Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34830094

RESUMEN

Cancer is one of the leading causes of death in the world. A cancer-targeted multifunctional probe labeled with the radionuclide has been developed to provide multi-modalities for NIR fluorescence and nuclear imaging (PET, SPECT), for photothermal therapy (PTT), and targeted radionuclide therapy of cancer. In this study, synthesis, characterization, in vitro, and in vivo biological evaluation of the cyanine-based probe (DOTA-NIR790) were demonstrated. The use of cyanine dyes for the selective accumulation of cancer cells were used to achieve the characteristics of tumor markers. Therefore, all kinds of organ tumors can be targeted for diagnosis and treatment. The DOTA-NIR790 labeled with lutetium-111 could detect original or metastatic tumors by using SPECT imaging and quantify tumor accumulation. The ß-emission of 177Lu-DOTA-NIR790 can be used for targeted radionuclide therapy of tumors. The DOTA-NIR790 enabled imaging by NIR fluorescence and by nuclear imaging (SPECT) to monitor in real-time the tumor accumulation and the situation of cancer therapy, and to guide the surgery or the photothermal therapy of the tumor. The radionuclide-labeled heptamethine cyanine based probe (DOTA-NIR790) offers multifunctional modalities for imaging and therapies of cancer.


Asunto(s)
Lutecio , Neoplasias Experimentales , Imagen Óptica , Compuestos de Quinolinio , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Nanomedicina Teranóstica , Células A549 , Animales , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Células HCT116 , Humanos , Lutecio/química , Lutecio/farmacología , Ratones , Ratones Desnudos , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/terapia , Compuestos de Quinolinio/química , Compuestos de Quinolinio/farmacología , Radioisótopos/química , Radioisótopos/farmacología
2.
Int J Mol Sci ; 22(2)2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33440880

RESUMEN

Osteoarthritis (OA) remains one of the common degenerative joint diseases and a major cause of pain and disability in older adult individuals. Oral administration of non-steroidal anti-inflammatory drugs (NSAIDs) (such as diclofenac, DIC) or intra-articular injected gluco-corticosteroids (such as dexamethasone, DEX) were the conventional treatment strategies for OA to reduce joint pain. Current limitations for both drugs including severe adverse effects with risks of toxicity were noted. The aim of the present study was to generate a novel OA treatment formulation hyaluronic acid (HA)-Liposomal (Lipo)-DIC/DEX to combat joint pain. The formulation was prepared by constructing DIC with DEX-loaded nanostructured lipid carriers Lipo-DIC/DEX mixed with hyaluronic acid (HA) for prolonged OA application. The prepared Lipo-DIC/DEX nanoparticles revealed the size as 103.6 ± 0.3 nm on average, zeta potential as -22.3 ± 4.6 mV, the entrapment efficiency of 90.5 ± 5.6%, and the DIC and DEX content was 22.5 ± 4.1 and 2.5 ± 0.6%, respectively. Evidence indicated that HA-Lipo-DIC/DEX could reach the effective working concentration in 4 h and sustained the drug-releasing time for at least 168 h. No significant toxicities but increased cell numbers were observed when HA-Lipo-DIC/DEX co-cultured with articular chondrocytes cells. Using live-animal In vivo imaging system (IVIS), intra-articular injection of each HA-Lipo-DIC/DEX sufficed to reduce knee joint inflammation in OA mice over a time span of four weeks. Single-dose injection could reduce the inflammation volume down to 77.5 ± 5.1% from initial over that time span. Our results provided the novel drug-releasing formulation with safety and efficiency which could be a promising system for osteoarthritis pain control.


Asunto(s)
Dexametasona/administración & dosificación , Diclofenaco/química , Ácido Hialurónico/química , Liposomas , Nanopartículas/química , Animales , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Cinética , Elastasa de Leucocito/metabolismo , Ratones , Estructura Molecular , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo
3.
Int J Mol Sci ; 22(8)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33918011

RESUMEN

Nanoliposomes are one of the leading potential nano drug delivery systems capable of targeting chemotherapeutics to tumor sites because of their passive nano-targeting capability through the enhanced permeability and retention (EPR) effect for cancer patients. Recent advances in nano-delivery systems have inspired the development of a wide range of nanotargeted materials and strategies for applications in preclinical and clinical usage in the cancer field. Nanotargeted 188Re-liposome is a unique internal passive radiotheranostic agent for nuclear imaging and radiotherapeutic applications in various types of cancer. This article reviews and summarizes our multi-institute, multidiscipline, and multi-functional studied results and achievements in the research and development of nanotargeted 188Re-liposome from preclinical cells and animal models to translational clinical investigations, including radionuclide nanoliposome formulation, targeted nuclear imaging, biodistribution, pharmacokinetics, radiation dosimetry, radiation tumor killing effects in animal models, nanotargeted radionuclide and radio/chemo-combination therapeutic effects, and acute toxicity in various tumor animal models. The systemic preclinical and clinical studied results suggest 188Re-liposome is feasible and promising for in vivo passive nanotargeted radionuclide theranostics in future cancer care applications.


Asunto(s)
Liposomas , Nanopartículas , Radioisótopos , Radiofármacos , Renio , Investigación Biomédica Traslacional , Animales , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Liposomas/química , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/mortalidad , Neoplasias/terapia , Evaluación de Resultado en la Atención de Salud , Radiometría , Radiofármacos/química , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Nanomedicina Teranóstica/métodos , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Investigación Biomédica Traslacional/métodos
4.
Int J Mol Sci ; 22(9)2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066508

RESUMEN

Ovarian cancer (OC) metastases frequently occur through peritoneal dissemination, and they contribute to difficulties in treatment. While photodynamic therapy (PDT) has the potential to treat OC, its use is often limited by tissue penetration depth and tumor selectivity. Herein, we combined Cerenkov radiation (CR) emitted by 18F-FDG accumulated in tumors as an internal light source and several photosensitizer (PS) candidates with matched absorption bands, including Verteporfin (VP), Chlorin e6 (Ce6) and 5'-Aminolevulinic acid (5'-ALA), to evaluate the anti-tumor efficacy. The in vitro effect of CR-induced PDT (CR-PDT) was evaluated using a cell viability assay, and the efficiency of PS was assessed by measuring the singlet oxygen production. An intraperitoneal ES2 OC mouse model was used for in vivo evaluation of CR-PDT. Positron emission tomography (PET) imaging and bioluminescence-based imaging were performed to monitor the biologic uptake of 18F-FDG and the therapeutic effect. The in vitro studies demonstrated Ce6 and VP to be more effective PSs for CR-PDT. Moreover, VP was more efficient in the generation of singlet oxygen and continued for a long time when exposed to fluoro-18 (18F). Combining CR emitted by 18F-FDG and VP treatment not only significantly suppressed tumor growth, but also prolonged median survival times compared to either monotherapy.


Asunto(s)
Fluorodesoxiglucosa F18/uso terapéutico , Neoplasias Ováricas/terapia , Fotoquimioterapia , Radiación , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Femenino , Inyecciones Intraperitoneales , Ratones Endogámicos BALB C
5.
Mol Ther ; 26(2): 404-419, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29248428

RESUMEN

Ionizing radiation therapy is a well-established method of eradicating locally advanced tumors. Here, we examined whether local RT enhanced the potency of an antigen-specific DNA vaccine, and we investigated the possible underlying mechanism. Using the HPV16 E6/E7+ syngeneic TC-1 tumor, we evaluated the combination of CTGF/E7 vaccination with local irradiation with regard to synergistic antigen-specific immunity and anti-tumor effects. Tumor-bearing mice treated with local RT (6 Gy twice weekly) and CTGF/E7 DNA vaccination exhibited dramatically increased numbers of E7-specific CD8+ cytotoxic T cell precursors, higher titers of anti-E7 Abs, and significantly reduced tumor size. The combination of local RT and CTGF/E7 vaccination also elicited abscopal effects on non-irradiated local subcutaneous and distant pulmonary metastatic tumors. Local irradiation induced the expression of high-mobility group box 1 protein (HMGB-1) in apoptotic tumor cells and stimulated dendritic cell (DC) maturation, consequently inducing antigen-specific immune responses. Additionally, local irradiation eventually increased the effector-to-suppressor cell ratio in the tumor microenvironment. Overall, local irradiation enhanced the antigen-specific immunity and anti-tumor effects on local and distant metastatic tumors generated by an antigen-specific DNA vaccine. These findings suggest that the combination of irradiation with antigen-specific immunotherapy is a promising new clinical strategy for cancer therapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Inmunoterapia/métodos , Ratones , Neoplasias/genética , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Microambiente Tumoral/genética , Irradiación Corporal Total , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Pharmaceuticals (Basel) ; 17(3)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38543093

RESUMEN

Chitinase-3-like protein-1 (CHI3L1), also known as YKL40, is a glycoprotein that belongs to the chitinase protein family. It is involved in various biological functions, including cell proliferation and tissue remodeling, with inflammatory and immunomodulatory capabilities. Several studies have shown that CHI3L1(YKL40) is upregulated in various diseases, such as cancer, asthma, and inflammatory bowel disease, among others. Although the expression level of CHI3L1(YKL40) is associated with disease activity, severity, and prognosis, its potential as a therapeutic target is still under investigation. In this review, we summarize the biological functions, pathological roles, and potential clinical applications of specific inhibitors and targeted therapies related to CHI3L1(YKL40).

7.
Gynecol Oncol ; 131(1): 63-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23917082

RESUMEN

OBJECTIVE: The ovarian cancer-associated ascites is an ideal material for evaluating the interaction between the host immune system and cancer cells in the tumor micro-environment. The aim of this study was to investigate whether the selected target cytokine expression levels in ascites could serve as an immune biomarker for predicting outcomes in ovarian cancer. METHODS: Eighty-eight specimens of ovarian cancer-associated ascites were evaluated to select the target cytokine by a cytokine profiling kit. The 144 total samples were subsequently analyzed for this target cytokine. The correlation between the target cytokine and clinical characteristics was analyzed. RESULTS: Interferon-gamma (IFN-γ) was identified as the target cytokine. Higher levels of IFN-γ in the ascites of the tumor micro-environment were associated with advanced disease (p=0.012), higher tumor histological grading (p=0.004), and sub-optimal surgical status (p=0.040). By multivariate analysis, the adjusted hazard ratios (HRs) were 2.74 (95% confidence interval (CI) 1.85-4.05, p<0.001) for disease-free survival (DFS) and 1.72 (95% CI 1.01-2.93, p=0.048) for overall survival (OS) for a 10-fold increase in IFN-γ concentration in the ascites. An inverse dose-response relationship between IFN-γ level and survival was also noted (Ptrend<0.001 for DFS and Ptrend<0.042 for OS). CONCLUSIONS: Patients with ovarian cancer and higher IFN-γ expression levels in cancer-associated ascites will have shorter DFS and OS. IFN-γ levels in the ascites may be a prognostic marker and a potential reference for immunotherapy targeting IFN-γ.


Asunto(s)
Ascitis/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma/inmunología , Carcinoma/patología , Interferón gamma/metabolismo , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Adulto , Anciano , Ascitis/etiología , Carcinoma/complicaciones , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Neoplasias Ováricas/complicaciones , Modelos de Riesgos Proporcionales
8.
Biochem J ; 442(2): 293-302, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21999204

RESUMEN

Ovarian cancer has one of the highest mortalities in malignancies in women, but little is known of its tumour progression properties and there is still no effective molecule that can monitor its growth or therapeutic responses. MSLN (mesothelin), a secreted protein that is overexpressed in ovarian cancer tissues with a poor clinical outcome, has been previously identified to activate PI3K (phosphoinositide 3-kinase)/Akt signalling and inhibit paclitaxel-induced apoptosis. The present study investigates the correlation between MSLN and MMP (matrix metalloproteinase)-7 in the progression of ovarian cancer, and the mechanism of MSLN in enhancing ovarian cancer invasion. The expression of MSLN correlated well with MMP-7 expression in human ovarian cancer tissues. Overexpressing MSLN or ovarian cancer cells treated with MSLN showed enhanced migration and invasion of cancer cells through the induction of MMP-7. MSLN regulated the expression of MMP-7 through the ERK (extracellular-signal-regulated kinase) 1/2, Akt and JNK (c-Jun N-terminal kinase) pathways. The expression of MMP-7 and the migrating ability of MSLN-treated ovarian cancer cells were suppressed by ERK1/2- or JNK-specific inhibitors, or a decoy AP-1 (activator protein 1) oligonucleotide in in vitro experiments, whereas in vivo animal experiments also demonstrated that mice treated with MAPK (mitogen-activated protein kinase)/ERK- or JNK-specific inhibitors could decrease intratumour MMP-7 expression, delay tumour growth and extend the survival of the mice. In conclusion, MSLN enhances ovarian cancer invasion by MMP-7 expression through the MAPK/ERK and JNK signal transduction pathways. Blocking the MSLN-related pathway could be a potential strategy for inhibiting the growth of ovarian cancer.


Asunto(s)
Proteínas Ligadas a GPI/metabolismo , Metaloproteinasa 7 de la Matriz/biosíntesis , Neoplasias Ováricas/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Cartilla de ADN/genética , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 7 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Mesotelina , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/genética , Invasividad Neoplásica/fisiopatología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Interferente Pequeño/genética , Factor de Transcripción AP-1/metabolismo
9.
Clin Sci (Lond) ; 123(6): 347-60, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22264240

RESUMEN

The aim of the present study was to investigate whether CBSCs [(umbilical) cord blood stem cells] can be a new source of DCs (dendritic cells), which can generate more potent antigen-specific immune responses and anti-tumour effects. CBSCs and PBMCs (peripheral blood mononuclear cells) were collected, cultured and differentiated into DCs. Surface markers, secreting cytokines, antigen-presentation activity, antigen-specific cell-mediated immunity and cytotoxic killing effects induced by these two DC origins were evaluated and compared. CBSCs were expanded ~17-fold by ex vivo culture. The expression of surface markers in CBSC-derived DCs were higher than those in PBMC-derived DCs treated with LPS (lipopolysaccharide). The CBSC-derived DCs mainly secreted IL (interleukin)-6, IL-10 and TNF (tumour necrosis factor)-α, whereas PBMC-derived DCs mainly secreted IL-5 and IFN (interferon)-γ. The CBSC-derived DCs had better antigen-presentation abilities when stimulated with LPS or TNF-α, induced higher numbers of IFN-γ-secreting antigen-specific CD8+ T-cells, as assessed using an ELISpot (enzyme-linked immunosorbent spot) assay, and stimulated more potent antigen-specific CTL (cytotoxic T-cell) activities (P<0.01, one-way ANOVA). CBSC-derived DCs had quicker and greater ERK (extracellular-signal-regulated kinase) and Akt phosphorylation, and weaker p38 phosphorylation, than PBMC-derived DCs when stimulated with LPS. In conclusion, CBSC-derived DCs have the ability to induce stronger antigen-specific immunity and more potent anti-tumour effects and therefore could be a good source of DCs for use in DC-based cancer vaccines and immunotherapy.


Asunto(s)
Células Dendríticas/citología , Sangre Fetal/citología , Inmunidad Celular , Neoplasias/terapia , Células Madre/citología , Presentación de Antígeno , Diferenciación Celular , Línea Celular Tumoral , Medios de Cultivo , Citocinas/metabolismo , Células Dendríticas/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sangre Fetal/inmunología , Células HeLa , Humanos , Inmunoterapia , Células K562 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células Madre/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Biomed Pharmacother ; 155: 113668, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36088858

RESUMEN

Epithelial ovarian cancer (EOC) is usually diagnosed at an advanced stage and has poor prognosis. Theranostic agents are the current trend in drug development, but are lacking in EOC. YKL40 is predominantly expressed and involved in tumorigenesis in EOC. In this study, we developed a companion theranostic agent targeting YKL40. We measured YKL40 expression levels in ascites using ELISA and correlated them with the clinical outcomes of patients with EOC. We developed radionuclide labeled In-111/Lu-177-DTPA-YKL40 neutralizing antibodies and investigated their radiochemical purity, SPECT/CT imaging, bio-distribution, and therapeutic responses in ovarian cancer xenograft mice. We demonstrated that YKL40 expression levels in ascites were significantly higher in EOC patients with serous histological type, high tumor grade, advanced stage, tumor recurrence, chemoresistance, and tumor-related death. The radiochemical purity of In-111/Lu-177-DTPA-YKL40 neutralizing antibodies reached more than 90% after 24 h of labeling. SPECT/CT imaging showed significant accumulation of In-111-DTPA-YKL40 and Lu-177-DTPA-YKL40 antibodies at the tumor site of ovarian cancer xenograft mice 24 h after administration. Lu-177-DTPA-YKL40 antibodies significantly inhibited tumor growth in ovarian cancer xenograft mice. Our study indicated that In-111/Lu-177-DTPA-YKL40 neutralizing antibodies could be potential companion theranostic agents for patients with EOC.


Asunto(s)
Neoplasias Ováricas , Radiofármacos , Femenino , Humanos , Animales , Ratones , Carcinoma Epitelial de Ovario/patología , Proteína 1 Similar a Quitinasa-3 , Ascitis , Medicina de Precisión , Recurrencia Local de Neoplasia , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Radioisótopos , Ácido Pentético/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Línea Celular Tumoral
11.
Mol Ther ; 18(6): 1233-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20372107

RESUMEN

Although cancer vaccines are emerging as innovative methods for cancer treatment, these alone have limited potential for treating measurable tumor burden. Thus, the importance of identifying anticancer strategies with greater potency is necessary. The chimeric DNA vaccine CTGF/E7 (connective tissue growth factor linked to the tumor antigen human papillomavirus 16 E7) generates potent E7-specific immunity and antitumor effects. We tested immune-modulating doses of chemotherapy in combination with the CTGF/E7 DNA vaccine to treat existing tumors in mice. Metronomic low doses of paclitaxel, not the maximal tolerable dose, are synergistic with the antigen-specific DNA vaccine. Paclitaxel, given in metronomic sequence with the CTGF/E7 DNA vaccine enhanced the vaccine's potential to delay tumor growth and decreased metastatic tumors in vivo better than the CTGF/E7 DNA vaccine alone. The two possible mechanisms of metronomic paclitaxel chemotherapy are the depletion of regulatory T cells and the inhibition of tumor angiogenesis rather than direct cancer cell cytolytic effects. Results indicate that combination treatment of metronomic chemotherapy and antigen-specific DNA vaccine can induce more potent antigen-specific immune responses and antitumor effects. This provides an immunologic basis for further testing in cancer patients.


Asunto(s)
Antineoplásicos/administración & dosificación , Vacunas contra el Cáncer , Depleción Linfocítica , Neoplasias/terapia , Neovascularización Patológica , Linfocitos T Reguladores/inmunología , Animales , Humanos , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación
12.
J Women Aging ; 23(3): 233-45, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21767087

RESUMEN

PURPOSE: This study investigated the effects of gender differences on the association of chronic stress and depressive symptoms in middle-aged and older adults in Taiwan. METHODS: The population base was of adults aged 50 and older in Taiwan. This study included 2,889 participants and examined the gender differences on the impacts of life stress that exhibited depressive symptoms. RESULTS: Females were more susceptible to depressive symptoms when they felt constant stress from finances, increasing stress from jobs, and fluctuating stress from family relationships. DISCUSSION: Gender differences were evident when assuming social roles, as were psychological susceptibilities.


Asunto(s)
Depresión/etiología , Estrés Psicológico/complicaciones , Anciano , Relaciones Familiares , Femenino , Estado de Salud , Humanos , Masculino , Persona de Mediana Edad , Factores Sexuales , Factores Socioeconómicos , Taiwán
13.
Biochem J ; 424(3): 449-58, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19747165

RESUMEN

Mesothelin, a secreted protein, is overexpressed in some cancers, but its exact function remains unclear. The aim of the present study was to evaluate the possible function of mesothelin. Real-time PCR, RT (reverse transcription)-PCR, cytotoxicity assays, proliferative assays, apoptotic assays by Hoechst staining, detection of active caspases 3 and 7 by flow cytometric analysis, and immunoprecipitation and immunoblotting were performed. Cancer tissues in paclitaxel-resistant ovarian cancer patients expressed higher levels of mesothelin as assessed using real-time PCR than paclitaxel-sensitive ovarian cancer patients (the mean crossing point value change of mesothelin was 26.9+/-0.4 in the resistant group and 34.3+/-0.7 for the sensitive group; P<0.001). Mesothelin also protected cells from paclitaxel-induced apoptosis. The protein expression of Bcl-2 family members, such as Bcl-2 and Mcl-1, was significantly increased regardless of whether cells were treated with exogenous mesothelin or were mesothelin-transfectants. Furthermore, mesothelin-treated cells revealed rapid tyrosine phosphorylation of the p85 subunit of PI3K (phosphoinositide 3-kinase) and ERK (extracellular-signal-regulated kinase) 1/2 for enhancing MAPK (mitogen-activated protein kinase) activity. The anti-apoptotic ability was suppressed and the expression of Bcl-2 family in response to mesothelin was altered by inhibiting PI3K activity, but not by inhibiting MAPK activity. Thus mesothelin can inhibit paclitaxel-induced cell death mainly by involving PI3K signalling in the regulation of Bcl-2 family expression. Mesothelin is a potential target in reducing resistance to cytotoxic drugs.


Asunto(s)
Apoptosis/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Paclitaxel/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , Resistencia a Antineoplásicos , Femenino , Proteínas Ligadas a GPI , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Mesotelina , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
14.
Biomed Phys Eng Express ; 6(4): 045017, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-33444277

RESUMEN

Eye drops are a conventional method of drug delivery to the eye, accounting for 90% of currently accessible ophthalmic formulations. The major problem with eye drop treatments is rapid pre-corneal drug loss. Furthermore, the need for frequent administration of eye drops can profoundly affect the quality of life of ophthalmological patients. In the current study, we developed a liposomal nanoparticle encapsulated with chloramphenicol mixed with biodegradable materials against ophthalmological disease. We first established a protocol for chloramphenicol (CAP) loaded into liposomal nanoparticle (LipoCAP). We also established the collagen/gelatin/sodium alginate (CGA) as the component of biodegradable polymers and calibrated the novel drug-releasing formulation. Finally, we combined LipoCAP with CGA to generate an 8-h degradable ophthalmic chloramphenicol gel, CGA-LipoCAP-8. CGA-LipoCAP-8 reached the effective working concentration in 75 min and prolonged the drug-releasing time for at least 12 h. In addition, CGA-LipoCAP-8 could stably and continuously inhibit E. coli proliferation. The inhibiting phenomenon was more pronounced over time. Furthermore, there were no significant toxicities observed when CGA-LipoCAP-8 co-cultured with ocular epithelial cells. In conclusion, CGA-LipoCAP-8 achieved effective CAP dose concentrations in a short time and sustained CAP release for a prolonged period. Our results provide an innovative concept in relation to novel drug-release formulations, with safety and efficiency supporting use in future treatments for ophthalmological diseases.


Asunto(s)
Administración Oftálmica , Alginatos/química , Materiales Biocompatibles/química , Cloranfenicol/administración & dosificación , Colágeno/química , Ojo/efectos de los fármacos , Gelatina/química , Liposomas/química , Antibacterianos/administración & dosificación , Bacillus , Calibración , Córnea/efectos de los fármacos , Medios de Cultivo , Sistemas de Liberación de Medicamentos/métodos , Células Epiteliales/efectos de los fármacos , Escherichia coli/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Transmisión , Soluciones Oftálmicas , Polímeros/química , Reproducibilidad de los Resultados
15.
Biomed Mater ; 15(5): 055022, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32434164

RESUMEN

Infectious keratitis is still one of the major causes of visual impairment and blindness, often affecting developing countries. Eye-drop therapy to reduce disease progression is the first line of treatment for infectious keratitis. The current limitations in controlling ophthalmic infections include rapid precorneal drug loss and the inability to provide long-term extraocular drug delivery. The aim of the present study was to develop a novel ophthalmic formulation to treat corneal infection. The formulation was prepared by constructing moxifloxacin (MFX) and dexamethasone (DEX)-loaded nanostructured lipid carriers (Lipo-MFX/DEX) mixed with a collagen/gelatin/alginate (CGA) biodegradable material (CGA-Lipo-MFX/DEX) for prolonged ocular application. The characteristics of the prepared Lipo-MFX/DEX nanoparticles were as follows: average size, 132.1 ± 73.58 nm; zeta potential, -6.27 ± 4.95 mV; entrapment efficiency, 91.5 ± 3.5%; drug content, 18.1 ± 1.7%. Our results indicated that CGA-Lipo-MFX/DEX could release an effective working concentration in 60 min and sustain the drug release for at least 12 h. CGA-Lipo-MFX/DEX did not produce significant toxicities, but it increased cell numbers when co-cultured with ocular epithelial cells. An animal study also confirmed that CGA-Lipo-MFX/DEX could inhibit pathogen microorganism growth and improve corneal wound healing. Our results suggest that CGA-Lipo-MFX/DEX could be a useful anti-inflammatory formulation for ophthalmological disease treatment.


Asunto(s)
Alginatos/química , Colágeno/química , Córnea/efectos de los fármacos , Enfermedades de la Córnea/tratamiento farmacológico , Dexametasona/administración & dosificación , Gelatina/química , Hidrogeles , Liposomas/química , Moxifloxacino/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Bacillus , Materiales Biocompatibles/química , Sistemas de Liberación de Medicamentos , Edema/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Escherichia coli , Humanos , Inflamación/tratamiento farmacológico , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Factores de Tiempo
16.
Toxicology ; 256(1-2): 83-91, 2009 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19070643

RESUMEN

Morphine has been widely used for pain management. Other than analgesia, it has effects on vascular endothelial cells, including angiogenesis and apoptosis. An in vitro model of human umbilical vein endothelial cells (HUVECs) was made to investigate the effects and comprehensive mechanisms of morphine on vascular endothelial cells. Morphine enhanced apoptosis of HUVECs, increased intracellular reactive oxygen species (ROS), and reduced mitochondrial membrane potentials (MMPs). It also induced the release of NO and activated NF-kappaB in HUVECs. Naloxone, the opioid receptor antagonist, could reverse cell apoptosis and ROS generation, NO production, and MMP loss. Expression levels of Bak and Bax, and the activation of caspases 3 and 7 in HUVECs significantly increased when treated with morphine. Inhibition of NO production by NO synthase inhibitor reduced morphine-induced apoptosis. Morphine could induce apoptosis of HUVECs through both the NO and ROS pathways. Thus, inhibiting NO or ROS may be a potential target in blocking morphine-induced apoptosis of endothelial cells.


Asunto(s)
Analgésicos Opioides/farmacología , Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Morfina/farmacología , Óxido Nítrico/fisiología , Especies Reactivas de Oxígeno/metabolismo , Western Blotting , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/metabolismo , Inmunoprecipitación , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , FN-kappa B/metabolismo
17.
Cancers (Basel) ; 11(2)2019 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-30823599

RESUMEN

Epithelial ovarian cancer patients usually relapse after primary management. We utilized the support vector machine algorithm to develop a model for the chemo-response using the Cancer Cell Line Encyclopedia (CCLE) and validated the model in The Cancer Genome Atlas (TCGA) and the GSE9891 dataset. Finally, we evaluated the feasibility of the model using ovarian cancer patients from our institute. The 10-gene predictive model demonstrated that the high response group had a longer recurrence-free survival (RFS) (log-rank test, p = 0.015 for TCGA, p = 0.013 for GSE9891 and p = 0.039 for NTUH) and overall survival (OS) (log-rank test, p = 0.002 for TCGA and p = 0.016 for NTUH). In a multivariate Cox hazard regression model, the predictive model (HR: 0.644, 95% CI: 0.436⁻0.952, p = 0.027) and residual tumor size < 1 cm (HR: 0.312, 95% CI: 0.170⁻0.573, p < 0.001) were significant factors for recurrence. The predictive model (HR: 0.511, 95% CI: 0.334⁻0.783, p = 0.002) and residual tumor size < 1 cm (HR: 0.252, 95% CI: 0.128⁻0.496, p < 0.001) were still significant factors for death. In conclusion, the patients of high response group stratified by the model had good response and favourable prognosis, whereas for the patients of medium to low response groups, introduction of other drugs or clinical trials might be beneficial.

18.
Epigenomics ; 10(11): 1397-1413, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30324802

RESUMEN

AIM: To investigate the CDH1, DLEC1 and SFRP5 gene methylation panel for advanced epithelial ovarian carcinoma (EOC). MATERIALS & METHODS: One hundred and seventy-seven advanced EOC specimens were evaluated by methylation-specific PCR. We also used The Cancer Genome Atlas dataset to evaluate the panel. RESULTS: The presence of two or more methylated genes was significant in recurrence (hazard ratio [HR]: 1.91 [1.33-2.76]; p = 0.002) and death (HR: 1.96 [1.26-3.06]; p = 0.006) in our cohort. In The Cancer Genome Atlas dataset, the presence of two or three methylated genes was significant in death (HR: 1.59 [1.15-2.18]; p = 0.0047) and close to the significance level in recurrence (HR: 1.37 [0.99-1.88]; p = 0.058). CONCLUSION: The CDH1, DLEC1 and SFRP5 methylation panel is a potential prognostic biomarker for advanced EOC.


Asunto(s)
Antígenos CD/genética , Biomarcadores de Tumor/genética , Cadherinas/genética , Carcinoma Epitelial de Ovario/genética , Metilación de ADN , Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Neoplasias Ováricas/genética , Proteínas Supresoras de Tumor/genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Carcinoma Epitelial de Ovario/patología , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/patología , Análisis de Supervivencia
19.
Cancer Lett ; 425: 152-163, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29596890

RESUMEN

As a tumor antigen, mesothelin (MSLN) can be identified in various malignancies. MSLN is potential for antigen-specific cancer vaccines. We generated a novel chimeric DNA vaccine using antigen-specific connective tissue growth factor lined with MSLN (CTGF/MSLN). The anti-tumor effects of the CTGF/MSLN DNA vaccine combined with anti-CD40 Ab and toll-like receptor 3 ligand-poly(I:C) were validated in an MSLN-expressing model. CTGF/MSLN DNA with anti-CD40Ab and poly(I:C) vaccinated mice demonstrated potent anti-tumor effects with longer survival and less tumor volumes. An increase in MSLN-specific CD8+ T cells and anti-MSLN Ab titers was also noted in CTGF/MSLN DNA with anti-CD40Ab and poly(I:C) vaccinated mice. The CTGF/MSLN DNA vaccine combined with immuno-modulator EGCG also generated potent anti-tumor effects. Immuno-modulators could enhance the antigen-specific anti-tumor effects of CTGF/MSLN DNA vaccine through promoting the DC maturation. In addition, MSLN-specific cell-based vaccine with AAV-IL-12 and the CTGF/MSLN DNA vaccine with anti-CD40Ab/polyp(I:C) generated more potent anti-tumor effects than the other combinational regimens. The results indicate that an MSLN-specific DNA vaccine combined with immuno-modulators may be an effective immunotherapeutic strategy to control MSLN-expressing tumors including ovarian and pancreastic cancers, and malignant mesothelioma.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Catequina/análogos & derivados , Factor de Crecimiento del Tejido Conjuntivo/genética , Proteínas Ligadas a GPI/genética , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Poli I-C/administración & dosificación , Vacunas de ADN/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Catequina/administración & dosificación , Catequina/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Sinergismo Farmacológico , Femenino , Neoplasias Pulmonares/inmunología , Mesotelina , Mesotelioma/inmunología , Mesotelioma Maligno , Ratones , Poli I-C/inmunología , Resultado del Tratamiento , Vacunas de ADN/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Gynecol Oncol ; 107(3): 404-12, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17905417

RESUMEN

OBJECTIVE: Vaccination based on tumor antigen is an attractive strategy for cancer prevention and therapy. Cervical cancer is highly associated with human papillomavirus, especially type 16. We developed DNA vaccines encoding heat shock protein 60 (HSP60) linked to HPV16 E6 or E7 to test if HSP60 chimeric DNA vaccines may generate strong E6 and/or E7-specific immune response and anti-tumor effects in vaccinated mice. METHODS: In vivo antitumor effects such as preventive, therapeutic, and antibody depletion experiments were performed. In vitro assays such as intracellular cytokine stainings, ELISA for Ab responses, and direct and cross-priming effects, were also performed. RESULTS: HSP60 chimeric DNA vaccines generated strong E6- or E7-specific immune responses and anti-tumor effects in vaccinated mice via direct and cross-priming effects. HSP60 was also linked with both E6 and E7 antigens and the HSP60/E6/E7 chimeric DNA vaccine generated more potent immunotherapeutic effects on E6- and E7-expressing tumors than HSP60/E6 or HSP60/E7 chimeric DNA vaccine alone. CONCLUSION: Utilization of both E6 and E7 tumor antigens can advance the therapy of tumors associated with HPV-infections. The DNA vaccine encoding heat shock protein 60 co-linked to HPV16 E6 and E7 tumor antigens can generate more potent immunotherapeutic effects than E6 or E7 tumor antigens alone.


Asunto(s)
Chaperonina 60/inmunología , Proteínas Oncogénicas Virales/inmunología , Proteínas Represoras/inmunología , Vacunas de ADN/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Chaperonina 60/genética , ADN/administración & dosificación , ADN/genética , ADN/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Proteínas Represoras/genética , Linfocitos T Citotóxicos/inmunología , Transfección , Vacunas de ADN/genética , Vacunas de ADN/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA