Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
1.
J BUON ; 14 Suppl 1: S111-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19785053

RESUMEN

PURPOSE: Dendritic cell (DC)-based tumor vaccines have been tested extensively to treat cancer patients. However, the results of several DC-based clinical trials have been disappointing. Amelioration of such a modality for cancer treatment seems warranted, i.e. by improving DC immunogenicity and polarization. The goal of our study was to evaluate the potential for immune activation of human DCs by incubating them in vitro with the Newcastle Disease Virus (NDV), a paramyxovirus with strong immunostimulatory properties. RESULTS: In vitro infection with NDV of human monocyte-derived DCs--generated from peripheral monocytes cultured with IL-4 and GM-CSF--induces the generation of viral M gene transcripts and RIG-I expression within DCs. Expression of both genes was increased upon co-stimulation with LPS. Surprisingly, LPS and NDV had opposite effects on induction of interferon (IFN)-alpha. Furthermore, NDV induced DC maturation (as measured by TNF-alpha production and CD80 cell surface expression) only in the presence of LPS. Most interestingly, an optimal combination of NDV and LPS caused polarization of the DCs to Th1 type cytokines with a high ratio of interleukin (IL)-12 to IL-10. CONCLUSION: These in vitro results provide a means and protocol for maturation and activation of DCs with enhanced and sustained T helper type 1-polarizing capacity. Such pretreated DCs may significantly improve DC-based cancer immunotherapy. The data encourage the use of RNA-based viral vectors as potential novel and powerful gene transfer modality for cytoplasmic gene expression in professional antigen-presenting cells (APC) to overcome immunosuppression in cancer patients.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/inmunología , Inmunoterapia/métodos , Monocitos/inmunología , Neoplasias/inmunología , Virus de la Enfermedad de Newcastle/inmunología , Animales , ADN Complementario/genética , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Citometría de Flujo , Humanos , Inmunidad Innata , Lipopolisacáridos/farmacología , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/virología , Neoplasias/terapia , Reacción en Cadena de la Polimerasa , ARN/genética , ARN/aislamiento & purificación
2.
Cancer Immunol Immunother ; 58(1): 61-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18488223

RESUMEN

PURPOSE: Metastatic disease is a major cause of mortality in colorectal cancer patients. Even after complete resection of isolated liver metastases, recurrence develops in the majority of patients. Therefore, development of strategies to prevent recurrent liver metastases is of major clinical importance. The present prospectively randomised phase III trial investigates the efficiency of active specific immunotherapy (ASI) after liver resection for hepatic metastases of colorectal cancer. METHODS: Patients with histologically confirmed liver metastases from colorectal cancer were randomised to the vaccination or control group. After complete resection of liver metastases, patients randomised to the vaccination group received six doses of Newcastle disease virus (NDV) infected autologous tumour cell vaccine (ATV-NDV). The primary end-point was overall survival, secondary end-points were disease-free survival and metastases-free survival. RESULTS: Fifty-one patients were enrolled in the study with 50 patients available for analysis. The follow-up period was 116.1 +/- 23.8 month in the vaccination arm and 112.4 +/- 18.5 month in the control group. In the total patient group, no differences in the primary and secondary end-points were detected. Most interestingly, subgroup analysis revealed a significant advantage for vaccinated colon cancer patients with respect to overall survival [hazard ratio: 3.3; 95%, confidence interval (CI): 1.0-10.4; P = 0.042] and metastases-free survival (hazard ratio: 2.7; 95%, CI: 1.0-7.4; P = 0.047) in the intention-to-treat analysis. CONCLUSION: Active specific immunotherapy in unselected colorectal cancer patients was not effective for prevention of recurrent metastatic disease. However, in colon cancer patients, ASI with ATV-NDV appears to be beneficial prolonging overall and metastases-free survival.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/tratamiento farmacológico , Hepatectomía , Inmunoterapia Activa , Neoplasias Hepáticas/tratamiento farmacológico , Virus de la Enfermedad de Newcastle , Adulto , Anciano , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Terapia Combinada , Femenino , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Virus de la Enfermedad de Newcastle/inmunología
4.
Gene Ther ; 14(23): 1639-49, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17914407

RESUMEN

This is the first report describing recombinant (rec) Newcastle disease virus (NDV) as vector for gene therapy of cancer. The gene encoding granulocyte/macrophage colony-stimulating factor (GM-CSF) was inserted as an additional transcription unit at two different positions into the NDV genome. The rec virus with the strongest production of the gene product (rec(GM-CSF)) was selected for our study. The insertion of the new foreign gene did neither affect the main features of NDV replication nor its tumor selectivity. The gene product was biologically active and stable. Tumor vaccine cells infected by rec(GM-CSF) stimulated human peripheral blood mononuclear cells (PBMC) to exert antitumor bystander effects in vitro in a tumor neutralization assay. These effects were significantly increased when compared to vaccine infected by rec(-) virus. Furthermore, rec(GM-CSF) led to a much higher interferon-alpha (IFN-alpha) production than rec(-) when added as virus or as virus-modified vaccine to PBMC. Two distinct cell types, monocytes and plasmacytoid dendritic cells were shown to contribute to the augmented IFN-alpha response of PBMC. In conclusion, the already inherent anti-neoplastic and immunostimulatory properties of NDV could be further augmented by the introduction of a therapeutic gene whose product initiates a broad cascade of immunological effects in the microenvironment of the vaccine.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/administración & dosificación , Vectores Genéticos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Virus de la Enfermedad de Newcastle/genética , Viroterapia Oncolítica/métodos , Apoptosis , Neoplasias de la Mama/inmunología , Efecto Espectador , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/virología , Citometría de Flujo , Expresión Génica , Ingeniería Genética , Humanos , Interferón-alfa/inmunología , Monocitos/inmunología , Monocitos/virología , Vacunas Sintéticas/administración & dosificación
5.
Int J Oncol ; 28(6): 1419-28, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16685444

RESUMEN

Pancreatic carcinoma is a very aggressive disease and little is known about its immunobiology. We here describe the presence in pancreatic cancer patients of spontaneously induced functional CD4 and CD8 memory/effector T cells reactive to autologous tumor cells or to the pancreatic cancer associated antigen, MUC-1. Such specific cells were present in the bone marrow or peripheral blood of most of the 23 tested patients. Low dose stimulation of primary cultures of pancreatic cancer cells with 500 IU/ml IFN-gamma for 72 h enhanced HLA-I expression and induced the de novo expression of HLA-II molecules. This led to a much better immune recognition by autologous HLA-I restricted and purified CD8 T cells and allowed tumor cell recognition by HLA-II restricted purified CD4 T-helper cells. Thus, interferon-gamma appears to be a useful adjuvant cytokine to enhance the immunogenicity of a patients' tumor cells and their recognition by tumor reactive immune cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/farmacología , Neoplasias Pancreáticas/inmunología , Anciano , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Antígenos HLA-D/análisis , Antígenos de Histocompatibilidad Clase I/análisis , Humanos , Memoria Inmunológica/efectos de los fármacos , Interleucina-4/farmacología , Complejo Mayor de Histocompatibilidad , Masculino , Persona de Mediana Edad
6.
Genomics ; 85(2): 231-7, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15676281

RESUMEN

The product of the oncosuppressor adenomatous polyposis coli (APC) gene is involved in cell cycle arrest and apoptosis and its loss of function is associated with the development of colorectal carcinogenesis. Its transcriptional regulation seems rather complex and has not been completely elucidated up to now. In an attempt to identify the transcription start sites for the mouse Apc gene we have detected a novel transcript in mouse embryonic stem (ES) cells and colon tissue. This transcript contains an untranslated exon, whose flanking sequences exhibited strong promoter activity in transient transfection experiments. These results suggest that we have identified a novel promoter for the mouse Apc gene, localized about 40 kb upstream of the initiating methionine, which drives expression of the unique Apc transcript type detected in undifferentiated totipotent ES cells. Transcripts bearing the novel exon combined either with exon 1 or with exon 2 were detected in all mouse tissues tested.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Metionina/genética , Regiones Promotoras Genéticas , Sitio de Iniciación de la Transcripción , Región de Flanqueo 3' , Región de Flanqueo 5' , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Colon/fisiología , Exones , Orden Génico , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Células Madre Totipotentes/fisiología
7.
Int J Oncol ; 21(4): 685-94, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12239606

RESUMEN

We demonstrated before that primary operated breast cancer patients contain in their bone marrow (BM) cancer reactive memory T cells (MTC) which have to be re-activated to become tumor infiltrating effector cells. The aim of this study was to optimize an ex vivo stimulation protocol for MTC based on autologous dendritic cells (DC). As source of tumor antigens we used lysates from unmodified tumor cells or from tumor cells infected with Newcastle Disease Virus (NDV) which contain IFN-alpha inducing viral dsRNA as one danger signal. DC from breast cancer patients were pulsed with lysates from the MCF-7 breast cancer line (Tu-L) or from NDV infected MCF-7 cells (TuN-L, viral oncolysates) and compared for stimulatory capacity in an ELISPOT response of autologous BM derived MTC. To analyze potential further danger signals derived from NDV infection, we employed MALDI mass spectrometry, Western blots, FACS cytometry and ELISA tests. DC pulsed with viral oncolysates showed increased expression of co-stimulatory molecules in comparison to Tu-L pulsed DC and induced significantly higher ELISPOT MTC responses. Supernatants from co-cultures of MTC and TuN-L pulsed DC contained increased titers of IFN-alpha and IL-15. NDV infection of tumor cells resulted in a number of differences in protein expression including a heat-shock protein (HSP27) which became phosphorylated. The results suggest that a DC preparation pulsed with viral oncolysate includes danger signals (e.g. dsRNA, cytokines, HSP molecules) and is superior for MTC stimulation to a DC preparation pulsed with lysate from non-infected tumor cells.


Asunto(s)
Neoplasias de la Mama/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Proteínas de Choque Térmico , Linfocitos T/metabolismo , Western Blotting , Neoplasias de la Mama/metabolismo , Separación Celular , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Proteínas de Choque Térmico HSP27 , Humanos , Técnicas para Inmunoenzimas , Memoria Inmunológica , Interferón-alfa/metabolismo , Interleucina-15/sangre , Chaperonas Moleculares , Proteínas de Neoplasias/sangre , Virus de la Enfermedad de Newcastle/metabolismo , Fosforilación , ARN Viral/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Tiempo , Células Tumorales Cultivadas , Regulación hacia Arriba
8.
Int J Oncol ; 21(1): 85-93, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12063554

RESUMEN

In order to analyse immune-stimulatory effects of infection of human tumor cells with Newcastle Disease Virus (NDV), gamma-irradiated human breast carcinoma, colon-carcinoma or glioblastoma cells from defined cell lines were modified either by true infection with live virus or by cell surface adsorption of UV-inactivated replication deficient virus. Modification with live but not inactive NDV induced in all human tumor cells IFN-beta and the chemokines RANTES and IFN-gamma-inducible protein-10 (IP-10). In addition, infection by live NDV induced upregulation of HLA-ABC-molecules in all tumor lines tested and HLA-DR molecules in breast carcinoma lines. Two cell adhesion molecules, ICAM-I (CD54) and LFA-3 (CD58), were also upregulated on human tumor cells after infection with live NDV. When infection of MCF-7 breast carcinoma cells by NDV was performed in the presence of neutralizing anti-IFN-beta antibodies no upregulation of HLA molecules was observed suggesting an important role of IFN-beta in this process. Forty-eight to 72 hours after infection of the irradiated tumor cells with live NDV, many tumor cells were dead or in early or late stages of apoptosis. These results provide explanations for the function of the virus-modified autologous tumor vaccine ATV-NDV with which promising clinical results have already been obtained.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/virología , Citocinas/biosíntesis , Antígenos HLA-DR/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón Tipo I/biosíntesis , Virus de la Enfermedad de Newcastle/fisiología , Neoplasias de la Mama/patología , Antígenos CD58/metabolismo , Quimiocina CCL5/metabolismo , Quimiocina CXCL10 , Quimiocinas CXC/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferón beta/metabolismo , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/virología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Replicación Viral
9.
Parasitol Res ; 88(4): 368-75, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11999027

RESUMEN

DNA-based vaccine technology was used to immunize against the schistosome digestive enzyme, cathepsin D aspartic proteinase. The cDNA coding for Schistosomajaponicum aspartic proteinase was cloned in a mammalian expression vector under control of the CMV promoter/enhancer and expressed for the first time in transfected mammalian cells as well as in mice immunized--by means of intra-ear pinna injection--with the aspartic proteinase-encoding DNA construct. Mice developed antibodies which recognized the native protein in homogenates of S. japonicum worms and reacted with the gut and, to a much lesser degree, with the parenchyma of the parasites in cryostat sections. It was noteworthy that the vaccinated mouse sera did not detectably cross-react with S. mansoni antigens either in homogenates or on cryostat sections. By contrast, infection sera of mice or humans strongly cross-reacted with both schistosome species. We conclude that DNA vaccination can induce species-restricted antibody responses against schistosome proteins. The implications of this previously unrecognized specificity are discussed.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Catepsina D/inmunología , ADN Complementario/inmunología , ADN de Helmintos/inmunología , Schistosoma japonicum/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antihelmínticos/biosíntesis , Células COS , Catepsina D/biosíntesis , Catepsina D/genética , Chlorocebus aethiops , Reacciones Cruzadas , ADN Complementario/genética , ADN de Helmintos/genética , Femenino , Técnica del Anticuerpo Fluorescente Directa , Humanos , Ratones , Ratones Endogámicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Transfección , Vacunación
10.
Parasitology ; 124(Pt 3): 301-6, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11922431

RESUMEN

A DNA-construct coding for the elastase of the parasite Schistosoma mansoni was prepared from adult S. mansoni worm RNA which was reverse transcribed into cDNA. The gene coding for the elastase was amplified using primers specific for the sequence of cercarial elastase and was cloned into a mammalian expression vector. Expression of the elastase gene at the transcriptional level was achieved for the first time in transfected mammalian cells (COS-7) and was also successful in muscle tissue of mice injected with the DNA-construct. These mice developed antibodies recognizing in Western blots the elastase from cercarial secretions. Also, these antibodies reacted in immunofluorescence tests with the preacetabular glands of cercariae, i.e. the site of origin for elastase. Thus, the DNA-construct induced the expression of elastase in mice and formation of antibodies that recognized the native antigen.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , ADN Complementario/inmunología , Elastasa Pancreática/genética , Elastasa Pancreática/inmunología , Schistosoma mansoni/enzimología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Biomphalaria/parasitología , Western Blotting , Células COS , Chlorocebus aethiops , ADN Complementario/genética , Femenino , Ratones , Hibridación de Ácido Nucleico , Elastasa Pancreática/biosíntesis , ARN Protozoario/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Schistosoma mansoni/genética , Schistosoma mansoni/inmunología , Organismos Libres de Patógenos Específicos , Transfección
11.
Eur J Clin Invest ; 32(3): 207-17, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895473

RESUMEN

BACKGROUND: Fusion of tumour cells with dendritic cells (DC) is a powerful new technology to increase tumour vaccine immunogenicity. The aim of this study was to compare fusion protocols with syngenic DCs with respect to the efficiency of polyethylen-glycol-(PEG) and electric pulse-mediated fusions for induction of protective anti-tumour immune responses. As a model we chose a low immunogenic and metastatic murine mammary carcinoma cell line, which mimics clinically relevant tumour features. METHODS: FACS-staining, chromium release assay, therapeutic immunization, adoptive transfer. RESULTS: We show that the parental line with low cell surface expression of MHC molecules as well as a lacZ transfectant becomes highly immunogenic upon fusion with DCs. This was true for PEG- as well as for electro-fused cells. Immunization with products of DCs and tumour cells cocultivated for 16 h without the fusing agent PEG also caused induction of profound anti-tumour immunity, while this was not the case when using parental tumour cells or their lacZ transfectants as vaccines. Immune protection against the parental tumour cells after vaccination with fused cells was long-lasting and could be transferred via immune spleen cells into immuno-incompetent nude (nu/nu) mice. CONCLUSION: Fusion products of DA3(hi) mammary carcinoma cells and DCs produced by an electric pulse were similar to those produced by PEG fusion with regard to vaccine potency in prophylactic antitumour immunization assays in vivo. Therefore, both techniques seem to be promising for clinical application.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Fusión Celular/métodos , Células Dendríticas/citología , Inmunoterapia/métodos , Neoplasias Mamarias Experimentales/terapia , Animales , Antineoplásicos/farmacología , Técnicas de Cocultivo , Células Dendríticas/inmunología , Electroporación , Expresión Génica/efectos de los fármacos , Inmunocompetencia , Interferón-alfa/farmacología , Operón Lac , Complejo Mayor de Histocompatibilidad/genética , Neoplasias Mamarias Experimentales/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Desnudos , Ratones SCID , Polietilenglicoles , beta-Galactosidasa/genética
12.
Br J Cancer ; 86(5): 828-36, 2002 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-11875749

RESUMEN

The endogenous viral superantigen 7 in DBA/2 mice serves as a target antigen on syngeneic ESb-MP lymphoma cells for allogeneic graft-vs-leukaemia reactive cells. Allogeneic viral superantigen 7 reactive Vbeta6+ T cells are able to transfer graft-vs-leukaemia reactivity and to kill specifically viral superantigen 7+ ESb-MP tumour cells in vitro. Here we elucidate the mechanism of this superantigen specific cell lysis. Already 10 min after co-incubation with in vitro stimulated Vbeta6+ T cells, viral superantigen 7+ ESb-MP tumour cells show an apoptotic phenotype (Annexin V-positivity, DNA-fragmentation). This extremely rapid type of cell death is not mediated by the death inducing ligands CD95L, TRAIL and TNF but by perforin and granzyme B. Surprisingly, neither mitochondria nor any of the known caspases appear to be involved in this type of tumour cell killing. In contrast, nitric oxide, released by activated macrophages and endothelial cells, induces in the same tumour cells another type of apoptosis which is much slower and involves mitochondria and caspase activation. A synergistic effect between the two different effector mechanisms of superantigen reactive donor cytotoxic T lymphocytes and nitric oxide releasing host macrophages and endothelial cells might explain the effective immune rejection of even advanced metastasised cancer in this graft-vs-leukaemia animal model.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Leucemia de Células T/patología , Linfoma de Células T/patología , Glicoproteínas de Membrana/biosíntesis , Serina Endopeptidasas/biosíntesis , Superantígenos/inmunología , Linfocitos T/inmunología , Animales , Granzimas , Ligandos , Glicoproteínas de Membrana/farmacología , Ratones , Perforina , Proteínas Citotóxicas Formadoras de Poros , Serina Endopeptidasas/farmacología , Células Tumorales Cultivadas
13.
Int J Oncol ; 20(2): 247-53, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11788884

RESUMEN

Dendritic cells (DCs) currently used for vaccination in clinical studies to induce immunity against malignant cells are normally generated from peripheral blood-derived monocytes. Here we studied conditions for the generation of DCs from unseparated human bone marrow (BM) mononuclear cells and compared them functionally with DCs from blood. The two types of DCs, from bone marrow (BM-DC) and peripheral blood (BL-DC), were generated in parallel from the same normal healthy donors by culturing in serum-free X-VIVO 20 medium containing GM-CSF and IL-4, and then the phenotypes and functions were compared. BM-DC generation occurred in 14 days and involved proliferative expansion from CD34 stem cells and differentiation while BL-DC generation occurred in 7 days from CD14 monocytes and involved only differentiation. A 7- to 25-fold higher number of DCs could be obtained from BM than from blood. BM-DC had similar phenotypes as BL-DC. The capacity to stimulate MLR reactivity in allogeneic T lymphocytes was higher with BM-DC than that with BL-DC. Also, the capacity to stimulate autologous memory T cell responses to tetanus toxoid (TT) or tuberculin (PPD) was higher with BM-DC than with BL-DC. These results suggest that BM-DC as produced here may be a very economic and useful source of professional antigen-presenting cells for anti-tumor immunotherapeutic protocols.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Células Dendríticas/citología , Leucocitos Mononucleares/citología , Presentación de Antígeno , Tamaño de la Célula , Células Dendríticas/fisiología , Endocitosis , Citometría de Flujo , Humanos , Prueba de Cultivo Mixto de Linfocitos , Fenotipo
14.
Vaccine ; 20(3-4): 439-47, 2001 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-11672907

RESUMEN

DNA-based vaccine technology was used to induce an immune response in mice against a schistosome cysteine proteinase, asparaginyl endopeptidase (Sm32). The cDNA coding for Sm32 was cloned in a mammalian expression vector under control of the CMV promoter/enhancer and expressed for the first time in transfected mammalian cells as well as in mice immunized with the Sm32-encoding DNA construct. These mice developed antibodies which recognized the native protein not only in homogenates of Schistosoma mansoni worms but also in the gut on cryostat sections of the parasites. This DNA vaccine led to an anti-fecundity effect: female worms of a challenge infection produced 37% less eggs than those growing in naïve mice. The results suggest that Sm32 may be a candidate antigen for the generation of an anti-pathology vaccine against schistosomes.


Asunto(s)
Cisteína Endopeptidasas/inmunología , Fertilidad , Proteínas del Helminto , Schistosoma mansoni/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antihelmínticos/sangre , Células COS , Cisteína Endopeptidasas/genética , Femenino , Ratones , Transcripción Genética , Vacunación
15.
Semin Cancer Biol ; 11(4): 285-95, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11513564

RESUMEN

We conclude from animal tumour model studies that T cell immunity can play an essential role in the induction and maintenance of tumour dormancy. Evidence was found in tumour dormancy situations for active control of proliferating tumour cells by CD8 memory T cells leading to a long-term balance in the bone marrow between low numbers of tumour cells and immunological memory. In breast cancer patients, too, the bone marrow may represent a privileged compartment for tumour dormancy and immunological memory. Upon restimulation with tumour antigen pulsed autologous dendritic cells, bone marrow-derived memory T cells from cancer patients could be shown to exist and to become activated into potent anti-tumour effector cells.


Asunto(s)
Supervivencia Celular/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Células Dendríticas/inmunología , Humanos , Inmunidad Celular , Memoria Inmunológica
16.
Adv Cancer Res ; 82: 107-31, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11447761

RESUMEN

Nitric oxide (NO), an important molecule involved in neurotransmission, vascular homeostasis, immune regulation, and host defense, is generated from a guanido nitrogen of L-arginine by the family of NO synthase enzymes. Large amounts of NO produced for relatively long periods of time (days to weeks) by inducible NO synthase in macrophages and vascular endothelial cells after challenge with lipopolysaccharide or cytokines (such as interferons, tumor necrosis factor-alpha, and interleukin-1), are cytotoxic for various pathogens and tumor cells. This cytotoxic effect against tumor cells was found to be associated with apoptosis (programmed cell death). The mechanism of NO-mediated apoptosis involves accumulation of the tumor suppressor protein p53, damage of different mitochondrial functions, alterations in the expression of members of the Bcl-2 family, activation of the caspase cascade, and DNA fragmentation. Depending on the amount, duration, and the site of NO production, this molecule may not only mediate apoptosis in target cells but also protect cells from apoptosis induced by other apoptotic stimuli. In this review, we will concentrate on the current knowledge about the role of NO as an effector of apoptosis in tumor cells and discuss the mechanisms of NO-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias/patología , Óxido Nítrico/farmacología , Animales , Humanos
17.
Nat Med ; 7(4): 452-8, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283672

RESUMEN

In an analysis of 84 primary-operated breast cancer patients and 11 healthy donors, we found that the bone marrow of most patients contained memory T cells with specificity for tumor-associated antigens. Patients' bone marrow and peripheral blood contained CD8+ T cells that specifically bound HLA/peptide tetramers. In short-term culture with autologous dendritic cells pre-pulsed with tumor lysates, patients' memory T cells from bone marrow (but not peripheral blood) could be specifically reactivated to interferon-gamma-producing and cytotoxic effector cells. A single transfer of restimulated bone-marrow T cells into NOD/SCID mice caused regression of autologous tumor xenotransplants associated with infiltration by human T cells and tumor-cell apoptosis and necrosis. T cells from peripheral blood showed much lower anti-tumor reactivity. Our findings reveal an innate, specific recognition of breast cancer antigens and point to a possible novel cancer therapy using patients' bone-marrow-derived memory T cells.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Apoptosis , Trasplante de Médula Ósea , Neoplasias de la Mama/patología , Femenino , Antígeno HLA-A2/metabolismo , Humanos , Memoria Inmunológica , Técnicas In Vitro , Interferón gamma/biosíntesis , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mucina-1/química , Mucina-1/inmunología , Necrosis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Receptor ErbB-2/química , Receptor ErbB-2/inmunología , Linfocitos T Citotóxicos/inmunología , Trasplante Autólogo , Trasplante Heterólogo
18.
Int J Cancer ; 92(1): 96-105, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11279612

RESUMEN

Previous studies with animal tumors showed that bone marrow (BM) is a privileged site where potentially lethal tumor cells are controlled in a dormant state by the immune system. Here, we investigated BM of breast cancer patients with respect to tumor cell content, immune activation status and memory T-cell content. BM-derived cells from primary operated breast cancer patients (n = 90) were compared with those from healthy donors (n = 10) and also with cells from respective blood samples. Cytokeratin 19-positive tumor cells were detected by nested polymerase chain reaction. Three-color flow cytometry was used to identify numbers and activation state of T cells, natural killer (NK) cells, monocytes/macrophages and subsets by a panel of monoclonal antibodies (mAbs). The proportion of memory T cells among the CD4 and CD8 T cells was much higher in BM of cancer patients than in healthy donors (p < 0.001). The extent of memory T-cell increase was related to the size of the primary tumor. Patient-derived BM memory CD8 T cells could be shown to contain specific HLA-A2/Her-2/neu(369-377) tetramer binding cells. Patients with disseminated tumor cells in their BM had more memory CD4 T cells and more CD56(+) CD8(+) cells than patients with tumor cell-negative BM. Only some of the immunological changes seen in BM samples of cancer patients were also detectable in peripheral blood samples. Our hypothesis that BM is a special compartment for immunological memory and tumor dormancy is supported by the above findings. The overall results reveal that BM is a valuable additional compartment for immune diagnosis in pathological conditions and possibly for follow-up treatment strategies.


Asunto(s)
Médula Ósea/inmunología , Neoplasias de la Mama/inmunología , Memoria Inmunológica , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Médula Ósea/patología , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Antígeno CD56/análisis , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Recuento de Células , Femenino , Citometría de Flujo , Antígeno HLA-A2/análisis , Humanos , Queratinas/análisis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Recuento de Linfocitos , Macrófagos/inmunología , Macrófagos/patología , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/patología , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Receptor ErbB-2/análisis , Linfocitos T/patología , Células Tumorales Cultivadas
19.
Exp Cell Res ; 265(2): 274-82, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11302692

RESUMEN

We have recently shown that nitric-oxide (NO)-induced apoptosis in Jurkat human leukemia cells requires degradation of mitochondria phospholipid cardiolipin, cytochrome c release, and activation of caspase-9 and caspase-3. Moreover, an inhibitor of lipid peroxidation, Trolox, suppressed apoptosis in Jurkat cells induced by NO donor glycerol trinitrate. Here we demonstrate that this antiapoptotic effect of Trolox occurred despite massive release of the mitochondrial protein cytochrome c into the cytosol and mitochondrial damage. Incubation with Trolox caused a profound reduction of intracellular ATP concentration in Jurkat cells treated by NO. Trolox prevented cardiolipin degradation and caused its accumulation in Jurkat cells. Furthermore, Trolox markedly downregulated the NO-mediated activation of caspase-9 and caspase-3. Caspase-9 is known to be activated by released cytochrome c and together with caspase-3 is considered the most proximal to mitochondria. Our results suggest that the targets of the antiapoptotic effect of Trolox are located downstream of the mitochondria and that caspase activation and subsequent apoptosis could be blocked even in the presence of cytochrome c released from the mitochondria.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/fisiología , Cromanos/farmacología , Grupo Citocromo c/metabolismo , Mitocondrias/efectos de los fármacos , Óxido Nítrico/metabolismo , Adenosina Trifosfato/metabolismo , Cardiolipinas/metabolismo , Caspasas/metabolismo , Separación Celular , Citometría de Flujo , Humanos , Células Jurkat , Microscopía Fluorescente , Mitocondrias/metabolismo , Vitamina E/análogos & derivados
20.
Int J Oncol ; 18(5): 945-52, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11295039

RESUMEN

Newcastle Disease Virus (NDV) has interesting anti-neoplastic and pleiotropic immune stimulatory properties. The virus preferentially replicates in and kills tumor cells and appears to be safe and to varying degrees effective in phase II-clinical studies in the US and in Europe. Here we have compared various lytic and non-lytic strains of NDV with regard to their antitumor effects after local or systemic application. As tumor models we used human metastatic melanoma xenotransplants in nude mice and murine metastatic colon carcinoma (CT26), renal carcinoma (Renca) and lymphoma (ESb) cell lines. Intra or peri-tumoral application of NDV or NDV infected tumor cells showed more pronounced antitumor activity than systemic application even when in the latter case much higher dose ranges were used. In the CT26 colon carcinoma model the non-lytic strain Ulster showed stronger antitumor activity than the lytic strain 73T. In the human MeWo melanoma xentransplant model strong antitumor bystander effects were observed by 20% admixture of melanoma cells pre-infected in vitro with NDV (either strain Ulster or Italien). Virus therapy of pre-established human melanomas by intra-tumoral injection of NDV was effective with the lytic strain Italien but not with the non-lytic strain Ulster. Systemic anti-metastatic effects were never observed with NDV alone in contrast to previous results obtained with NDV modified tumor vaccines.


Asunto(s)
Inmunoterapia Activa , Neoplasias/terapia , Virus de la Enfermedad de Newcastle/fisiología , Animales , Antígenos Virales/inmunología , Citotoxicidad Inmunológica , Femenino , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias/virología , Análisis de Supervivencia , Linfocitos T Citotóxicos/fisiología , Células Tumorales Cultivadas/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...