Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Thromb Thrombolysis ; 57(2): 212-219, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37839022

RESUMEN

Atrial fibrillation (AF) can increase thrombosis, especially arterial thrombosis, and some studies show that AF patients have a higher risk of developing pulmonary embolism (PE). The objective of our study is to investigate whether there is a direct causal effect of AF on PE. A two-sample Mendelian randomization (MR) approach was utilized to determine whether there is a causal relationship between AF and PE. European population-based consortia provided statistical data on the associations between Single Nucleotide Polymorphisms (SNPs) and relevant traits. The AF dataset was obtained from genome-wide association studies (GWAS) comprising 60,620 cases and 970,216 controls, while a GWAS of 1846 cases and 461,164 controls identified genetic variations associated with PE. Estimation of the causal effect was mainly performed using the random effects inverse-variance weighted method (IVW). Additionally, other tests such as MR-Egger intercept, MR-PRESSO, Cochran's Q test, "Leave-one-out," and funnel plots were conducted to assess the extent of pleiotropy and heterogeneity. Using 70 SNPs, there was no evidence to suggest an association between genetically predicted AF and risk of PE with multiplicative random-effects IVW MR analysis (odds ratio = 1.0003, 95% confidence interval: 0.9998-1.0008, P = 0.20). A null association was also observed in other methods. MR-Egger regression and MR-PRESSO respectively showed no evidence of directional (intercept, - 2.25; P = 0.94) and horizontal(P-value in the global heterogeneity test = 0.99) pleiotropic effect across the genetic variants. No substantial evidence was found to support the causal role of AF in the development of PE.


Asunto(s)
Fibrilación Atrial , Embolia Pulmonar , Trombosis , Humanos , Fibrilación Atrial/genética , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Nonoxinol , Embolia Pulmonar/genética
2.
J Transl Med ; 11: 86, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23552524

RESUMEN

BACKGROUND: The use of adenoviral vector for gene therapy is still an important strategy for advanced cancers, however, the lack of the requisite coxsackie-adenovirus receptor in cancer cells and host immune response to adenovirus limit the application of adenoviral vector in vivo. METHOD: We designed the antiangiogenic gene therapy with recombinant PEDF adenovirus (Ad-PEDF) encapsulated in cationic liposome (Ad-PEDF/Liposome), and investigated the anti-tumor efficacy of Ad-PEDF/Liposome complex on inhibition of tumor metastasis. RESULTS: We found that systemic administration of Ad-PEDF/liposome was well tolerated and resulted in marked suppression of tumor growth, and was more potent than uncoated Ad-PEDF to induce apoptosis in B16-F10 melanoma cells and inhibit murine pulmonary metastases in vivo. After Ad-luciferase was encapsulated with liposome, its distribution decreased in liver and increased in lung. The anti-Ad IgG level of Ad-PEDF/Liposome was significantly lower than Ad-PEDF used alone. CONCLUSION: The present findings provide evidences of systematic administration of cationic liposome-encapsulated Ad-PEDF in pulmonary metastatic melanoma mice model, and show an encouraging therapeutic effect for further exploration and application of more complexes based on liposome-encapsulated adenovirus for more cancers.


Asunto(s)
Adenoviridae/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Liposomas/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Melanoma/patología , Melanoma/terapia , Factores de Crecimiento Nervioso/genética , Serpinas/genética , Animales , Cationes , Femenino , Vectores Genéticos , Inmunoglobulina G/química , Liposomas/química , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 44(1): 46-8, 75, 2013 Jan.
Artículo en Zh | MEDLINE | ID: mdl-23600207

RESUMEN

OBJECTIVE: To prepare water soluble curcumin liposome and investigate its anti-tumour and antiangiogenic effects. METHODS: Liposomal curcumin was prepared by alcohol injection method. Proliferation inhibition to murin Lewis lung cancer cell line LL/2 of curcumin liposome was evaluated by MTT assay. Apoptosis and cell cycle arrest induced by liposomal curcumin were analysed by flow cytometry. Anti-tumour effects were investigated in a murine lung cancer model, and the anti-angiogenic effect was determined by aginate encapsulation assay. RESULTS: In vitro, liposomal curcumin inhibits the proliferation of LL/2 cells and induces apoptosis and cell cycle arrest. In vivo, the systemic administration of liposomal curcumin resulted in the inhibition of tumour. Aginate encapsulation assay revealed angiogenesis was decreased by curcumin liposome. CONCLUSION: The curcumin liposome treatment can significantly inhibit tumour growth in vivo.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Curcumina/farmacología , Liposomas , Neoplasias Pulmonares/patología , Neovascularización Patológica/prevención & control , Animales , Apoptosis , Línea Celular Tumoral/efectos de los fármacos , Ratones
4.
Nanotechnology ; 22(9): 095102, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21270494

RESUMEN

In an attempt to improve anticancer activity and reduce systemic toxicity of doxorubicin (Dox), we encapsulated Dox in monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) (MPEG-PCL) micelles by a novel self-assembly procedure without using surfactants, organic solvents or vigorous stirring. These Dox encapsulated MPEG-PCL (Dox/MPEG-PCL) micelles with drug loading of 4.2% were monodisperse and ∼ 20 nm in diameter. The Dox can be released from the Dox/MPEG-PCL micelles; the Dox-release at pH 5.5 was faster than that at pH 7.0. Encapsulation of Dox in MPEG-PCL micelles enhanced the cellular uptake and cytotoxicity of Dox on the C-26 colon carcinoma cell in vitro, and slowed the extravasation of Dox in the transgenic zebrafish model. Compared to free Dox, Dox/MPEG-PCL micelles were more effective in inhibiting tumor growth in the subcutaneous C-26 colon carcinoma and Lewis lung carcinoma models, and prolonging survival of mice bearing these tumors. Dox/MPEG-PCL micelles also induced lower systemic toxicity than free Dox. In conclusion, incorporation of Dox in MPEG-PCL micelles enhanced the anticancer activity and decreased the systemic toxicity of Dox; these Dox/MPEG-PCL micelles are an interesting formulation of Dox and may have potential clinical applications in cancer therapy.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Doxorrubicina/química , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Poliésteres/química , Polietilenglicoles/química , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/efectos adversos , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Micelas , Resultado del Tratamiento
5.
BMC Cancer ; 10: 402, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20678220

RESUMEN

BACKGROUND: Colorectal peritoneal carcinomatosis (CRPC) is a common form of systemic metastasis of intra-abdominal cancers. Intraperitoneal chemotherapy is a preferable option for colorectal cancer. Here we reported that a new system, 5-FU-loaded hydrogel system, can improve the therapeutic effects of intraperitoneal chemotherapy. METHODS: A biodegradable PEG-PCL-PEG (PECE) triblock copolymer was successfully synthesized. The biodegradable and temperature sensitive hydrogel was developed to load 5-FU. Methylene blue-loaded hydrogel were also developed for visible observation of the drug release. The effects and toxicity of the 5-FU-hydrogel system were evaluated in a murine CRPC model. RESULTS: The hydrogel system is an injectable flowing solution at ambient temperature and forms a non-flowing gel depot at physiological temperature. 5-FU-hydrogel was subsequently injected into abdominal cavity in mice with CT26 cancer cells peritoneal dissemination. The results showed that the hydrogel delivery system prolonged the release of methylene blue; the 5-FU-hydrogel significantly inhibited the peritoneal dissemination and growth of CT26 cells. Furthermore, intraperitoneal administration of the 5-FU-hydrogel was well tolerated and showed less hematologic toxicity. CONCLUSIONS: Our data indicate that the 5-FU-hydrogel system can be considered as a new strategy for peritoneal carcinomatosis, and the hydrogel may provide a potential delivery system to load different chemotherapeutic drugs for peritoneal carcinomatosis of cancers.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Sistemas de Liberación de Medicamentos , Fluorouracilo/uso terapéutico , Hidrogeles , Neoplasias Peritoneales/tratamiento farmacológico , Polímeros/química , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Peritoneales/secundario , Poliésteres/química , Polietilenglicoles/química , Polímeros/síntesis química , Polímeros/farmacología
6.
Int J Nanomedicine ; 10: 2051-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25844036

RESUMEN

Colorectal cancer, a type of malignant neoplasm originating from the epithelial cells lining the colon and/or rectum, has been the third most frequent malignancy and one of the leading causes of cancer-related deaths in the US. As a bioflavonoid with high anticancer potential, quercetin (Qu) has been proved to have a prospective applicability in chemotherapy for a series of cancers. However, quercetin is a hydrophobic drug, the poor hydrophilicity of which hinders its clinical usage in cancer therapy. Therefore, a strategy to improve the solubility of quercetin in water and/or enhance the bioavailability is desired. Encapsulating the poorly water-soluble, hydrophobic agents into polymer micelles could facilitate the dissolution of drugs in water. In our study, nanotechnology was employed, and quercetin was encapsulated into the biodegradable nanosized amphiphilic block copolymers of monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) (MPEG-PCL), attempting to present positive evidences that this drug delivery system of polymeric micelles is effective. The quercetin-loaded MPEG-PCL nanomicelles (Qu-M), with a high drug loading of 6.85% and a minor particle size of 34.8 nm, completely dispersed in the water and released quercetin in a prolonged period in vitro and in vivo. At the same time, compared with free quercetin, Qu-M exhibited improved apoptosis induction and cell growth inhibition effects in CT26 cells in vitro. Moreover, the mice subcutaneous CT26 colon cancer model was established to evaluate the therapy efficiency of Qu-M in detail, in which enhanced anti-colon cancer effect was proved in vivo: Qu-M were more efficacious in repressing the growth of colon tumor than free quercetin. In addition, better effects of Qu-M on inducing cell apoptosis, inhibiting tumor angiogenesis, and restraining cell proliferation were observed by immunofluorescence analysis. Our study indicated that Qu-M were a novel nanoagent of quercetin with an enhanced antitumor activity, which could serve as a promising potential candidate for colon cancer chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Portadores de Fármacos/química , Quercetina/química , Quercetina/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Apoptosis/efectos de los fármacos , Materiales Biocompatibles/química , Línea Celular Tumoral , Neoplasias del Colon/patología , Portadores de Fármacos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Micelas , Nanopartículas/química , Neovascularización Patológica/tratamiento farmacológico , Tamaño de la Partícula , Poliésteres/química , Polietilenglicoles/química , Quercetina/administración & dosificación , Quercetina/farmacocinética , Ratas Sprague-Dawley , Solubilidad , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
J Biomed Nanotechnol ; 11(4): 578-89, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26310065

RESUMEN

Paclitaxel (PTX) is efficacious in treating various solid tumors. However, the severe adverse effects of its present formulation (Cremophor EL and ethanol) and the development of drug resistance by the activation of nuclear factor-κB (NF-κB) reduce the anti-tumor activities of PTX. Curcumin (Cur) demonstrates anti-tumor activity by means of antiangiogenesis and induction of apoptosis as well as suppression of the activity of NF-κB. Therefore, to improve its antitumor activity and eliminate the toxicity of the commercial formulation of PTX, we prepared biodegradable monomethoxy poly(ethyleneglycol)-poly(ε-caprolactone) (MPEG-PCL) micelles to co-deliver PTX and Cur using a solid dispersion method. The mixed PTX and Cur polymeric micelles (PTX-Cur-M) produced were monomorphous micelles of 38 nm in diameter that released PTX and Cur for an extended period of time and induced cell apoptosis in vitro. In addition, the PTX-Cur-M exhibited anti-angiogenic activity in vitro and in vivo. Furthermore, the therapeutic efficacy of PTX-Cur-M in a mouse model of colon cancer was evaluated. PTX-Cur-M micelles produced significantly more inhibition of tumor growth than Cur micelles (Cur-M) and PTX micelles (PTX-M) alone at the same dose (P < 0.05 and P < 0.05, respectively). Immunohistochemical and immunofluorescent analyses demonstrated that PTX-Cur-M enhanced tumor cell apoptosis and inhibited angiogenesis to a greater extent than control treatment. Our data suggested that PTX-Cur-M may have potential clinical applications in cancer therapy.


Asunto(s)
Antineoplásicos/química , Neoplasias del Colon/tratamiento farmacológico , Curcumina/administración & dosificación , Curcumina/química , Sistemas de Liberación de Medicamentos , Micelas , Paclitaxel/administración & dosificación , Animales , Apoptosis , Neoplasias del Colon/patología , Portadores de Fármacos , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Microcirculación , Microscopía Fluorescente , FN-kappa B/metabolismo , Neovascularización Patológica , Polímeros/química , Pez Cebra
8.
Curr Cancer Drug Targets ; 14(2): 167-80, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24410728

RESUMEN

FUS1 is one of the most important tumor suppressor genes in lung cancer, as well as an important immunomodulatory molecule. Interleukin (IL)-12 has attracted considerable interest as a potential anti-tumor cytokine. Cationic liposome has been shown to effectively deliver therapeutic genes to the lungs and control metastatic lung tumors when administered intravenously. Here we evaluated the enhanced efficacy of cationic liposome-mediated delivery of FUS1 and human IL (hIL)-12 eukaryotic coexpression plasmid (pVITRO2-FUS1-hIL-12) against the human lung cancer in HuPBL-NOD/SCID mice model by local and systemic administration, and explored the related molecular mechanism. Our study demonstrated that FUS1-hIL-12 coexpression could more sufficiently inhibit tumor growth and experimental lung metastasis, significantly prolong the survival of experimental lung metastasis mice. Moreover, FUS1-hIL-12 coexpression performed higher antitumor activity and lower toxicity in the inhibition of experimental lung metastatic tumor compared to cisplatin. We further identified that FUS1-hIL-12 coexpression could induce strong antitumor immune response by secreting much higher levels of human interferon-γ (hIFN-γ) and hIL-15, enhancing expression of MHC-I and Fas, increasing infiltration of activated human CD4+ and CD8+ T lymphocytes. FUS1-hIL-12 coexpression could also obviously induce tumor cell apoptosis and inhibit tumor cell proliferation partly by higher activation of STAT1 signal pathway and upregulation of p53. In addition, FUS1-hIL-12 coexpression also superiorly reduced the angiogenesis in tumors, which might be associated with downregulation of VEGF and VEGFR, and upregulation of human IP-10. Our results therefore suggest that cationic liposome-mediated FUS1-hIL-12 coexpression may be a new promising strategy for lung cancer treatment in clinical studies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Interleucina-12/biosíntesis , Neoplasias Pulmonares/terapia , Plásmidos , Proteínas Supresoras de Tumor/biosíntesis , Animales , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/secundario , Cationes , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Femenino , Humanos , Interleucina-12/genética , Liposomas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Patológica , Factores de Tiempo , Carga Tumoral , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Asian Pac J Cancer Prev ; 14(4): 2307-10, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23725132

RESUMEN

Curcumin previously was proven to inhibit angiogenesis and display potent antitumor activity in vivo and in vitro. In the present study, we investigated whether a combination curcumin with hyperthermia would have a synergistic antitumor effect in the LL/2 model. The results indicated that combination therapy significantly inhibited cell proliferation of MS-1 and LL/2 in vitro. LL/2 experiment model also demonstrated that the combination therapy inhibited tumor growth and prolonged the life span in vivo. Furthermore, combination therapy reduced angiogenesis and increased tumor apoptosis. Our findings suggest that the combination therapy exerted synergistic antitumor effects, providing a new perspective fpr clinical tumor therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma Pulmonar de Lewis/prevención & control , Curcumina/uso terapéutico , Hipertermia Inducida , Neovascularización Patológica/prevención & control , Animales , Western Blotting , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular , Terapia Combinada , Femenino , Técnica del Anticuerpo Fluorescente , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
10.
Integr Cancer Ther ; 12(3): 264-70, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22740083

RESUMEN

Thermotherapy and thermochemotherapy have been used in clinics to treat patients with malignant diseases, including colon cancer, and their efficacy has been well proved. Heat shock proteins (HSPs), especially Hsp70, play important roles in neutralizing their efficacy. It has been reported that quercetin can suppress cancer by inhibiting the intratumoral expression of Hsp70. This study was designed to investigate whether quercetin could enhance sensitivity to thermotherapy and thermochemotherapy. Soluble quercetin liposome was used in this study. The effects of quercetin were investigated in vitro and in mouse colon cancer models of subcutaneous tumor and peritoneal carcinomatosis. The results showed that quercetin liposome inhibited the upregulation of Hsp70 and enhanced apoptosis induced by hyperthermia and thermochemotherapy. Systemic administration of quercetin liposome can sensitize CT26 cells to thermotherapy and chemothermotherapy. This study suggests that quercetin liposome might be potentially applied for clinical cancer therapy.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/terapia , Hipertermia Inducida/métodos , Liposomas/farmacología , Quercetina/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Terapia Combinada , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Ratones Endogámicos BALB C , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
11.
Int J Nanomedicine ; 8: 2453-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23885172

RESUMEN

Adhesions can form after almost any type of abdominal surgery. Postoperative adhesions can be prevented by improved surgical techniques, such as reducing surgical trauma, preventing ischemia, and avoiding exposure of the peritoneal cavity to foreign materials. Although improved surgical techniques can potentially reduce formation of adhesions, they cannot be eliminated completely. Therefore, finding more effective methods to prevent postoperative adhesions is imperative. Recently, we found that a novel thermosensitive hydrogel, ie, poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCEC) had the potential to prevent postoperative adhesions. Using the ring-opening polymerization method we prepared a PCEC copolymer which could be dissolved and assembled at 55°C into PCEC micelles with mean size of 25 nm. At body temperature, a solution containing PCEC micelles could convert into a hydrogel. The PCEC copolymer was biodegradable and had low toxicity in vitro and in vivo. We found that most animals in a hydrogel-treated group (n = 10) did not develop adhesions. In contrast, 10 untreated animals developed adhesions that could only be separated by sharp dissection (P < 0.001). The hydrogel could adhere to peritoneal wounds and degraded gradually over 7-9 days, transforming into a viscous fuid that was completely absorbed within 12 days. The injured parietal and visceral peritoneum remesothelialized over about seven and nine days, respectively. This study confirms that PCEC hydrogel has potential application in the prevention of postoperative adhesions.


Asunto(s)
Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapéutico , Poliésteres/uso terapéutico , Polietilenglicoles/uso terapéutico , Adherencias Tisulares , Abdomen/patología , Animales , Línea Celular , Femenino , Fibrosis/patología , Células HEK293 , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Ratones , Ratones Endogámicos BALB C , Micelas , Nanopartículas/química , Poliésteres/química , Poliésteres/farmacología , Polietilenglicoles/química , Polietilenglicoles/farmacología , Ratas Wistar , Estadísticas no Paramétricas , Temperatura , Adherencias Tisulares/tratamiento farmacológico , Adherencias Tisulares/prevención & control
12.
Int J Mol Med ; 31(1): 33-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23128378

RESUMEN

Cisplatin is one of the most effective antitumor drugs for non-small cell lung carcinoma (NSCLC) patients. However, its efficacy has encountered a plateau due to its side effects and drug resistance. Inducible nitric oxide (NO) synthase (iNOS) gene therapy has been reported to have antitumor effects in several types of cancers and enhances sensitivity to cisplatin, but the effects of iNOS gene therapy alone or its combination with cisplatin in lung cancer remain unclear. In the current study, we evaluated the effects of cationic liposome (LP)-mediated iNOS gene transfection on enhancing low-dose cisplatin-mediated antitumor effects in the A549 human lung adenocarcinoma cell line in vitro. Furthermore, we examined whether iNOS gene therapy enhances the antitumor effects of low-dose cisplatin in two A549 human lung cancer cell xenograft mouse models. The results revealed that iNOS gene therapy may significantly enhance low-dose cisplatin-mediated inhibition of cell proliferation, invasion, migration and promotion of cell apoptosis in A549 cells. Intratumoral administration of the LP-pVAX-iNOS complex significantly enhanced low-dose cisplatin-mediated suppression of subcutaneous tumor growth. Moreover, intravenous injection of the LP-pVAX-iNOS complex greatly enhanced low-dose cisplatin-mediated inhibition of experimental lung metastasis and prolonged the life span of mice without significant organ-related toxicity in a nude mouse model of lung metastasis compared to the cisplatin alone-treated group. Furthermore, iNOS gene-mediated enhancement of cisplatin-mediated antitumor effects in lung cancer may be related to the attenuation of p-mTOR, MMP2 and the activation of p-p53. Thus, the combination treatment with iNOS gene therapy and cisplatin may be a novel and effective therapeutic strategy for lung cancer.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Terapia Genética , Liposomas/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Óxido Nítrico Sintasa de Tipo II/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Int J Nanomedicine ; 8: 3521-31, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24101869

RESUMEN

Systemic administration of chemotherapy for cancer often has toxic side effects, limiting the doses that can be used in its treatment. In this study, we developed methoxy poly(ethylene glycol)-poly(caprolactone) (MPEG-PCL) micelles loaded with curcumin and doxorubicin (Cur-Dox/MPEG-PCL) that were tolerated by recipient mice and had enhanced antitumor effects and fewer side effects. It was shown that these Cur-Dox/MPEG-PCL micelles could release curcumin and doxorubicin slowly in vitro. The long circulation time of MPEG-PCL micelles and the slow rate of release of curcumin and doxorubicin in vivo may help to maintain plasma concentrations of active drug. We also demonstrated that Cur-Dox/MPEG-PCL had improved antitumor effects both in vivo and in vitro. The mechanism by which Cur-Dox/MPEG-PCL micelles inhibit lung cancer might involve increased apoptosis of tumor cells and inhibition of tumor angiogenesis. We found advantages using Cur-Dox/MPEG-PCL micelles in the treatment of cancer, with Cur-Dox/MPEG-PCL achieving better inhibition of LL/2 lung cancer growth in vivo and in vitro. Our study indicates that Cur-Dox/MPEG-PCL micelles may be an effective treatment strategy for cancer in the future.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/síntesis química , Neoplasias Pulmonares/tratamiento farmacológico , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Poliésteres/química , Polietilenglicoles/química , Animales , Línea Celular Tumoral , Curcumina/administración & dosificación , Curcumina/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Femenino , Inyecciones Intravenosas , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Micelas , Nanocápsulas/ultraestructura , Tamaño de la Partícula , Resultado del Tratamiento
14.
Carbohydr Polym ; 89(4): 1101-9, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24750920

RESUMEN

The need to enhance the immunogenicity of tumor-associated antigens and modulate the resulting immune responses has prompted the development of new adjuvants. We prepared a novel adjuvant, lipopolysaccharides (LPS) loaded thermosensitive hydrogel (LPS-Hydrogel), for truncated basic fibroblast growth factor (tbFGF) peptide to enhance immunological responses and improve therapeutic effects in cancer. When co-formulated with tbFGF, LPS-Hydrogel formed antigen-adjuvant complexes, which enhanced antibody and cell-mediated responses in mice, thus promoting a more balanced antibody-mediated and cytotoxic T lymphocyte (CTL)-mediated immune response to inhibit tumor growth and metastases in vivo. Furthermore, the secretion of IFN-γ and IL-4 was detected, confirming activation of the two immune responses in vivo. There were no significant systemic toxicities observed with tbFGF-LPS-Hydrogel treatment. These results suggested that the thermosensitive and biodegradable LPS-Hydrogel was a novel adjuvant and carrier for peptide vaccines in cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos , Carcinoma Pulmonar de Lewis , Factor 2 de Crecimiento de Fibroblastos , Hidrogel de Polietilenoglicol-Dimetacrilato , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Lipopolisacáridos , Adyuvantes Inmunológicos/síntesis química , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/farmacología , Hidrogel de Polietilenoglicol-Dimetacrilato/síntesis química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Interferón gamma/inmunología , Interleucina-4/inmunología , Lipopolisacáridos/química , Lipopolisacáridos/farmacología , Ratones , Células 3T3 NIH , Linfocitos T/inmunología , Linfocitos T/patología
15.
Mol Med Rep ; 6(6): 1309-14, 2012 12.
Artículo en Inglés | MEDLINE | ID: mdl-22971976

RESUMEN

Adjuvants are important components of recombinant protein vaccines which are often poorly immunogenic. For decades, the search for new vaccine adjuvants has been predominantly empirical. In addition, combinations of more than one adjuvant plus antigen have been systematically studied. Plasmid DNA containing additional oligodeoxynucleotides with unmethylated CpG motifs (CpG ODN) entrapped in liposomes has been used as an adjuvant for DNA vaccines and has shown powerful immunostimulatory functions. In our study, the combination of plasmid DNA containing 16 additional CpG ODNs (pv-16CpG) and aluminum hydroxide (AL) or incomplete Freund's adjuvant (IFA) was used as an adjuvant for a hepatitis B surface antigen (HBsAg) vaccine to immunize C57BL/6J mice. ELISA and ELISPOT assays were used to analyze the immunological effects of the novel vaccine. A significant enhancement of the anti-HBs titer and seroconversion was observed when the CpG plasmid was combined with IFA, but not with AL. In addition, anti-HBs antibody isotype analysis revealed that the combination of CpG plasmid and IFA induced a strong HBsAg-specific IgG2a response. Moreover, the ELISPOT assays suggested that pv-16CpG suspended in IFA evoked a strong T helper 1 (Th1) immune response and high IFN-γ production. These results demonstrate that pv-16CpG suspended in IFA is able to induce cellular and humoral immune responses to HBsAg, and confirm its potential as an adjuvant for use in protein vaccines.


Asunto(s)
Hidróxido de Aluminio/inmunología , Adyuvante de Freund/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Lípidos/inmunología , Oligodesoxirribonucleótidos/genética , Plásmidos/metabolismo , Animales , Antígenos de Superficie de la Hepatitis B/metabolismo , Inmunoglobulina G/metabolismo , Isotipos de Inmunoglobulinas/metabolismo , Liposomas/química , Liposomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Vacunas de ADN/inmunología
16.
Oncol Res ; 20(12): 579-87, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-24139416

RESUMEN

Metastasis is the most lethal attribute of human malignancy. High-level expression of survivin is involved in both carcinogenesis and angiogenesis in cancer. Previous studies indicate that a mutation of the threonine residue at position 34 (Thr34Ala) of survivin generates a dominant-negative mutant that induces apoptosis, inhibits angiogenesis, and suppresses highly metastatic breast carcinoma in mouse models. We investigated the efficacy of gene therapy with a survivin dominant-negative mutant and possible factors related to lymph node metastasis. The metastasis rate was compared between each group in order to find a survivin-targeted therapy against lymphangiogenesis in its earliest stages. We established lymph node metastasis models and treated animals with H22 tumors with Lip-mSurvivinT34A (Lip-mS), Lip-plasmid (Lip-P), or normal saline (NS). Eight days after the last dose, five randomly chosen mice from each group were sacrificed. We detected the apoptotic index, microvessel density (MVD), lymphatic microvessel density (LMVD), and the expression of VEGF-D with immunohistochemistry. After the remaining animals were sacrificed, we compared the tumor-infiltrated lymph nodes in each group. Administration of mSurvivinT34A plasmid complexed with cationic liposome (DOTAP/chol) resulted in the efficacious inhibition of tumor growth and lymph node metastasis within the mouse H22 tumor model. These responses were associated with tumor cell apoptosis, and angiogenesis and lymphangiogenesis inhibition. Our results suggested that Lip-mSurvivinT34A induced apoptosis and inhibited tumor angiogenesis and lymphangiogenesis, thus suppressing tumor growth and lymphatic metastasis. The mSurvivinT34A survivin mutant is a promising strategy of gene therapy to inhibit lymphatic metastasis.


Asunto(s)
Terapia Genética/métodos , Proteínas Inhibidoras de la Apoptosis/genética , Plásmidos/administración & dosificación , Plásmidos/genética , Proteínas Represoras/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Liposomas/administración & dosificación , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/terapia , Linfangiogénesis/genética , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Mutación , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Proteínas Represoras/biosíntesis , Survivin
17.
Int J Nanomedicine ; 7: 2601-11, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22679371

RESUMEN

Radiation pneumonitis (RP) is an important dose-limiting toxicity during thoracic radiotherapy. Previous investigations have shown that curcumin is used for the treatment of inflammatory conditions and cancer, suggesting that curcumin may prevent RP and sensitize cancer cells to irradiation. However, the clinical advancement of curcumin is limited by its poor water solubility and low bioavailability after oral administration. Here, a water-soluble liposomal curcumin system was developed to investigate its prevention and sensitizing effects by an intravenous administration manner in mice models. The results showed that liposomal curcumin inhibited nuclear factor-κB pathway and downregulated inflammatory factors including tumor necrosis factor-α, interleukin (IL)-6, IL-8, and transforming growth factor-ß induced by thoracic irradiation. Furthermore, the combined treatment with liposomal curcumin and radiotherapy increased intratumoral apoptosis and microvessel responses to irradiation in vivo. The significantly enhanced inhibition of tumor growth also was observed in a murine lung carcinoma (LL/2) model. There were no obvious toxicities observed in mice. The current results indicate that liposomal curcumin can effectively mitigate RP, reduce the fibrosis of lung, and sensitize LL/2 cells to irradiation. This study also suggests that the systemic administration of liposomal curcumin is safe and deserves to be investigated for further clinical application.


Asunto(s)
Adenocarcinoma/radioterapia , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Curcumina/farmacología , Liposomas/farmacología , Neoplasias Pulmonares/radioterapia , Neumonitis por Radiación/terapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Análisis de Varianza , Animales , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/radioterapia , Línea Celular Tumoral , Terapia Combinada , Curcumina/química , Curcumina/toxicidad , Citocinas/sangre , Histocitoquímica , Liposomas/química , Liposomas/toxicidad , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Neumonitis por Radiación/diagnóstico por imagen , Neumonitis por Radiación/tratamiento farmacológico , Neumonitis por Radiación/prevención & control , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/toxicidad , Radiografía Torácica
18.
Int J Mol Med ; 28(1): 3-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21503566

RESUMEN

Basic fibroblast growth factor (bFGF), an angiogenic factor, exhibits pro-angiogenic abilities by interacting with tyrosine kinase receptors and heparin-sulfated proteoglycan receptors. Here, we designed an N-, C-terminally truncated basic fibroblast growth factor (tbFGF) for immuno-therapy of murine lung carcinoma with PCEC hydrogel as adjuvant, comparing it with the wild-type bFGF. In vitro, tbFGF did not stimulate NIH-3T3 fibroblast proliferation. In vivo, after immunization, both tbFGF and bFGF were able to induce a robust bFGF-specific immune response. The protective anti-tumor investigation showed a significant inhibition of tumor growth and reduction of tumor vascularization detected by immunohistochemical staining and the alginate-encapsulated tumor cell assay in the tbFGF or the bFGF group. These data suggested that tbFGF can be used in the immunotherapy of tumors, without the risks associated with bFGF, which induces neovascularization in normal tissues.


Asunto(s)
Carcinoma Pulmonar de Lewis/terapia , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Neoplasias Pulmonares/terapia , Neovascularización Patológica/terapia , Animales , Vacunas contra el Cáncer , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Inmunoterapia/métodos , Neoplasias Pulmonares/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Poliésteres/farmacología , Polietilenglicoles/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico
19.
Nanoscale ; 3(4): 1558-67, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21283869

RESUMEN

Curcumin is an effective and safe anticancer agent, but its hydrophobicity inhibits its clinical application. Nanotechnology provides an effective method to improve the water solubility of hydrophobic drug. In this work, curcumin was encapsulated into monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) (MPEG-PCL) micelles through a single-step nano-precipitation method, creating curcumin-loaded MPEG-PCL (Cur/MPEG-PCL) micelles. These Cur/MPEG-PCL micelles were monodisperse (PDI = 0.097 ± 0.011) with a mean particle size of 27.3 ± 1.3 nm, good re-solubility after freeze-drying, an encapsulation efficiency of 99.16 ± 1.02%, and drug loading of 12.95 ± 0.15%. Moreover, these micelles were prepared by a simple and reproducible procedure, making them potentially suitable for scale-up. Curcumin was molecularly dispersed in the PCL core of MPEG-PCL micelles, and could be slow-released in vitro. Encapsulation of curcumin in MPEG-PCL micelles improved the t(1/2) and AUC of curcumin in vivo. As well as free curcumin, Cur/MPEG-PCL micelles efficiently inhibited the angiogenesis on transgenic zebrafish model. In an alginate-encapsulated cancer cell assay, intravenous application of Cur/MPEG-PCL micelles more efficiently inhibited the tumor cell-induced angiogenesis in vivo than that of free curcumin. MPEG-PCL micelle-encapsulated curcumin maintained the cytotoxicity of curcumin on C-26 colon carcinoma cells in vitro. Intravenous application of Cur/MPEG-PCL micelle (25 mg kg(-1) curcumin) inhibited the growth of subcutaneous C-26 colon carcinoma in vivo (p < 0.01), and induced a stronger anticancer effect than that of free curcumin (p < 0.05). In conclusion, Cur/MPEG-PCL micelles are an excellent intravenously injectable aqueous formulation of curcumin; this formulation can inhibit the growth of colon carcinoma through inhibiting angiogenesis and directly killing cancer cells.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Curcumina/administración & dosificación , Curcumina/química , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/síntesis química , Polímeros/química , Implantes Absorbibles , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Neoplasias del Colon/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Micelas , Resultado del Tratamiento
20.
ACS Nano ; 4(10): 5573-84, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20839784

RESUMEN

Biodegradable cationic nanoparticles have promising application as a gene delivery system. In this article, heparin-polyethyleneimine (HPEI) nanogels were prepared, and these nanogels were developed as a nonviral gene vector. The transfection efficiency of HPEI nanogels was comparable with that of PEI25K, while the cytotoxicity was lower than that of PEI2K and much lower than that of PEI25K in vitro. These HPEI nanogels also had better blood compatibility than PEI25K. After intravenous administration, HPEI nanogels degraded, and the degradation products were excreted through urine. The plasmid expressing vesicular stomatitis virus matrix protein (pVSVMP) could be efficiently transfected into C-26 colon carcinoma cells by HPEI nanogels in vitro, inhibiting the cell proliferation through apoptosis induction. Intraperitoneal injection of pVSVMP/HPEI complexes efficiently inhibited the abdominal metastases of C-26 colon carcinoma through apoptosis induction (mean tumor weight in mice treated with pVSVMP/HPEI complex = 0.93 g and in control mice = 3.28 g, difference = 2.35 g, 95% confidence interval [CI] = 1.75-2.95 g, P < 0.001) and prolonged the survival of treated mice. Moreover, intravenous application of pVSVMP/HPEI complexes also inhibited the growth of pulmonary metastases of C-26 colon carcinoma through apoptosis induction. The HPEI nanogels delivering pVSVMP have promising application in treating colon carcinoma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/terapia , Geles/química , Terapia Genética/métodos , Nanotecnología/métodos , Polietileneimina/farmacología , Vesiculovirus/genética , Proteínas de la Matriz Viral/genética , Animales , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Ratones , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA