Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 303: 122382, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37977005

RESUMO

Anti-epidermal growth factor receptor (EGFR) antibody, cetuximab, therapy has significantly improved the clinical outcomes of patients with colorectal cancer, but the response to cetuximab can vary widely among individuals. We thus need strategies for predicting the response to this therapy. However, the current methods are unsatisfactory in their predictive power. Cetuximab can promote the internalization and degradation of EGFR, and its therapeutic efficacy is significantly correlated with the degree of EGFR degradation. Here, we present a new approach to predict the response to anti-EGFR therapy, cetuximab by evaluating the degree of EGFR internalization and degradation of colorectal cancer cells in vitro and in vivo. Our newly developed fluorogenic cetuximab-conjugated probe (Cetux-probe) was confirmed to undergo EGFR binding, internalization, and lysosomal degradation to yield fluorescence activation; it thus shares the action mechanism by which cetuximab exerts its anti-tumor effects. Cetux-probe-activated fluorescence could be used to gauge EGFR degradation and showed a strong linear correlation with the cytotoxicity of cetuximab in colorectal cancer cells and tumor-bearing mice. The predictive ability of Cetux-probe-activated fluorescence was much higher than those of EGFR expression or KRAS mutation status. The Cetux-probes may become useful tools for predicting the response to cetuximab therapy by assessing EGFR degradation.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Humanos , Camundongos , Animais , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Receptores ErbB/metabolismo , Neoplasias Colorretais/patologia , Mutação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
2.
ACS Nano ; 15(7): 11369-11384, 2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34191497

RESUMO

MicroRNAs (miRNAs), a recently discovered class of noncoding RNAs, play pivotal roles in regulating fundamental biological processes by suppressing the expression of target genes. Aberrant miRNA expression is commonly correlated with human diseases, including cancers. Anti-miRNA oligonucleotides provide an innovative therapeutic strategy for silencing disease-associated miRNAs. However, the clinical application of anti-miRNA therapy has been limited by formulation challenges and physiological delivery barriers. Here, to provide the safe and effective tumor-targeted delivery of anti-miRNAs, we designed carrier-free maleimide-functionalized anti-miRNAs (MI-Anti-miRNAs) that enable "piggybacking" onto albumin in vivo. These functionalized MI-Anti-miRNAs covalently bind to cysteine-34 of endogenous albumin within minutes. In addition to resulting in a markedly extended blood circulation lifetime, this strategy allows MI-Anti-miRNAs to "hitchhike" to the tumor site. Importantly, in situ-generated albumin-Anti-miRNAs are capable of intracellularly internalizing highly negatively charged anti-miRNA molecules and knocking down target miRNAs. In particular, MI-Anti-miRNAs that targeted miRNA-21, which is involved in tumor initiation, progression, invasion, and metastasis in several types of cancer, successfully repressed miRNA-21 activity, resulting in a superior antitumor activity in both solid and metastatic tumor models without causing systemic toxicity. This endogenous albumin-piggybacking approach using MI-Anti-miRNAs provides a simple and broadly applicable platform strategy for the systemic delivery of anti-miRNA therapeutics.


Assuntos
MicroRNAs , Neoplasias , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinogênese , Neoplasias/genética , Neoplasias/patologia , Oligonucleotídeos , Albuminas
3.
Biomaterials ; 226: 119543, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31634653

RESUMO

Inflammasome plays a critical role in diverse inflammatory disorders, including cancers and Alzheimer's disease. It is induced by various pathogenic insults and activates caspase-1, a hallmark executor of inflammasome. Here, we developed an activatable fluorescence probe for visualization of active caspase-1. This caspase-1 probe is biocompatible, efficiently delivered into cells and tissues, and specifically emits fluorescence upon caspase-1 activation as assessed in in vitro and in vivo models of inflammatory conditions. We demonstrated efficient in vivo imaging of caspase-1 activation in early stages of various inflammatory conditions of mice models, including endotoxin shock, inflammatory bowel disorder, transplanted cancer, and Alzheimer's disease. Notably, the caspase-1 probe enables detection of neuroinflammation in vivo two months earlier than cognitive impairments occur in Alzheimer's disease model. We detected significant fluorescence emitted from inflamed sites, as well as their draining lymph nodes, by macroscopic imaging analysis within 30 min after systemic injection of the probe. This novel synthetic probe could be applied for efficient and rapid detection of caspase-1 activity in a spatiotemporal way by non-invasive imaging.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença de Alzheimer/diagnóstico por imagem , Animais , Caspase 1/metabolismo , Caspases/metabolismo , Inflamassomos/metabolismo , Camundongos , Proteólise
4.
Biomaterials ; 226: 119550, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31645012

RESUMO

Unresolved inflammation is a hallmark of many deadly diseases including atherosclerosis, a silent pathological condition behind majority of cardiovascular diseases. Yet, anti-inflammatory drugs are not clinically used in the treatment of patients with atherosclerosis. The currently approved treatment regimen against atherosclerosis is mainly focused on lowering the cholesterol/lipid levels in blood and has little to do with controlling inflammation, the underlying cause. Recent preclinical and clinical data suggest that effective alleviation of inflammation in the atherosclerosis plaque could reduce the risk of cardiovascular disease. In this work, we have encapsulated interleukin-10 (IL10), a multipotent anti-inflammatory cytokine into cRGD conjugated pluronic based nano-carriers (NC) for targeted delivery to atherosclerotic plaques. The NC could encapsulate the therapeutic protein with a high loading efficiency in a mild condition and showed sustained release capabilities. The efficacy of cytokine encapsulated NC was analyzed in vitro using the lipopolysaccharide stimulated macrophage cells and in vivo using an established apolipoprotein E-knockout (ApoE-/-) C57BL/6 mouse model. Compared to free IL10, intravenous administration of NC encapsulated IL10 resulted in vastly improved pharmacokinetic profile and profoundly high accumulation of the cytokine in the atherosclerosis lesions. IL10 delivered by NC was bioactive and reduced the production of pro-inflammatory cytokine IL-1ß in the lesion and led to significant regression in the plaque size. These results signify the prospect of nanoparticle based cytokine delivery for preventing atherosclerotic through inflammation modulation in near future.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Anti-Inflamatórios/uso terapêutico , Aterosclerose/tratamento farmacológico , Citocinas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE
5.
J Mater Chem B ; 3(2): 198-206, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-32261940

RESUMO

Current theranostic approaches in cancer therapy demand delivery systems that can carry multiple drugs or imaging agents in a single nanoplatform with uniform biodistribution and improved target specificity. In this study, we have developed amphiphilized poly(ethyleneimine) nanoparticles (aPEI NPs) as a versatile multi-cargo delivery platform. The aPEI NPs were engineered to have the loading capacity for both hydrophobic molecules and negatively charged hydrophilic colloidal cargos through amphiphilic modification, i.e., octadecylation and subsequent PEGylation of poly(ethyleneimine). In the aqueous phase, the resulting aPEIs underwent amphiphilic self-assembly into spherical nanoparticles whose structure is constituted of the hydrophobic core with the positively charged surface and the hydrophilic neutral corona. The high degree of PEGylation resulted in the tiny colloidal size (<15 nm in diameter) and rendered the outmost surface coated with an antifouling corona which minimizes general shortcomings of poly(ethyleneimine)-based nanocarriers (e.g., cytotoxicity and liver filtration) while keeping its advantage (loading capability for negatively charged drugs). The unique nanostructure of aPEI NPs allowed for facile loading of hydrophobic model drugs (rubrene and IR780) in the core as well as negatively charged colloids (Pdots, proteins and DNA) on the inner surface via the hydrophobic and electrostatic interactions, respectively. Fluorescence imaging experiments demonstrated that the highly PEGylated aPEI-25 NPs showed prolonged blood circulation with minimal liver filtration and efficient delivery of the loaded cargos to the tumor. These combined merits, along with negligible toxicity profiles both in vitro and in vivo, validate the potential of aPEI-25 NPs as versatile nanocarriers for multi-cargo delivery.

6.
Biomaterials ; 34(37): 9475-85, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24050874

RESUMO

The safe and effective systemic delivery of siRNA is a prerequisite for the successful development of siRNA-based cancer therapeutics. For the enhanced delivery of siRNA, cationic lipids and polymers have been widely used as siRNA carriers to form electrolyte complexes with anionic siRNA. However, the considerable toxicity of strong cationic-charged molecules hampers their clinical use. In this study, we utilized human serum albumin (HSA), which is the most abundant of the plasma proteins, as a siRNA carrier for systemic tumor-targeted siRNA delivery. Both HSA and siRNA molecules were thiol-introduced to improve the binding affinity for each other. The resulting thiolated HSA (tHSA) and polymerized siRNA (psi) formed stable nanosized complexes (psi-tHSAs) by chemical crosslinking and self-crosslinking. After internalization, the psi-tHSAs showed target gene silencing activity in vitro comparable to conventional Lipofectamine™-siRNA complexes, without remarkable cytotoxicity. After intravenous injection in tumor-bearing mice, psi-tHSAs accumulated specifically at the tumor sites, leading to efficient gene silencing in the tumors in a sequential manner. The therapeutic VEGF siRNA was loaded into psi-tHSAs, which significantly inhibited tumor-related angiogenesis in PC-3 tumor xenografts and resulted in retarding the growth of PC-3 tumors. The results showed that self-crosslinked psi-tHSA nanocarriers might provide a promising approach for the systemic siRNA therapy of various human cancers.


Assuntos
Portadores de Fármacos/química , Neoplasias/terapia , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , Albumina Sérica/química , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Moleculares , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Polimerização , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Compostos de Sulfidrila/química , Fator A de Crescimento do Endotélio Vascular/genética
7.
Biomaterials ; 34(21): 5273-80, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23591396

RESUMO

One of the major hurdles of the nanoparticles as drug carriers is the unintended burst release of loaded drugs during blood circulation. To surmount this issue, we developed photo-crosslinked hyaluronic acid nanoparticles (c-HANPs) with improved stability for tumor-targeted drug delivery. They were readily prepared via UV-triggered chemical crosslinking with the acrylate groups in the polymer backbone. The size of c-HANPs was not much different from that of uncrosslinked HANPs. However, c-HANPs exhibited significantly high stability in a physiological buffer and released the loaded drug, paclitaxel (PTX), in a sustained manner. It is noteworthy that the drug release rate from c-HANPs remarkably increased in the presence of hyaluronidase, an enzyme abundant at the intracellular compartments of the tumor cells. It was found from in vitro cellular uptake tests that c-HANPs were rapidly taken up by the tumor cells via the receptor (CD44)-mediated endocytosis, which was not inhibited by photo-crosslinking. In non-invasive animal imaging results, they showed higher tumor-targeting ability than uncrosslinked HANPs because high stability of c-HANPs enabled their long circulation in the body. Owing to the sustained release of the drug and enhanced tumor-targeting ability, c-HANPs showed higher therapeutic efficacy compared to free PTX and uncrosslinked HANPs. These data implied the promising potential of c-HANP as tumor-targeting drug carriers and demonstrated the remarkable effect of the improved stability upon the biodistribution and therapeutic efficacy of drug-loaded nanoparticles.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Ácido Hialurônico/química , Luz , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Ácido Hialurônico/síntese química , Ácido Hialurônico/ultraestrutura , Camundongos , Camundongos Nus , Nanopartículas/ultraestrutura , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Tamanho da Partícula , Distribuição Tecidual/efeitos dos fármacos
8.
J Mater Chem B ; 1(28): 3437-3442, 2013 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32260933

RESUMO

Hydrogen bonding is a major intermolecular interaction for self-assembly occurring in nature. Here we report novel polymeric carbohydrates, i.e., poly(oxyethylene galactaramide)s (PEGAs), as biomimetic building blocks to construct hydrogen bond-mediated self-assembled nanoparticles that are useful for biomedical in vivo applications. PEGAs were conceptually designed as a biocompatible hybrid between polysaccharide and poly(ethylene glycol) (PEG) to attain multivalent hydrogen bonding as well as fully hydrophilic, non-ionic and antifouling characteristics. It was revealed that PEGAs are capable of homospecies hydrogen bonding in water and constructing multi-chain assembled nanoparticles whose structural integrity is highly stable with varying concentration, temperature and pH. Using near-infrared fluorescence imaging we demonstrate facile blood circulation and efficient tumor accumulation of the self-assembled PEGA nanoparticles that were intravenously injected into mice. These in vivo behaviors elucidate the combined merits of our design strategy, i.e., biocompatible chemical constitution capable of multivalent hydrogen bonding, antifouling properties, minimal cell interaction and mesoscopic colloidal self-assembly, as well as size-motivated tumor targeting.

9.
Biomaterials ; 31(34): 9057-64, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20813405

RESUMO

The clinical applications of tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL), an emerging therapeutic protein for cancer and rheumatoid arthritis (RA), are limited by its instability and short biological half-life. In this study, efficient therapeutic modalities for RA treatment were developed in the form of nano-sized complexes (nanocomplexes) based on hyaluronic acid (HA) and polyethylene glycol (PEG)-derivatized TRAIL (PEG-TRAIL) formed by N-terminal specific PEGylation. The nanocomplexes were prepared by simply mixing the positively charged PEG-TRAIL and negatively charged HA, and showed negligible loss of bioactivity compared with the PEG-TRAIL. The in vivo biodistribution and diffusion kinetics of Cy5.5-labeled PEG-TRAIL in mice were observed using a near-infrared optical imaging system after subcutaneous injection of three different formulations: PEG-TRAIL in phosphate-buffered saline (PBS, pH 7.4), nanocomplex in PBS, or nanocomplex in 1% HA solution. The results suggested that PEG-TRAIL is released slowly in vivo from the nanocomplex in 1% HA. Experiments in a collagen-induced arthritis mouse model demonstrated that the magnitudes of therapeutic effects, as judged by clinical scores and histology, were significantly enhanced by the sustained delivery of PEG-TRAIL, with the order of nanocomplex in 1% HA>nanocomplex in PBS>PEG-TRAIL in PBS. In addition, sustained delivery of PEG-TRAIL from the nanocomplex in 1% HA resulted in significant reduction of serum inflammatory cytokines and collagen-specific antibodies that are responsible for the pathogenesis of RA. These results imply that HA/PEG-TRAIL nanocomplex formulations are promising therapeutic modalities for the treatment of RA.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Ácido Hialurônico/uso terapêutico , Polietilenoglicóis/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Artrite Reumatoide/sangue , Proliferação de Células/efeitos dos fármacos , Citocinas/sangue , Difusão/efeitos dos fármacos , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Íons , Cinética , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/patologia , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Nanoestruturas/uso terapêutico , Tamanho da Partícula , Baço/citologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Distribuição Tecidual/efeitos dos fármacos
10.
Biomaterials ; 31(19): 5191-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20359742

RESUMO

Caged proteins have been utilized as a biological container in a wide range of applications from material science to biomedicine, and GALA peptide has been known to undergo coil-to-helix transition upon the increased acidity. In this study, GALA synthetic peptide is incorporated to cage protein by genetic modification. Our engineered caged scaffold retains intact at the physiological pH but dissociate completely at pH 6.0, and the dissociated subunits are re-assembled simply by neutralization to biological pH. This acid-induced dissociation has the potential as molecular switch in vivo as well as in vitro so that the acid-sensitive caged proteins are applicable to drug delivery system for acidic target sites such as tumor. Since our design depends on the conformational transition of GALA peptide, not on removal of characteristic interface observed only in viral capsid-like protein, non-viral caged proteins can also be engineered to have molecular switching function. Therefore, this design for acid-sensitive scaffold would broaden the width of applications in nanotechnology including biomimetic material synthesis and biomedicine.


Assuntos
Ferritinas/química , Complexos Multiproteicos/química , Peptídeos/química , Concentração de Íons de Hidrogênio
11.
Biomaterials ; 29(12): 1920-30, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18289669

RESUMO

Antiangiogenic peptide drugs have received much attention in the fields of tumor therapy and tumor imaging because they show promise in the targeting of integrins such as alpha(v)beta(3) on angiogenic endothelial cells. However, systemic antiangiogenic peptide drugs have short half-lives in vivo, resulting in fast serum clearance via the kidney, and thus the therapeutic effects of such drugs remain modest. In this study, we prepared self-assembled glycol chitosan nanoparticles and explored whether this construct might function as a prolonged and sustained drug delivery system for RGD peptide, used as an antiangiogenic model drug in cancer therapy. Glycol chitosan hydrophobically modified with 5beta-cholanic acid (HGC) formed nanoparticles with a diameter of 230 nm, and RGD peptide was easily encapsulated into HGC nanoparticles (yielding RGD-HGC nanoparticles) with a high loading efficiency (>85%). In vitro work demonstrated that RGD-HGC showed prolonged and sustained release of RGD, lasting for 1 week. RGD-HGC also inhibited HUVEC adhesion to a beta ig-h3 protein-coated surface, indicating an antiangiogenic effect of the RGD peptide in the HGC nanoparticles. In an in vivo study, the antiangiogenic peptide drug formulation of RGD-HGC markedly inhibited bFGF-induced angiogenesis and decreased hemoglobin content in Matrigel plugs. Intratumoral administration of RGD-HGC significantly decreased tumor growth and microvessel density compared to native RGD peptide injected either intravenously or intratumorally, because the RGD-HGC formulation strongly enhanced the antiangiogenic and antitumoral efficacy of RGD peptide by affording prolonged and sustained RGD peptide delivery locally and regionally in solid tumors.


Assuntos
Quitosana/química , Preparações de Ação Retardada/química , Portadores de Fármacos/química , Melanoma/tratamento farmacológico , Nanopartículas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/administração & dosagem , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Animais , Preparações de Ação Retardada/administração & dosagem , Portadores de Fármacos/administração & dosagem , Glicóis/administração & dosagem , Glicóis/química , Masculino , Teste de Materiais , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/ultraestrutura , Neovascularização Patológica/patologia , Oligopeptídeos/química , Resultado do Tratamento
12.
Biomaterials ; 28(16): 2667-76, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17335894

RESUMO

The inhibitory efficacies of new bile acid acylated-heparin derivative (heparin-DOCA) were evaluated on experimental lung metastasis. We evaluated the effect of heparin-DOCA on intercellular interactions including those between B16F10 and thrombin-activated platelets and TNF-alpha-activated HUVECs, and between B16F10 and immobilized mouse P-selectin. In addition, the inhibitory effects of heparin-DOCA on adhesion and invasion of B16F10 to Matrigel were studied. In an animal mouse study, the blood clot formation and the retention of red fluorescence protein (RFP)-B16F10 in lungs were assessed after heparin-DOCA and RFP-B16F10 intravenous administration. Furthermore, we investigated the anti-metastatic effect of heparin-DOCA against lung metastasis induced by B16F10 and SCC7. Heparin-DOCA inhibited intercellular interactions between B16F10 and activated platelets or activated HUVECs by blocking P- and E-selectin-mediated interactions. Moreover, it reduced adhesion and invasion of B16F10 to ECM, thereby affecting the reduction of early retention of B16F10 in the lung. Heparin-DOCA attenuated lung colony formation on the surfaces and in interior of the lung, and attenuated metastasis by B16F10 and SCC7. These results suggest that heparin-DOCA may have potentials as therapeutic agent that prevents tumor metastasis and progression.


Assuntos
Antineoplásicos , Ácidos e Sais Biliares , Heparina , Neoplasias Pulmonares , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Ácidos e Sais Biliares/química , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/uso terapêutico , Adesão Celular , Linhagem Celular , Selectina E/metabolismo , Matriz Extracelular/metabolismo , Heparina/análogos & derivados , Heparina/química , Heparina/metabolismo , Heparina/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Teste de Materiais , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias Experimentais , Selectina-P/metabolismo
13.
Biomaterials ; 27(30): 5178-85, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16797693

RESUMO

A pH- and thermo-sensitive block copolymer was synthesized by adding pH-sensitive sulfamethazine oligomers (SMOs) to either end of a thermo-sensitive poly(epsilon-caprolactone-co-lactide)-poly(ethylene glycol)-poly(epsilon-caprolactone-co-lactide) (PCLA-PEG-PCLA) block copolymer. The resulting pH- and thermo-sensitive SMO-PCLA-PEG-PCLA-SMO block copolymer solution did not form a gel at high pH (pH 8.0) or at increased temperatures (ca. 70 degrees C), but did form a stable gel under physiological conditions (pH 7.4 and 37 degrees C). The degradation rate of the pH- and thermo-sensitive block copolymer decreased substantially compared with the control block copolymer of PCLA-PEG-PCLA, due to the buffering effect of the SMO-PCLA-PEG-PCLA-SMO sulfonamide groups on the acidic monomer-induced rapid degradation of PCLA-PEG-PCLA. This suitable sol-gel transition and sustained biodegradability of the pH- and thermo-sensitive SMO-PCLA-PEG-PCLA-SMO block copolymer resolves two of the major drawbacks associated with thermo-sensitive block copolymers, namely premature gelation and rapid degradation. Interestingly, SMO-PCLA-PEG-PCLA-SMO showed no evidence of cytotoxicity in vitro. However, subcutaneous injection of the pH- and thermo-sensitive block copolymer solution (20wt% in PBS at pH 8.0) into Sprague-Dawley (SD) rats resulted in rapid, stable gel formation, with the injected hydrogel being completely degraded in vivo in just 6 weeks. The injected hydrogel in vivo presented a typical acute inflammation within 2 weeks, although chronic inflammation was not observed during the first 6-week period. As such, the pH- and thermo-sensitive hydrogel of the SMO-PCLA-PEG-PCLA-SMO block copolymer is a suitable candidate for use in drug delivery systems and cell therapy.


Assuntos
Materiais Biocompatíveis/química , Materiais Biocompatíveis/toxicidade , Caproatos/química , Caproatos/toxicidade , Hidrogéis/química , Hidrogéis/toxicidade , Lactonas/química , Lactonas/toxicidade , Poliésteres/química , Poliésteres/toxicidade , Animais , Materiais Biocompatíveis/metabolismo , Biodegradação Ambiental , Caproatos/metabolismo , Temperatura Alta , Humanos , Hidrogéis/metabolismo , Concentração de Íons de Hidrogênio , Lactonas/metabolismo , Masculino , Poliésteres/metabolismo , Ratos , Ratos Sprague-Dawley , Soluções/química , Temperatura
14.
J Control Release ; 89(3): 437-46, 2003 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12737846

RESUMO

Polymeric micelles based on amphiphilic block copolymers of poly(2-ethyl-2-oxazoline) (PEtOz) and poly(epsilon -caprolactone) (PCL) were prepared in an aqueous phase. The loading of paclitaxel into PEtOz-PCL micelles was confirmed by 1H-NMR spectra. Paclitaxel was efficiently loaded into PEtOz-PCL micelles using dialysis method, and the loading content of paclitaxel in micelles was in the range 0.5-7.6 wt.% depending on the block composition of block copolymers, organic solvent used in the dialysis, and feed weight ratio of paclitaxel to block copolymer. The higher the content of hydrophobic block in the block copolymers, the higher the loading efficiency of micelles for paclitaxel. When acetonitrile was used as solvent, a higher drug loading efficiency was obtained than with THF. The loading efficiency decreased with increasing feed weight ratio of paclitaxel to block copolymer from 0.1:1 to 0.2:1. The hydrodynamic diameters of paclitaxel-loaded micelles were in the range 18.3-23.4 nm with narrow size distribution. The hemolysis test of PEtOz-PCL performed in vitro indicated that the toxicity of PEtOz-PCLs to lipid membrane was not significant compared with Tween 80, and was comparable to that observed with Cremophore EL. The proliferation inhibition activity of paclitaxel-loaded micelles for KB human epidermoid carcinoma cells was also evaluated in vitro. Paclitaxel-entrapped polymeric micelles exhibited comparable activity to that observed with Cremophore EL-based paclitaxel formulations in inhibiting the growth of KB cells.


Assuntos
Caproatos/química , Caproatos/farmacocinética , Lactonas/química , Lactonas/farmacocinética , Micelas , Oxazóis/química , Oxazóis/farmacocinética , Paclitaxel/química , Paclitaxel/farmacocinética , Polímeros/química , Polímeros/farmacocinética , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/farmacocinética , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...