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

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(15): 4725-30, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825750

RESUMEN

Nonresolving chronic inflammation at the neoplastic site is consistently associated with promoting tumor progression and poor patient outcomes. However, many aspects behind the mechanisms that establish this tumor-promoting inflammatory microenvironment remain undefined. Using bladder cancer (BC) as a model, we found that CD14-high cancer cells express higher levels of numerous inflammation mediators and form larger tumors compared with CD14-low cells. CD14 antigen is a glycosyl-phosphatidylinositol (GPI)-linked glycoprotein and has been shown to be critically important in the signaling pathways of Toll-like receptor (TLR). CD14 expression in this BC subpopulation of cancer cells is required for increased cytokine production and increased tumor growth. Furthermore, tumors formed by CD14-high cells are more highly vascularized with higher myeloid cell infiltration. Inflammatory factors produced by CD14-high BC cells recruit and polarize monocytes and macrophages to acquire immune-suppressive characteristics. In contrast, CD14-low BC cells have a higher baseline cell division rate than CD14-high cells. Importantly, CD14-high cells produce factors that further increase the proliferation of CD14-low cells. Collectively, we demonstrate that CD14-high BC cells may orchestrate tumor-promoting inflammation and drive tumor cell proliferation to promote tumor growth.


Asunto(s)
Proliferación Celular/genética , Citocinas/genética , Receptores de Lipopolisacáridos/genética , Microambiente Tumoral/genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Queratina-14/genética , Queratina-14/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
2.
Proc Natl Acad Sci U S A ; 110(9): 3501-6, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23382202

RESUMEN

Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract and arises from the interstitial cells of Cajal. It is characterized by expression of the receptor tyrosine kinase CD117 (KIT). In 70-80% of GIST cases, oncogenic mutations in KIT are present, leading to constitutive activation of the receptor, which drives the proliferation of these tumors. Treatment of GIST with imatinib, a small-molecule tyrosine kinase inhibitor, inhibits KIT-mediated signaling and initially results in disease control in 70-85% of patients with KIT-positive GIST. However, the vast majority of patients eventually develop resistance to imatinib treatment, leading to disease progression and posing a significant challenge in the clinical management of these tumors. Here, we show that an anti-KIT monoclonal antibody (mAb), SR1, is able to slow the growth of three human GIST cell lines in vitro. Importantly, these reductions in cell growth were equivalent between imatinib-resistant and imatinib-sensitive GIST cell lines. Treatment of GIST cell lines with SR1 reduces cell-surface KIT expression, suggesting that mAb-induced KIT down-regulation may be a mechanism by which SR1 inhibits GIST growth. Furthermore, we also show that SR1 treatment enhances phagocytosis of GIST cells by macrophages, indicating that treatment with SR1 may enhance immune cell-mediated tumor clearance. Finally, using two xenotransplantation models of imatinib-sensitive and imatinib-resistant GIST, we demonstrate that SR1 is able to strongly inhibit tumor growth in vivo. These results suggest that treatment with mAbs targeting KIT may represent an alternative, or complementary, approach for treating GIST.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Resistencia a Antineoplásicos , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/inmunología , Pirimidinas/uso terapéutico , Animales , Anticuerpos Monoclonales/farmacología , Benzamidas , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Mesilato de Imatinib , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis/efectos de los fármacos , Piperazinas/farmacología , Pirimidinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 109(17): 6656-61, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22451919

RESUMEN

Antibodies against CD47, which block tumor cell CD47 interactions with macrophage signal regulatory protein-α, have been shown to decrease tumor size in hematological and epithelial tumor models by interfering with the protection from phagocytosis by macrophages that intact CD47 bestows upon tumor cells. Leiomyosarcoma (LMS) is a tumor of smooth muscle that can express varying levels of colony-stimulating factor-1 (CSF1), the expression of which correlates with the numbers of tumor-associated macrophages (TAMs) that are found in these tumors. We have previously shown that the presence of TAMs in LMS is associated with poor clinical outcome and the overall effect of TAMs in LMS therefore appears to be protumorigenic. However, the use of inhibitory antibodies against CD47 offers an opportunity to turn TAMs against LMS cells by allowing the phagocytic behavior of resident macrophages to predominate. Here we show that interference with CD47 increases phagocytosis of two human LMS cell lines, LMS04 and LMS05, in vitro. In addition, treatment of mice bearing subcutaneous LMS04 and LMS05 tumors with a novel, humanized anti-CD47 antibody resulted in significant reductions in tumor size. Mice bearing LMS04 tumors develop large numbers of lymph node and lung metastases. In a unique model for neoadjuvant treatment, mice were treated with anti-CD47 antibody starting 1 wk before resection of established primary tumors and subsequently showed a striking decrease in the size and number of metastases. These data suggest that treatment with anti-CD47 antibodies not only reduces primary tumor size but can also be used to inhibit the development of, or to eliminate, metastatic disease.


Asunto(s)
Anticuerpos/uso terapéutico , Antígeno CD47/inmunología , Modelos Animales de Enfermedad , Inmunoterapia , Leiomiosarcoma/terapia , Animales , Anticuerpos/inmunología , Línea Celular Tumoral , Leiomiosarcoma/patología , Ratones , Metástasis de la Neoplasia , Fagocitosis/inmunología
4.
Sci Transl Med ; 9(381)2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28298418

RESUMEN

Morbidity and mortality associated with pediatric malignant primary brain tumors remain high in the absence of effective therapies. Macrophage-mediated phagocytosis of tumor cells via blockade of the anti-phagocytic CD47-SIRPα interaction using anti-CD47 antibodies has shown promise in preclinical xenografts of various human malignancies. We demonstrate the effect of a humanized anti-CD47 antibody, Hu5F9-G4, on five aggressive and etiologically distinct pediatric brain tumors: group 3 medulloblastoma (primary and metastatic), atypical teratoid rhabdoid tumor, primitive neuroectodermal tumor, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Hu5F9-G4 demonstrated therapeutic efficacy in vitro and in vivo in patient-derived orthotopic xenograft models. Intraventricular administration of Hu5F9-G4 further enhanced its activity against disseminated medulloblastoma leptomeningeal disease. Notably, Hu5F9-G4 showed minimal activity against normal human neural cells in vitro and in vivo, a phenomenon reiterated in an immunocompetent allograft glioma model. Thus, Hu5F9-G4 is a potentially safe and effective therapeutic agent for managing multiple pediatric central nervous system malignancies.


Asunto(s)
Anticuerpos/uso terapéutico , Antígenos de Diferenciación/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Antígeno CD47/inmunología , Fagocitosis , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos/farmacología , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Niño , Modelos Animales de Enfermedad , Humanos , Inmunocompetencia , Inyecciones Intraventriculares , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/patología , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/secundario , Ratones Endogámicos C57BL , Modelos Biológicos , Metástasis de la Neoplasia , Fagocitosis/efectos de los fármacos , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Clin Invest ; 126(7): 2610-20, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27294525

RESUMEN

Small-cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer with limited treatment options. CD47 is a cell-surface molecule that promotes immune evasion by engaging signal-regulatory protein alpha (SIRPα), which serves as an inhibitory receptor on macrophages. Here, we found that CD47 is highly expressed on the surface of human SCLC cells; therefore, we investigated CD47-blocking immunotherapies as a potential approach for SCLC treatment. Disruption of the interaction of CD47 with SIRPα using anti-CD47 antibodies induced macrophage-mediated phagocytosis of human SCLC patient cells in culture. In a murine model, administration of CD47-blocking antibodies or targeted inactivation of the Cd47 gene markedly inhibited SCLC tumor growth. Furthermore, using comprehensive antibody arrays, we identified several possible therapeutic targets on the surface of SCLC cells. Antibodies to these targets, including CD56/neural cell adhesion molecule (NCAM), promoted phagocytosis in human SCLC cell lines that was enhanced when combined with CD47-blocking therapies. In light of recent clinical trials for CD47-blocking therapies in cancer treatment, these findings identify disruption of the CD47/SIRPα axis as a potential immunotherapeutic strategy for SCLC. This approach could enable personalized immunotherapeutic regimens in patients with SCLC and other cancers.


Asunto(s)
Antígeno CD47/metabolismo , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Macrófagos/inmunología , Carcinoma Pulmonar de Células Pequeñas/terapia , Animales , Anticuerpos Monoclonales/farmacología , Antígeno CD56/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Neoplasias Pulmonares/inmunología , Ratones , Fagocitosis , Receptores Inmunológicos/metabolismo , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas/inmunología
6.
Oncoimmunology ; 2(6): e24452, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23894705

RESUMEN

Acquired resistance to imatinib is a significant problem for the clinical management of gastrointestinal stromal tumor (GIST) patients, and second-line small molecules have shown limited efficacy in this setting. We have recently demonstrated that a monoclonal antibody targeting KIT could potentially bypass imatinib resistance in preclinical models of GIST.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA