Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Med Rep ; 9(6): 2069-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24714917

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a potent inhibitor of angiogenesis, and the antitumor effect of adeno-associated virus (AAV)-mediated PEDF expression has been demonstrated in a range of animal models. The combined treatment of low-dose chemotherapy and gene therapy inhibits the growth of solid tumors more effectively than current traditional therapies or gene therapy alone. In the present study, the effect of treatment with an AAV2 vector harboring the human PEDF (hPEDF) gene in combination with low-dose cisplatin on the growth of Lewis lung carcinoma (LLC) in mice was assessed. LLC cells were infected with AAV-enhanced green fluorescent protein (EGFP) in the presence or absence of cisplatin, and then the effect of cisplatin on AAV-mediated gene expression was evaluated by image and flow cytometric analysis. Tumor growth, survival time, vascular endothelial growth factor (VEGF) expression, microvessel density (MVD) and apoptotic index were analyzed in C57BL/6 mice treated with AAV-hPEDF, cisplatin or cisplatin plus AAV-hPEDF. The results of the present study provide evidence that cisplatin treatment is able to enhance AAV-mediated gene expression in LLC cells. In addition, the combined treatment of cisplatin plus AAV­hPEDF markedly prolonged the survival time of the mice and inhibited tumor growth, resulting in significant suppression of tumor angiogenesis and induction of tumor apoptosis in vivo, and also protected against cisplatin-related toxicity. These findings suggest that combination of AAV-hPEDF and cisplatin has potential as a novel therapeutic strategy for lung cancer.


Subject(s)
Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/therapy , Cisplatin/pharmacology , Dependovirus/genetics , Eye Proteins/genetics , Genetic Vectors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Nerve Growth Factors/genetics , Serpins/genetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cisplatin/administration & dosage , Combined Modality Therapy , Disease Models, Animal , Eye Proteins/metabolism , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Lung Neoplasms/pathology , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Nerve Growth Factors/metabolism , Serpins/metabolism
2.
J Biomed Nanotechnol ; 9(12): 1984-95, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24266254

ABSTRACT

Postsurgical peritoneal adhesion is a major concern in clinical practice which causes significant morbidity and mortality. In this study, we investigated the efficacy of biodegradable and injectable thermosensitive poly(ethylene glycol)-poly(epsilon-caprolactone)-poly(ethylene glycol) (PEG-PCL-PEG) micelles in preventing postsurgical cauterization-induced peritoneal adhesion. The biodegradable PEG-PCL-PEG copolymer could form nano-sized micelles in water, which instantly turned into a non-flowing gel at body temperature due to micellar aggregation. Moreover, a novel sidewall and cecum cauterization rat model was developed and the micelles were assigned for adhesion prevention tests. The PEG-PCL-PEG micelles could be administered by an ordinary syringe and provided unrestricted coverage of the cauterized peritoneum. The micelles instantly formed a gel in situ at body temperature and the formed gel could adhere to the cauterized sites as a durable barrier during critical time of adhesion formation. All rats from the control group (n = 10) developed score 5 adhesion, whereas, eight out of ten rats in the micelle-treated group showed no adhesion at all. Besides, cauterization-induced adhesion formation, adhesiveness and degradation of micelles, remesothelization of peritoneum, and restoration of cauterized tissue were investigated in detail. Our results thus indicated that, it was feasible to use biodegradable and injectable thermosensitive PEG-PCL-PEG micelles for prevention of peritoneal adhesions after surgery.


Subject(s)
Absorbable Implants , Cautery/adverse effects , Micelles , Peritoneal Diseases/prevention & control , Polyesters/therapeutic use , Polyethylene Glycols/therapeutic use , Tissue Adhesions/prevention & control , Animals , Cautery/methods , Female , Materials Testing , Mice , NIH 3T3 Cells , Peritoneum/drug effects , Peritoneum/surgery , Polyesters/pharmacokinetics , Polyesters/pharmacology , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Rats , Rats, Wistar , Temperature , Treatment Outcome
3.
BMC Cancer ; 12: 129, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22462776

ABSTRACT

BACKGROUND: Angiogenesis plays an important role in tumor growth and metastasis, therefore antiangiogenic therapy was widely investigated as a promising approach for cancer therapy. Recently, pigment epithelium-derived factor (PEDF) has been shown to be the most potent inhibitor of angiogenesis. Adeno-associated virus (AAV) vectors have been intensively studied due to their wide tropisms, nonpathogenicity, and long-term transgene expression in vivo. The objective of this work was to evaluate the ability of AAV-mediated human PEDF (hPEDF) as a potent tumor suppressor and a potential candidate for cancer gene therapy. METHODS: Recombinant AAV2 encoding hPEDF (rAAV2-hPEDF) was constructed and produced, and then was assigned for in vitro and in vivo experiments. Conditioned medium from cells infected with rAAV2-hPEDF was used for cell proliferation and tube formation tests of human umbilical vein endothelial cells (HUVECs). Subsequently, colorectal peritoneal carcinomatosis (CRPC) mouse model was established and treated with rAAV2-hPEDF. Therapeutic efficacy of rAAV2-hPEDF were investigated, including tumor growth and metastasis, survival time, microvessel density (MVD) and apoptosis index of tumor tissues, and hPEDF levels in serum and ascites. RESULTS: rAAV2-hPEDF was successfully constructed, and transmission electron microscope (TEM) showed that rAAV2-hPEDF particles were non-enveloped icosahedral shape with a diameter of approximately 20 nm. rAAV2-hPEDF-infected cells expressed hPEDF protein, and the conditioned medium from infected cells inhibited proliferation and tube-formation of HUVECs in vitro. Furthermore, in CRPC mouse model, rAAV2-hPEDF significantly suppressed tumor growth and metastasis, and prolonged survival time of treated mice. Immunofluorescence studies indicated that rAAV2-hPEDF could inhibit angiogenesis and induce apoptosis in tumor tissues. Besides, hPEDF levels in serum and ascites of rAAV2-hPEDF-treated mice were significant higher than those in rAAV2-null or normal saline (NS) groups. CONCLUSIONS: Thus, our results suggest that rAAV2-hPEDF may be a potential candidate as an antiangiogenic therapy agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma/therapy , Eye Proteins/administration & dosage , Genetic Therapy/methods , Nerve Growth Factors/administration & dosage , Peritoneal Neoplasms/therapy , Serpins/administration & dosage , Adenoviridae/genetics , Animals , Blotting, Western , Carcinoma/pathology , Carcinoma/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Genetic Vectors , Mice , Mice, Inbred BALB C , Microvessels/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Peritoneal Neoplasms/pathology
4.
Oncol Rep ; 27(4): 1142-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22218393

ABSTRACT

Pigment epithelium-derived factor (PEDF) is the most potent inhibitor of angiogenesis in the mammalian eye, and mechanisms through which PEDF exerts its antitumour activity have recently been defined. The aim of our research was to evaluate the ability of adeno-associated virus (AAV) vector-mediated transfer of human PEDF to inhibit lewis lung carcinoma (LCC) cell growth. Intratumoural injection of AAV-PEDF caused significant reduction of the tumour volume and prolonged the survival time of mice bearing LLC cells, which were associated with decreased microvessel density and increased apoptosis in the tumours. AAV vectors represent a very promising tool for cancer gene therapy. No noticeable toxicity concerning AAV was detected as inferred from monitoring changes in animal body weight as well as basic organ structure and histological morphology, and by analyzing mouse liver and kidney function. Our findings indicate that AAV-mediated PEDF gene expression may offer an active approach to inhibit LLC growth and that treatment with AAV-PEDF may provide a promising therapeutic strategy in lung cancer treatment.


Subject(s)
Carcinoma, Lewis Lung/therapy , Dependovirus/genetics , Eye Proteins/genetics , Genetic Therapy/methods , Genetic Vectors , Nerve Growth Factors/genetics , Serpins/genetics , Animals , Apoptosis , Capillaries/pathology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Dependovirus/metabolism , Eye Proteins/metabolism , Genetic Therapy/adverse effects , Genetic Vectors/toxicity , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Nerve Growth Factors/metabolism , Serpins/metabolism , Time Factors , Transfection , Tumor Burden
5.
Growth Factors ; 29(6): 290-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21981422

ABSTRACT

In this work, a novel vaccine delivery system, biodegradable nanoparticles (NPs) in thermosensitive hydrogel, was investigated. Human basic fibroblast growth factor (bFGF)-loaded NPs (bFGF-NPs) were prepared, and then bFGF-NPs were incorporated into thermosensitive hydrogel to form bFGF-NPs in a hydrogel composite (bFGF-NPs/hydrogel). bFGF-NPs/hydrogel was an injectable sol at ambient temperature, but was converted into a non-flowing gel at body temperature. The in vitro release profile showed that bFGF could be released from bFGF-NPs or bFGF-NPs/hydrogel at an extended period, but the release rate of bFGF-NPs/hydrogel was much lower. In vivo experiments suggested that immunogenicity of bFGF improved significantly after being incorporated into the NPs/hydrogel composite, and strong humoral immunity was maintained for longer than 12 weeks. Furthermore, an in vivo protective anti-tumor immunity assay indicated that immunization with bFGF-NPs/hydrogel could induce significant suppression of the growth and metastases of tumors. Thus, the NPs/hydrogel composite may have great potential application as a novel vaccine delivery system.


Subject(s)
Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/therapy , Fibroblast Growth Factor 2/immunology , Hydrogels , Nanocapsules , Vaccination/methods , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/prevention & control , Female , Fibroblast Growth Factor 2/administration & dosage , Humans , Mice , Mice, Inbred C57BL
6.
J Biomed Mater Res B Appl Biomater ; 91(1): 26-36, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19365823

ABSTRACT

In this work, a biodegradable poly(ethylene glycol)-poly(epsilon-caprolactone)-poly (ethylene glycol) (PEG-PCL-PEG, PECE) triblock copolymer was successfully synthesized. The aqueous solution of such PECE copolymer displayed special sol-gel-sol transition as temperature increase, which is a flowing sol at low-temperature and turns into a nonflowing gel at body temperature. The cytotoxicity of PECE copolymer was evaluated by cell viability assay using HEK 293 cells. In vivo gel formation and degradation test based on intraperitoneal and subcutaneous administration was conducted, respectively. The acute toxicity test and histopathological study were performed in BALB/c mice by intrapleural, intraperitoneal, or subcutaneous administration of PECE hydrogel (30 Wt %), respectively. The dose of intrapleural, intraperitoneal, or subcutaneous administration was up to 10 g/kg body weight (b.w.), 25 g/kg b.w., and 25 g/kg b.w., respectively, and the mice were observed continuously for 14 days. For histopathologic study, samples including heart, liver, lung, kidneys, spleen, stomach, intestine, and tissue of injection site were prepared for histochemical analysis and were stained with hematoxylin-eosin. No mortality or significant signs of acute toxicity was observed during the whole observation period and there is no significant lesion to be shown in histopathologic study of major organs. Therefore, the maximum tolerance dose of PECE hydrogel by intrapleural, intraperitoneal, or subcutaneous administration was calculated to be higher than 10 g/kg b.w., 25 g/kg b.w., and 25 g/kg b.w., respectively. The results indicated that the prepared PECE hydrogel was nontoxic after intrapleural, intraperitoneal, or subcutaneous administration, and it could be a safe candidate for in situ gel-forming controlled drug delivery system.


Subject(s)
Biocompatible Materials , Drug Carriers , Drug Delivery Systems , Hydrogel, Polyethylene Glycol Dimethacrylate , Polyesters , Polyethylene Glycols , Absorbable Implants , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/toxicity , Cell Line , Drug Carriers/chemistry , Drug Carriers/toxicity , Female , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/toxicity , Infusions, Subcutaneous , Male , Materials Testing , Mice , Mice, Inbred BALB C , Molecular Structure , Phase Transition , Polyesters/chemistry , Polyesters/toxicity , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...