Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Apoptosis ; 16(4): 370-81, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21197579

RESUMEN

Multiple myelomas (MMs) are etiologically heterogeneous and there are limited treatment options; indeed, current monoclonal antibody therapies have had limited success, so more effective antibodies are urgently needed. Polyclonal antibodies are a possible alternative because they target multiple antigens simultaneously. In this study, we produced polyclonal rabbit anti-murine plasmacytoma cell immunoglobulin (PAb) by immunizing rabbits with the murine plasmacytoma cell line MPC-11. The isolated PAb bound to plasma surface antigens in several MM cell lines, inhibited their proliferation as revealed by MTT assay, and induce apoptosis as indicated by flow cytometry, microscopic observation of apoptotic changes in morphology, and DNA fragmentation on agarose gels. The cytotoxicity of PAb on MPC-11 cell lines was both dose-dependent and time-dependent; PAb exerted a 50% inhibitory effect on MPC-11 cell viability at a concentration of 200 µg/ml in 48 h. Flow cytometry demonstrated that PAb treatment significantly increased the number of apoptotic cells (48.1%) compared with control IgG (8.3%). Apoptosis triggered by PAb was confirmed by activation of caspase-3, -8, and -9. Serial intravenous or intraperitoneal injections of PAb inhibited tumour growth and prolonged survival in mice bearing murine plasmacytoma, while TUNEL assay demonstrated that PAb induced statistically significant apoptosis (P < 0.05) compared to control treatments. We conclude that PAb is an effective agent for in vitro and in vivo induction of apoptosis in multiple myeloma and that exploratory clinical trials may be warranted.


Asunto(s)
Apoptosis/efectos de los fármacos , Inmunoglobulinas/farmacología , Mieloma Múltiple/patología , Plasmacitoma/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Unión Proteica/efectos de los fármacos , Conejos
2.
AAPS J ; 22(4): 92, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32676788

RESUMEN

In this study, we characterized the pharmacokinetics of OSU-2S, a fingolimod-derived, non-immunosuppressive phosphatase activator, in mice, rats, and dogs, as well as tolerability and food effects in dogs. Across all species tested, plasma protein binding for OSU-2S was > 99.5%, and metabolic stability and hepatic intrinsic clearance were in the moderate range. OSU-2S did not significantly modulate CYP enzyme activity up until 50 µM, and Caco-2 data suggested low permeability with active efflux at 2 µM. Apparent oral bioavailability in mice was 16% and 69% at 10 and 50 mg/kg, respectively. In rats, bioavailability was 24%, 35%, and 28% at 10, 30, and 100 mg/kg, respectively, while brain/plasma ratio was 36 at 6-h post-dose at 30 mg/kg. In dogs, OSU-2S was well tolerated with oral capsule bioavailability of 27.5%. Plasma OSU-2S exposures increased proportionally over a 2.5-20 mg/kg dose range. After 4 weeks of 3 times weekly, oral administration (20 mg/kg), plasma AUClast (26.1 µM*h), and Cmax (0.899 µM) were nearly 2-fold greater than those after 1 week of dosing, and no food effects were observed. The elimination half-life (29.7 h), clearance (22.9 mL/min/kg), and plasma concentrations of repeated oral doses support a 3-times weekly dosing schedule in dogs. No significant CBC, serum biochemical, or histopathological changes were observed. OSU-2S has favorable oral PK properties similar to fingolimod in rodents and dogs and is well tolerated in healthy animals. This work supports establishing trials of OSU-2S efficacy in dogs with spontaneous tumors to guide its clinical development as a cancer therapeutic for human patients.


Asunto(s)
Análisis de Datos , Clorhidrato de Fingolimod/farmacocinética , Inmunosupresores/farmacocinética , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Perros , Relación Dosis-Respuesta a Droga , Clorhidrato de Fingolimod/administración & dosificación , Haplorrinos , Humanos , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Glicoles de Propileno/administración & dosificación , Ratas , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Esfingosina/farmacocinética
3.
Oncol Rep ; 24(3): 661-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20664971

RESUMEN

Colon carcinoma is one of the common malignant tumors and has high morbidity and mortality in the world. Pigment epithelial-derived factor (PEDF) has been found to be the most potent natural inhibitor of angiogenesis and PEDF gene has been extensively used for the therapy of tumors, which suggests a potential approach to the therapy of colon carcinoma. However, the transfer of PEDF gene largely depends on the effective gene delivery systems. Poly (lactic-co-glycolic acid) nanoparticles (PLGANPs) have been extensively used for gene therapy due to its low-toxicity, biocompatibility and biodegradability, due to its potential to be an excellent carrier of the PEDF gene. We investigated the effect of PEDF gene loaded in PLGA nanoparticles (PEDF-PLGANPs) on the mouse colon carcinoma cells (CT26s) in vitro and in vivo. Blank PLGANPs (bPLGANPs) showed lower cytotoxicity than PEI to the CT26s. In vitro, PEDF-PLGANPs directly induced CT26 apoptosis and inhibit human umbilical vein endothelial cell (HUVEC) proliferation. In vivo, PEDF-PLGANPs inhibited CT26 tumors growth by inducing CT26 apoptosis, decreasing MVD and inhibiting angiogenesis. Our present study demonstrates the inhibitory effect of PEDF-PLGANPs on the growth of CT26s in vitro and in vivo for the first time. PLGANP-mediated PEDF gene could provide an innovative strategy for the therapy of colon carcinoma.


Asunto(s)
Carcinoma/terapia , Neoplasias del Colon/terapia , Proteínas del Ojo/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Ácido Láctico/química , Nanopartículas , Factores de Crecimiento Nervioso/genética , Ácido Poliglicólico/química , Serpinas/genética , Animales , Apoptosis/efectos de los fármacos , Carcinoma/irrigación sanguínea , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Dependovirus/genética , Células Endoteliales/efectos de los fármacos , Proteínas del Ojo/biosíntesis , Vectores Genéticos , Humanos , Ácido Láctico/toxicidad , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Factores de Crecimiento Nervioso/biosíntesis , Ácido Poliglicólico/toxicidad , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Serpinas/biosíntesis , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
4.
J Exp Clin Cancer Res ; 28: 75, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19500366

RESUMEN

BACKGROUND: Angiogenesis plays an important role in tumor growth, invasion, and eventually metastasis. Antiangiogenic strategies have been proven to be a promising approach for clinical therapy for a variety of tumors. As a potent inhibitor of tumor angiogenesis, pigment epithelium-derived factor (PEDF) has recently been studied and used as an anticancer agent in several tumor models. METHODS: A recombined adenovirus carrying PEDF gene (Ad-PEDF) was prepared, and its expression by infected cells and in treated animals was confirmed with Western blotting and ELISA, respectively. Its activity for inhibiting human umbilical vein endothelial cell (HUVEC) proliferation was tested using the MTT assay. C57BL/6 mice bearing B16-F10 melanoma were treated with i.v. administration of 5 x 108 IU/mouse Ad-PEDF, or 5 x 108 IU/mouse Ad-Null, or normal saline (NS), every 3 days for a total of 4 times. Tumor volume and survival time were recorded. TUNEL, CD31 and H&E stainings of tumor tissue were conducted to examine apoptosis, microvessel density and histological morphology changes. Antiangiogenesis was determined by the alginate-encapsulated tumor cell assay. RESULTS: The recombinant PEDF adenovirus is able to transfer the PEDF gene to infected cells and successfully produce secretory PEDF protein, which exhibits potent inhibitory effects on HUVEC proliferation. Through inhibiting angiogenesis, reducing MVD and increasing apoptosis, Ad-PEDF treatment reduced tumor volume and prolonged survival times of mouse bearing B16-F10 melanoma. CONCLUSION: Our data indicate that Ad-PEDF may provide an effective approach to inhibit mouse B16-F10 melanoma growth.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/administración & dosificación , Proteínas del Ojo/genética , Terapia Genética , Melanoma Experimental/terapia , Factores de Crecimiento Nervioso/genética , Serpinas/genética , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Proliferación Celular , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Neoplasias del Colon/virología , Endotelio Vascular/metabolismo , Proteínas del Ojo/metabolismo , Femenino , Técnicas de Transferencia de Gen , Humanos , Luciferasas/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microvasos , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA