multidrug resistance (MDR) is one of the main challenges in the treatment of breast cancer. A new microsphere formulation able to generate reactive oxygen species (ROS) locally was thus investigated for circumventing MDR in breast cancer cells in this work. Glucose oxidase (GOX) was encapsulated in alginate/chitosan hydrogel microspheres (ACMS-GOX). The in vitro cytotoxicity of ACMS-GOX to murine breast cancer EMT6/AR1.0 cells, which overexpress P-glycoprotein (P-gp), was evaluated by a clonogenic assay. The mechanism of the cytotoxicity of ACMS-GOX was investigated by using various extracellular and intracellular ROS scavengers and antioxidant enzyme inhibitors. The effect of lipid peroxidation and cellular uptake of GOX was also evaluated. ACMS-GOX exhibited similar dose and time-dependent cytotoxicity to EMT6/AR1.0 cells as to their wild-type EMT6/WT parent cells, in effect circumventing the MDR phenotype of EMT6/AR1.0 cells. Extracellular H(2)O(2) and intracellular hydroxyl radical were found to play critical roles in the cytotoxicity of ACMS-GOX. Cellular uptake of GOX was negligible and thus not responsible for intracellular ROS generation. Combining ACMS-GOX with intracellular antioxidant inhibitors-enhanced cytoxicity. This work demonstrates that the ACMS-GOX are effective in circumventing P-gp-mediated MDR in breast cancer cells.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/metabolismo , Glucose Oxidase/administração & dosagem , Microesferas , Espécies Reativas de Oxigênio/administração & dosagem , Animais , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Peróxido de Hidrogênio/metabolismo , Camundongos
10.
Pharm Res ; 26(10): 2343-57, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19685212

RESUMO

PURPOSE: This work focused on the characterization and in vitro/in vivo evaluation of an alginate/chitosan microsphere (ACMS) formulation of glucose oxidase (GOX) for the locoregional delivery of reactive oxygen species for the treatment of solid tumors. METHODS: The GOX distribution and ACMS composition were determined by confocal laser scanning microscopy and X-ray photoelectron spectroscopy. The mechanism of GOX loading and GOX-polymer interactions were examined with Fourier transform infrared spectroscopy and differential scanning calorimetry. In vitro cytotoxicity and in vivo efficacy of GOX-encapsulated ACMS (ACMS-GOX) were evaluated in EMT6 breast cancer cells and solid tumors. RESULTS: GOX was loaded into calcium alginate (CaAlg) gel beads via electrostatic interaction and the CaAlg-GOX-chitosan complexation likely stabilized GOX. Higher concentrations of GOX near the surface of ACMS were detected. GOX retained its integrity upon adsorption to CaAlg gel beads during the coating and after release from ACMS. ACMS-GOX exhibited cytotoxicity to the breast cancer cells in vitro and their efficacy increased with increasing incubation time. Intratumorally delivered ACMS-GOX significantly delayed tumor growth with much lower general toxicity than free GOX. CONCLUSION: The results suggest that the ACMS-GOX formulation has the potential for the intratumoral delivery of therapeutic proteins to treat solid tumors.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Modelos Animais de Doenças , Glucose Oxidase/administração & dosagem , Glucose Oxidase/farmacocinética , Microesferas , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Feminino , Glucose Oxidase/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Int J Pharm ; 339(1-2): 148-56, 2007 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-17398046

RESUMO

The purpose of this study was to develop a novel microsphere formulation of glucose oxidase (GOX) with high drug loading, encapsulation efficiency and bioactivity. GOX was encapsulated in alginate/chitosan microspheres (ACMS) using an emulsification-internal gelation, followed by GOX adsorption and polyelectrolyte coating method. The factors influencing GOX loading, encapsulation efficiency and activity of the loaded GOX were investigated. The resultant ACMS in wet state were spherical with a mean diameter of about 138 microm. GOX loading was found to be pH dependent. High GOX loading and encapsulation efficiency were achieved when the pH of the adsorption medium was lower than the isoelectric point (pI) of GOX. GOX loading and encapsulation efficiency increased with increasing GOX concentration in the loading solution, but decreased with increasing chitosan concentration in the coating solution. The activity of loaded GOX increased and then decreased with increasing chitosan concentration. The activity of GOX in ACMS was maintained and showed sustained production of H(2)O(2) as compared to free GOX. Around 90% of the original activity of immobilized GOX remained after lyophilization and storage at -20 degrees C for a month. These results suggest that the ACMS and the fabrication method are suitable for microencapsulation of proteins like GOX.


Assuntos
Enzimas Imobilizadas/administração & dosagem , Glucose Oxidase/administração & dosagem , Microesferas , Adsorção , Alginatos/química , Química Farmacêutica , Quitosana/química , Emulsões , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Hidrogéis/química , Concentração de Íons de Hidrogênio , Tamanho da Partícula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...