Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Virol ; 87(18): 10283-94, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864625

RESUMEN

Vesicular stomatitis virus (VSV) is a promising oncolytic agent against various malignancies. Here, for the first time, we tested VSV in vitro and in vivo in a clinically relevant, immunocompetent mouse model of pancreatic ductal adenocarcinoma (PDA). Our system allows the study of virotherapy against PDA in the context of overexpression (80% of PDA patients) or no expression of human mucin 1 (MUC1), a major marker for poor prognosis in patients. In vitro, we tested three VSV recombinants, wild-type VSV, VSV-green fluorescent protein (VSV-GFP), and a safe oncolytic VSV-ΔM51-GFP, against five mouse PDA cell lines that either expressed human MUC1 or were MUC1 null. All viruses demonstrated significant oncolytic abilities independent of MUC1 expression, although VSV-ΔM51-GFP was somewhat less effective in two PDA cell lines. In vivo administration of VSV-ΔM51-GFP resulted in significant reduction of tumor growth for tested mouse PDA xenografts (+MUC1 or MUC1 null), and antitumor efficacy was further improved when the virus was combined with the chemotherapeutic drug gemcitabine. The antitumor effect was transient in all tested groups. The developed system can be used to study therapies involving various oncolytic viruses and chemotherapeutics, with the goal of inducing tumor-specific immunity while preventing premature virus clearance.


Asunto(s)
Adenocarcinoma/terapia , Terapia Biológica/métodos , Carcinoma Ductal Pancreático/terapia , Mucina-1/biosíntesis , Virus Oncolíticos/crecimiento & desarrollo , Vesiculovirus/crecimiento & desarrollo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Resultado del Tratamiento
2.
BMC Cancer ; 14: 225, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24674692

RESUMEN

BACKGROUND: IL-17A is a pro-inflammatory cytokine that is normally associated with autoimmune arthritis and other pro-inflammatory conditions. Recently, IL-17A has emerged as a critical factor in enhancing breast cancer (BC)-associated metastases. We generated immune competent arthritic mouse models that develop spontaneous BC-associated bone and lung metastasis. Using these models, we have previously shown that neutralization of IL-17A resulted in significant reduction in metastasis. However, the underlying mechanism/s remains unknown. METHODS: We have utilized two previously published mouse models for this study: 1) the pro-arthritic mouse model (designated SKG) injected with metastatic BC cell line (4T1) in the mammary fat pad, and 2) the PyV MT mice that develop spontaneous mammary gland tumors injected with type II collagen to induce autoimmune arthritis. Mice were treated with anti-IL-17A neutralizing antibody and monitored for metastasis and assessed for pro-inflammatory cytokines and chemokines associated with BC-associated metastasis. RESULTS: We first corroborate our previous finding that in vivo neutralization of IL-17A significantly reduced metastasis to the bones and lungs in both models. Next, we report that treatment with anti-IL17A antibody significantly reduced the expression of a key chemokine, CXCL12 (also known as stromal derived factor-1 (SDF - 1)) in the bones and lungs of treated mice. CXCL12 is a ligand for CXCR4 (expressed on BC cells) and their interaction is known to be critical for metastasis. Interestingly, levels of CXCR4 in the tumor remained unchanged with treatment. Consequently, protein lysates derived from the bones and lungs of treated mice were significantly less chemotactic for the BC cells than lysates from untreated mice; and addition of exogenous SDF-1 to the lysates from treated mice completely restored BC cell migration. In addition, cytokines such as IL-6 and M-CSF were significantly reduced in the lung and bone lysates following treatment. The data presented suggests that systemic neutralization of IL-17A can block the CXCR4/SDF-1 signaling pathway by reducing the expression of SDF-1 in the metastatic niches and significantly reducing metastasis in both mouse models. CONCLUSION: In our model, neutralization of IL-17A regulates SDF-1 expression in the metastatic niches either directly or indirectly via reducing levels of IL-6 and M-CSF.


Asunto(s)
Artritis/complicaciones , Neoplasias Óseas/patología , Quimiocina CXCL12/metabolismo , Interleucina-17/antagonistas & inhibidores , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Artritis/inducido químicamente , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interleucina-6/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Factor Estimulante de Colonias de Macrófagos/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Receptores CXCR4/metabolismo
3.
Breast Cancer Res ; 15(2): R32, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23577751

RESUMEN

INTRODUCTION: Breast cancer remains the second leading cause of cancer-related deaths for women in the United States. Metastasis is regulated not only by intrinsic genetic changes in malignant cells, but also by the microenvironment, especially those associated with chronic inflammation. We recently reported that mice with autoimmune arthritis have significantly increased incidence of bone and lung metastasis and decreased survival associated with breast cancer. In this study, we evaluated the mechanism underlying the increased metastasis. METHODS: We used two mouse models; one that develops spontaneous autoimmune arthritis (SKG mice) injected with metastatic breast cancer cells (4T1), and another that develops spontaneous breast cancer (MMTV-PyV MT mice) injected with type II collagen to induce autoimmune arthritis. Mast cell levels and metastasis were monitored. RESULTS: First, we confirmed that breast tumor-bearing arthritic mice have a significantly higher incidence of bone and lung metastasis than do their nonarthritic counterparts. Next, we showed increased recruitment of mast cells within the primary tumor of arthritic mice, which facilitates metastasis. Next, we report that arthritic mice without any tumors have higher numbers of mast cells in the bones and lungs, which may be the underlying cause for the enhanced lung and bone metastases observed in the arthritic mice. Next, we showed that once the tumor cells populate the metastatic niches (bones and lungs), they further increase the mast cell population within the niche and assist in enhancing metastasis. This may primarily be due to the interaction of c-Kit receptor present on mast cells and stem cell factor (SCF, the ligand for ckit) expressed on tumor cells. Finally, we showed that targeting the SCF/cKit interaction with an anti-ckit antibody reduces the differentiation of mast cells and consequently reduces metastasis. CONCLUSION: This is the first report to show that mast cells may play a critical role in remodeling not only the tumor microenvironment but also the metastatic niche to facilitate efficient metastasis through SCF/cKit interaction in breast cancer with arthritis.


Asunto(s)
Artritis Experimental/fisiopatología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Neoplasias Pulmonares/secundario , Mastocitos/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Animales , Apoptosis , Artritis Experimental/complicaciones , Western Blotting , Neoplasias Óseas/etiología , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Movimiento Celular , Proliferación Celular , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Mastocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Microambiente Tumoral
4.
J Virol ; 86(6): 3073-87, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238308

RESUMEN

Vesicular stomatitis virus (VSV) is a promising oncolytic agent against a variety of cancers. However, it has never been tested in any pancreatic cancer model. Pancreatic ductal adenocarcinoma (PDA) is the most common and aggressive form of pancreatic cancer. In this study, the oncolytic potentials of several VSV variants were analyzed in a panel of 13 clinically relevant human PDA cell lines and compared to conditionally replicative adenoviruses (CRAds), Sendai virus and respiratory syncytial virus. VSV variants showed oncolytic abilities superior to those of other viruses, and some cell lines that exhibited resistance to other viruses were successfully killed by VSV. However, PDA cells were highly heterogeneous in their susceptibility to virus-induced oncolysis, and several cell lines were resistant to all tested viruses. Resistant cells showed low levels of very early VSV RNA synthesis, indicating possible defects at initial stages of infection. In addition, unlike permissive PDA cell lines, most of the resistant cell lines were able to both produce and respond to interferon, suggesting that intact type I interferon responses contributed to their resistance phenotype. Four cell lines that varied in their permissiveness to VSV-ΔM51 and CRAd dl1520 were tested in mice, and the in vivo results closely mimicked those in vitro. While our results demonstrate that VSV is a promising oncolytic agent against PDA, further studies are needed to better understand the molecular mechanisms of resistance of some PDAs to oncolytic virotherapy.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Neoplasias Pancreáticas/terapia , Virus de la Estomatitis Vesicular Indiana/fisiología , Animales , Carcinoma Ductal Pancreático/inmunología , Línea Celular Tumoral , Humanos , Interferón Tipo I/inmunología , Masculino , Ratones , Ratones Desnudos , Viroterapia Oncolítica/instrumentación , Virus Oncolíticos/genética , Neoplasias Pancreáticas/inmunología , Virus de la Estomatitis Vesicular Indiana/genética
5.
J Surg Oncol ; 107(7): 713-22, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23335066

RESUMEN

BACKGROUND AND OBJECTIVES: MUC1 is over-expressed and aberrantly glycosylated in >60% of human pancreatic cancer (PC). Development of novel approaches for detection and/or targeting of MUC1 are critically needed and should be able to detect MUC1 on PC cells (including cancer stem cells) and in serum. METHODS: The sensitivity and specificity of the anti-MUC1 antibody, TAB 004, was determined. CSCs were assessed for MUC1 expression using TAB 004-FITC on in vitro PC cell lines, and on lineage(-) cells from in vivo tumors and human samples. Serum was assessed for shed MUC1 via the TAB 004 EIA. RESULTS: In vitro and in vivo, TAB 004 detected MUC1 on >95% of CSCs. Approximately, 80% of CSCs in patients displayed MUC1 expression as detected by TAB 004. Shed MUC1 was detected serum in mice with HPAF-II (MUC1(high) ) but not BxPC3 tumors (MUC1(low)). The TAB 004 EIA was able to accurately detect stage progression in PC patients. CONCLUSIONS: The TAB 004 antibody may be explored as a therapeutic targeting agent for CSCs in PC. The TAB 004 EIA detected circulating MUC1 in a stage-dependent manner in patients with PC and thus may be explored as a PC stage diagnostic biomarker.


Asunto(s)
Adenocarcinoma/metabolismo , Mucina-1/inmunología , Mucina-1/aislamiento & purificación , Células Madre Neoplásicas/inmunología , Neoplasias Pancreáticas/metabolismo , Antígeno AC133 , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/inmunología , Glicosilación , Humanos , Técnicas para Inmunoenzimas , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Péptidos/inmunología , Sensibilidad y Especificidad , Regulación hacia Arriba
6.
Cancer Immunol Immunother ; 61(11): 2055-65, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22543528

RESUMEN

Monoclonal antibodies (mAbs) against tumor-associated antigens are useful anticancer agents. Antibody-dependent cellular cytotoxicity (ADCC) is one of the major mechanisms responsible for initiating natural killer cell (NK)-mediated killing of tumors. However, the regulation of ADCC via NK cells is poorly understood. We have investigated the cytolytic activity of NK cells against pancreatic cancer cells that were coated with an antibody directed against the human tumor antigen, Mucin-1 designated HMFG-2, either alone or conjugated to CpG oligodeoxynucleotide (CpG ODN). Conjugated antibodies were tested for their ability to elicit ADCC in vitro and in vivo against pancreatic cancer cells. NK cells cultured in the presence of immobilized CpG ODN, HMFG-2 Ab, or CpG ODN-conjugated HMFG-2 Ab were able to up-regulate perforin similarly. Interestingly, a significant higher ADCC was observed when CpG ODN-conjugated HMFG-2-coated tumor cells were co-cultured with NK cells compared to unconjugated HMFG-2 Ab or CpG ODN alone. Moreover, MyD88-deficient NK cells can perform ADCC in vitro. Furthermore, intratumoral injections of CpG ODN-conjugated HMFG-2 induced a significant reduction in tumor burden in vivo in an established model of pancreatic tumor in nude mice compared to CpG ODN or the HMFG-2 alone. Depletion of macrophages or NK cells before treatment confirmed that both cells were required for the anti-tumor response in vivo. Results also suggest that CpG ODN and HMFG-2 Ab could be sensed by NK cells on the mAb-coated tumor cells triggering enhanced ADCC in vitro and in vivo.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Carcinoma Ductal Pancreático/terapia , Islas de CpG/inmunología , Células Asesinas Naturales/inmunología , Mucina-1/inmunología , Neoplasias Pancreáticas/terapia , Adyuvantes Inmunológicos/uso terapéutico , Animales , Carcinoma Ductal Pancreático/inmunología , Línea Celular Tumoral , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/inmunología , Oligodesoxirribonucleótidos/inmunología , Neoplasias Pancreáticas/inmunología , Perforina/biosíntesis , Perforina/inmunología , Regulación hacia Arriba/inmunología
7.
PLoS One ; 14(11): e0224309, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31693710

RESUMEN

In recent years, vaccines against tumor antigens have shown potential for combating invasive cancers, including primary tumors and metastatic lesions. This is particularly pertinent for breast cancer, which is the second-leading cause of cancer-related death in women. MUC1 is a glycoprotein that is normally expressed on glandular epithelium, but is overexpressed and under-glycosylated in most human cancers, including the majority of breast cancers. This under-glycosylation exposes the MUC1 protein core on the tumor-associated form of the protein. We have previously shown that a vaccine consisting of MUC1 core peptides stimulates a tumor-specific immune response. However, this immune response is dampened by the immunosuppressive microenvironment within breast tumors. Thus, in the present study, we investigated the effectiveness of MUC1 vaccination in combination with four different drugs that inhibit different components of the COX pathway: indomethacin (COX-1 and COX-2 inhibitor), celecoxib (COX-2 inhibitor), 1-methyl tryptophan (indoleamine 2,3 dioxygenase inhibitor), and AH6809 (prostaglandin E2 receptor antagonist). These treatment regimens were explored for the treatment of orthotopic MUC1-expressing breast tumors in mice transgenic for human MUC1. We found that the combination of vaccine and indomethacin resulted in a significant reduction in tumor burden. Indomethacin did not increase tumor-specific immune responses over vaccine alone, but rather appeared to reduce the proliferation and increase apoptosis of tumor cells, thus rendering them susceptible to immune cell killing.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/administración & dosificación , Inhibidores de la Ciclooxigenasa/administración & dosificación , Indometacina/administración & dosificación , Neoplasias Mamarias Experimentales/terapia , Mucina-1/inmunología , Animales , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Terapia Combinada/métodos , Femenino , Humanos , Inmunogenicidad Vacunal/efectos de los fármacos , Inmunogenicidad Vacunal/inmunología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Transgénicos , Mucina-1/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
8.
Cancer Res ; 71(13): 4432-42, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558393

RESUMEN

MUC1 is overexpressed and aberrantly glycosylated in more than 60% of pancreatic ductal adenocarcinomas. The functional role of MUC1 in pancreatic cancer has yet to be fully elucidated due to a dearth of appropriate models. In this study, we have generated mouse models that spontaneously develop pancreatic ductal adenocarcinoma (KC), which are either Muc1-null (KCKO) or express human MUC1 (KCM). We show that KCKO mice have significantly slower tumor progression and rates of secondary metastasis, compared with both KC and KCM. Cell lines derived from KCKO tumors have significantly less tumorigenic capacity compared with cells from KCM tumors. Therefore, mice with KCKO tumors had a significant survival benefit compared with mice with KCM tumors. In vitro, KCKO cells have reduced proliferation and invasion and failed to respond to epidermal growth factor, platelet-derived growth factor, or matrix metalloproteinase 9. Further, significantly less KCKO cells entered the G(2)-M phase of the cell cycle compared with the KCM cells. Proteomics and Western blotting analysis revealed a complete loss of cdc-25c expression, phosphorylation of mitogen-activated protein kinase (MAPK), as well as a significant decrease in nestin and tubulin-α2 chain expression in KCKO cells. Treatment with a MEK1/2 inhibitor, U0126, abrogated the enhanced proliferation of the KCM cells but had minimal effect on KCKO cells, suggesting that MUC1 is necessary for MAPK activity and oncogenic signaling. This is the first study to utilize a Muc1-null PDA mouse to fully elucidate the oncogenic role of MUC1, both in vivo and in vitro.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Mucina-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Animales , Butadienos/farmacología , Carcinoma Ductal Pancreático/enzimología , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Factor de Crecimiento Epidérmico , Humanos , Proteínas de Filamentos Intermediarios/biosíntesis , Metaloproteinasa 9 de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mucina-1/genética , Metástasis de la Neoplasia , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Nitrilos/farmacología , Neoplasias Pancreáticas/enzimología , Factor de Crecimiento Derivado de Plaquetas , Inhibidores de Proteínas Quinasas/farmacología , Tubulina (Proteína)/biosíntesis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda