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












Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 14(8)2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35454801

RESUMEN

Prostate cancer (PCa) poses a major public health problem in men. Metastatic PCa is incurable, and ultimately threatens the life of many patients. Mutations in tumor suppressor genes and oncogenes are important for PCa progression, whereas the role of epigenetic factors in prostate carcinogenesis is insufficiently examined. The histone demethylase KDM5C exerts important roles in tumorigenesis. KDM5C has been reported to be highly expressed in various cancer cell types, particularly in primary PCa. Here, we could show that KDM5C is highly upregulated in metastatic PCa. Functionally, in KDM5C knockdown cells migratory and invasion capacity was reduced. Interestingly, modulation of KDM5C expression influences several EMT signaling pathways (e.g., Akt/mTOR), expression of EMT transcription factors, epigenetic modifiers, and miR-205, resulting in increased expression of E-cadherin and reduced expression of N-cadherin. Mouse xenografts of KDM5C knockdown cells showed reduced tumor growth. In addition, the Akt/mTOR pathway is one of the classic signaling pathways to mediate tumor metabolic homeostasis, which is beneficial for tumor growth and metastasis. Taken together, our findings indicate that a combination of a selective KDM5C- and Akt/mTOR-inhibitor might be a new promising therapeutic strategy to reduce metastatic burden in PCa.

2.
Clin Cancer Res ; 23(7): 1829-1840, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27678455

RESUMEN

Purpose: The Mediator complex is a multiprotein assembly, which serves as a hub for diverse signaling pathways to regulate gene expression. Because gene expression is frequently altered in cancer, a systematic understanding of the Mediator complex in malignancies could foster the development of novel targeted therapeutic approaches.Experimental Design: We performed a systematic deconvolution of the Mediator subunit expression profiles across 23 cancer entities (n = 8,568) using data from The Cancer Genome Atlas (TCGA). Prostate cancer-specific findings were validated in two publicly available gene expression cohorts and a large cohort of primary and advanced prostate cancer (n = 622) stained by immunohistochemistry. The role of CDK19 and CDK8 was evaluated by siRNA-mediated gene knockdown and inhibitor treatment in prostate cancer cell lines with functional assays and gene expression analysis by RNAseq.Results: Cluster analysis of TCGA expression data segregated tumor entities, indicating tumor-type-specific Mediator complex compositions. Only prostate cancer was marked by high expression of CDK19 In primary prostate cancer, CDK19 was associated with increased aggressiveness and shorter disease-free survival. During cancer progression, highest levels of CDK19 and of its paralog CDK8 were present in metastases. In vitro, inhibition of CDK19 and CDK8 by knockdown or treatment with a selective CDK8/CDK19 inhibitor significantly decreased migration and invasion.Conclusions: Our analysis revealed distinct transcriptional expression profiles of the Mediator complex across cancer entities indicating differential modes of transcriptional regulation. Moreover, it identified CDK19 and CDK8 to be specifically overexpressed during prostate cancer progression, highlighting their potential as novel therapeutic targets in advanced prostate cancer. Clin Cancer Res; 23(7); 1829-40. ©2016 AACR.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/genética , Complejo Mediador/genética , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Estadificación de Neoplasias , Neoplasias de la Próstata/patología , Transcriptoma/genética
3.
Exp Cell Res ; 348(2): 123-131, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27646113

RESUMEN

Cell migration is a central process in the development and maintenance of multicellular organisms. Tissue formation during embryonic development, wound healing, immune responses and invasive tumors all require the orchestrated movement of cells to specific locations. Histone demethylase proteins alter transcription by regulating the chromatin state at specific gene loci. FBXL10 is a conserved and ubiquitously expressed member of the JmjC domain-containing histone demethylase family and is implicated in the demethylation of H3K4me3 and H3K36me2 and thereby removing active chromatin marks. However, the physiological role of FBXL10 in vivo remains largely unknown. Therefore, we established an inducible gain of function model to analyze the role of Fbxl10 and compared wild-type with Fbxl10 overexpressing mouse embryonic fibroblasts (MEFs). Our study shows that overexpression of Fbxl10 in MEFs doesn't influence the proliferation capability but leads to an enhanced migration capacity in comparison to wild-type MEFs. Transcriptome and ChIP-seq experiments demonstrated that Fbxl10 binds to genes involved in migration like Areg, Mdk, Lmnb1, Thbs1, Mgp and Cxcl12. Taken together, our results strongly suggest that Fbxl10 plays a critical role in migration by binding to the promoter region of migration-associated genes and thereby might influences cell behaviour to a possibly more aggressive phenotype.


Asunto(s)
Movimiento Celular , Embrión de Mamíferos/citología , Proteínas F-Box/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Doxiciclina/farmacología , Proteínas F-Box/genética , Femenino , Fibroblastos/efectos de los fármacos , Perfilación de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Interferente Pequeño/metabolismo , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Transgenes
4.
Int J Cancer ; 137(11): 2578-88, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26084847

RESUMEN

Liposarcomas (LS) are the most common malignant mesenchymal tumors, with an overall long-term mortality rate of 60%. LS comprise three major subtypes, i.e., well-differentiated/dedifferentiated liposarcoma (WDLS/DDLS), myxoid/round cell liposarcoma (MLS) and pleomorphic liposarcoma (PLS). Aiming at the preclinical identification of novel therapeutic options, we here investigate the functional significance of SRC in primary human LS and in LS-derived cell lines. Immunohistochemical and Western blot analyses reveal relevant levels of activated p-(Tyr416)-SRC in LS of the different subtypes with particular activation in MLS and PLS. Dysregulation of the SRC modifiers CSK and PTP1B was excluded as major reason for the activation of the kinase. Consistent siRNA-mediated knockdown of SRC or inhibition by the SRC inhibitor Dasatinib led to decreased proliferation of LS cell lines of the different subtypes, with MLS cells reacting particularly sensitive in MTT assays. Flow cytometric analyses revealed that this effect was due to a significant decrease in mitotic activity and an induction of apoptosis. SRC inhibition by Dasatinib resulted in dephosphorylation of SRC itself, its interacting partners FAK and IGF-IR as well as its downstream target AKT. Consistent with a particular role of SRC in cell motility, Dasatinib reduced the migratory and invasive potential of MLS cells in Boyden chamber and Matrigel chamber assays. In summary, we provide evidence that SRC activation plays an important role in LS biology and therefore represents a potential therapeutic target, particularly in MLS and PLS.


Asunto(s)
Liposarcoma Mixoide/tratamiento farmacológico , Liposarcoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Mitosis/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1 , Receptores de Somatomedina/metabolismo
5.
Liver Int ; 34(3): 447-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23998316

RESUMEN

BACKGROUND: Interleukin 12 (IL-12), one of the most potent Th1-cytokines, has been used to improve dendritic cells (DC)-based immunotherapy of cancer. However, it failed to achieve clinical response in patients with hepatocellular carcinoma (HCC). In this study, improved conditions of immunotherapy with DC engineered to express IL-12 were studied in murine subcutaneous HCC. METHODS: Tumour-lysate pulsed DC were transduced with IL-12-encoding adenoviruses or cultivated with recombinant (r)IL-12. DC were injected intratumourally, subcutaneously or intravenously at different stages of tumour-development. RESULTS: Dendritic cell overexpressing IL-12 by adenoviruses showed enhanced expression of costimulatory molecules and stronger priming of HCC-specific effector cells than DC cultured with rIL-12. Intratumoural but not systemic injections of IL-12-DC induced the strongest antitumoural effects reaching complete regressions in 75% of early-staged tumours and in 33% of advanced tumours. Importantly, antitumoural effects could be further enhanced through combination with sorafenib. Analysing the tumour-environment, IL-12-DC increased the levels of Th1-cytokines/chemokines and of CD4(+) -, CD8(+) -T- and NK-cells. Induced immunity was tumour-specific and sustained since all tumour-free animals were protected towards hepatic tumour-cell rechallenge. However, IL-12-DC also enhanced immunosuppressive cytokines, regulatory T cells and even myeloid-derived suppressor cells within the tumours. CONCLUSIONS: Induced IL-12-overexpression by adenoviral vectors can effectively immunostimulate DC. Intratumoural but not systemic injection of activated IL-12-DC was crucial for effective tumour regression. The mechanism of this approach seems to be the induction of a sufficient Th1 tumour-environment allowing the recruitment of effector cells rather than the inhibition of tumour immunosuppression. Thus, improved immunotherapy with IL-12-DC represents a promising approach towards HCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/terapia , Células Dendríticas/inmunología , Interleucina-12/genética , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Humanos , Inmunoterapia , Ratones , Ratones Endogámicos C3H , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Sorafenib
6.
Cancer Res ; 73(8): 2518-28, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23580575

RESUMEN

Synovial sarcoma is a soft-tissue malignancy characterized by a reciprocal t(X;18) translocation encoding a chimeric transcriptional modifier. Several receptor tyrosine kinases have been found activated in synovial sarcoma; however, no convincing therapeutic concept has emerged from these findings. On the basis of the results of phosphokinase screening arrays, we here investigate the functional and therapeutic relevance of the SRC kinase in synovial sarcoma. Immunohistochemistry of phosphorylated SRC and its regulators CSK and PTP1B (PTPN1) was conducted in 30 synovial sarcomas. Functional aspects of SRC, including dependence of SRC activation on the SS18/SSX fusion proteins, were analyzed in vitro. Eventually, synovial sarcoma xenografts were treated with the SRC inhibitor dasatinib in vivo. Activated phospho (p)-(Tyr416)-SRC was detected in the majority of tumors; dysregulation of CSK or PTP1B was excluded as the reason for the activation of the kinase. Expression of the SS18/SSX fusion proteins in T-REx-293 cells was associated with increased p-(Tyr416)-SRC levels, linked with an induction of the insulin-like growth factor pathway. Treatment of synovial sarcoma cells with dasatinib led to apoptosis and inhibition of cellular proliferation, associated with reduced phosphorylation of FAK (PTK2), STAT3, IGF-IR, and AKT. Concurrent exposure of cells to dasatinib and chemotherapeutic agents resulted in additive effects. Cellular migration and invasion were dependent on signals transmitted by SRC involving regulation of the Rho GTPases Rac and RhoA. Treatment of nude mice with SYO-1 xenografts with dasatinib significantly inhibited tumor growth in vivo. In summary, SRC is of crucial biologic importance and represents a promising therapeutic target in synovial sarcoma.


Asunto(s)
Sarcoma Sinovial/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dasatinib , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Mitosis/efectos de los fármacos , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Fosfotransferasas/metabolismo , Análisis por Matrices de Proteínas , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Interferencia de ARN , Proteínas Represoras/genética , Sarcoma Sinovial/genética , Sarcoma Sinovial/patología , Tiazoles/farmacología , Translocación Genética , Carga Tumoral/efectos de los fármacos , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
7.
Biochim Biophys Acta ; 1833(5): 1190-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23391410

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer deaths, worldwide. MicroRNAs, inhibiting gene expression by targeting various transcripts, are involved in genomic dysregulation during hepatocellular tumorigenesis. In previous studies, microRNA-198 (miR-198) was shown to be significantly downregulated in HCV-positive hepatocellular carcinoma (HCC). Herein, the function of miR-198 in hepatocellular carcinoma cell growth and gene expression was studied. In hepatoma cell-types with low levels of liver-specific transcription factor HNF1α indicating a low differentiation grade, miR-198 expression was most downregulated. However, miR-198 treatment did not restore the expression of the liver-specific transcription factors HNF1α or HNF4α. Importantly, overexpression of miR-198 in Pop10 hepatoma cells markedly reduced cell growth. In agreement, comprehensive gene expression profiling by microarray hybridisation and real-time quantification revealed that central signal transducers of proliferation pathways were downregulated by miR-198. In contrast, genes mediating cellular adherence were highly upregulated by miR-198. Thus, the low expression of E-cadherin and claudin-1, involved in cell adhesion and cell-cell contacts, was abolished in hepatoma cells after miR-198 overexpression. This definite induction of both proteins by miR-198 was shown to be accompanied by a significantly impaired migration activity of hepatoma Pop10 cells. In conclusion, miR-198 acts as a tumor suppressor by repression of mitogenic and motogenic pathways diminishing cell growth and migration.


Asunto(s)
Carcinoma Hepatocelular , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , MicroARNs , Cadherinas/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Claudina-1/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo
8.
Int J Cancer ; 129(7): 1564-75, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21128248

RESUMEN

Synovial sarcomas account for 5-10% of all malignant soft tissue tumors. They have been shown to express different membranous growth factor receptors, many of them signaling via intracellular kinase cascades. In our study, the functional role of PI3K/AKT signals in synovial sarcoma is analyzed with regard to tumor biology and therapeutic applicability. Immunohistochemical stainings of (Ser473)-phosphorylated (p)-AKT, its targets p-(Ser9)-GSK-3ß and p-(Ser2448)-mTOR and the cell cycle regulators Cyclin D1 and p27(KIP1) were performed in 36 synovial sarcomas. The PIK3CA gene was screened for mutations. In vitro, four synovial sarcoma cell lines were treated with the PI3K inhibitor LY294002. Phosphorylation of AKT, GSK-3ß and mTOR was assessed, and cellular proliferation and apoptosis were analyzed to functionally characterize the effects of PI3K inhibition. Finally, coincubations of LY294002 with cytotoxic drugs were performed. Most tumors showed significant expression levels of p-AKT, p-GSK-3ß and p-mTOR, indicating activation of the PI3K/AKT signaling cascade in synovial sarcomas; Cyclin D1 and p27(KIP1) were differentially expressed. Mutations in the PIK3CA gene could be excluded. In vitro, PI3K inhibition diminished synovial sarcoma cell growth accompanied by reduced phosphorylation of AKT, GSK-3ß and mTOR. Mechanistically, PI3K pathway inhibition lead to enhanced apoptosis and decreased cellular proliferation linked to reduced Cyclin D1 and increased p27(KIP1) levels. Simultaneous treatment of synovial sarcoma cell lines with LY294002 and cytotoxic drugs resulted in additive effects. In summary, PI3K signaling plays an essential role in growth control of synovial sarcomas and might be successfully targeted in multimodal therapeutic strategies.


Asunto(s)
Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sarcoma Sinovial/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Nucleares/genética , Fosforilación , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética
9.
In Vivo ; 24(1): 45-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20133974

RESUMEN

BACKGROUND: In a large number of patients with multiple myeloma, chemotherapy is the only therapeutic option. During recent years, major effort has been put into immunotherapeutic approaches for this malignancy. MATERIALS AND METHODS: In this study, wild-type (wt) myeloma cells (5x10(5)) were injected subcutaneously into Balb/c mice. CD40L-transfected myeloma cells (5x10(5)) were subsequently injected intratumorally into the established (>100 mm(3)) wt tumor nodules. Overall survival and tumor growth were measured. RESULTS: Out of eight animals receiving wt tumor cells, one died prior to the formation of a solid tumor nodule. Following the CD40L-transfected myeloma cell injection, stable complete remission at day 60 with all the animals surviving resulted. On day 60, a re-challenge was performed with wt myeloma cells. No tumor growth was observed after 120 days out of seven remaining animals, one died. CONCLUSION: Intratumoral injection of CD40L-transfected myeloma cells induces complete tumor remission and long lasting immunity against tumor recurrence.


Asunto(s)
Ligando de CD40/genética , Plasmacitoma/terapia , Animales , Ligando de CD40/inmunología , Línea Celular Tumoral , Citometría de Flujo , Terapia Genética , Longevidad , Ratones , Ratones Endogámicos BALB C , Plasmacitoma/inmunología , Plasmacitoma/patología , Inducción de Remisión , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Pancreas ; 38(7): 758-65, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19546834

RESUMEN

OBJECTIVES: Fusion of dendritic cells (DC) with tumor cells is an approach in immunotherapy combining antigenicity and capacity of antigen presentation to activate T cells for the induction of tumor-specific cytotoxic immunity. Although there have been reports of clinical benefit, response rates have been limited and further improvements are warranted. METHODS: We used murine DC and a novel protocol for an effective fusion of those cells with the murine pancreatic cell line Panc02. RESULTS: We observed 2 events: only moderate in vitro and in vivo cytotoxicity of tumor cell/DC hybrids and a down-regulation of costimulatory molecules on fused cells. Therefore, we transfected tumor cell/DC hybrids with an adenovirus expressing CD154 to improve DC activation and generating antitumor immune response without the need of CD4 T cells. High CD154 expression could be obtained by transfection of DC and Panc02 cells prior fusion. Furthermore, vaccination with CD154-transfected tumor cell/DC hybrid led to a significantly increased induction of cytotoxic T cells in vitro and to an improved antitumoral effect in an orthotopic in vivo mouse model. CONCLUSIONS: CD154-transfected tumor cell/DC hybrids are a promising approach to increase the efficiency of antitumoral response.


Asunto(s)
Ligando de CD40/metabolismo , Células Dendríticas/metabolismo , Células Híbridas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Ligando de CD40/genética , Ligando de CD40/inmunología , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Células Híbridas/inmunología , Células Híbridas/trasplante , Inmunoterapia Adoptiva/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Transfección
12.
Pancreas ; 37(3): 321-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18815556

RESUMEN

OBJECTIVES: The major histocompatibility complex class II chaperone invariant chain (Ii) is widely used as a carrier for inserted antigenic sequences and their introduction into the class II processing pathway. The tumor-associated antigen core 2beta 1,6 N-acetylglucosaminyltransferase (C2GnT), a glycosyltransferase present in human pancreatic tumor cells, is not expressed by normal pancreatic tissues. METHODS: A set of expression vectors was engineered where the class II binding region of Ii was replaced by C2GnT-derived sequences. We investigated in vitro whether dendritic cells transfected with Ii-C2GnT constructs were capable to stimulate proliferation of CD4 T cells. We also tested whether vaccination with Ii-C2GnT would protect mice from tumor development. RESULTS: Invariant chain-C2GnT fusion proteins bind to human DR1, DR3, DR4 and to mouse I-A molecules. Our results demonstrate that the plasmid DNA encoding the C2GnT epitope embedded in Ii induces tumor-specific T-cell responses. Mice immunized with the Ii constructs showed reduced growth of Panc02 pancreatic tumor cells. CONCLUSIONS: Therefore, Ii clipped with the tumor-associated antigen C2GnT shows promise for the treatment of pancreatic cancer.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , N-Acetilglucosaminiltransferasas/inmunología , Neoplasias Pancreáticas/terapia , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Células COS , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Chlorocebus aethiops , Antígenos HLA-DR/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , N-Acetilglucosaminiltransferasas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Proteínas Recombinantes de Fusión/inmunología , Factores de Tiempo , Transfección
13.
Hepatology ; 48(1): 157-68, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18537185

RESUMEN

UNLABELLED: Dendritic cells (DCs) are professional antigen-presenting cells able to prime T-cells against tumor-associated antigens (TAA), but their potential to induce hepatocellular carcinoma (HCC) regression is still limited. CD40/CD40L interaction is essential for DC activation and induction of antigen-specific T-cells. In this study, transduction of TAA-pulsed DC with a CD40L-encoding adenovirus (Ad-CD40L) was used to improve the immune response induced by DC toward HCC. Bone marrow-derived DC from C3H/HeNcrl mice were cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4. On day 6, tumor-lysate pulsed DCs were infected with adenoviruses. HCCs were induced by inoculation of mice with Hepa129-cells subcutaneously. When tumor-volume was 100 to 400 mm(3), DCs were injected intratumorally, subcutaneously, or intravenously. Ad-CD40L transduction exerted CD40/CD40L interactions between DCs, increasing DC immunostimulation with up-regulation of CD80/CD86- and interleukin-12 (IL-12) expression. Intratumoral injection of CD40L-DC was superior to intravenous or subcutaneous treatments, yielding tumor elimination in almost 70% of mice. Moreover, all tumor-free animals were protected against hepatic tumor cell rechallenge. In a preventive setting, subcutaneous injection of CD40L-expressing DCs protected 50% of mice for more than 3 months toward tumor cell challenge. The induced immune response seemed to be dependent on cross-priming with Th1-lymphocytes in the lymph nodes, because transduced DCs were redetected in lymphoid tissues. In addition, immunohistochemistry of tumors indicated a significant tumor infiltration with CD4+, CD8+ T cells and natural killer (NK) cells. Tumor-infiltrating lymphocytes were tumor-specific, as shown in interferon-gamma (IFN-gamma) enzyme-linked immunosorbent spot and T-cell proliferation assays. CONCLUSION: Transduction of DCs with Ad-CD40L increases significantly the stimulatory capacity of DCs. Intratumoral injection of DCs activates both acquired and innate immunity, inducing complete regression of established tumors and long-term immunity against tumor recurrence. This approach improves the antitumoral potential of DCs.


Asunto(s)
Ligando de CD40/metabolismo , Carcinoma Hepatocelular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Neoplasias Hepáticas Experimentales/inmunología , Animales , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Línea Celular Tumoral , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Inmunidad Activa , Inmunidad Innata , Inyecciones Intralesiones , Interleucina-12/sangre , Interleucina-12/metabolismo , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/cirugía , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C3H , Recurrencia Local de Neoplasia/prevención & control , Fenotipo , Bazo/citología , Transducción Genética , Vacunación
14.
Urol Int ; 79(3): 235-43, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17940356

RESUMEN

PURPOSE: For a better understanding of the factors contributing to tumor progression in metastatic renal cell carcinoma and to identify possible targets for immunotherapeutic approaches, we characterized several primary cultures from renal cell carcinoma. MATERIALS AND METHODS: Cell cultures were tested for activity of telomerase, secretion of immunosuppressive cytokines and others. The induction of cytotoxic activity against the autologous tumor was tested in a cytotoxicity assay after coculture of immunological effector cells with antigen-pulsed dendritic cells. The data were tested for influence on survival. RESULTS: We were able to establish primary cell cultures from 58 patients with renal cell carcinoma and their metastasis. 48/58 were positive for telomerase activity and all secreted IL-6, TGF-beta, VEGF and IL-8. High TGF-beta secretion, the activity of telomerase and the induction of a telomerase-specific immune response against telomerase peptides in telomerase-positive tumors had a significant impact on survival. CONCLUSION: TGF-beta secretion, activity of telomerase in telomerase-positive tumors and the ability to generate a telomerase-specific immune response might serve as a prognostic marker for RCC. New approaches might focus on attacking the TGF-beta pathway and on induction of telomerase-specific immune cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Citocinas/metabolismo , Neoplasias Renales/metabolismo , Telomerasa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/inmunología , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/terapia , Técnicas de Cultivo de Célula , Citotoxicidad Inmunológica , Femenino , Humanos , Inmunoterapia/métodos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Estimación de Kaplan-Meier , Neoplasias Renales/enzimología , Neoplasias Renales/inmunología , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Neoplasias Renales/terapia , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Telomerasa/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Resultado del Tratamiento , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Arch Immunol Ther Exp (Warsz) ; 54(6): 403-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17122879

RESUMEN

INTRODUCTION: Ex vivo expansion of monocyte-derived dendritic cells (mDCs) and subsequent coculture with autologous cytokine-induced killer (CIK) cells is an established system to create specific and non-specific anti-tumoral immunity. mDCs constitute the most frequently applied DC subset in clinical studies. One recently published approach to optimize the immunological functions of the DC/CIK cell system is the replacement of interleukin (IL)-4 by interferon (IFN)-alpha in the maturation process of the DCs. MATERIALS AND METHODS: The expressions of relevant surface antigens of IL-4-DCs and IFNalpha-DCs by flow cytometry and the anti-tumoral activation of effector cells cocultured with both types of DCs using cytotoxicity assays were compared. In addition, short-term coculture experiments with both types of DCs and IFNgamma-LAK effector cells were performed and compared with standard CIK cell coculture experiments. RESULTS: Regarding the expressions of functionally relevant surface markers, no differences could be detected for CD80, CD83, and HLA-DR between IFNalpha-DCs and IL-4-DCs, whereas the mean fluorescence intensities of CD40, CD86, CD54, and HLA-ABC were decreased and the expression of CD14 was increased for IFNgamma-DCs. Moreover, no enhancement of cytotoxicity of cocultured CIK cells against tumor cell lines (A498 and SW480) was detected by the use of IFNalpha-DCs. Additionally, coculture experiments with IFNgamma-LAK cells were performed and unexpectedly higher lysis rates in comparison with the established IL-4-DC/CIK coculture model was observed. Early incubation of the mDCs with several CpG-ODNs failed to increase the anti-tumoral cytotoxicity of the cocultured IFNgamma-LAK cells. CONCLUSIONS: These results demonstrate that in the mDC/CIK cell system, IFNalpha-DCs are not superior in inducing anti-tumoral cytotoxicity and even moderately inferior regarding the expression of functionally relevant surface markers compared with IL-4-DCs.


Asunto(s)
Islas de CpG , Células Dendríticas/efectos de los fármacos , Interferón-alfa/farmacología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Neoplasias/terapia , Antígenos de Superficie/metabolismo , Comunicación Celular/inmunología , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-4/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/fisiología , Oligonucleótidos/farmacología
16.
Liver Int ; 26(3): 369-79, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16584401

RESUMEN

BACKGROUND/AIMS: Breaking immunologic tolerance towards the hepatocellular carcinoma (HCC)-associated alpha-fetoprotein (AFP) antigen is possible. The use of this potential for the treatment of immunocompromised HCC patients is limited. In this study, we analyzed whether dendritic cells (DCs) from HCC patients transduced with a human AFP (hAFP)-expressing adenovirus and co-cultured with cytokine-induced killer (CIK) cells can induce a strong specific immune response against HCC-cells. METHODS: An hAFP-encoding adenovirus (Ad-hAFP) was generated. DCs from healthy donors or patients were transduced at a very high efficacy. Afterwards, DCs were co-cultured with autologous CIK-cells, and their ability to lyse HCC-cells was analyzed. RESULTS: AFP-transduced DCs stimulated CIK cells strongly to lyse about 70% of AFP-expressing HCC cells. Cytotoxicity was significantly higher when lymphocytes were co-cultured with Ad-hAFP-transduced DCs than with Ad-mock-transduced DCs, indicating an AFP-specific immune response. More interestingly, CIK cells from patients with AFP-positive HCC could be stimulated to lyse AFP-expressing HCC cells as effectively as CIK cells from healthy individuals and stronger than CIK cells from patients without AFP-expressing HCC. CONCLUSIONS: The data demonstrate that patient-derived DCs that were transduced with an AFP-expressing adenovirus and co-cultured with autologous CIK cells induce an AFP-specific, strong immune response against HCC cells. Therefore, this approach may have a potential for an adoptive and/or DC-based immunotherapy for HCC patients.


Asunto(s)
Carcinoma Hepatocelular/terapia , Citocinas/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/terapia , Transducción Genética , alfa-Fetoproteínas/inmunología , Adenoviridae , Traslado Adoptivo , Presentación de Antígeno/inmunología , Antígenos de Diferenciación de Linfocitos T , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Carcinoma Hepatocelular/inmunología , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Citometría de Flujo , Antígenos HLA/inmunología , Antígenos HLA/metabolismo , Humanos , Inmunofenotipificación , Células Asesinas Naturales/metabolismo , Neoplasias Hepáticas/inmunología , Activación de Linfocitos , Trasplante Autólogo , Células Tumorales Cultivadas , alfa-Fetoproteínas/metabolismo
17.
Cancer ; 106(4): 759-64, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16369992

RESUMEN

BACKGROUND: Adoptive cell transfer is described as an innovative and challenging option for the treatment of malignant melanoma. In the current study, the generation and expansion of telomerase-specific T-cells for adoptive cell transfer and their use in a syngeneic pancreatic carcinoma mouse model was investigated. METHODS: Telomerase-specific T-cells were generated either in vitro by coculture of human lymphocytes with telomerase-peptide-pulsed dendritic cells or in vivo by injection of peptide plus adjuvant into C57BL/6 mice. Spleens were harvested after immunization and lymphocytes were expanded in the presence of feeder cells. T-cells were tested in vitro against human leukocyte antigen (HLA)-matched, telomerase-positive pancreatic carcinoma cells. Tumor-bearing (subcutaneous) mice pretreated with cyclophosphamide were injected intravenously with the expanded cells. RESULTS: It was possible to generate and expand telomerase-specific T-cells with cytotoxic activity. The protocol did not work as well in the murine setting. However, adoptive cell transfer with murine antigen-specific T-cells delayed disease progression in tumor-bearing mice significantly. CONCLUSIONS: Generation of antigen-specific T-cells is feasible; the expansion of these cells could be accomplished without loss of function. Antigen-specific T-cells demonstrated significant cytotoxic activity in a syngeneic, subcutaneous mouse model. However, further optimization of the expansion protocol is warranted.


Asunto(s)
Traslado Adoptivo , Carcinoma/terapia , Neoplasias Pancreáticas/terapia , Linfocitos T/inmunología , Telomerasa/metabolismo , Animales , Antígenos de Neoplasias , Antineoplásicos Alquilantes/administración & dosificación , Carcinoma/inmunología , Ciclofosfamida/administración & dosificación , Células Dendríticas , Progresión de la Enfermedad , Infusiones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/inmunología , Bazo/citología , Linfocitos T/enzimología , Células Tumorales Cultivadas
18.
Ger Med Sci ; 4: Doc02, 2006 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19675693

RESUMEN

OBJECTIVE: Therapeutic vaccination with dendritic cells (DC) showed promising results in first clinical trials in cases of metastatic renal cell carcinoma (RCC). Human telomerase reverse transcriptase (hTERT) could be a potential target because it is detectable in more than 85% of human tumors including RCC. DESIGN: 10 patients with progressive metastatic RCC were enrolled in a clinical phase I/II trial using DC pulsed with hTERT-peptide. Beside toxicity and feasibility aspects, a complex immune monitoring including in vitro data were evaluated. In addition to detection of tumor-specific effector cells we investigated their functionality like IFN-gamma secretion and cytotoxic activity against tumor cells. RESULTS: The vaccine was well tolerated. Two patients showed a mixed response (MR) and one patient a stable disease (SD). Interestingly, responders showed cytotoxic activity already before start of therapy and there was a significant increase in cytotoxic activity of effector cells from all responders (SD and MR patients) after the first vaccination. In contrast non-responders showed no cytotoxic activity before and during treatment. Therefore, cytotoxic activity might be used as a predictive marker in the future. Tetramer staining detected higher amounts of tumor-specific cytotoxic cells in responding patients compared to non-responders. Also, responders possessed increasing amounts of IFN-gamma producing immunological effector cells. CONCLUSION: Telomerase-pulsed DC could enhance a tumor-specific immune response against RCC.

19.
J Urol ; 171(1): 114-9, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14665857

RESUMEN

PURPOSE: Renal cell carcinoma (RCC) is known for its immunological susceptibility. Unfortunately RCC lacks specific tumor antigens for the induction of specific immunotherapy. We investigated the role of telomerase as a tumor antigen and pulsed dendritic cells (DCs) as antigen presenting cells with an immunogenic peptide from telomerase. MATERIAL AND METHODS: DCs and immunological effector cells, that is cytokine induced killer (CIK) cells, from patients with RCC or healthy donors were generated. CIK cells were co-cultured with telomerase peptide pulsed DCs. CIK cells were tested for cytotoxic activity against primary cultures. Using the dimer technique we determined the percent of telomerase specific T cells. Activation status was identified using interferon-gamma secretion assay. RESULTS: After pulsing DCs with telomerase peptide co-cultured CIK cells had a significant increase in cytotoxic activity against tumor cells compared with CIK cells without co-culture, that is 100% at an effector-to-target ratio of 60:1 vs 41.7% (p <0.05). Using a complete autologous model with immunological cells derived from patients with metastatic RCC we were able to induce cytotoxicity against autologous, telomerase positive primary cell cultures. We could detect 2.4% telomerase specific effector cells after co-culture with peptide pulsed DCs, which secreted interferon-gamma after re-stimulation. CONCLUSIONS: Telomerase could serve as a specific tumor associated antigen for RCC. The presentation of telomerase peptide by DCs to lymphocytes allows the generation of antigen specific cytotoxic effector cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Carcinoma de Células Renales/terapia , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias Renales/terapia , Telomerasa/inmunología , Carcinoma de Células Renales/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Neoplasias Renales/inmunología
20.
Immunology ; 107(1): 129-35, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12225371

RESUMEN

Major histocompatibility complex class I (MHCI) molecules are major targets of virus evasion strategies because they introduce antigens from the biosynthesis pathway into the antigen-processing and presentation pathways for immune recognition by CD8+ T cells. Little is known about viral strategies that interfere with the MHC class II (MHCII) antigen presentation pathway. We identified a six amino acid sequence from type I herpes simplex virus (HSV-1) glycoprotein B (gB) that is identical to a sequence of human leucocyte antigen D (HLA-D) -associated invariant chain (Ii). In addition, this gB sequence is adjacent to a highly conserved HLA-DR1 binding motif. Both viral sequences together resemble the class II binding site of human Ii, consisting of a MHCII groove binding segment and a promiscuous binding site. We cloned gB from HSV-1 strain 17 and demonstrate association of the virus envelope protein to three HLA-DR allotypes. With chimeric Ii/gB fusion proteins we identified gB sequences that mediate promiscuous or allotype-specific binding to the HLA-DR peptide-binding domain. Mutation of two Lys residues in the viral segment of Ii/gB abolished promiscuous binding to HLA-DR heterodimers. The result indicates promiscuous binding of the virus sequence to HLA-DR molecules and suggests a potential for HSV-1 to manipulate antigen processing and presentation.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Antígenos HLA-DR/metabolismo , Herpesvirus Humano 1/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Antígenos Virales/metabolismo , Sitios de Unión , Unión Competitiva , Células COS , Herpesvirus Humano 1/clasificación , Herpesvirus Humano 1/inmunología , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...