Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biomacromolecules ; 14(2): 302-10, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23327610

RESUMEN

meso-Tetra(N-methyl-4-pyridyl) porphine tetra tosylate (TMP) is a photosensitizer that can be used in photodynamic therapy (PDT) to induce cell death through generation of reactive oxygen species in targeted tumor cells. However, TMP is highly hydrophilic, and therefore, its ability to accumulate intracellularly is limited. In this study, a strategy to improve TMP uptake into cells has been investigated by encapsulating the compound in a hydrogel-based chitosan/alginate nanoparticle formulation. Nanoparticles of 560 nm in diameter entrapping 9.1 µg of TMP per mg of formulation were produced and examined in cell-based assays. These particles were endocytosed into human colorectal carcinoma HCT116 cells and elicited a more potent photocytotoxic effect than free drug. Antibodies targeting death receptor 5 (DR5), a cell surface apoptosis-inducing receptor up-regulated in various types of cancer and found on HCT116 cells, were then conjugated onto the particles. The conjugated antibodies further enhanced uptake and cytotoxic potency of the nanoparticle. Taken together, these results show that antibody-conjugated chitosan/alginate nanoparticles significantly enhanced the therapeutic effectiveness of entrapped TMP. This novel approach provides a strategy for providing targeted site-specific delivery of TMP and other photosensitizer drugs to treat colorectal tumors using PDT.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/uso terapéutico , Alginatos , Anticuerpos/inmunología , Transporte Biológico , Caspasa 8/metabolismo , Línea Celular Tumoral , Quitosano/inmunología , Ácido Glucurónico/inmunología , Ácidos Hexurónicos/inmunología , Humanos , Nanopartículas , Fotoquimioterapia , Interferencia de ARN , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología
2.
Biochimie ; 94(2): 487-93, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21896304

RESUMEN

Cathepsin S is a lysosomal cysteine protease implicated in tumourigenesis with key roles in invasion and angiogenesis. We have previously shown that the specific inhibition of Cathepsin S using a monoclonal antibody (Fsn0503) blocks colorectal carcinoma tumour growth and angiogenesis in vivo. We investigated whether Cathepsin S expression levels were affected by chemotherapy in human cancer cell lines by RT-PCR. Using colorectal xenograft models, we examined the therapeutic benefit of Cathepsin S inhibition using Fsn0503 in combination with a metronomic dosing regimen of CPT-11. We analysed the effects of the combination therapy on tumour progression and on tumour vascularisation by immunohistochemical staining of tumours. Cathepsin S expression levels are upregulated in HCT116, LoVo, Colo205 cell lines and HUVECs after exposure to CPT-11 in vitro. The administration of Fsn0503 in combination with CPT-11 significantly attenuated tumour growth in comparison to CPT-11 alone in colorectal HCT116 xenograft models. Furthermore, analysis of tumour vascularisation revealed that this was also significantly disrupted by the combination treatment. These results show that the combination of Cathepsin S inhibition with CPT-11 enhances the therapeutic effect of the chemotherapy. This rationale may have clinical application in the treatment of colorectal cancer upon further evaluation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Catepsinas/antagonistas & inhibidores , Colon/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Catepsinas/genética , Catepsinas/metabolismo , Línea Celular Tumoral , Colon/efectos de los fármacos , Colon/patología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Inhibidores de Cisteína Proteinasa/administración & dosificación , Expresión Génica/efectos de los fármacos , Humanos , Irinotecán , Ratones , Ratones Desnudos , Neovascularización Patológica , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Cancer ; 10: 147, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22168338

RESUMEN

BACKGROUND: Proteolytic enzymes have been implicated in driving tumor progression by means of their cancer cell microenvironment activity where they promote proliferation, differentiation, apoptosis, migration, and invasion. Therapeutic strategies have focused on attenuating their activity using small molecule inhibitors, but the association of proteases with the cell surface during cancer progression opens up the possibility of targeting these using antibody dependent cellular cytotoxicity (ADCC). Cathepsin S is a lysosomal cysteine protease that promotes the growth and invasion of tumour and endothelial cells during cancer progression. Our analysis of colorectal cancer patient biopsies shows that cathepsin S associates with the cell membrane indicating a potential for ADCC targeting. RESULTS: Here we report the cell surface characterization of cathepsin S and the development of a humanized antibody (Fsn0503h) with immune effector function and a stable in vivo half-life of 274 hours. Cathepsin S is expressed on the surface of tumor cells representative of colorectal and pancreatic cancer (23%-79% positive expression). Furthermore the binding of Fsn0503h to surface associated cathepsin S results in natural killer (NK) cell targeted tumor killing. In a colorectal cancer model Fsn0503h elicits a 22% cytotoxic effect. CONCLUSIONS: This data highlights the potential to target cell surface associated enzymes, such as cathepsin S, as therapeutic targets using antibodies capable of elicitingADCC in tumor cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Catepsinas/inmunología , Citotoxicidad Inmunológica , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacocinética , Catepsinas/química , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Femenino , Citometría de Flujo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Masculino , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Ratas , Ratas Sprague-Dawley
4.
PLoS One ; 5(9)2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20824056

RESUMEN

BACKGROUND: Angiogenesis is a key hallmark of tumourigenesis and its inhibition is a proven strategy for the development of novel anti-cancer therapeutics. An important aspect of early angiogenesis is the co-ordinated migration and invasion of endothelial cells through the hypoxic tumour tissue. Cathepsin S has been shown to play an important role in angiogenesis as has vascular endothelial growth factor (VEGF). We sought to assess the anti-angiogenic effect of Fsn0503, a novel cathepsin S inhibitory antibody, when combined with anti-VEGF on vascular development. METHODOLOGY/PRINCIPAL FINDINGS: Cathepsin S expression and secretion from endothelial cells was characterised using RT-PCR and western blotting. We further show that cathepsin S promotes pericellular hydrolysis of extracellular matrix components in the tumour microenvironment and facilitates endothelial invasion. The cathepsin S inhibitory antibody, Fsn0503, blocks extracellular proteolysis, inhibiting endothelial invasion and tube formation in cell-based assays. The anti-angiogenic effects of Fsn0503 were also shown in vivo where it significantly retarded the development of vasculature in human xenograft models. Furthermore, when Fsn0503 was combined with an anti-VEGF antibody, a synergistic inhibition of microvascular development was observed. CONCLUSIONS/SIGNIFICANCE: Taken together, this data demonstrates that the antibody-mediated targeting of cathepsin S represents a novel method of inhibiting angiogenesis. Furthermore, when used in combination with anti-VEGF therapies, Fsn0503 has the potential to significantly enhance current treatments of tumour neovascularisation and may also be of use in the treatment of other conditions associated with inappropriate angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Anticuerpos/administración & dosificación , Catepsinas/antagonistas & inhibidores , Catepsinas/metabolismo , Regulación hacia Abajo , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/inmunología , Inhibidores de la Angiogénesis/farmacología , Animales , Anticuerpos/farmacología , Catepsinas/inmunología , Línea Celular , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/metabolismo
5.
Clin Cancer Res ; 15(19): 6042-51, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19789302

RESUMEN

PURPOSE: Cathepsin S is a cysteine protease that promotes the invasion of tumor and endothelial cells during cancer progression. Here we investigated the potential to target cathepsin S using an antagonistic antibody, Fsn0503, to block these tumorigenic effects. EXPERIMENTAL DESIGN: A panel of monoclonal antibodies was raised to human cathepsin S. The effects of a selected antibody were subsequently determined using invasion and proteolysis assays. Endothelial cell tube formation and aorta sprouting assays were done to examine antiangiogenic effects. In vivo effects were also evaluated using HCT116 xenograft studies. RESULTS: A selected cathepsin S antibody, Fsn0503, significantly blocked invasion of a range of tumor cell lines, most significantly HCT116 colorectal carcinoma cells, through inhibition of extracellular cathepsin S-mediated proteolysis. We subsequently found enhanced expression of cathepsin S in colorectal adenocarcinoma biopsies when compared with normal colon tissue. Moreover, Fsn0503 blocked endothelial cell capillary tube formation and aortic microvascular sprouting. We further showed that administration of Fsn0503 resulted in inhibition of tumor growth and neovascularization of HCT116 xenograft tumors. CONCLUSIONS: These results show that blocking the invasive and proangiogenic effects of cathepsin S with antibody inhibitors may have therapeutic utility upon further preclinical and clinical evaluation.


Asunto(s)
Adenocarcinoma/patología , Anticuerpos Monoclonales/farmacología , Catepsinas/antagonistas & inhibidores , Neoplasias Colorrectales/patología , Neovascularización Patológica/prevención & control , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Catepsinas/inmunología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/terapia , Femenino , Células HCT116 , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Ratas , Ratas Wistar , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cell Death ; 2: 45-51, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-26124680

RESUMEN

The study of death receptor family induced apoptosis has gained momentum in recent years with the knowledge that therapeutic antibodies targeting DR4 and DR5 (death receptor's 4 and 5) have proved efficacious in multiple clinical trials. The therapeutic rationale is based on targeting and amplifying a tumour tissues normal cell death programme (apoptosis). While advances in the targeting of DR4 and DR5 have been successful the search for an agonistic antibody to another family member, the Fas receptor, has proven more elusive. This is partly due to the differing in vitro and in vivo characteristics of individual antibodies. In order to induce Fas targeted cell death an antibody must be capable of binding to and trimerising the receptor. It has been shown that antibodies capable of performing this function in vivo, with the assistance of tumour associated cells, do not always induce apoptosis in vitro. As a result the use of current methodologies to detect functional antibodies in vitro may have dismissed potential therapeutic candidates ('false negative'). Here we report a novel high throughput screening technique which artificially cross-links antibodies bound to the Fas receptor. By combining this process with Annexin-V and Prodidium Iodide (PI) staining we can select for antibodies which have the potential to induce apoptosis in vivo.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA