Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Carcinogenesis ; 35(3): 613-23, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24233838

RESUMEN

Previous studies have implicated cancer stem cells in tumor recurrence and revealed that the stem cell gene SOX2 plays an important role in the tumor cell resistance to apoptosis. Nonetheless, the mechanism by which SOX2 regulates apoptosis signals remained undefined. Here, we demonstrated the surprising finding that silencing of the SOX2 gene effectively induces apoptosis via the activation of death receptor and mitochondrial signaling pathways in human non-small cell lung cancer cells. Unexpectedly, reverse transcription-PCR analysis suggested that downregulation of SOX2 leads to activation of MAP4K4, previously implicated in cell survival. Evaluation of the apoptotic pathways revealed an increased expression of key inducers of apoptosis, including tumor necrosis factor-α and p53, with concurrent attenuation of Survivin. Although p53 appeared dispensable for this pathway, the loss of Survivin in SOX2-deficient cells appeared critical for the observed MAP4K4 induced cell death. Rescue experiments revealed that SOX2-silencing-mediated killing was blocked by ectopic expression of Survivin, or by reduction of MAP4K4 expression. Clinically, expressions of Survivin and SOX2 were highly correlated with each other. The results reveal a key target of SOX2 expression and highlight the unexpected context-dependent role for MAP4K4, a pluripotent activator of several mitogen-activated protein kinase pathways, in regulating tumor cell survival.


Asunto(s)
Apoptosis/fisiología , Proteínas Inhibidoras de la Apoptosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Silenciador del Gen , Humanos , Ratones , Ratones SCID , Factores de Transcripción SOXB1/genética , Survivin
2.
Stem Cells ; 31(2): 248-58, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169551

RESUMEN

The cancer stem cell (CSC) hypothesis has gained significant recognition as a descriptor of tumorigenesis. Additionally, tumor-associated macrophages (TAMs) are known to promote growth and metastasis of breast cancer. However, it is not known whether TAMs mediate tumorigenesis through regulation of breast CSCs. Here, we report that TAMs promote CSC-like phenotypes in murine breast cancer cells by upregulating their expression of Sox-2. These CSC-like phenotypes were characterized by increased Sox-2, Oct-4, Nanog, AbcG2, and Sca-1 gene expression, in addition to increased drug-efflux capacity, resistance to chemotherapy, and increased tumorigenicity in vivo. Downregulation of Sox-2 in tumor cells by siRNA blocked the ability of TAMs to induce these CSC-like phenotypes and inhibited tumor growth in vivo. Furthermore, we identified a novel epidermal growth factor receptor (EGFR)/signal transducers and activators of transcription 3 (Stat3)/Sox-2 paracrine signaling pathway between macrophages and mouse breast cancer cells that is required for macrophage-induced upregulation of Sox-2 and CSC phenotypes in tumor cells. We showed that this crosstalk was effectively blocked by the small molecule inhibitors AG1478 or CDDO-Im against EGFR and Stat3, respectively. Therefore, our report identifies a novel role for TAMs in breast CSC regulation and establishes a rationale for targeting the EGFR/Stat3/Sox-2 signaling pathway for CSC therapy.


Asunto(s)
Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/genética , Factor de Transcripción STAT3/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteína Homeótica Nanog , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Factores de Transcripción SOXB1/antagonistas & inhibidores , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
3.
J Immunol ; 189(5): 2656-64, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22844125

RESUMEN

hu14.18-IL-2 (IC) is an immunocytokine consisting of human IL-2 linked to hu14.18 mAb, which recognizes the GD2 disialoganglioside. Phase 2 clinical trials of i.v. hu14.18-IL-2 (i.v.-IC) in neuroblastoma and melanoma are underway and have already demonstrated activity in neuroblastoma. We showed previously that intratumoral hu14.18-IL-2 (IT-IC) results in enhanced antitumor activity in mouse models compared with i.v.-IC. The studies presented in this article were designed to determine the mechanisms involved in this enhanced activity and to support the future clinical testing of intratumoral administration of immunocytokines. Improved survival and inhibition of growth of both local and distant tumors were observed in A/J mice bearing s.c. NXS2 neuroblastomas treated with IT-IC compared with those treated with i.v.-IC or control mice. The local and systemic antitumor effects of IT-IC were inhibited by depletion of NK cells or T cells. IT-IC resulted in increased NKG2D receptors on intratumoral NKG2A/C/E⁺ NKp46⁺ NK cells and NKG2A/C/E⁺ CD8⁺ T cells compared with control mice or mice treated with i.v.-IC. NKG2D levels were augmented more in tumor-infiltrating lymphocytes compared with splenocytes, supporting the localized nature of the intratumoral changes induced by IT-IC treatment. Prolonged retention of IC at the tumor site was seen with IT-IC compared with i.v.-IC. Overall, IT-IC resulted in increased numbers of activated T and NK cells within tumors, better IC retention in the tumor, enhanced inhibition of tumor growth, and improved survival compared with i.v.-IC.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neuroblastoma/inmunología , Neuroblastoma/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inyecciones Intralesiones , Interleucina-2/administración & dosificación , Interleucina-2/metabolismo , Interleucina-2/uso terapéutico , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Neuroblastoma/patología , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
4.
N Engl J Med ; 363(14): 1324-34, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20879881

RESUMEN

BACKGROUND: Preclinical and preliminary clinical data indicate that ch14.18, a monoclonal antibody against the tumor-associated disialoganglioside GD2, has activity against neuroblastoma and that such activity is enhanced when ch14.18 is combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-2. We conducted a study to determine whether adding ch14.18, GM-CSF, and interleukin-2 to standard isotretinoin therapy after intensive multimodal therapy would improve outcomes in high-risk neuroblastoma. METHODS: Patients with high-risk neuroblastoma who had a response to induction therapy and stem-cell transplantation were randomly assigned, in a 1:1 ratio, to receive standard therapy (six cycles of isotretinoin) or immunotherapy (six cycles of isotretinoin and five concomitant cycles of ch14.18 in combination with alternating GM-CSF and interleukin-2). Event-free survival and overall survival were compared between the immunotherapy group and the standard-therapy group, on an intention-to-treat basis. RESULTS: A total of 226 eligible patients were randomly assigned to a treatment group. In the immunotherapy group, a total of 52% of patients had pain of grade 3, 4, or 5, and 23% and 25% of patients had capillary leak syndrome and hypersensitivity reactions, respectively. With 61% of the number of expected events observed, the study met the criteria for early stopping owing to efficacy. The median duration of follow-up was 2.1 years. Immunotherapy was superior to standard therapy with regard to rates of event-free survival (66±5% vs. 46±5% at 2 years, P=0.01) and overall survival (86±4% vs. 75±5% at 2 years, P=0.02 without adjustment for interim analyses). CONCLUSIONS: Immunotherapy with ch14.18, GM-CSF, and interleukin-2 was associated with a significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma. (Funded by the National Institutes of Health and the Food and Drug Administration; ClinicalTrials.gov number, NCT00026312.)


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Gangliósidos/inmunología , Inmunoterapia , Neuroblastoma/terapia , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/uso terapéutico , Niño , Preescolar , Terapia Combinada , Quimioterapia Combinada , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunoterapia/efectos adversos , Lactante , Análisis de Intención de Tratar , Interleucina-2/uso terapéutico , Isotretinoína/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Trasplante de Células Madre , Análisis de Supervivencia
5.
Cancer Immunol Immunother ; 62(8): 1303-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23661160

RESUMEN

Hu14.18-IL2 is an immunocytokine (IC) consisting of human IL-2 linked to hu14.18 mAb, which recognizes GD2 disialoganglioside. Phase II clinical trials of intravenous-hu14.18-IL2 (IV-IC) in neuroblastoma and melanoma are underway, and have already demonstrated activity in neuroblastoma. In our Phase II trial, lower neuroblastoma burden at the time of treatment was associated with a greater likelihood of clinical response to IV-IC. We have previously shown that intratumoral-hu14.18-IL2 (IT-IC) compared to IV-IC results in enhanced local and systemic antitumor activity in tumor-bearing mice. We utilized a mouse model to investigate the impact of tumor burden on hu14.18-IL2 treatment efficacy in IV- versus IT-treated animals. Studies presented here describe the analyses of tumor burden at the initiation of treatment and its effects on treatment efficacy, survival, and tumor-infiltrating leukocytes in A/J mice bearing subcutaneous NXS2 neuroblastoma. We show that smaller tumor burden at treatment initiation is associated with increased infiltration of NK and CD8+ T cells and increased overall survival. NXS2 tumor shrinkage shortly after completion of the 3 days of hu14.18-IL2 treatment is necessary for long-term survival. This model demonstrates that tumor size is a strong predictor of hu14.18-IL2-induced lymphocyte infiltration and treatment outcome.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Neuroblastoma/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Interleucina-2/administración & dosificación , Interleucina-2/genética , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Pronóstico , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
6.
Immunol Rev ; 222: 117-28, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18363997

RESUMEN

Four novel oral DNA vaccines provide protection against melanoma, colon, breast, and lung carcinoma in mouse models. Vaccines are delivered by attenuated Salmonella typhimurium to secondary lymphoid organs and respectively target vascular endothelial growth factor receptor-2, transcription factor Fos-related antigen-1, anti-apoptosis protein survivin and Legumain, an asparaginyl endopeptidase specifically overexpressed on tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). These vaccines are all capable of inducing potent cell-mediated protective immunity against self-antigens, resulting in marked suppression of tumor growth and dissemination. Key mechanisms induced by these DNA vaccines include efficient suppression of angiogenesis in the tumor vasculature and marked activation of cytotoxic T cells, natural killer cells, and antigen-presenting dendritic cells. The vaccine targeting Legumain establishes the new paradigm whereby a reduction in the density of TAMs in the TME decreases the release of factors potentiating tumor growth and angiogenesis. This, in turn, remodels the TME and decreases its immunosuppressive milieu and thereby potentiates the DNA vaccine's ability to effectively suppress tumor cell proliferation, vascularization, and metastasis. It is anticipated that such research efforts will lead to novel DNA-based vaccines that will be effective for the treatment of cancer.


Asunto(s)
Inmunoterapia , Neoplasias Experimentales/terapia , Neovascularización Patológica/prevención & control , Vacunas de ADN/administración & dosificación , Administración Oral , Animales , Linfocitos T CD8-positivos/inmunología , Quimiocina CCL21/inmunología , Técnicas de Transferencia de Gen , Memoria Inmunológica , Proteínas Inhibidoras de la Apoptosis , Interleucina-18/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Ratones , Proteínas Asociadas a Microtúbulos/inmunología , Invasividad Neoplásica , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/inmunología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-fos/inmunología , Proteínas Represoras , Salmonella typhimurium , Autotolerancia/inmunología , Survivin , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
7.
Cancer Immunol Immunother ; 60(5): 731-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21340652

RESUMEN

We investigated the anti-tumor effect of peritumoral resveratrol in combination with immunotherapy in vivo in neuroblastoma-bearing mice. Subcutaneous NXS2 tumors were induced in A/J mice. On day 10, some mice received 15 mcg of intravenous immunocytokine for 5 days, mice received 20 mg of peritumoral resveratrol twice a week (starting on day 12) for a total of 5 injections, and a separate group received a combination of both regimens. Tumor progression and survival were assessed every 3-4 days. Blood and primary tumor tissue samples were collected on day 20 for Complete Blood Count and CD45 immunohistochemistry and histology, respectively. The primary tumor regressed in all mice receiving peritumoral resveratrol. Most of these mice receiving peritumoral resveratrol alone developed metastatic tumors and recurrence of the primary tumor after cessation of therapy. When resveratrol and immunocytokine regimens were combined, 61% of the mice receiving this combination therapy resolved their primary tumors and survived without developing metastatic tumors, compared to 15 and 13% receiving resveratrol or immunocytokine alone, respectively. None of the therapeutic regimes prevented lymphocyte infiltration or affected the complete blood count. Greater necrosis was observed microscopically in tumors from mice receiving the combination therapy. These results demonstrate that the combination therapy of peritumoral resveratrol plus intravenous immunocytokine provides better anti-tumor effects in this model than either therapy alone.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Interleucina-2/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/terapia , Estilbenos/uso terapéutico , Animales , Antineoplásicos Fitogénicos/sangre , Línea Celular Tumoral , Terapia Combinada , Citometría de Flujo , Gangliósidos/análisis , Gangliósidos/sangre , Inmunoterapia , Antígenos Comunes de Leucocito , Leucocitos/efectos de los fármacos , Ratones , Ratones Endogámicos A , Recurrencia Local de Neoplasia , Neuroblastoma/inmunología , Resveratrol , Estilbenos/sangre , Sobrevida , Carga Tumoral/efectos de los fármacos
8.
Nat Med ; 8(12): 1369-75, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12415261

RESUMEN

Tumor cells are elusive targets for immunotherapy due to their heterogeneity and genetic instability. Here we describe a novel, oral DNA vaccine that targets stable, proliferating endothelial cells in the tumor vasculature rather than tumor cells. Targeting occurs through upregulated vascular-endothelial growth factor receptor 2 (FLK-1) of proliferating endothelial cells in the tumor vasculature. This vaccine effectively protected mice from lethal challenges with melanoma, colon carcinoma and lung carcinoma cells and reduced growth of established metastases in a therapeutic setting. CTL-mediated killing of endothelial cells indicated breaking of peripheral immune tolerance against this self antigen, resulting in markedly reduced dissemination of spontaneous and experimental pulmonary metastases. Angiogenesis in the tumor vasculature was suppressed without impairment of fertility, neuromuscular performance or hematopoiesis, albeit with a slight delay in wound healing. Our strategy circumvents problems in targeting of genetically unstable tumor cells. This approach may provide a new strategy for the rational design of cancer therapies.


Asunto(s)
Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/prevención & control , Vacunas de ADN/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , Citotoxicidad Inmunológica , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Vacunación , Cicatrización de Heridas
9.
Nanomedicine ; 7(6): 665-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21419870

RESUMEN

Unresolved problems associated with ligand-targeting of liposomal nanoparticles (NPs) to solid tumors include variable target receptor expression due to genetic heterogeneity and insufficient target specificity, leading to systemic toxicities. This study addresses these issues by developing a novel ligand-targeting strategy for liposomal NPs using RR-11a, a synthetic enzyme inhibitor of Legumain, an asparaginyl endopeptidase. Cell-surface expression of Legumain is driven by hypoxic stress, a hallmark of solid tumors. Legumain-targeted RR-11a-coupled NPs revealed high ligand-receptor affinity, enhanced solid-tumor penetration and uptake by tumor cells. Treatment of tumor-bearing mice with RR-11a-coupled NPs encapsulating doxorubicin resulted in improved tumor selectivity and drug sensitivity, leading to complete inhibition of tumor growth. These antitumor effects were achieved while eliminating systemic drug toxicity. Therefore, synthetic enzyme inhibitors, such as RR-11a, represent a new class of compounds that can be used for highly specific ligand-targeting of NPs to solid tumors. FROM THE CLINICAL EDITOR: This study addresses the problems associated with ligand-targeting of liposomal nanoparticles to solid tumors with variable target receptor expression. A novel and efficacious targeting strategy has been developed towards a synthetic enzyme inhibitor of Legumain. The authors demonstrate successful tumor growth inhibiting effect while eliminating systemic drug toxicity in an animal model using this strategy.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisteína Endopeptidasas/metabolismo , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liposomas/química , Nanopartículas/química , Inhibidores de Proteasas/metabolismo , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Ligandos , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteasas/química
10.
Exp Dermatol ; 19(2): 157-64, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19849715

RESUMEN

Complex biological processes often require in vivo analysis, and many important research advances have been made using mice as a model for the study of various biological systems. Cutaneous melanomas are tumors originating from skin melanocytes, which are present in hair follicles, and interfollicular epidermal and dermal layers. Until recently, mouse melanoma models were largely based on transplantation models, i.e. transplantation of either syngeneic or xenogeneic melanoma cells into wild type or genetically modified animals. More recently, however, the use of novel technologies specifically modifying the genome allows for the generation of mouse strains, which may develop spontaneous melanoma. Nevertheless, it should be kept in mind that animal models provide only an approximation of reality in humans. In this review, we will discuss a representative selection of currently available transplantation and transgenic melanoma models; despite the fact that this selection will be biased by personal experience, we are confident to demonstrate how the use of mouse melanoma models facilitates translational research in several biomedical disciplines.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma Experimental , Ratones , Animales , Humanos , Melanoma Experimental/genética , Trasplante Heterólogo , Trasplante Isogénico
11.
J Clin Invest ; 116(7): 1955-62, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16794736

RESUMEN

Tumor-associated fibroblasts are key regulators of tumorigenesis. In contrast to tumor cells, which are genetically unstable and mutate frequently, the presence of genetically more stable fibroblasts in the tumor-stromal compartment makes them an optimal target for cancer immunotherapy. These cells are also the primary source of collagen type I, which contributes to decreased chemotherapeutic drug uptake in tumors and plays a significant role in regulating tumor sensitivity to a variety of chemotherapies. To specifically kill tumor-associated fibroblasts, we constructed an oral DNA vaccine targeting fibroblast activation protein (FAP), which is specifically overexpressed by fibroblasts in the tumor stroma. Through CD8+ T cell-mediated killing of tumor-associated fibroblasts, our vaccine successfully suppressed primary tumor cell growth and metastasis of multidrug-resistant murine colon and breast carcinoma. Furthermore, tumor tissue of FAP-vaccinated mice revealed markedly decreased collagen type I expression and up to 70% greater uptake of chemotherapeutic drugs. Most importantly, pFap-vaccinated mice treated with chemotherapy showed a 3-fold prolongation in lifespan and marked suppression of tumor growth, with 50% of the animals completely rejecting a tumor cell challenge. This strategy opens a new venue for the combination of immuno- and chemotherapies.


Asunto(s)
Vacunas contra el Cáncer , Fibroblastos/metabolismo , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias , Serina Endopeptidasas/metabolismo , Vacunas de ADN , Animales , Linfocitos T CD8-positivos/inmunología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Endopeptidasas , Femenino , Gelatinasas/genética , Gelatinasas/uso terapéutico , Proteínas de la Membrana/genética , Proteínas de la Membrana/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Serina Endopeptidasas/genética , Serina Endopeptidasas/uso terapéutico , Tasa de Supervivencia
12.
J Clin Invest ; 116(8): 2132-2141, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16862213

RESUMEN

Tumor-associated macrophages (TAMs) are associated with tumor progression and metastasis. Here, we demonstrate for the first time that legumain, a member of the asparaginyl endopeptidase family functioning as a stress protein, overexpressed by TAMs, provides an ideal target molecule. In fact, a legumain-based DNA vaccine served as a tool to prove this point, as it induced a robust CD8+ T cell response against TAMs, which dramatically reduced their density in tumor tissues and resulted in a marked decrease in proangiogenic factors released by TAMs such as TGF-beta, TNF-alpha, MMP-9, and VEGF. This, in turn, led to a suppression of both tumor angiogenesis and tumor growth and metastasis. Importantly, the success of this strategy was demonstrated in murine models of metastatic breast, colon, and non-small cell lung cancers, where 75% of vaccinated mice survived lethal tumor cell challenges and 62% were completely free of metastases. In conclusion, decreasing the number of TAMs in the tumor stroma effectively altered the tumor microenvironment involved in tumor angiogenesis and progression to markedly suppress tumor growth and metastasis. Gaining better insights into the mechanisms required for an effective intervention in tumor growth and metastasis may ultimately lead to new therapeutic targets and better anticancer strategies.


Asunto(s)
Macrófagos/fisiología , Neoplasias Mamarias Animales/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Progresión de la Enfermedad , Femenino , Inmunohistoquímica , Activación de Linfocitos , Neoplasias Mamarias Animales/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neovascularización Patológica/prevención & control , Linfocitos T/inmunología
13.
Nat Rev Drug Discov ; 5(2): 147-59, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16424916

RESUMEN

Treatment of cancer is a double-edged sword: it should be as aggressive as possible to completely destroy the tumour, but it is precisely this aggressiveness which often causes severe side effects - a reason why some promising therapeutics can not be applied systemically. In addition, therapeutics such as cytokines that physiologically function in a para- or autocrine fashion require a locally enhanced level to exert their effect appropriately. An elegant way to accumulate therapeutic agents at the tumour site is their conjugation/fusion to tumour-specific antibodies. Here, we discuss recent preclinical and clinical data for antibody-drug conjugates and fusion proteins with a special focus on drug components that exert their antitumour effects through normal biological processes.


Asunto(s)
Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Inmunotoxinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Sistemas de Liberación de Medicamentos , Humanos , Inmunotoxinas/inmunología , Profármacos , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico
14.
Cancer Res ; 67(5): 2331-8, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332365

RESUMEN

Fractalkine (FKN) is a unique CX3C chemokine (CX3CL1) known to induce both adhesion and migration of leukocytes mediated by a membrane-bound and a soluble form, respectively. Its function is mediated through CX3C receptor (CX3CR), which is expressed by T(H)1 immune cells including T cells and natural killer (NK) cells. FKN was shown to be expressed in >90% of 68 neuroblastoma samples as determined by cDNA microarray analysis. Here, we characterized the effect of FKN in the neuroblastoma microenvironment using a syngeneic model genetically engineered to secrete FKN. We show FKN-mediated migration, adhesion, and IFN-gamma secretion of immune effector cells, but limited antineuroblastoma activity, in vitro and in vivo. Therefore, we tested the hypothesis that a combined increase of FKN and interleukin-2 (IL-2) in the neuroblastoma microenvironment induces an effective antitumor immune response. For this purpose, IL-2 was targeted to ganglioside GD2, which is highly expressed on neuroblastoma tissue, using an anti-GD2 antibody IL-2 immunocytokine (ch14.18-IL-2). Only mice bearing FKN- and IL-2-enriched neuroblastoma tumors exhibited a reduction in primary tumor growth and a complete eradication of experimental liver metastases. The depletion of T cells and NK cells in vivo abrogated the effect, and these effector cells showed the highest cytolytic activity in vitro. Finally, only the FKN- and IL-2-enriched neuroblastoma microenvironment resulted in T-cell activation and the release of proinflammatory cytokines. In summary, we showed for the first time the immunologic mechanisms by which targeted IL-2 treatment of neuroblastoma with an FKN-rich microenvironment induces an effective antitumor response.


Asunto(s)
Quimiocinas CX3C/metabolismo , Interleucina-2/uso terapéutico , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Quimiocina CX3CL1 , Quimiocinas CX3C/genética , Quimiocinas CX3C/fisiología , Femenino , Marcación de Gen , Humanos , Inmunidad Celular , Inmunoterapia/métodos , Interleucina-2/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 27(5): 1095-100, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17303776

RESUMEN

OBJECTIVE: Similarities between neovascular ingrowth in atherosclerotic plaques and angiogenesis in tumors suggest that antiangiogenic factors that target tumor expansion may prove efficacious in the treatment of atherosclerosis. This study examined whether an oral DNA vaccine against the murine VEGF receptor 2 (Flk-1) with demonstrated antitumor effect through inhibition of pathological neovascularization can prevent or retard progression of atherosclerosis in hyperlipidemic low density lipoprotein receptor-deficient (LDLr-/-) mice. METHODS AND RESULTS: Vaccination against Flk-1 resulted in T cell activation, suppression of neoangiogenesis, and a marked reduction in atherosclerosis which was independent of hypercholesterolemia in both male and female mice. Immunohistochemical characterization of aortic sinus lesions showed that the decreased lesion area was not associated with reduced plaque stability and had a lower density of microvessels. CONCLUSIONS: These findings demonstrate for the first time that a DNA vaccine targeting activated endothelial cells in atherosclerotic lesions provides direct atheroprotective effects.


Asunto(s)
Aterosclerosis/terapia , Receptores de LDL/deficiencia , Vacunación/métodos , Vacunas de ADN/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Resultado del Tratamiento , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
16.
Arterioscler Thromb Vasc Biol ; 27(9): 2050-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17600223

RESUMEN

OBJECTIVE: Vascular endothelial growth factor receptor 2 (VEGFR2)-overexpressing cells may form an interesting target for the treatment of atherosclerosis because of their involvement in processes that contribute to this disease, such as angiogenesis. METHODS AND RESULTS: We vaccinated mice against VEGFR2 by an orally administered DNA vaccine, comprising a plasmid, encoding murine VEGFR2, carried by live attenuated Salmonella typhimurium. This vaccine induces cellular immunity against cells that overexpress VEGFR2. Vaccination of hypercholesterolemic mice against VEGFR2 resulted in a marked induction of CD8+ cytotoxic T cells specific for VEGFR2 and led to an inhibition of angiogenesis in a hindlimb ischemia model. Interestingly, VEGFR2 vaccination attenuated the progression of preexisting advanced atherosclerotic lesions in the brachiocephalic artery of apoE-/- mice. Furthermore, VEGFR2 vaccination strongly reduced the initiation of collar-induced atherosclerosis in the carotid arteries of LDLr-/- mice. In addition, denudation of the carotid artery, as a model for postinterventional lesion formation, resulted in delayed endothelial replacement and significantly increased neointima formation on VEGFR2 vaccination. CONCLUSIONS: These data indicate the prominent role of VEGFR2+ cells in cardiovascular diseases and show that induction of cellular immunity against atherosclerosis-associated cells by means of DNA vaccination may contribute to the development of novel therapies against atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Hipercolesterolemia/terapia , Inmunoterapia Activa/métodos , Neovascularización Patológica/prevención & control , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Administración Oral , Animales , Aterosclerosis/patología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cocultivo , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Miembro Posterior , Histocitoquímica , Inmunidad Celular/inmunología , Isquemia/terapia , Ratones , Vacunas de ADN/administración & dosificación
17.
Clin Cancer Res ; 24(1): 189-196, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972044

RESUMEN

Purpose: In 2010, a Children's Oncology Group (COG) phase III randomized trial for patients with high-risk neuroblastoma (ANBL0032) demonstrated improved event-free survival (EFS) and overall survival (OS) following treatment with an immunotherapy regimen of dinutuximab, GM-CSF, IL2, and isotretinoin compared with treatment with isotretinoin alone. Dinutuximab, a chimeric anti-GD2 monoclonal antibody, acts in part via natural killer (NK) cells. Killer immunoglobulin-like receptors (KIR) on NK cells and their interactions with KIR-ligands can influence NK cell function. We investigated whether KIR/KIR-ligand genotypes were associated with EFS or OS in this trial.Experimental Design: We genotyped patients from COG study ANBL0032 and evaluated the effect of KIR/KIR-ligand genotypes on clinical outcomes. Cox regression models and log-rank tests were used to evaluate associations of EFS and OS with KIR/KIR-ligand genotypes.Results: In this trial, patients with the "all KIR-ligands present" genotype as well as patients with inhibitory KIR2DL2 with its ligand (HLA-C1) together with inhibitory KIR3DL1 with its ligand (HLA-Bw4) were associated with improved outcome if they received immunotherapy. In contrast, for patients with the complementary KIR/KIR-ligand genotypes, clinical outcome was not significantly different for patients who received immunotherapy versus those receiving isotretinoin alone.Conclusions: These data show that administration of immunotherapy is associated with improved outcome for neuroblastoma patients with certain KIR/KIR-ligand genotypes, although this was not seen for patients with other KIR/KIR-ligand genotypes. Further investigation of KIR/KIR-ligand genotypes may clarify their role in cancer immunotherapy and may enable KIR/KIR-ligand genotyping to be used prospectively for identifying patients likely to benefit from certain cancer immunotherapy regimens. Clin Cancer Res; 24(1); 189-96. ©2017 AACRSee related commentary by Cheung and Hsu, p. 3.


Asunto(s)
Genotipo , Neuroblastoma/genética , Neuroblastoma/mortalidad , Receptores KIR/genética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Ensayos Clínicos Fase III como Asunto , Femenino , Humanos , Inmunoterapia , Ligandos , Masculino , Neuroblastoma/inmunología , Neuroblastoma/terapia , Receptores KIR2DL1/genética , Receptores KIR2DL1/metabolismo , Receptores KIR3DL1/genética , Receptores KIR3DL1/metabolismo
18.
J Clin Invest ; 113(12): 1792-8, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15199414

RESUMEN

A lack of relevant animal models has hampered preclinical screening and critical evaluation of the efficacy of human vaccines in vivo. Carcinoembryonic antigen-A2Kb (CEA-A2Kb) double transgenic mice provide a biologically relevant model for preclinical screening and critical evaluation of human CEA vaccine efficacy in vivo, particularly because such animals are peripherally tolerant of CEA. We established the utility of this model by demonstrating that an oral DNA minigene vaccine induces effective HLA-A2-restricted, CEA-specific antitumor CTL responses. This finding is supported by three lines of evidence: (a). an effective HLA-A2-restricted, CEA(691)-specific CTL response; (b). specific in vitro killing of CEA-A2Kb transduced MC-38 colon carcinoma cells; and (c). protective immunity induced in vaccinated mice against challenges of these tumor cells. Importantly, peripheral T cell tolerance against CEA in CEA-A2Kb double transgenic mice was broken by the CEA(691) (IMIGVLVGV) minigene vaccine. In conclusion, CEA-A2Kb double transgenic mice were demonstrated to be good candidates for in vivo testing of human CEA-based vaccines. This result suggests a potential for these vaccines in future human vaccine development. The feasibility of using nonmutated self-antigens as targets for therapeutic vaccinations was indicated, provided that such antigens are presented in an immunogenic context; that is, as a DNA minigene in a bacterial carrier system.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Vectores Genéticos/genética , Vacunas de ADN , Animales , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Vectores Genéticos/metabolismo , Antígeno HLA-A2/inmunología , Humanos , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Citotóxicos/inmunología
19.
J Clin Invest ; 110(2): 185-92, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12122110

RESUMEN

Development of tumor immunotherapies focuses on inducing autoimmune responses against tumor-associated self-antigens primarily encoded by normal, unmutated genes. We hypothesized that such responses could be elicited by T cell homeostatic proliferation in the periphery, involving expansion of T cells recognizing self-MHC/peptide ligands. Herein, we demonstrate that sublethally irradiated lymphopenic mice transfused with autologous or syngeneic T cells showed tumor growth inhibition when challenged with melanoma or colon carcinoma cells. Importantly, the antitumor response depended on homeostatic expansion of a polyclonal T cell population within lymph nodes. This response was effective even for established tumors, was characterized by CD8(+) T cell-mediated tumor-specific cytotoxicity and IFN-gamma production, and was associated with long-term memory. The results indicate that concomitant induction of the physiologic processes of homeostatic T cell proliferation and tumor antigen presentation in lymph nodes triggers a beneficial antitumor autoimmune response.


Asunto(s)
Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Antígenos de Neoplasias , Autoantígenos , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Citotoxicidad Inmunológica , Homeostasis , Memoria Inmunológica , Inmunoterapia , Interferón gamma/biosíntesis , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/trasplante , Vitíligo/inmunología , Vitíligo/patología
20.
Clin Cancer Res ; 12(6): 1750-9, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16551859

RESUMEN

PURPOSE: Evaluate the clinical safety, toxicity, immune activation/modulation, and maximal tolerated dose of hu14.18-IL2 (EMD 273063) in pediatric patients with recurrent/refractory neuroblastoma and other GD2-positive solid tumors. EXPERIMENTAL DESIGN: Twenty-seven pediatric patients with recurrent/refractory neuroblastoma and one with melanoma were treated with a humanized anti-GD2 monoclonal antibody linked to human interleukin 2 (IL-2). Cohorts of patients received hu14.18-IL2, administered i.v. over 4 hours for three consecutive days, at varying doses. Patients with stable disease, partial, or complete responses were eligible to receive up to three additional courses of therapy. RESULTS: Most of the clinical toxicities were anticipated and similar to those reported with IL-2 and anti-GD2 monoclonal antibody therapy and to those noted in the initial phase I study of hu14.18-IL2 in adults with metastatic melanoma. The maximal tolerated dose was determined to be 12 mg/m2/d, with agent-related dose-limiting toxicities of hypotension, allergic reaction, blurred vision, neutropenia, thrombocytopenia, and leukopenia. Three patients developed dose-limiting toxicity during course 1; seven patients in courses 2 to 4. Two patients required dopamine for hypotension. There were no treatment-related deaths, and all toxicity was reversible. Treatment with hu14.18-IL2 led to immune activation/modulation as evidenced by elevated serum levels of soluble IL-2 receptor alpha (sIL2Ralpha) and lymphocytosis. The median half-life of hu14.18-IL2 was 3.1 hours. There were no measurable complete or partial responses to hu14.18-IL2 in this study; however, three patients did show evidence of antitumor activity. CONCLUSION: Hu14.18-IL2 (EMD 273063) can be administered safely with reversible toxicities in pediatric patients at doses that induce immune activation. A phase II clinical trial of hu14.18-IL2, administered at a dose of 12 mg/m2/d x 3 days repeated every 28 days, will be done in pediatric patients with recurrent/refractory neuroblastoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Interleucina-2/uso terapéutico , Melanoma/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Adolescente , Anemia/inducido químicamente , Anticuerpos/sangre , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Área Bajo la Curva , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Femenino , Fiebre/inducido químicamente , Humanos , Hipotensión/inducido químicamente , Infusiones Intravenosas , Interleucina-2/efectos adversos , Interleucina-2/farmacocinética , Masculino , Melanoma/patología , Tasa de Depuración Metabólica , Recurrencia Local de Neoplasia , Neuroblastoma/patología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA