Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Pharm ; 16(9): 3957-3967, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31381352

RESUMEN

Curcumin exhibits potent anticancer activity via various mechanisms, but its in vivo efficacy has been hampered by poor solubility. Nanotechnology has been employed to deliver curcumin, but most of the reported systems suffered from low drug loading capacity and poor stability. Here, we report the development and optimization of a liposomal formulation for curcumin (Lipo-Cur) using an automated microfluidic technology. Lipo-Cur exhibited a mean diameter of 120 nm with a low polydispersity index (<0.2) and superior loading capacity (17 wt %) compared to other reported liposomal systems. Lipo-Cur increased the water solubility of curcumin by 700-fold, leading to 8-20-fold increased systemic exposure compared to the standard curcumin suspension formulation. When coadministered with cisplatin to tumor-bearing mice, Lipo-Cur augmented the antitumor efficacy of cisplatin in multiple mouse tumor models and decreased the nephrotoxicity. This is the first report demonstrating the dual effects of curcumin enabled by a nanoformulation in enhancing the efficacy and reducing the toxicity of a chemo-drug in animal models under a single and low dose administration.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Curcumina/química , Dimiristoilfosfatidilcolina/química , Sistemas de Liberación de Medicamentos/métodos , Liposomas/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Cisplatino/administración & dosificación , Curcumina/administración & dosificación , Curcumina/farmacocinética , Dimiristoilfosfatidilcolina/administración & dosificación , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Liberación de Fármacos , Quimioterapia Combinada , Femenino , Liposomas/administración & dosificación , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/química , Nanotecnología/métodos , Solubilidad , Distribución Tisular
2.
Biol Pharm Bull ; 41(7): 1107-1111, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29962407

RESUMEN

Liposomes have been used as targeting carriers for drug delivery systems (DDSs), and the carriers are able to be modified with targeting ligands, such as antibodies and peptides. To evaluate the targetability of DDS carriers modified with a targeting ligand, culture cells expressing the targeting molecules as well as small animals are used. Furthermore, in vitro and in vivo screening analyses must be repeatedly performed. Therefore, it is important to establish an easy and high-precision screening system for targeting carriers. With this aim, we focused that whether this ex vivo system could easily support assessment of interaction between targeting ligand and its receptor under physiological environment and further screen the DDS carrier-modified with targeting moiety. We examined targeting ability via in vitro, ex vivo, and in vivo analyses using integrin αvß3-targeting C16Y-L. For the in vitro analysis, the cellular uptake of C16Y-L was higher than that of control liposomes in colon26 cells. For the ex vivo analysis, we performed an immunohistochemical analysis using colon26 tumor sections. C16Y-L was specifically attached to the tumor sections, as found in the in vitro analysis. Moreover, to evaluate the ex vivo-in vivo correlation, we examined the intratumoral localization of C16Y-L. This result showed that C16Y-L was accumulated not only in the tumor tissue but also in the tumor vasculature after the intravenous injection of C16Y-L, suggesting that the ex vivo peptide-modified liposomal analysis was correlated with the in vivo analysis. Thus, the ex vivo peptide-modified liposomal analysis may be an easy and rapid screening system with high-precision and for consideration in in vivo conditions.


Asunto(s)
Química Farmacéutica/métodos , Sistemas de Liberación de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Nanopartículas/química , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/metabolismo , Ligandos , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Oligopéptidos/química , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Liver Int ; 36(8): 1151-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26353075

RESUMEN

BACKGROUND & AIMS: Clinical studies suggest that splenectomy improves liver function in cirrhotic patients, but the influence of splenectomy on stem cell transplantation is poorly understood. This study investigated the effect of splenectomy on stem cell infusion and elucidated its mechanism. METHODS: Rat adipose tissue-derived mesenchymal stem cells were infused into cirrhosis rats with or without splenectomy, followed by the assessment of the in vivo distribution of stem cells and pathological changes. Stromal cell-derived factor-1 and hepatocyte growth factor expression were also investigated in splenectomized cirrhosis patients and rats. RESULTS: Splenectomy, prior to cell infusion, improved liver function and suppressed fibrosis progression more efficiently than cell infusion alone in the experimental cirrhosis model. Stromal cell-derived factor-1 and hepatocyte growth factor levels after splenectomy were increased in patients and rats. These upregulated cytokines significantly facilitated stem cell motility, migration and proliferation in vitro. C-X-C chemokine receptor type 4 neutralization weakened the promotion of cell migration by these cytokines. The infused cells integrated into liver fibrosis septa and participated in regeneration more efficiently in splenectomized rats. Direct coculture with stem cells led to inhibition of hepatic stellate cell proliferation. In addition, hepatocyte growth factor induced hepatic stellate cell apoptosis via the c-jun N-terminal kinase-p53 pathway. CONCLUSIONS: Splenectomy prior to cell infusion enhanced the therapeutic effect of stem cells on cirrhosis, which involved upregulation of stromal cell-derived factor-1 and hepatocyte growth factor after splenectomy.


Asunto(s)
Quimiocina CXCL12/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Cirrosis Hepática Experimental/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Esplenectomía , Tejido Adiposo/patología , Animales , Proliferación Celular , Células Cultivadas , Hepatocitos/patología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/patología , Cirrosis Hepática Experimental/cirugía , Regeneración Hepática , Masculino , Ratas , Ratas Endogámicas F344
4.
Mol Pharm ; 12(5): 1422-30, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25811429

RESUMEN

We describe the development of neuropilin 1-binding peptide (iRGD)-nanocages that specifically target human pancreatic cancer cells in which an iRGD is joined to the surface of naturally occurring heat shock protein (HSP) cages. Using a genetic engineering approach, the iRGD domain was joined to the C-terminal region of the HSP cage using flexible linker moieties. The characteristics of the interdomain linkages between the nanocage and the iRGD domain play an important role in the specificity and affinity of the iRGD-nanocages for their target cells. An engineered L30-iRGD-nanocage with 30 amino acid linkers, (GGS)10, showed greater binding affinity for pancreatic cancer cells relative to that of other linkers. Furthermore, a moderately hydrophobic anticancer drug, OSU03012, was successfully incorporated into the L30-iRGD-nanocage by heating the mixture. The OSU03012-loaded L30-iRGD-nanocage induced cell death of pancreatic cancer cells by activating the caspase cascade more effectively than the same concentrations of free OSU03012. The iRGD-nanocages show great potential as a novel nanocarrier for pancreatic cancer-targeted drug delivery.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Neuropilina-1/química , Neoplasias Pancreáticas/metabolismo , Línea Celular Tumoral , Humanos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Protein Expr Purif ; 110: 52-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25497224

RESUMEN

Lipid modification of proteins plays key roles in cellular signaling pathways. We describe the development of myristoylated preS1-nanocages (myr-preS1-nanocages) that specifically target human hepatocyte-like HepaRG cells in which a specific receptor-binding peptide (preS1) is joined to the surface of naturally occurring ferritin cages. Using a genetic engineering approach, the preS1 peptide was joined to the N-terminal regions of the ferritin cage via flexible linker moieties. Myristoylation of the preS1 peptide was achieved by co-expression with yeast N-myristoyltransferase-1 in the presence of myristic acid in Escherichia coli cells. The myristoylated preS1-nanocages exhibited significantly greater specificity for human hepatocyte-like HepaRG cells than the unmyristoylated preS1-nanocages. These results suggest that the lipid-modified nanocages have great potential for effective targeted delivery to specific cells.


Asunto(s)
Ferritinas/genética , Antígenos de Superficie de la Hepatitis B/genética , Hepatocitos/química , Plásmidos/química , Precursores de Proteínas/genética , Proteínas del Envoltorio Viral/genética , Aciltransferasas/química , Aciltransferasas/genética , Línea Celular , Clonación Molecular , Sistemas de Liberación de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Ferritinas/química , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Expresión Génica , Ingeniería Genética , Antígenos de Superficie de la Hepatitis B/química , Antígenos de Superficie de la Hepatitis B/aislamiento & purificación , Hepatocitos/patología , Hepatocitos/virología , Humanos , Terapia Molecular Dirigida , Ácido Mirístico/química , Ácido Mirístico/metabolismo , Plásmidos/metabolismo , Unión Proteica , Precursores de Proteínas/química , Precursores de Proteínas/aislamiento & purificación , Estructura Terciaria de Proteína , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/aislamiento & purificación
6.
J Gastroenterol Hepatol ; 30(6): 1065-74, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25639333

RESUMEN

BACKGROUND AND AIM: Recent studies show that adipose tissue-derived mesenchymal stem cells have potential clinical applications. However, the mechanism has not been fully elucidated yet. Here, we investigated the effect of basic fibroblast growth factor-treated adipose tissue-derived mesenchymal stem cells infusion on a liver fibrosis rat model and elucidated the underlying mechanism. METHODS: Adipose tissue-derived mesenchymal stem cells were infused into carbon tetrachloride-induced hepatic fibrosis rats through caudal vein. Liver functions and pathological changes were assessed. A co-culture model was used to clarify the potential mechanism. RESULTS: Basic fibroblast growth factor treatment markedly improved the proliferation, differentiation, and hepatocyte growth factor expression ability of adipose tissue-derived mesenchymal stem cells. Although adipose tissue-derived mesenchymal stem cells infusion alone slightly ameliorated liver functions and suppressed fibrosis progression, basic fibroblast growth factor-treatment significantly enhanced the therapeutic effect in association with elevated hepatocyte growth factor expression. Moreover, double immunofluorescence staining confirmed that the infused cells located in fibrosis area. Furthermore, co-culture with adipose tissue-derived mesenchymal stem cell led to induction of hepatic stellate cell apoptosis and enhanced hepatocyte proliferation. However, these effects were significantly weakened by knockdown of hepatocyte growth factor. Mechanism investigation revealed that co-culture with adipose tissue-derived mesenchymal stem cells activated c-jun N-terminal kinase-p53 signaling in hepatic stellate cell and promoted apoptosis. CONCLUSIONS: Basic fibroblast growth factor treatment enhanced the therapeutic effect of adipose tissue-derived mesenchymal stem cells, and secretion of hepatocyte growth factor from adipose tissue-derived mesenchymal stem cells plays a critical role in amelioration of liver injury and regression of fibrosis.


Asunto(s)
Tejido Adiposo/citología , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Factor de Crecimiento de Hepatocito/fisiología , Cirrosis Hepática/etiología , Cirrosis Hepática/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas , Comunicación Paracrina/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Factor de Crecimiento de Hepatocito/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas Endogámicas F344
7.
Mol Pharm ; 11(3): 1053-61, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24433046

RESUMEN

Duchenne muscular dystrophy (DMD) is a genetic disorder that is caused by mutations in the DMD gene that lead to an absence of functional protein. The mdx dystrophic mouse contains a nonsense mutation in exon 23 of the dystrophin gene; a phosphorodiamidate morpholino oligomer (PMO) designed to skip this mutated exon in the mRNA induces dystrophin expression. However, an efficient PMO delivery method is needed to improve treatment strategies for DMD. We previously developed polyethylene glycol (PEG)-modified liposomes (Bubble liposomes) that entrap ultrasound contrast gas and demonstrated that the combination of Bubble liposomes with ultrasound exposure is an effective gene delivery tool in vitro and in vivo. In this study, to evaluate the ability of Bubble liposomes as a PMO delivery tool, we tested the potency of the Bubble liposomes combined with ultrasound exposure to boost the delivery of PMO and increase the skipping of the mutated exon in the mdx mouse. The results indicated that the combination of Bubble liposomes and ultrasound exposure increased the uptake of the PMO targeting a nonsense mutation in exon 23 of the dystrophin gene and consequently increased the PMO-mediated exon-skipping efficiency compared with PMO injection alone, leading to significantly enhanced dystrophin expression. This increased efficiency indicated the potential of the combination of Bubble liposomes with ultrasound exposure to enhance PMO delivery for treating DMD. Thus, this ultrasound-mediated Bubble liposome technique may provide an effective, noninvasive, nonviral method for PMO therapy for DMD muscle as well as for other muscular dystrophies.


Asunto(s)
Distrofina/antagonistas & inhibidores , Técnicas de Transferencia de Gen , Morfolinos/administración & dosificación , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , Oligonucleótidos Antisentido/administración & dosificación , Ultrasonido , Animales , Apoptosis , Western Blotting , Proliferación Celular , Células Cultivadas , Distrofina/fisiología , Terapia Genética , Técnicas para Inmunoenzimas , Liposomas , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Morfolinos/farmacología , Músculo Esquelético/patología , Músculo Esquelético/efectos de la radiación , Distrofia Muscular de Duchenne/genética , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Biol Pharm Bull ; 37(1): 174-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24389493

RESUMEN

Ultrasound (US) is used in the clinical setting not only for diagnosis but also for therapy. As a therapeutic US technique, high-intensity focused ultrasound (HIFU) can be applied to treat cancer in a clinical setting. Microbubbles increased temperature and improved the low therapeutic efficiency under HIFU; however, microbubbles have room for improvement in size, stability, and targeting ability. To solve these issues, we reported that "Bubble liposomes" (BLs) containing the US imaging gas (perfluoropropane gas) liposomes were suitable for ultrasound imaging and gene delivery. In this study, we examined whether BLs and HIFU could enhance the ablation area of the tumor and the antitumor effect. First, we histologically analyzed the tumor after BLs and HIFU. The ablation area of the treatment of BLs and HIFU was broader than that of HIFU alone. Next, we monitored the temperature of the tumor, and examined the antitumor effect. The temperature increase with BLs and HIFU treatment was faster and higher than that with HIFU alone. Moreover, treatment with BLs and HIFU enhanced the antitumor effect, which was better than with HIFU alone. Thus, the combination of BLs and HIFU could be efficacious for cancer therapy.


Asunto(s)
Técnicas de Ablación/métodos , Técnicas de Transferencia de Gen , Terapia Genética , Calor , Microburbujas , Neoplasias/diagnóstico por imagen , Ultrasonido/métodos , Animales , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Ultrasonografía
9.
Int J Mol Sci ; 16(1): 148-58, 2014 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-25547485

RESUMEN

Matrix metalloproteinase 2 (MMP-2) in metastatic cancer tissue, which is associated with a poor prognosis, is a potential target for tumor imaging in vivo. Here, we describe a metastatic cancer cell-targeted protein nanocage. An MMP-2-binding peptide, termed CTT peptide (CTTHWGFTLC), was conjugated to the surface of a naturally occurring heat shock protein nanocage by genetic modification. The engineered protein nanocages showed a binding affinity for MMP-2 and selective uptake in cancer cells that highly expressed MMP-2 in vitro. In near-infrared fluorescence imaging, the nanocages showed specific and significant accumulation in tumor tissue after intravenous injection in vivo. These protein nanocages conjugated with CTT peptide could be potentially applied to a noninvasive near-infrared fluorescence detection method for imaging gelatinase activity in metastatic tumors in vivo.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias/metabolismo , Péptidos Cíclicos/farmacocinética , Animales , Células HT29 , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Nanoconjugados , Metástasis de la Neoplasia , Imagen Óptica , Péptidos Cíclicos/administración & dosificación , Unión Proteica , Distribución Tisular
10.
Biopolymers ; 100(4): 402-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23532952

RESUMEN

Targeted gene delivery to neovascular vessels in tumors is considered a promising strategy for cancer therapy. We previously reported that "Bubble liposomes" (BLs), which are ultrasound (US) imaging gas-encapsulating liposomes, were suitable for US imaging and gene delivery. When BLs are exposed to US, the bubble is destroyed, creating a jet stream by cavitation, and resulting in the instantaneous ejection of extracellular plasmid DNA (pDNA) or other nucleic acids into the cytosol. We developed AG73 peptide-modified Bubble liposomes (AG73-BL) as a targeted US contrast agent, which was designed to attach to neovascular tumor vessels and to allow specific US detection of angiogenesis (Negishi et al., Biomaterials 2013, 34, 501-507). In this study, to evaluate the effectiveness of AG73-BL as a gene delivery tool for neovascular vessels, we examined the gene transfection efficiency of AG73-BL with US exposure in primary human endothelial cells (HUVEC). The transfection efficiency was significantly enhanced if the AG73-BL attached to the HUVEC was exposed to US compared to the BL-modified with no peptide or scrambled peptide. In addition, the cell viability was greater than 80% after transfection with AG73-BL. These results suggested that after the destruction of the AG73-BL with US exposure, a cavitation could be effectively induced by the US exposure against AG73-BL binding to the cell surface of the HUVEC, and the subsequent gene delivery into cells could be enhanced. Thus, AG73-BL may be useful for gene delivery as well as for US imaging of neovascular vessels.


Asunto(s)
Técnicas de Transferencia de Gen , Liposomas , Terapia Genética , Humanos , Plásmidos , Transfección , Ultrasonido
11.
Mol Pharm ; 10(2): 774-9, 2013 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-23210449

RESUMEN

Encapsulating anticancer drugs in liposomes improves their therapeutic window by enhancing antitumor efficacy and reducing side effects. To devise more effective liposomal formulations for antitumor therapy, many research groups have tried to develop tumor-targeting liposomes with enhanced drug release. Previously, we developed doxorubicin (Dox)-encapsulated AG73 peptide-modified liposomes (AG73-Dox), which targeted cancer and endothelial cells, and ultrasound (US) imaging gas-entrapping liposomes, called "Bubble liposomes" (BLs). In this study, to enhance the antitumor effect of AG73-Dox, we combined AG73-Dox with BLs and US. First, to determine whether the addition of BLs and application of US could enhance the cytotoxicity of AG73-Dox, we evaluated the cytotoxicity of the combination of AG73-Dox with BLs and US. BLs and US enhanced cytotoxicity of AG73-Dox more than they enhanced nontargeted Dox-encapsulated liposomes. Next, we examined the intracellular behavior of Dox after treatment with BLs and US. The combination of AG73-Dox with BLs and US did not enhance cellular uptake of Dox, but it did promote drug release in the cytoplasm. To further elucidate the release of Dox in the cytoplasm, we blocked cellular uptake via endosomes at a low temperature. As a result, BLs and US did not have an enhanced drug-release effect until AG73-Dox was taken up into cells. Thus, the combination of AG73-Dox with BLs and US may be useful for cancer therapy as a dual-function drug delivery system with targeted and controlled release.


Asunto(s)
Antineoplásicos/química , Liposomas/química , Ultrasonido , Línea Celular , Citometría de Flujo , Humanos , Microscopía Confocal , Modelos Biológicos
12.
J Immunol Methods ; 521: 113554, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37661049

RESUMEN

Antibodies are essential components of the immune system with a wide range of molecular targets. They have been recognized as modalities for treating several diseases and more than 130 approved antibody-based therapeutics are available for clinical use. However, limitations remain associated with its efficacy, tissue permeability, and safety, especially in cancer treatment. Nanoparticles, particularly those responsive to external stimuli, have shown promise in improving the efficacy of antibody-based therapeutics and tissue-selective delivery. In this study, we developed a reliable and accurate method for quantifying the amount of antibody loaded onto lipid nanoparticles modified with Herceptin® (Trastuzumab), an antibody-based therapeutic used to treat HER2-positive cancers, using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) followed by silver staining. This method proved to be a suitable alternative to commonly used protein quantification techniques, which are limited by lipid interference present in the samples. Furthermore, the amount of Herceptin modified on the liposomes, measured by this method, was confirmed by Herceptin's antibody-dependent cell-mediated cytotoxicity activity. Our results demonstrate the potential of this method as a critical tool for developing tissue-selective antibody delivery systems, leading to improved efficacy and reduced side effects of antibody-based therapeutics.


Asunto(s)
Liposomas , Nanopartículas , Trastuzumab , Anticuerpos
13.
Pharmaceutics ; 15(6)2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37376113

RESUMEN

Strategies for gene and nucleic acid delivery to skeletal muscles have been extensively explored to treat Duchenne muscular dystrophy (DMD) and other neuromuscular diseases. Of these, effective intravascular delivery of naked plasmid DNA (pDNA) and nucleic acids into muscles is an attractive approach, given the high capillary density in close contact with myofibers. We developed lipid-based nanobubbles (NBs) using polyethylene-glycol-modified liposomes and an echo-contrast gas and found that these NBs could improve tissue permeability by ultrasound (US)-induced cavitation. Herein, we delivered naked pDNA or antisense phosphorodiamidate morpholino oligomers (PMOs) into the regional hindlimb muscle via limb perfusion using NBs and US exposure. pDNA encoding the luciferase gene was injected with NBs via limb perfusion into normal mice with application of US. High luciferase activity was achieved in a wide area of the limb muscle. DMD model mice were administered PMOs, designed to skip the mutated exon 23 of the dystrophin gene, with NBs via intravenous limb perfusion, followed by US exposure. The number of dystrophin-positive fibers increased in the muscles of mdx mice. Combining NBs and US exposure, which can be widely delivered to the hind limb muscles via the limb vein, could be an effective therapeutic approach for DMD and other neuromuscular disorders.

14.
J Ultrasound Med ; 31(12): 1909-16, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23197543

RESUMEN

OBJECTIVES: The purpose of this study was to quantitatively assess the contrast kinetics of vascular endothelial growth factor receptor 2 (VEGFR2)-targeted microbubbles (BR55; Bracco Suisse, Geneva, Switzerland) compared to clinically used microbubbles (SonoVue; Bracco SpA, Milan, Italy) in both normal liver and human hepatocellular carcinoma xenograft tumors in mice. METHODS: Microbubbles were injected intravenously into healthy mice (n = 5) and mice bearing hepatocellular carcinoma xenograft tumors (n = 10). Cine loops of contrast enhancement in normal liver and the tumors were acquired for 10 minutes. Quantitative perfusion parameters were derived by fitting time-intensity curves using a dedicated mathematical model combining a bolus function and a ramp function. Immunohistochemical examinations were also performed for normal liver and tumor specimens to determine the level of VEGFR2 expression. RESULTS: The peak contrast enhancement observed in normal liver with BR55 was comparable to that with SonoVue, whereas a significant difference was observed in latephase enhancement at 10 minutes (ramp slope: P < .01). In the tumor model, SonoVue was rapidly cleared from the circulation, without any noticeable binding in the tumor, whereas BR55 showed a gradual decline, resulting in a longer wash-out period (mean transit time: P < .01). Immunohistochemical examinations showed that intratumoral vascular endothelial cells had sparse and weak VEGFR2 expression, whereas the sinusoidal capillaries in normal liver had much more diffuse and much stronger expression. CONCLUSIONS: Our results suggest that BR55 accurately reflects the VEGFR2 status in human hepatocellular carcinoma xenograft tumors. We showed that quantification applied to molecular ultrasound imaging may provide an objective method for measuring the degree of microbubble binding.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Neoplasias Hepáticas/diagnóstico por imagen , Hígado/diagnóstico por imagen , Microburbujas , Técnicas de Diagnóstico Molecular/métodos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias/diagnóstico por imagen , Ultrasonografía
15.
Pharmaceutics ; 15(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36678759

RESUMEN

A key challenge in treating solid tumors is that the tumor microenvironment often inhibits the penetration of therapeutic antibodies into the tumor, leading to reduced therapeutic efficiency. It has been reported that the combination of ultrasound-responsive micro/nanobubble and therapeutic ultrasound (TUS) enhances the tissue permeability and increases the efficiency of delivery of macromolecular drugs to target tissues. In this study, to facilitate efficient therapeutic antibody delivery to tumors using this combination system, we developed therapeutic antibody-modified nanobubble (NBs) using an Fc-binding polypeptide that can quickly load antibodies to nanocarriers; since the polypeptide was derived from Protein G. TUS exposure to this Herceptin®-modified NBs (Her-NBs) was followed by evaluation of the antibody's own ADCC activity, resulting the retained activity. Moreover, the utility of combining therapeutic antibody-modified NBs and TUS exposure as an antibody delivery system for cancer therapy was assessed in vivo. The Her-NBs + TUS group had a higher inhibitory effect than the Herceptin and Her-NBs groups. Overall, these results suggest that the combination of therapeutic antibody-modified NBs and TUS exposure can enable efficient antibody drug delivery to tumors, while retaining the original antibody activity. Hence, this system has the potential to maximize the therapeutic effects in antibody therapy for solid cancers.

16.
J Control Release ; 329: 988-996, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33091529

RESUMEN

Therapeutic strategies based on antisense oligonucleotides and therapeutic genes are being extensively investigated for the treatment of hereditary muscle diseases and hold great promise. However, the cellular uptake of these polyanions to the muscle cells is inefficient. Therefore, it is necessary to develop more effective methods of gene delivery into the muscle tissue. The A2G80 peptide (VQLRNGFPYFSY) from the laminin α2 chain has high affinity for α-dystroglycan (α-DG) which is expressed on the membrane of muscle cells. In this study, we designed a peptide-modified A2G80 with oligoarginine and oligohistidine (A2G80-R9-H8), and prepared peptide/plasmid DNA (pDNA) complex, to develop an efficient gene delivery system for the muscle tissue. The peptide/pDNA complex showed α-DG-dependent cellular uptake of the A2G80 sequence and significantly improved gene transfection efficiency mediated by the oligohistidine sequence in C2C12 myoblast cells. Further, the peptide/pDNA complex promoted efficient and sustained gene expression in the Duchenne muscular dystrophy mouse models. The A2G80-R9-H8 peptide has the potential for use as a specific carrier for targeting muscle in gene therapy in muscular dystrophy.


Asunto(s)
Laminina , Células Musculares , Animales , Técnicas de Transferencia de Gen , Ratones , Péptidos , Plásmidos
17.
Pharmaceutics ; 13(7)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34371694

RESUMEN

In brain-targeted delivery, the transport of drugs or genes across the blood-brain barrier (BBB) is a major obstacle. Recent reports found that focused ultrasound (FUS) with microbubbles enables transient BBB opening and improvement of drug or gene delivery. We previously developed nano-sized bubbles (NBs), which were prepared based on polyethylene glycol (PEG)-modified liposomes containing echo-contrast gas, and showed that our NBs with FUS could also induce BBB opening. The aim of this study was to enhance the efficiency of delivery of pDNA into neuronal cells following transportation across the BBB using neuron-binding peptides. This study used the RVG-R9 peptide, which is a chimeric peptide synthesized by peptides derived from rabies virus glycoprotein and nonamer arginine residues. The RVG peptide is known to interact specifically with the nicotinic acetylcholine receptor in neuronal cells. To enhance the stability of the RVG-R9/pDNA complex in vivo, PEGylated polyethyleneimine (PEG-PEI) was also used. The ternary complexes composed of RVG-R9, PEG-PEI, and pDNA could interact with mouse neuroblastoma cells and deliver pDNA into the cells. Furthermore, for the in vivo experiments using NBs and FUS, gene expression was observed in the FUS-exposed brain hemispheres. These results suggest that this systemic gene delivery system could be useful for gene delivery across the BBB.

18.
J Control Release ; 329: 1037-1045, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33080271

RESUMEN

Safe and efficient gene therapy for the treatment of Duchenne muscular dystrophy (DMD), a genetic disorder, is required. For this, the muscle-targeting delivery system of genes and nucleic acids is ideal. In this study, we focused on the A2G80 peptide, which has an affinity for α-dystroglycan expressed on muscle cell membranes, as a muscle targeted nanocarrier for DMD and developed A2G80-modified liposomes. We also prepared A2G80-modified liposomes coated with long- and short-chain PEG, called A2G80-LSP-Lip, to improve the blood circulation of liposomes using microfluidics. The liposomes had a particle size of approximately 80 nm. A2G80-LSP-Lip showed an affinity for the muscle tissue section of mice by overlay assay. When the liposomes were administered to DMD model mice (mdx mice) via the tail vein, A2G80-LSP-Lip accumulated efficiently in muscle tissue compared to control liposomes. These results suggest that A2G80-LSP-Lip can function as a muscle-targeting liposome for DMD via systemic administration, and may be a useful tool for DMD treatment.


Asunto(s)
Distrofia Muscular de Duchenne , Animales , Modelos Animales de Enfermedad , Distroglicanos , Liposomas , Ratones , Ratones Endogámicos mdx , Músculo Esquelético , Músculos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Péptidos
19.
Biol Pharm Bull ; 33(10): 1766-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20930391

RESUMEN

Targeted gene delivery to cancer cells is considered as a promising strategy for cancer therapy. Since, several targeting ligands have been studied for cancer gene therapy, such as transferrin, folate, anisamide, RGD-peptide, and antibodies. We have focused on AG73 peptide, which is derived from the globular domain of the laminin α1 chain. AG73 peptide is known as a ligand for syndecans, one of the major heparin sulfate-containing transmembrane proteoglycans. Syndecan-2 is highly expressed in various cancer cells and plays a role in angiogenesis. In this study, we prepared AG73-labeled polyethyleneglycol-modified liposomes (AG73-PEG liposomes) for gene delivery tool to syndecan-2 overexpressing cancer cells, and assessed the characterization of AG73-PEG liposomes. We confirmed the conjugation of AG73 peptide to PEG liposomes by reverse-phase high-performance liquid chromatography analysis. Electron microscopy analysis showed that monodiseperse AG73-labeled lipsomes were prepared. We also assessed the gene transfection efficiency of AG73-PEG liposomes in syndecan-2 overexpressing cancer cells or syndecan-2 less expressing cancer cells. As a result, AG73-mediated liposomal gene transfection efficiency was increased by 100-fold in syndecan-2 overexpressing cancer cells compared to syndecan-2 less expressing cancer cells. These results suggested that AG73-PEG liposomes were successfully prepared from a point of view of the modification of AG73 peptide to PEG-liposomes and the particle size of liposomes, which presented nano size. Furthermore, our results suggest that AG73-PEG liposomes can be a useful targeted gene delivery vehicle for syndecan-2 overexpressing cancer cells.


Asunto(s)
ADN/administración & dosificación , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Laminina , Liposomas , Neoplasias/tratamiento farmacológico , Fragmentos de Péptidos , Sindecano-2/metabolismo , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , ADN/uso terapéutico , Humanos , Liposomas/administración & dosificación , Liposomas/síntesis química , Microscopía Electrónica , Neoplasias/metabolismo , Tamaño de la Partícula , Transfección/métodos
20.
Pharmaceutics ; 11(6)2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-31208098

RESUMEN

Ultrasound (US) imaging is a widely used imaging technique. The use of US contrast agents such as microbubbles, which consist of phospholipids and are filled with perfluorocarbon gases, has become an indispensable component of clinical US imaging, while molecular US imaging has recently attracted significant attention in combination with efficient diagnostics. The avidin-biotin interaction method is frequently used to tether antibodies to microbubbles, leading to the development of a molecular targeting US imaging agent. However, avidin still has limitations such as immunogenicity. We previously reported that lipid-based nanobubbles (NBs) containing perfluorocarbon gas are suitable for US imaging and gene delivery. In this paper, we report on the development of a novel antibody modification method for NBs using Fc-region-binding polypeptides derived from protein A/G. First, we prepared anti-CD146 antibody-modified NBs using this polypeptide, resulting in high levels of attachment to human umbilical vein endothelial cells expressing CD146. To examine their targeting ability and US imaging capability, the NBs were administered to tumor-bearing mice. The contrast imaging of antibody-modified NBs was shown to be prolonged compared with that of non-labeled NBs. Thus, this antibody modification method using an Fc-binding polypeptide may be a feasible tool for developing a next-generation antibody-modified US imaging agent.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA