Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 15(1): 390-9, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118070

RESUMEN

PURPOSE: CTL-associated antigen 4 (CTLA4)-blocking monoclonal antibodies induce long-term regression of metastatic melanoma in some patients, but the exact mechanism is unknown. In this study, biopsies of selected accessible tumor lesions from patients treated with tremelimumab were examined to further elucidate the mechanism of its antitumor activity. EXPERIMENTAL DESIGN: Fifteen tumor biopsies from 7 patients who had been treated with tremelimumab (CP-675,206) were collected. Samples were analyzed for melanoma markers, immune cell subset markers, the presence of the T regulatory-specific transcription factor FoxP3 and the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO). RESULTS: Clinically responding lesions had diffuse intratumoral infiltrates of CD8(+) T cells that were markedly increased in cases where comparison with a baseline biopsy was available. Nonregressing lesions had sparse, patchy CD8(+) intratumoral infiltrates. Patients with regressing lesions had an increased frequency of CD8(+) cells with or without a concomitant increase in CD4(+) cells. Two of 3 responding patients with paired samples showed a slight increase in the number of FoxP3(+) cells in the postdosing biopsies. In patients with regressing lesions who had paired samples, the intensity of IDO staining in macrophages and/or melanoma cells showed no clear pattern of change postdosing. CONCLUSIONS: Administration of tremelimumab was associated with massive intratumoral infiltrates of CD8(+) CTLs in patients with regressing tumors but had varying effects on intratumoral infiltrates of CD4(+) and FoxP3(+) cells or intratumoral expression of IDO.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Factores de Transcripción Forkhead/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Linfocitos T Citotóxicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados , Biopsia , Linfocitos T CD4-Positivos/inmunología , Antígeno CTLA-4 , Femenino , Humanos , Masculino , Melanoma/inmunología , Melanoma/terapia , Persona de Mediana Edad
2.
J Immunother ; 31(3): 294-309, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18317358

RESUMEN

We performed a phase 1/2 trial testing the safety, toxicity, and immune response of a vaccine consisting of autologous dendritic cells (DCs) transduced with a replication-defective adenovirus (AdV) encoding the full-length melanoma antigen MART-1/Melan-A (MART-1). This vaccine was designed to activate MART-1-specific CD+8 and CD4+ T cells. Metastatic melanoma patients received 3 injections of 10(6) or 10(7) DCs, delivered intradermally. Cell surface phenotype and cytokine production of the DCs used for the vaccines were tested, and indicated intermediate maturity. CD8+ T-cell responses to MART-1 27-35 were assessed by both major histocompatibility complex class I tetramer and interferon (IFN)-gamma enzyme-linked immunosorbent spot (ELISPOT) before, during, and after each vaccine and CD4+ T-cell responses to MART-1 51-73 were followed by IFN-gamma ELISPOT. We also measured antigen response breadth. Determinant spreading from the immunizing antigen MART-1 to other melanoma antigens [gp100, tyrosinase, human melanoma antigen-A3 (MAGE-A3)] was assessed by IFN-gamma ELISPOT. Twenty-three patients were enrolled and 14 patients received all 3 scheduled DC vaccines. Significant CD8+ and/or CD4+ MART-1-specific T-cell responses were observed in 6/11 and 2/4 patients evaluated, respectively, indicating that the E1-deleted adenovirus encoding the cDNA for MART-1/Melan-A (AdVMART1)/DC vaccine activated both helper and killer T cells in vivo. Responses in CD8+ and CD4+ T cells to additional antigens were noted in 2 patients. The AdVMART1-transduced DC vaccine was safe and immunogenic in patients with metastatic melanoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Inmunoterapia Activa , Melanoma/terapia , Proteínas de Neoplasias/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias/química , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Femenino , Humanos , Epítopos Inmunodominantes/inmunología , Inmunoterapia Activa/efectos adversos , Antígeno MART-1 , Masculino , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/química , Estadificación de Neoplasias , Ingeniería de Proteínas , Transducción Genética , Resultado del Tratamiento
3.
Clin Cancer Res ; 12(9): 2817-25, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16675576

RESUMEN

Alpha-fetoprotein (AFP) is a self protein expressed by fetal liver at high levels, but is transcriptionally repressed at birth. AFP is up-regulated in hepatocellular carcinomas, and patients with active disease could have plasma levels as high as 1 mg/mL. We previously identified four immunodominant HLA-A*0201-restricted peptides [hAFP(137-145) (PLFQVPEPV), hAFP(158-166) (FMNKFIYEI), hAFP(325-334) (GLSPNLNRFL), and hAFP(542-550) (GVALQTMKQ)] derived from human AFP that could stimulate specific T cell responses in healthy donor peripheral blood lymphocytes in vitro. We conducted a phase I/II clinical trial in which HLA-A*0201 patients with AFP-positive hepatocellular carcinoma were immunized with three biweekly intradermal vaccinations of the four AFP peptides pulsed onto autologous dendritic cells (DC). DCs were prepared from adherent peripheral blood mononuclear cells cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4 for 7 days. Sixteen subjects were enrolled and 10 were treated. Peripheral blood lymphocytes were isolated from these patients before, during, and after AFP peptide/DC immunization and were tested ex vivo with MHC tetramer and IFNgamma ELISPOT analysis. Six of 10 subjects expanded statistically significant levels of AFP-specific T cells postvaccine to at least one peptide by MHC tetramer. Also, 6 of 10 subjects increased IFNgamma producing AFP-specific T cell responses to at least one of the peptides postvaccination, by ELISPOT. We conclude that the human T cell repertoire is capable of responding to the AFP self antigen after the administration of AFP peptide-pulsed DC even in an environment of high circulating levels of this oncofetal antigen.


Asunto(s)
Carcinoma Hepatocelular/terapia , Células Dendríticas/trasplante , Neoplasias Hepáticas/terapia , Fragmentos de Péptidos/uso terapéutico , alfa-Fetoproteínas/uso terapéutico , Adulto , Anciano , Secuencia de Aminoácidos , Carcinoma Hepatocelular/inmunología , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/inmunología , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/toxicidad , alfa-Fetoproteínas/toxicidad
4.
J Immunol ; 176(8): 4757-65, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16585569

RESUMEN

Proteasome inhibition results in proapoptotic changes in cancer cells, which may make them more sensitive to immune effector cells. We established a murine model to test whether the proteasome inhibitor bortezomib could sensitize established B16 melanoma tumors to dendritic cell (DC)-activated immune effector cells. Day 3-established s.c. B16 tumors had significantly decreased tumor outgrowth when treated with a combination of bortezomib and DC, regardless of whether the DC were loaded or not with a tumor Ag. In vivo Ab-depletion studies demonstrated that the effector cells were NK and CD8+ cells, but not CD4+ cells. NF-kappaB nuclear transcription factor assay and gene-expression profiling of B16 treated with bortezomib was consistent with inhibition of NF-kappaB target genes leading to a proapoptotic phenotype. In vitro lytic assays demonstrated that TNF-alpha, but not perforin, Fas-ligand, or TRAIL, was responsible for bortezomib-sensitized B16 cytotoxicity. In conclusion, the proteasome inhibitor bortezomib can pharmacologically sensitize tumor cells to the lytic effects of DC-activated immune effector cells.


Asunto(s)
Ácidos Borónicos/farmacología , Células Dendríticas/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Animales , Apoptosis , Bortezomib , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Inmunización , Inmunoterapia , Técnicas In Vitro , Células Asesinas Naturales/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Necrosis Tumoral alfa/farmacología
5.
Cancer Immunol Immunother ; 55(6): 663-71, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16133107

RESUMEN

Dendritic cell (DC) administration to CD8alpha knock-out (CD8alphaKO) mice results in a strong antigen-non-specific protection to a B16 murine melanoma tumor challenge. This response is mediated by lytic NK cells and cytokine-producing CD4 cells. We aimed to determine the signals that guide tumor targeting of this response. CD8alphaKO mice in the C57BL/6 background received subcutaneous (s.c.) injections of immature DC. Mice were challenged in vivo or assayed for lytic activity in vitro to a panel of syngeneic tumors with different levels of MHC class I expression. These studies support the following conclusions: (1) DC administration to CD8alphaKO mice results in protective in vivo responses to syngeneic tumors from epithelial, neuroectodermal and hematopoietic origin; in vivo protection is independent of the level of MHC classes I and II expression. (2) The in vitro lytic activity of DC-activated NK cells from CD8alphaKO mice has sensitive and insensitive targets, which is independent of the cell lineage or the level of inhibitory self-MHC surface molecules. (3) In sensitive targets a putative activating NK ligand in DC-stimulated NK cells from CD8alphaKO mice signals directly to PI3-K, but is distinct from NKG2D.


Asunto(s)
Antígenos CD8/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neoplasias Experimentales/inmunología , Animales , Antígenos CD8/genética , Línea Celular Tumoral , Antígenos de Histocompatibilidad/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/inmunología
6.
Cancer Gene Ther ; 12(6): 516-27, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15775996

RESUMEN

Tumor antigen gene-modified dendritic cells (DC) generates robust antigen-specific protective antitumor responses. Though the role of CD4 positive and CD8 positive cells in the immunological response to gene-modified DC has been well-characterized, the role of NK cells in this response has been somewhat less clear. Owing to the significant contribution of innate immunity in other model systems, we postulated that NK cells would hold a critical position in the generation of an immune response following immunization with tumor antigen-engineered DC. Immunization with MART-1 melanoma antigen-engineered DC in C57BL/6 mice resulted in the generation of antigen-specific cytotoxic T lymphocytes and in vivo protective responses to the murine B16 melanoma. These responses were dependent on the presence of functional NK cells, although NK cells alone were not sufficient in generating protective responses. Adoptive transfer of NK cells into an NK-deficient but T-cell-competent environment restored the protective response to gene-modified DC immunization. In conclusion, protective immunity after tumor antigen gene-modified DC immunization requires collaboration between CD4+ and CD8+ T cells and NK cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/trasplante , Células Asesinas Naturales/inmunología , Melanoma Experimental/prevención & control , Proteínas de Neoplasias/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Femenino , Antígeno MART-1 , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Mutantes , Proteínas de Neoplasias/genética , Células Tumorales Cultivadas
7.
J Immunother ; 27(5): 354-67, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15314544

RESUMEN

MART-1(27-35)-peptide-pulsed immature dendritic cells (DCs) resulted in immunologic and clinical activity in a prior phase 1 trial. A phase 2 cohort expansion was initiated to further characterize the phenotype and cytokine milieu of the DC vaccines and their immunologic activity in vitro and to further examine a possible link between clinical activity and determinant spreading. In an open-label phase 2 trial, 10(7) autologous ex vivo generated DCs pulsed with the HLA-A*0201 immunodominant peptide MART-1(27-35) were administered to 10 subjects with stage II-IV melanoma. The experimental vaccines were administered intradermally in a biweekly schedule for a total of three injections, and blood for immunologic assays was obtained before each administration and at three time points after. DC vaccine preparations had wide intra- and interpatient variability in terms of cell surface markers and preferential cytokine milieu, but they did not correlate with the levels of antigen-specific T cells after vaccination. Of four patients with measurable disease, one had stable disease for 6 months and another has a continued complete response for over 2 years, which is confounded by receiving a closely sequenced CTLA4 blocking antibody. The DC vaccines induced determinant spreading in this subject, and CTLA4 blockade reactivated T cells with prior antigen exposure. The DC phenotype and cytokine profile do not correlate with the ability to induce antigen-specific T cells, while determinant spreading after DC immunization may be a marker of an efficient antitumor response. Sequential CTLA4 blockade may enhance the immune activity of DC-based immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia , Isoantígenos/inmunología , Melanoma/inmunología , Melanoma/terapia , Fragmentos de Péptidos/inmunología , Adulto , Anciano , Anticuerpos Bloqueadores/uso terapéutico , Antígenos CD , Antígenos de Diferenciación/inmunología , Biomarcadores/análisis , Antígeno CTLA-4 , Vacunas contra el Cáncer/uso terapéutico , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Linfocitos T/inmunología , Resultado del Tratamiento
8.
J Immunother ; 27(3): 191-200, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15076136

RESUMEN

Dendritic cells (DCs) have been shown to require a degree of maturation to stimulate antigen-specific, type 1 cytotoxic T lymphocytes in numerous murine models. Limited data in humans suggest that immature DCs (DC) can induce tolerance, yet a variety of nonmatured DC used clinically have induced antigen-specific type 1 T cells in vivo to various tumor-associated antigens. Use of adenovirus to engineer DCs is an efficient method for delivery of entire genes to DC, but the data on the biologic effects of viral transduction are contradictory. The authors demonstrate that DCs transduced with adenovirus (AdV) clearly become more mature by the phenotypic criterion of upregulation of CD83 and downregulation of CD14. Transduced DCs also decrease production of IL-10, and a subset of transduced DCs produce increased levels of IL-12 p70. This level of maturation is superior to that achieved by treatment of these cells with tumor necrosis factor-alpha or interferon-alpha but less pronounced than with CD40L trimer or CD40L + interferon-gamma. Maturation by AdV transduction alone leads to efficient stimulation of antigen-specific T cells from both healthy donors and patients with advanced cancer using two defined human tumor-associated antigens, MART-1 and AFP. Given the pivotal role of DCs in immune activation, it is important to understand the direct biologic effects of AdV on DCs, as well as the impact these biologic changes have on the stimulation of antigen-specific T cells. This study has important implications for the design of DC-based clinical trials.


Asunto(s)
Adenoviridae/genética , Células Dendríticas/citología , Técnicas de Transferencia de Gen , Linfocitos T/inmunología , Antígenos CD , Antígenos de Neoplasias/química , Ligando de CD40/química , Ensayos Clínicos como Asunto , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulinas/química , Interleucina-10/química , Interleucina-12/química , Glicoproteínas de Membrana/química , Péptidos/química , Fenotipo , Subunidades de Proteína/química , Linfocitos T/química , Linfocitos T Citotóxicos/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba , Antígeno CD83
9.
J Immunol ; 172(8): 4762-9, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15067052

RESUMEN

Wild-type mice immunized with MART-1 melanoma Ag-engineered dendritic cells (DC) generate strong Ag-specific immunity that has an absolute requirement for both CD8(+) and CD4(+) T cells. DC administration to CD8 alpha knockout mice displayed unexpectedly enhanced levels of protection to tumor challenge despite this deficiency in CD8(+) T cells and the inability to mount MHC class I-restricted immune responses. This model has the following features: 1) antitumor protection is Ag independent; 2) had an absolute requirement for CD4(+) and NK1.1(+) cells; 3) CD4(+) splenocytes are responsible for cytokine production; 4) lytic cells in microcytotoxicity assays express NK, but lack T cell markers (NK1.1(+) alpha beta TCR(-) CD3(-)); and 5) the lytic phenotype can be transferred to naive CD8 alpha knockout mice by NK1.1(+) splenocytes. Elucidation of the signaling events that activate these effective cytotoxic cells and the putative suppressive mechanisms in a wild-type environment may provide means to enhance the clinical activity of DC-based approaches.


Asunto(s)
Antígenos CD8/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/prevención & control , Proteínas , Adenovirus Humanos/genética , Animales , Antígenos/biosíntesis , Antígenos Ly , Antígenos de Neoplasias , Antígenos de Superficie , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/virología , Epítopos de Linfocito T/genética , Prueba de Histocompatibilidad , Humanos , Inmunofenotipificación , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Lectinas Tipo C , Activación de Linfocitos/inmunología , Depleción Linfocítica , Linfopenia/genética , Linfopenia/inmunología , Antígeno MART-1 , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sitios Menores de Histocompatibilidad/genética , Subfamilia B de Receptores Similares a Lectina de Células NK , Proteínas de Neoplasias/administración & dosificación , Proteínas de Neoplasias/genética , Biosíntesis de Proteínas , Bazo/citología , Bazo/inmunología , Bazo/trasplante
10.
Clin Cancer Res ; 9(16 Pt 1): 5902-8, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14676113

RESUMEN

PURPOSE: An existing immunological paradigm is that high concentrations of soluble protein contribute to the maintenance of peripheral tolerance/ignorance to self protein. We tested this hypothesis in a clinical immunotherapy trial using class I-restricted peptide epitopes derived from alpha-fetoprotein (AFP). AFP is a self protein expressed by fetal liver at high levels, but transcriptionally repressed at birth. AFP is de-repressed in a majority of hepatocellular carcinomas (HCCs) and patients with active disease can have plasma levels in the mg/ml range. We previously identified four immunodominant HLA-A*0201-restricted peptides derived from human AFP that could stimulate specific T-cell responses in normal volunteer peripheral blood lymphocytes cultures. We wished to test the hypothesis that AFP peptide-reactive T cells could be expanded in vivo in HCC patients immunized with these four AFP peptides. EXPERIMENTAL DESIGN: We undertook a pilot Phase I clinical trial in which HLA-A*0201 patients with AFP-positive HCC were immunized with three biweekly intradermal vaccinations of the four AFP peptides (100 microg or 500 microg each) emulsified in incomplete Freund's adjuvant. RESULTS: All of the patients (n=6) generated T-cell responses to most or all of the peptides as measured by direct IFNgamma enzyme-linked immunospot (ELISPOT) and MHC class I tetramer assays. CONCLUSIONS: We conclude that the human T-cell repertoire is capable of recognizing AFP in the context of MHC class I even in an environment of high circulating levels of this oncofetal protein.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Antígenos HLA-A/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos T/inmunología , Vacunas de Subunidad/inmunología , alfa-Fetoproteínas/inmunología , Adulto , Anciano , Presentación de Antígeno/genética , Carcinoma Hepatocelular/secundario , Carcinoma Hepatocelular/terapia , Ensayo de Inmunoadsorción Enzimática , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Antígeno HLA-A2 , Humanos , Interferón gamma/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Persona de Mediana Edad , Proyectos Piloto , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/metabolismo
11.
Clin Cancer Res ; 9(3): 998-1008, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12631598

RESUMEN

PURPOSE: The purpose of this study was to determine the toxicity and immunological effects of three different doses and two routes of administration of autologous dendritic cells (DCs) pulsed with the MART-1(27-35) immunodominant epitope. EXPERIMENTAL DESIGN: Eighteen HLA-A*0201-positive subjects with stage III-IV melanoma received three biweekly i.v. or intradermal injections of ex vivo generated myeloid DCs pulsed with MART-1(27-35) epitope. Repeated blood samples were processed to obtain peripheral blood mononuclear cells for immunological analysis using IFN-gamma ELISPOT, MHC class I tetramer, intracellular cytokine staining, and microcytotoxicity assays. RESULTS: The frequency of MART-1/Melan-A (MART-1) antigen-specific T cells in peripheral blood increased in all dose levels as assessed by ELISPOT and MHC class I tetramer assays, but without a clear dose-response effect. The intradermal route generated stronger MART-1 immunity compared with the i.v. route. MART-1-specific immunity did not correlate with clinical outcome in any of the four immunological assays used. However, analysis of determinant spreading to other melanoma antigens was noted in the only subject with complete response to this single-epitope immunization. CONCLUSIONS: Intradermal immunization with MART-1 peptide-pulsed DCs results in an increase in circulating IFN-gamma-producing, antigen-specific T cells. The frequency of these cells did not correlate with response. In contrast, spreading of immune reactivity to other melanoma antigens was only evident in a subject with a complete response, suggesting that determinant spreading may be an important factor of clinical response to this form of immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Epítopos/uso terapéutico , Inmunoterapia/métodos , Melanoma/terapia , Proteínas de Neoplasias/uso terapéutico , Adulto , Anciano , Antígenos de Neoplasias , Antígenos CD8/biosíntesis , Vacunas contra el Cáncer , Citocinas/biosíntesis , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Femenino , Humanos , Inmunohistoquímica , Interferón gamma/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/química , Biosíntesis de Péptidos , Péptidos/química , Resultado del Tratamiento
12.
Cancer Gene Ther ; 9(11): 875-83, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12386826

RESUMEN

Genetic immunotherapy with tumor antigen gene-modified dendritic cells (DC) generates robust immunity, although antitumor protection is not complete in all models. Previous experience in a model in which C57BL/6 mice immunized with DC transduced with adenoviral vectors expressing MART-1 demonstrated a 20-40% complete protection to a tumor challenge with B16 melanoma cells. Tumors that did develop in immunized mice had slower growth kinetics compared to tumors implanted in naïve mice. In the present study, we wished to determine if the supraphysiological production of the Th1-skewing cytokine interleukin-12 (IL-12) could enhance immune activation and antitumor protection in this model. In a series of experiments immunizing mice with DC cotransduced with MART-1 and IL-12, antitumor protection and antigen-specific splenocyte cytotoxicity and interferon gamma production inversely correlated with the amount of IL-12 produced by DC. This adverse effect of IL-12 could not be explained by a direct cytotoxic effect of natural killer cells directed towards DC, nor the production of nitric oxide leading to down-regulation of the immune response - the two mechanisms previously recognized to explain immune-suppressive effects of IL-12-based vaccine therapy. In conclusion, in this animal model, IL-12 production by gene-modified DC leads to a cytokine-induced dose-dependent inhibition of antigen-specific antitumor protection.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas/inmunología , Inmunoterapia Adoptiva , Interleucina-12/inmunología , Melanoma Experimental/inmunología , Melanoma/inmunología , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/inmunología , Citocinas/análisis , Citotoxicidad Inmunológica , Humanos , Interleucina-12/genética , Interleucina-12/uso terapéutico , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...