Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancers (Basel) ; 14(6)2022 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-35326730

RESUMEN

Introduction: In this post hoc analysis we compared various response-assessment criteria in newly diagnosed glioblastoma (GB) patients treated with tumor lysate-charged autologous dendritic cells (Audencel) and determined the differences in prediction of progression-free survival (PFS) and overall survival (OS). Methods: 76 patients enrolled in a multicenter phase II trial receiving standard of care (SOC, n = 40) or SOC + Audencel vaccine (n = 36) were included. MRI scans were evaluated using MacDonald, RANO, Vol-RANO, mRANO, Vol-mRANO and iRANO criteria. Tumor volumes (T1 contrast-enhancing as well as T2/FLAIR volumes) were calculated by semiautomatic segmentation. The Kruskal-Wallis-test was used to detect differences in PFS among the assessment criteria; for correlation analysis the Spearman test was used. Results: There was a significant difference in median PFS between mRANO (8.6 months) and Vol-mRANO (8.6 months) compared to MacDonald (4.0 months), RANO (4.2 months) and Vol-RANO (5.4 months). For the vaccination arm, median PFS by iRANO was 6.2 months. There was no difference in PFS between SOC and SOC + Audencel. The best correlation between PFS/OS was detected for mRANO (r = 0.65) and Vol-mRANO (r = 0.69, each p < 0.001). A total of 16/76 patients developed a pure T2/FLAIR progressing disease, and 4/36 patients treated with Audencel developed pseudoprogression. Conclusion: When comparing different response-assessment criteria in GB patients treated with dendritic cell-based immunotherapy, the best correlation between PFS and OS was observed for mRANO and Vol-mRANO. Interestingly, iRANO was not superior for predicting OS in patients treated with Audencel.

2.
NPJ Vaccines ; 5(1): 5, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31969991

RESUMEN

Glioblastoma is the most prevalent and aggressive brain cancer. With a median overall survival of ~15-20 months under standard therapy, novel treatment approaches are desperately needed. A recent phase II clinical trial with a personalized immunotherapy based on tumor lysate-charged dendritic cell (DC) vaccination, however, failed to prolong survival. Here, we investigated tumor tissue from trial patients to explore glioblastoma survival-related factors. We followed an innovative approach of combining mass spectrometry-based quantitative proteomics (n = 36) with microRNA sequencing plus RT-qPCR (n = 38). Protein quantification identified, e.g., huntingtin interacting protein 1 (HIP1), retinol-binding protein 1 (RBP1), ferritin heavy chain (FTH1) and focal adhesion kinase 2 (FAK2) as factor candidates correlated with a dismal prognosis. MicroRNA analysis identified miR-216b, miR-216a, miR-708 and let-7i as molecules potentially associated with favorable tissue characteristics as they were enriched in patients with a comparably longer survival. To illustrate the utility of integrated miRNomics and proteomics findings, focal adhesion was studied further as one example for a pathway of potential general interest. Taken together, we here mapped possible drivers of glioblastoma outcome under immunotherapy in one of the largest DC vaccination tissue analysis cohorts so far-demonstrating usefulness and feasibility of combined proteomics/miRNomics approaches. Future research should investigate agents that sensitize glioblastoma to (immuno)therapy-potentially building on insights generated here.

3.
Cytotherapy ; 21(6): 643-658, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30975602

RESUMEN

BACKGROUND: Glioblastoma is the most aggressive type of brain cancer. Dendritic cell (DC)-based immunotherapy against glioblastoma depends on the effectiveness of loaded antigens. Sphere-inducing culture conditions are being studied by many as a potential antigen source. Here, we investigated two different in vitro conditions (spheroid culture versus adherent culture) in relation to DC immunotherapy: (1) We studied the specific spheroid-culture proteome and assessed the clinical importance of spheroid proteins. (2) We evaluated the immunogenicity of spheroid lysate - both compared to adherent conditions. METHODS: We used seven spheroid culture systems, three of them patient-derived. Stemness-related markers were studied in those three via immunofluorescence. Spheroid-specific protein expression was measured via quantitative proteomics. The Cancer Genome Atlas (TCGA) survival data was used to investigate the clinical impact of spheroid proteins. Immunogenicity of spheroid versus adherent cell lysate was explored in autologous ELISPOT systems (DCs and T cells from the three patients). RESULTS: (1) The differential proteome of spheroid versus adherent glioblastoma culture conditions could successfully be established. The top 10 identified spheroid-specific proteins were associated with significantly decreased overall survival (TCGA MIT/Harvard cohort; n = 350, P = 0.014). (2) In exploratory experiments, immunogenicity of spheroid lysate vis-á-vis interferon (IFN)γ production was lower than that of adherent cell lysate (IFNγ ELISPOT; P = 0.034). CONCLUSIONS: Spheroid culture proteins seem to represent survival-relevant targets, supporting the use of spheroid culture conditions as an antigen source for DC immunotherapy. However, immunogenicity enhancement should be considered for future research. Transferability of our findings in terms of clinical impact and regarding different spheroid-generation techniques needs further validation.


Asunto(s)
Neoplasias Encefálicas/inmunología , Técnicas de Cultivo de Célula/métodos , Células Dendríticas/inmunología , Glioblastoma/inmunología , Proteínas de Neoplasias/inmunología , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Humanos , Inmunoterapia/métodos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Proteínas de Neoplasias/metabolismo , Esferoides Celulares/patología , Linfocitos T/inmunología , Células Tumorales Cultivadas
4.
Acta Neuropathol Commun ; 6(1): 135, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518425

RESUMEN

Audencel is a dendritic cell (DC)-based cellular cancer immunotherapy against glioblastoma multiforme (GBM). It is characterized by loading of DCs with autologous whole tumor lysate and in vitro maturation via "danger signals". The recent phase II "GBM-Vax" trial showed no clinical efficacy for Audencel as assessed with progression-free and overall survival in all patients. Here we present immunological research accompanying the trial with a focus on immune system factors related to outcome and Audencel's effect on the immune system. Methodologically, peripheral blood samples (from apheresis before Audencel or venipuncture during Audencel) were subjected to functional characterization via enzyme-linked immunospot (ELISPOT) assays connected with cytokine bead assays (CBAs) as well as phenotypical characterization via flow cytometry and mRNA quantification. GBM tissue samples (from surgery) were subjected to T cell receptor sequencing and immunohistochemistry. As results we found: Patients with favorable pre-existing anti-tumor characteristics lived longer under Audencel than Audencel patients without them. Pre-vaccination blood CD8+ T cell count and ELISPOT Granzyme B production capacity in vitro upon tumor antigen exposure were significantly correlated with overall survival. Despite Audencel's general failure to induce a significant clinical response, it nevertheless seemed to have an effect on the immune system. For instance, Audencel led to a significant up-regulation of the Th1-related immunovariables ELISPOT IFNγ, the transcription factor T-bet in the blood and ELISPOT IL-2 in a dose-dependent manner upon vaccination. Post-vaccination levels of ELISPOT IFNγ and CD8+ cells in the blood were indicative of a significantly better survival. In summary, Audencel failed to reach an improvement of survival in the recent phase II clinical trial. No clinical efficacy was registered. Our concomitant immunological work presented here indicates that outcome under Audencel was influenced by the state of the immune system. On the other hand, Audencel also seemed to have stimulated the immune system. Overall, these immunological considerations suggest that DC immunotherapy against glioblastoma should be studied further - with the goal of translating an apparent immunological response into a clinical response. Future research should concentrate on investigating augmentation of immune reactions through combination therapies or on developing meaningful biomarkers.


Asunto(s)
Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos/fisiología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/fisiología , Glioblastoma/terapia , Antígenos CD/metabolismo , Compuestos de Boro/metabolismo , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Femenino , Glioblastoma/sangre , Glioblastoma/inmunología , Humanos , Estimación de Kaplan-Meier , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Estudios Longitudinales , Masculino , Resultado del Tratamiento , Regulación hacia Arriba
5.
Cancers (Basel) ; 10(10)2018 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-30301187

RESUMEN

Dendritic cells (DCs) are antigen-presenting cells that are capable of priming anti-tumor immune responses, thus serving as attractive tools to generate tumor vaccines. In this multicentric randomized open-label phase II study, we investigated the efficacy of vaccination with tumor lysate-charged autologous DCs (Audencel) in newly diagnosed glioblastoma multiforme (GBM). Patients aged 18 to 70 years with histologically proven primary GBM and resection of at least 70% were randomized 1:1 to standard of care (SOC) or SOC plus vaccination (weekly intranodal application in weeks seven to 10, followed by monthly intervals). The primary endpoint was progression-free survival at 12 months. Secondary endpoints were overall survival, safety, and toxicity. Seventy-six adult patients were analyzed in this study. Vaccinations were given for seven (3⁻20) months on average. No severe toxicity was attributable to vaccination. Seven patients showed flu-like symptoms, and six patients developed local skin reactions. Progression-free survival at 12 months did not differ significantly between the control and vaccine groups (28.4% versus 24.5%, p = 0.9975). Median overall survival was similar with 18.3 months (vaccine: 564 days, 95% CI: 436⁻671 versus control: 568 days, 95% CI: 349⁻680; p = 0.89, harzard ratio (HR) 0.99). Hence, in this trial, the clinical outcomes of patients with primary GBM could not be improved by the addition of Audencel to SOC.

6.
J Immunol ; 195(2): 541-52, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26078274

RESUMEN

Dendritic cell (DC)-mediated inflammation induced via TLRs is promoted by MAPK-activated protein kinase (MK)-2, a substrate of p38 MAPK. In this study we show an opposing role of MK2, by which it consolidates immune regulatory functions in DCs through modulation of p38, ERK1/2-MAPK, and STAT3 signaling. During primary TLR/p38 signaling, MK2 mediates the inhibition of p38 activation and positively cross-regulates ERK1/2 activity, leading to a reduction of IL-12 and IL-1α/ß secretion. Consequently, MK2 impairs secondary autocrine IL-1α signaling in DCs, which further decreases the IL-1α/p38 but increases the anti-inflammatory IL-10/STAT3 signaling route. Therefore, the blockade of MK2 activity enables human and murine DCs to strengthen proinflammatory effector mechanisms by promoting IL-1α-mediated Th1 effector functions in vitro. Furthermore, MK2-deficient DCs trigger Th1 differentiation and Ag-specific cytotoxicity in vivo. Finally, wild-type mice immunized with LPS in the presence of an MK2 inhibitor strongly accumulate Th1 cells in their lymph nodes. These observations correlate with a severe clinical course in DC-specific MK2 knockout mice compared with wild-type littermates upon induction of experimental autoimmune encephalitis. Our data suggest that MK2 exerts a profound anti-inflammatory effect that prevents DCs from prolonging excessive Th1 effector T cell functions and autoimmunity.


Asunto(s)
Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Células TH1/inmunología , Animales , Diferenciación Celular , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica , Humanos , Inmunización , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Células TH1/efectos de los fármacos , Células TH1/patología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
8.
Crit Rev Immunol ; 33(6): 489-547, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24266347

RESUMEN

Dendritic cells (DCs) are the most potent professional antigen-presenting cells, capable of initiating proper adaptive immune responses. Although tumor-infiltrating DCs are able to recognize cancer cells and uptake tumor antigens, they often have impaired functions because of the immunosuppressive tumor milieu. Therefore, DCs are targeted by therapeutic means either in vivo or ex vivo to facilitate tumor antigen presentation to T cells and induce or promote efficient antitumor immune responses in cancer patients. This immunotherapeutical approach is defined as specific active tumor immunotherapy or therapeutic cancer vaccination. In this review we briefly discuss general aspects of DC biology, followed by a thorough description of the current knowledge and optimization trends of DC vaccine production ex vivo, including various approaches for the induction of proper DC maturation and efficient loading with tumor antigens. We also discuss critical clinical aspects of DC vaccine application in cancer patients, including protocols of administration (routes and regimens), individualization of tumor immunotherapy, prediction and proper evaluation of immune and clinical responses to immunotherapy, and the critical role of combining tumor immunotherapy with other cancer treatment strategies to achieve maximal therapeutic effects.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas/inmunología , Inmunoterapia/métodos , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Ensayos Clínicos como Asunto , Células Dendríticas/trasplante , Humanos
9.
PLoS One ; 8(2): e54879, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23408948

RESUMEN

The dendritic cell (DC) coordinates innate and adaptive immunity to fight infections and cancer. Our observations reveal that DCs exposed to the microbial danger signal lipopolysaccharide (LPS) in the presence of interferon-γ (IFN-γ) acquire a continuously changing activation/maturation phenotype. The DCs' initial mode of action is pro-inflammatory via up-regulation among others of the signaling molecule interleukin (IL) 12, which polarizes IFN-γ secreting type 1 helper T-cells (Th1). Within 24 hours the same DC switches from the pro- into an anti-inflammatory phenotype. This is mediated by autocrine IL-10 release and secretion of soluble IL-2 receptor alpha (sIL-2RA) molecules. T-cells, when contacted with DCs during their anti-inflammatory phase loose their proliferative capacity and develop regulatory T-cell (Treg) -like anti-inflammatory functions indicated by IL-10 secretion and elevated FoxP3 levels. Studying the kinetics of IL-12 and IL-10 expression from LPS/IFN-γ activated myeloid DCs on a single cell level confirmed these observations. When T-cells are separated from DCs within 24 hours, they are spared from the anti-inflammatory DC activity. We conclude that, in addition to differentiation of DCs into distinct subsets, the observed sequential functional phases of DC differentiation permit the fine-tuning of an immune response. A better understanding of time-kinetic DC features is required for optimally exploiting the therapeutic capacity of DCs in cancer immune therapy.


Asunto(s)
Diferenciación Celular/fisiología , Células Dendríticas/citología , Inflamación/fisiopatología , Receptor Toll-Like 4/fisiología , Animales , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Ratones
10.
J Immunother ; 33(1): 40-52, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19952957

RESUMEN

In cancer patients undergoing immune therapy with lipopolysaccharides/interferon-gamma activated interleukin (IL)-12 secreting dendritic cells (DCs) we observed enhanced proliferative capacity of pyrophosphate-responsive peripheral blood (PB) gammadelta T-cells. This was not noted before as in other clinical trials DCs were used that were not enabled for IL-12 secretion and mice do not have a corresponding subset of PB gammadelta T-cells. In vitro examination of IL-12 DC/PB gammadelta T-cell interactions revealed a potential of PB gammadelta T-cells to negatively regulate the proliferative capacity of CD4 and CD8 T-cells. We further demonstrate that IL-12 is critical in the activation of PB gammadelta T-cells. In contrast, the regulatory activity of PB gammadelta T-cells in immune responses against strong recall antigens or alloantigens did not require activated DCs, but depended on pyrophosphate activation of PB gammadelta T-cells. Depletion of PB gammadelta T-cells abrogated the regulatory activity in IL-12 DC/peripheral blood mononuclear cell cocultures; adding back graded numbers of PB gammadelta T-cells restored it. Our observations revealed a potential PB gammadelta T cell-mediated negative regulatory feedback mechanism triggered by IL-12 DCs, which may critically impact on the design of DC cancer vaccines.


Asunto(s)
Células Dendríticas/trasplante , Interleucina-12/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos Fase I como Asunto , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Interleucina-12/metabolismo
11.
IDrugs ; 12(10): 629-32, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19790012

RESUMEN

The Chinese Medical Biotech Association's Fourth Medical Biotech Forum held in Dalian, China included topics covering the biotechnology industry in China and new therapeutic developments in the field of immunological approaches to cancer treatment. This conference report highlights selected presentations on China's biotechnology development policy, tumor-specific antigens, clinical applications of antitumor immune therapy, and novel photodynamic tumor therapy. Investigational therapeutics discussed include astuprotimut-r (GlaxoSmithKline plc) and the dendritic cell vaccine Trivax (Trimed Biotech).


Asunto(s)
Biotecnología , Sociedades Médicas , Animales , Antineoplásicos/administración & dosificación , Biotecnología/tendencias , China , Humanos , Sociedades Médicas/tendencias
12.
Blood ; 114(15): 3235-43, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19625705

RESUMEN

The role of the tryptophan-metabolizing enzyme indoleamine 2,3-dioxygenase (IDO) in down-regulating human alloresponses has recently been controversially debated. We here demonstrate that human monocyte-derived dendritic cells (mDCs) can be endowed with sustained IDO competence in vitro by 48-hour activation with lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma). IFN-gamma also amplified proinflammatory cytokine secretion during activation. Yet, on reculture after activation cytokine production ceased, whereas IDO enzymatic activity continued. Manipulation of tryptophan metabolism did not affect proinflammatory cytokine release, suggesting that IFN-gamma triggers IDO activity and proinflammatory cytokine release as distinct cellular programs. IDO-competent DCs down-regulated allogeneic T-cell responses, but this IDO-mediated effect was overcome by slightly modifying cell culture conditions. Nevertheless, the CD4(+)CD25(+) T-cell fraction stimulated by IDO-competent DCs displayed substantial suppressor activity. This suppressive activity (1) required allogeneic stimulation for its induction, (2) affected third-party T cells, and (3) was reduced by the IDO inhibitor methyl-thiohydantoin-tryptophan. It became also manifest when DC/T-cell cocultures were initiated with naive (CD4(+)CD25(-)CD45RA(+)) T cells, indicating the differentiation of adaptive regulatory T cells. Together, these findings suggest that IFN-gamma triggered IDO competence in human mDCs constitutes a critical factor for endowing allogeneic T cells with regulatory activity.


Asunto(s)
Células Dendríticas/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón gamma/inmunología , Monocitos/inmunología , Linfocitos T Reguladores/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/citología , Células Dendríticas/enzimología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indoles/farmacología , Mediadores de Inflamación/inmunología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Monocitos/citología , Monocitos/enzimología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/enzimología , Tiohidantoínas/farmacología
13.
Cancer Res ; 69(1): 203-11, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19118004

RESUMEN

We showed previously that Tyk2(-/-) natural killer cells lack the ability to lyse leukemic cells. As a consequence, the animals are leukemia prone. Here, we show that the impaired tumor surveillance extends to T cells. Challenging Tyk2(-/-) mice with EL4 thymoma significantly decreased disease latency. The crucial role of Tyk2 for CTL function was further characterized using the ovalbumin-expressing EG7 cells. Tyk2(-/-) OT-1 mice developed EG7-induced tumors significantly faster compared with wild-type (wt) controls. In vivo assays confirmed the defect in CD8(+) cytotoxicity on Tyk2 deficiency and clearly linked it to type I IFN signaling. An impaired CTL activity was only observed in IFNAR1(-/-) animals but not on IFNgamma or IL12p35 deficiency. Accordingly, EG7-induced tumors grew faster in IFNAR1(-/-) and Tyk2(-/-) but not in IFNgamma(-/-) or IL12p35(-/-) mice. Adoptive transfer experiments defined a key role of Tyk2 in CTL-mediated tumor surveillance. In contrast to wt OT-1 cells, Tyk2(-/-) OT-1 T cells were incapable of controlling EG7-induced tumor growth.


Asunto(s)
Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/inmunología , TYK2 Quinasa/inmunología , Timoma/inmunología , Neoplasias del Timo/inmunología , Animales , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Femenino , Vigilancia Inmunológica , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Interleucina-12/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , TYK2 Quinasa/deficiencia , TYK2 Quinasa/genética , TYK2 Quinasa/metabolismo , Timoma/enzimología , Neoplasias del Timo/enzimología
14.
J Cell Mol Med ; 13(1): 125-35, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18363835

RESUMEN

Manufacturing procedures for cellular therapies are continuously improved with particular emphasis on product safety. We previously developed a dendritic cell (DC) cancer vaccine technology platform that uses clinical grade lipopolysaccharide (LPS) and interferon (IFN)-y for the maturation of monocyte derived DCs. DCs are frozen after 6 hrs exposure at a semi-mature stage (smDCs) retaining the capacity to secret interleukin (IL)-12 and thus support cytolytic T-cell responses, which is lost at full maturation. We compared closed systems for monocyte enrichment from leucocyte apheresis products from healthy individuals using plastic adherence, CD14 selection, or CD2/19 depletion with magnetic beads, or counter flow centrifugation (elutriation) using a clinical grade in comparison to a research grade culture medium for the following DC generation. We found that elutriation was superior compared to the other methods showing 36 +/- 4% recovery, which was approximately 5-fold higher as the most frequently used adherence protocol (8 +/- 1%), and a very good purity (92 +/- 5%) of smDCs. Immune phenotype and IL-12 secretion (adherence: 1.4 +/- 0.4; selection: 20 +/- 0.6; depletion: 1 +/-0.5; elutriation: 3.6 +/- 1.5 ng/ml) as well as the potency of all DCs to stimulate T cells in an allogeneic mixed leucocyte reaction did not show statistically significant differences. Research grade and clinical grade DC culture media were equally potent and freezing did not impair the functions of smDCs. Finally, we assessed the functional capacity of DC cancer vaccines manufactured for three patients using this optimized procedure thereby demonstrating the feasibility of manufacturing DC cancer vaccines that secret IL-12 (9.4 +/- 6.4 ng/ml). We conclude that significant steps were taken here towards clinical grade DC cancer vaccine manufacturing.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Separación Celular/métodos , Células Dendríticas/inmunología , Monocitos , Eliminación de Componentes Sanguíneos , Células Dendríticas/citología , Humanos , Interferón gamma/inmunología , Interleucina-12/inmunología , Leucocitos/citología , Leucocitos/inmunología , Lipopolisacáridos/inmunología , Monocitos/citología , Monocitos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
15.
J Cell Mol Med ; 13(8B): 1741-1750, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20187300

RESUMEN

Inflammation triggered by microbial lipopolysaccharide (LPS) through Toll-like receptor (TLR) 4 in the presence of interferon (IFN)-gamma induces cytokine secretion in dendritic cells (DCs) tightly regulated by a defined differentiation program. This DC differentiation is characterized not only by a dynamic immune activating but also by tolerance-inducing phenotype associated with down-modulation of cytokines previously considered to be irreversible. CD40L on activated T cells further modifies DC differentiation. Using DNA micro-arrays, we showed down-regulated mRNA levels of TLR signalling molecules, whereas CD40/CD40L signalling molecules were up-regulated at a time when LPS/IFN-gamma-activated DCs had ceased cytokine expression. Accordingly, we demonstrated that CD40/CD40L but not TLR4 or TLR3 signalling mediated by LPS or poly (cytidylic-inosinic) acid (poly I:C) and dsRNA re-established the capacity for secreting interleukin (IL)-12 in primarily LPS/IFN-gamma-activated DCs, which have exhausted their potential for cytokine secretion. The resulting TH1 polarizing DC phenotype - which lacked accompanying secretion of the crucial immune suppressive factor IL-10 - maintained the potential for activation of cytotoxic T lymphocytes (CTLs). We therefore conclude that immune modulation is restricted to a secondary T-cell-mediated stimulus at an exhausted DC state, which prevents an immune tolerant DC phenotype. These findings impact on the rational design of TLR-activated DC-based cancer vaccines for the induction of anti-tumoural CTL responses.


Asunto(s)
Antígenos CD40/inmunología , Polaridad Celular , Células Dendríticas/inmunología , Interleucina-12/metabolismo , Receptor Toll-Like 4/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Prueba de Cultivo Mixto de Linfocitos , Transducción de Señal
16.
Int J Cancer ; 119(12): 2870-7, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17016825

RESUMEN

The potential immunogenicity of acute lymphoblastic leukemia of the T cell (T-ALL), a small subgroup of childhood leukemia with increased risk for treatment failure and early relapse, was addressed by serological identification of leukemia-derived antigens by recombinant expression cloning (SEREX). Thirteen antigens with homology to known genes that are involved in critical cellular processes were detected. Further characterization of the 4 novel isoforms revealed that 3 (HECTD1Delta, CX-ORF-15Delta and hCAP-EDelta) had restricted mRNA expression in more than 70% of T-ALLs (n = 22) and that specific antibodies against these isoforms were detected in up to 30% of patients (n = 16), with the highest frequency for HECTD1Delta. The latter protein was present at high abundance in T-ALLs but not in normal hematopoietic tissues. Given that the leukemia-associated antigens detected in this study have an intracellular localization, the generation of immune effector responses most likely requires antigen presentation. To test this assumption, dendritic cells were loaded with HECTD1Delta protein and used for T cell stimulation. A specific T cell response was induced in vitro in all 3 healthy donors studied, including a former T-ALL patient. These data suggest that T-ALL may induce a specific cellular and humoral antileukemia immune response in children, thereby supporting new approaches for immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Leucemia-Linfoma de Células T del Adulto/inmunología , Adolescente , Adulto , Anticuerpos/sangre , Anticuerpos/inmunología , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Niño , Preescolar , Clonación Molecular/métodos , ADN Complementario/química , ADN Complementario/genética , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Interferón gamma/biosíntesis , Células Jurkat , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/patología , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
17.
Cancer Immunol Immunother ; 54(1): 67-77, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15693141

RESUMEN

To induce cytolytic immunity, dendritic cells (DCs) need to release bioactive interleukin-12 (IL-12) p70 heterodimeric molecules. To study the role of IL-12 for the generation of an anti-tumor immune response, we generated two classes of DCs. (1) DCs were initiated to secrete IL-12 by exposure to LPS/IFN-gamma for 2 h resulting, as demonstrated in vitro, in continued IL-12 release for another 24 h (termed active DCs). (2) DCs were exposed to LPS/IFN-gamma for 24 h and injected into mice at a time point when IL-12 production had ceased (termed exhausted DCs). These two classes of DCs were probed for their capacity to induce a cytolytic anti-tumor immune response in vivo in a syngeneic mouse tumor model. The mouse tumor cell line K-Balb was engineered to express neomycin phosphotransferase (NPT) as a model tumor antigen. DCs were charged with various NPT-derived antigens, including recombinant NPT protein, whole tumor cell lysate and NPT-derived synthetic peptides, and the induction of in vivo anti-tumor immunity was determined by measuring tumor growth. Only the injection of active DCs, i.e., cells that maintained the capacity to secrete IL-12, but not exhausted DCs that had lost the ability to produce IL-12, resulted in a measurable deceleration of growth of K-Balb-NPT tumors. This anti-tumor immune response was most pronounced when using recombinant protein as an antigen source, which was evident in a prophylactic as well as in a therapeutic setting. The absence of a response to parental K-Balb tumors confirmed the antigen specificity of the anti-tumor immune response. Together these data provide evidence for the unique capacity of actively IL-12 secreting DCs to trigger effective anti-tumor immunity using exogenous tumor antigens.


Asunto(s)
Células Dendríticas/inmunología , Terapia Genética/métodos , Inmunoterapia/métodos , Interleucina-12/inmunología , Interleucina-12/metabolismo , Subunidades de Proteína/inmunología , Subunidades de Proteína/metabolismo , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/trasplante , Epítopos , Inmunofenotipificación , Interferón gamma/farmacología , Kanamicina Quinasa/biosíntesis , Kanamicina Quinasa/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo
18.
Cancer Immunol Immunother ; 54(8): 769-80, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15647926

RESUMEN

Dendritic cells (DC) are candidates for antigen-presenting cells that present exogenous antigen on MHC class I molecules to cytotoxic T lymphocytes (CTL), a process referred to as cross-priming. We triggered interleukin (IL)-12 release from DC, which was limited to the first day after maturation induction, by a combination of lipopolysaccharide (LPS) and interferon (IFN)-gamma. To stimulate T lymphocytes, we used soluble protein derived from lysis of Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines (LCL) or ovalbumin loaded onto DC. Co-culture was initiated 2-6 or 48 h after maturation corresponding to "semi-mature" actively IL-12-secreting type 1 DC (sm-DC1) or a "fully mature" DC1 that had lost the ability to release IL-12 (fm-DC1), respectively. IL-12-secreting sm-DC1 but not fm-DC1 efficiently triggered cytolytic activity in autologous T lymphocytes. The combination of IL-1beta, IL-6, TNF-alpha, and prostaglandin E2 generated type 2 DC that did not secrete IL-12 (DC2) and could not prime T-cell cytolytic activity. However, supplementation of cultures using DC2 with IL-12 resulted in CTL activity while the presence of anti-IL-12 monoclonal antibodies in cultures using IL-12 secreting sm-DC1 suppressed CTL activity. Thus, actively IL-12-secreting sm-DC1 are necessary and sufficient for the antigen-specific expansion of CTL in response to exogenously provided soluble antigen.


Asunto(s)
Presentación de Antígeno/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Interleucina-12/metabolismo , Linfocitos T Citotóxicos/inmunología , Anticuerpos Monoclonales , Linfocitos T CD8-positivos/inmunología , Técnicas de Cultivo de Célula , Muerte Celular , Ensayo de Inmunoadsorción Enzimática , Herpesvirus Humano 4/inmunología , Humanos , Ovalbúmina/inmunología
19.
Hum Immunol ; 64(8): 762-70, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12878354

RESUMEN

Cellular aggregation in response to surface antigen signaling is a regulated process important for cell-cell interaction and cell migration. We studied dendritic cell (DC) aggregation in response to major histocompatibility complex (MHC) class II antigen ligation using human monocyte derived mature DCs. Crosslinking of MHC class II antigens by monoclonal antibodies in DCs matured by different stimuli, but not in immature DCs, induced the formation of large and long-lived homotypic cell clusters within 1 hour. These aggregates were completely resistant to mechanical disruption and displayed no signs of increased cell death. Heterotypic conjugate formation of mature DCs with lymphocytes and monocytes following addition of MHC class II antibodies occurred as well. DC aggregation required antigen dimerization, the presence of serum and energy, an intact cytoskeleton, and could not be blocked by EDTA, mannan, and monoclonal antibodies specific for integrins. These data underscore the positive role of MHC class II signaling in mature DCs suggesting the possibility of reverse DC activation in the course of antigen specific interaction with lymphocytes.


Asunto(s)
Agregación Celular , Células Dendríticas/fisiología , Antígenos de Histocompatibilidad Clase II/inmunología , Monocitos/fisiología , Anticuerpos Monoclonales/inmunología , Presentación de Antígeno , Apoptosis , Adhesión Celular , Células Cultivadas , Reactivos de Enlaces Cruzados , Células Dendríticas/inmunología , Humanos , Linfocitos/fisiología , Monocitos/inmunología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...