Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Am Chem Soc ; 134(9): 4355-62, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22304702

RESUMO

We report a new strategy for differential delivery of antimicrobials to bacterial infection sites with a lipase-sensitive polymeric triple-layered nanogel (TLN) as the drug carrier. The TLN was synthesized by a convenient arm-first procedure using an amphiphilic diblock copolymer, namely, monomethoxy poly(ethylene glycol)-b-poly(ε-caprolactone), to initiate the ring-opening polymerization of the difunctional monomer 3-oxapentane-1,5-diyl bis(ethylene phosphate). The hydrophobic poly(ε-caprolactone) (PCL) segments collapsed and surrounded the polyphosphoester core, forming a hydrophobic and compact molecular fence in aqueous solution which prevented antibiotic release from the polyphosphoester core prior to reaching bacterial infection sites. However, once the TLN sensed the lipase-secreting bacteria, the PCL fence of the TLN degraded to release the antibiotic. Using Staphylococcus aureus (S. aureus) as the model bacterium and vancomycin as the model antimicrobial, we demonstrated that the TLN released almost all the encapsulated vancomycin within 24 h only in the presence of S. aureus, significantly inhibiting S. aureus growth. The TLN further delivered the drug into bacteria-infected cells and efficiently released the drug to kill intracellular bacteria. This technique can be generalized to selectively deliver a variety of antibiotics for the treatment of various infections caused by lipase-secreting bacteria and thus provides a new, safe, effective, and universal approach for the treatment of extracellular and intracellular bacterial infections.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Lipase/metabolismo , Polietilenoglicóis/química , Polietilenoimina/química , Polímeros/química , Antibacterianos/farmacologia , Portadores de Fármacos/metabolismo , Lipase/química , Testes de Sensibilidade Microbiana , Nanogéis , Polietilenoglicóis/metabolismo , Polietilenoimina/metabolismo , Polímeros/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Relação Estrutura-Atividade , Vancomicina/farmacologia
2.
Mol Pharm ; 9(10): 2863-74, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22924580

RESUMO

The particular characteristics of the tumor microenvironment have the potential to strongly promote tumor growth, metastasis and angiogenesis and induce drug resistance. Therefore, the development of effective, systemic therapeutic approaches specifically based on the tumor microenvironment is highly desirable. Hypoxia-inducible factor-1α (HIF-1α) is an attractive therapeutic target because it is a key transcription factor in tumor development and only accumulates in hypoxic tumors. We report here that a cationic mixed micellar nanoparticle (MNP) consisting of amphiphilic block copolymers poly(ε-caprolactone)-block-poly(2-aminoethylethylene phosphate) (PCL(29)-b-PPEEA(21)) and poly(ε-caprolactone)-block-poly(ethylene glycol) (PCL(40)-b-PEG(45)) was a suitable carrier for HIF-1α siRNA to treat hypoxic tumors, which showed an average diameter of 58.0 ± 3.4 nm. The complex MNP(siRNA), formed by the interaction of MNP and siRNA, was transfected into PC3 prostate cancer cells efficiently, while the inhibition of HIF-1α expression by MNP loaded with HIF-1α siRNA (MNP(siHIF)) blocked PC3 cell proliferation, suppressed cell migration and disturbed angiogenesis under in vitro hypoxic mimicking conditions. It was further demonstrated that systemic delivery of MNP(siHIF) effectively inhibited tumor growth in a PC3 prostate cancer xenograft murine model without activating innate immune responses. Moreover, delivery of MNP(siHIF) sensitized PC3 tumor cells to doxorubicin chemotherapy in vitro and in vivo by downregulating MDR1 gene expression which was induced by hypoxia. The underlying concept of use of MNP(siHIF) to block HIF-1α holds promise as an example of a clinical approach using specific siRNA therapy for cancer treatment aimed at the hypoxic tumor microenvironment.


Assuntos
Hipóxia Celular/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Nanopartículas/administração & dosagem , Neoplasias da Próstata/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Doxorrubicina/farmacologia , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Micelas , Nanopartículas/química , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Tamanho da Partícula , Polímeros/administração & dosagem , Polímeros/química , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/química , Transfecção/métodos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
3.
Biomacromolecules ; 9(1): 388-95, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18081252

RESUMO

A series of novel amphiphilic triblock copolymers of poly(ethyl ethylene phosphate) and poly(-caprolactone) (PEEP-PCL-PEEP) with various PEEP and PCL block lengths were synthesized and characterized. These triblock copolymers formed micelles composed of a hydrophobic core of poly(-caprolactone) (PCL) and a hydrophilic shell of poly(ethyl ethylene phosphate) (PEEP) in aqueous solution. The micelle morphology was spherical, determined by transmission electron microscopy. It was found that the size and critical micelle concentration values of the micelles depended on both hydrophobic PCL block length and PEEP hydrophilic block length. The in vitro degradation characteristics of the triblock copolymers were investigated in micellar form, showing that these copolymers were completely biodegradable under enzymatic catalysis of Pseudomonas lipase and phosphodiesterase I. These triblock copolymers were used for paclitaxel (PTX) encapsulation to demonstrate the potential in drug delivery. PTX was successfully loaded into the micelles, and the in vitro release profile was found to be correlative to the polymer composition. These biodegradable triblock copolymer micelles are potential as novel carriers for hydrophobic drug delivery.


Assuntos
Portadores de Fármacos , Micelas , Polímeros/química
4.
Adv Drug Deliv Rev ; 78: 63-76, 2014 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-24548540

RESUMO

Despite the wide use of antibiotics, bacterial infection is still one of the leading causes of hospitalization and mortality. The clinical failure of antibiotic therapy is linked with low bioavailability, poor penetration to bacterial infection sites, and the side effects of antibiotics, as well as the antibiotic resistance properties of bacteria. Antibiotics encapsulated in nanoparticles or microparticles made up of a biodegradable polymer have shown great potential in replacing the administration of antibiotics in their "free" form. Polymeric particles provide protection to antibiotics against environmental deactivation and alter antibiotic pharmacokinetics and biodistribution. Polymeric particles can overcome tissue and cellular barriers and deliver antibiotics into very dense tissues and inaccessible target cells. Polymeric particles can be modified to target or respond to particular tissues, cells, and even bacteria, and thereby facilitate the selective concentration or release of the antibiotic at infection sites, respectively. Thus, the delivery of antibiotics with polymeric particles augments the level of the bioactive drug at the site of infection while reducing the dosage and the dosing frequency. The end results are improved therapeutic effects as well as decreased "pill burden" and drug side effects in patients. The main objective of this review is to analyze recent advances and current perspectives in the use of polymeric antibiotic delivery systems in the treatment of bacterial infection.


Assuntos
Antibacterianos/administração & dosagem , Sistemas de Liberação de Medicamentos , Polímeros/química , Animais , Antibacterianos/farmacocinética , Antibacterianos/uso terapêutico , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/microbiologia , Farmacorresistência Bacteriana , Humanos , Microesferas , Nanopartículas , Distribuição Tecidual
5.
Adv Healthc Mater ; 3(11): 1792-803, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24947820

RESUMO

Patients with Her2-overexpressing (Her2(+)) breast cancers generally have a poorer prognosis due to the high aggressiveness and chemoresistance of the disease. Small interfering RNA (siRNA) targeting the gene encoding polo-like kinase 1 (Plk1; siPlk1) has emerged as an efficient therapeutic agent for Her2(+) breast cancers. Poly(ethylene glycol)-block-poly(D,L-lactide) (PEG-PLA)-based nanoparticles for siRNA delivery were previously developed and optimized. In this study, for targeted delivery of siPlk1 to Her2(+) breast cancer, anti-Her2 single-chain variable fragment antibody (ScFv(Her2))-decorated PEG-PLA-based nanoparticles with si Plk1 encapsulation (ScFv(Her2)-NP(si) Plk1) are developed. With the rationally designed conjugation site, ScFv(Her2)-NP(siRNA) can specifically bind to the Her2 antigen overexpressed on the surface of Her2(+) breast cancer cells. Therefore, ScFv(Her2)-NP(si) Plk1 exhibits improved cellular uptake, promoted Plk1 silencing efficiency, and induced enhanced tumor cell apoptosis in Her2(+) breast cancer cells, when compared with nontargeted NP(si) Plk1. More importantly, ScFv(Her2)-NP(siRNA) markedly enhances the accumulation of siRNA in Her2(+) breast tumor tissue, and remarkably improves the efficacy of tumor suppression. Dose-dependent anti-tumor efficacy further demonstrates that ScFvHer2 -decorated PEG-PLA-based nanoparticles with siPlk1 encapsulation can significantly enhance the inhibition of Her2(+) breast tumor growth and reduce the dose of injected siRNA. These results suggest that ScFvHer2 -decorated PEG-PLA-based nanoparticles show great potential for targeted RNA interference therapy of Her2(+) breast tumor.


Assuntos
Neoplasias da Mama/terapia , Nanopartículas/administração & dosagem , Polietilenoglicóis/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Anticorpos de Cadeia Única/administração & dosagem , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Feminino , Terapia Genética/métodos , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor ErbB-2/metabolismo , Quinase 1 Polo-Like
6.
J Control Release ; 166(2): 106-14, 2013 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-23266452

RESUMO

Due to its efficient and specific gene silencing ability, RNA interference has shown great potential in the treatment of liver diseases. However, achieving in vivo delivery of siRNA to critical liver cells remains the biggest obstacle for this technique to be a real clinic therapeutic modality. Here, we describe a promising liver targeting siRNA delivery system based on N-acetylgalactosamine functionalized mixed micellar nanoparticles (Gal-MNP), which can efficiently deliver siRNA to hepatocytes and silence the target gene expression after systemic administration. The Gal-MNP were assembled in aqueous solution from mixed N-acetylgalactosamine functionalized poly(ethylene glycol)-b-poly(ε-caprolactone) and cationic poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) (PCL-b-PPEEA); the properties of nanoparticles, including particle size, zeta potential and the density of poly(ethylene glycol) could be easily regulated. The hepatocyte-targeting effect of Gal-MNP was demonstrated by significant enriching of fluorescent siRNA in primary hepatocytes in vitro and in vivo. Successful down-regulation of liver-specific apolipoprotein B (apoB) expression was achieved in mouse liver, at both the transcriptional and protein level, following intravenous injection of Gal-MNP/siapoB to BALB/c mice. Systemic delivery of Gal-MNP/siRNA did not induce the innate immune response or positive hepatotoxicity. The results of this study suggested therapeutic potential for the Gal-MNP/siRNA system in liver disease.


Assuntos
Acetilgalactosamina/química , Sistemas de Liberação de Medicamentos/métodos , Fígado/efeitos dos fármacos , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , Células 3T3 , Animais , Apolipoproteínas B/genética , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos , Eletroquímica , Endocitose/efeitos dos fármacos , Etilenoglicóis , Inativação Gênica , Hepatócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Tamanho da Partícula , Poliésteres
7.
Biomaterials ; 34(26): 6284-98, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23714243

RESUMO

The interaction of nanocarriers with cells including their transcellular behavior is vital not only for a drug delivery system, but also for the safety of nanomaterials. In an attempt to clarify how the structures of polymers impact the transport mechanisms of their nanocarriers in epithelial cells, three amphiphilic polymers (PEEP-PCL, PEG-PCL and PEG-DSPE) with different hydrophilic or hydrophobic blocks were synthesized or chosen to form different micelle systems here. The endocytosis, exocytosis, intracellular colocalization, paracellular permeability and transcytosis of these micelle systems were compared using Förster resonance energy transfer analysis, real-time confocal images, colocalization assay, transepithelial electrical resistance study, and so on. All micelle systems were found intact during the studies with cells. The endocytosis and exocytosis studies with undifferentiated MDCK cells and the transcytosis study with differentiated MDCK monolayers all indicated the fact that PEG-DSPE micelles achieved the most and fastest transport, followed by PEG-PCL and PEEP-PCL in order. These might be because DSPE has higher hydrophobicity than PCL while PEG has lower hydrophilicity than PEEP. Different in hydrophilic or hydrophobic structures, all kinds of micelles demonstrated similar pathways during endocytosis and exocytosis, both caveolae- and clathrin-mediated but with difference in degree. The colocalization studies revealed different behaviors in intracellular trafficking among the three polymer micelles, suggesting the decisive role of hydrophilic shells on this process. Finally, all micelle systems did not impact the paracellular permeability of test cell monolayer. In conclusion, the hydrophilic and hydrophobic structures of test micelles could influence their transport ability, intracellular trafficking and the transport level under each pathway in MDCK cells.


Assuntos
Lactonas/metabolismo , Células Madin Darby de Rim Canino/citologia , Micelas , Fosfatidiletanolaminas/metabolismo , Poliésteres/metabolismo , Polietilenoglicóis/metabolismo , Animais , Cães , Endocitose , Exocitose , Interações Hidrofóbicas e Hidrofílicas , Lactonas/análise , Células Madin Darby de Rim Canino/metabolismo , Permeabilidade , Fosfatidiletanolaminas/análise , Poliésteres/análise , Polietilenoglicóis/análise , Tensoativos/análise , Tensoativos/metabolismo
8.
Adv Mater ; 24(46): 6175-80, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-22961974

RESUMO

Bacteria-Responsive Multifunctional Nanogel: We developed a bacteria-responsive multifunctional nanogel for targeted antibiotic delivery, in which bacterial enzymes are utilized to trigger antibiotic release by degrading the polyphosphoester core. The mannosylated nanogel preferentially delivers drugs to macrophages and leads to drug accumulation at bacterial infection sites through macrophage transport. This nanogel provides macrophage targeting and lesion site-activatable drug release properties, which enhances bacterial growth inhibition.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Portadores de Fármacos/química , Polietilenoglicóis/química , Polietilenoimina/química , Animais , Antibacterianos/química , Bactérias/enzimologia , Linhagem Celular , Embrião não Mamífero/metabolismo , Macrófagos/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Microscopia Confocal , Nanogéis , Fagocitose/efeitos dos fármacos , Fosfolipases/metabolismo , Vancomicina/química , Vancomicina/farmacologia , Peixe-Zebra/crescimento & desenvolvimento
9.
ACS Nano ; 6(6): 4955-65, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22646867

RESUMO

The clinical success of therapeutics of small interfering RNA (siRNA) is still hindered by its delivery systems. Cationic polymer or lipid-based vehicles as the major delivery systems of siRNA cannot sufficiently satisfy siRNA therapeutic applications. It is hypothesized that cationic lipid-polymer hybrid nanoparticles may take advantage of both polymeric and lipid-based nanoparticles for siRNA delivery, while diminishing the shortcomings of both. In this study, cationic lipid-polymer hybrid nanoparticles were prepared by a single-step nanoprecipitation of a cationic lipid (N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonyl aminoethyl) ammonium bromide, BHEM-Chol) and amphiphilic polymers for systemic delivery of siRNA. The formed hybrid nanoparticles comprised a hydrophobic polylactide core, a hydrophilic poly(ethylene glycol) shell, and a cationic lipid monolayer at the interface of the core and the shell. Such hybrid nanoparticles exhibited excellent stability in serum and showed significantly improved biocompatibility compared to that of pure BHEM-Chol particles. The hybrid nanoparticles were capable of delivering siRNA into BT474 cells and facilitated the escape of loaded siRNA from the endosome into the cytoplasm. The hybrid nanoparticles carrying polo-like kinase 1 (Plk1)-specific siRNA (siPlk1) remarkably and specifically downregulated expression of the oncogene Plk1 and induced cancer cell apoptosis both in vitro and in vivo and significantly suppressed tumor growth following systemic administration. We demonstrate that this system is stable, nontoxic, highly efficient, and easy to scale up, bringing the clinical application of siRNA therapy one important step closer to reality.


Assuntos
Lipídeos/química , Nanocápsulas/química , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Polímeros/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/química , Animais , Cátions , Linhagem Celular Tumoral , Terapia Genética/métodos , Camundongos , Nanocápsulas/administração & dosagem , Resultado do Tratamento
10.
J Control Release ; 128(1): 32-40, 2008 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-18395283

RESUMO

Cellular specific micellar systems from functional amphiphilic block copolymers are attractive for targeted intracellular drug delivery. In this study, we developed reactive micelles based on diblock copolymer of poly(ethyl ethylene phosphate) and poly(epsilon-caprolactone). The micelles were further surface conjugated with galactosamine to target asialoglycoprotein receptor (ASGP-R) of HepG2 cells. The size of micellar nanoparticles was about 70nm in diameter, and nanoparticles were negatively charged in aqueous solution. Through recognition between galactose ligands with ASGP-R of HepG2 cells, cell surface binding and internalization of galactosamine-conjugated micelles were significantly promoted, which were demonstrated by flow cytometric analyses using rhodamine 123 fluorescent dye. Paclitaxel-loaded micelles with galactose ligands exhibited comparable activity to free paclitaxel in inhibiting HepG2 cell proliferation, in contrast to the poor inhibition activity of micelles without galactose ligands particularly at lower paclitaxel doses. In addition, population of HepG2 cells arrested in G2/M phase was in positive response to paclitaxel dose when cells were incubated with paclitaxel-loaded micelles with galactosamine conjugation, which was against the performance of micelles without galactose ligand, owing to the ligand-receptor interaction. The surface functionalized micellar system is promising for specific anticancer drug transportation and intracellular drug release.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Receptor de Asialoglicoproteína/metabolismo , Portadores de Fármacos/farmacologia , Paclitaxel/farmacologia , Poliésteres/farmacologia , Polietilenos/farmacologia , Antineoplásicos Fitogênicos/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Galactosamina/química , Glucosamina/química , Humanos , Micelas , Paclitaxel/química , Poliésteres/química , Poliésteres/metabolismo , Polietilenos/química , Polietilenos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA