Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 107(4): 707-12, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22828609

RESUMEN

BACKGROUND: Despite focused research in conventional therapies and considerable advances in the understanding of the molecular carcinogenesis of head and neck squamous cell carcinoma (HNSCC), the 5-year survival rate for patients with advanced disease remains ∼15-20%. The major causes of HNSCC-related deaths are cervical node and distant metastasis. E-cadherin has a key role in epithelial intercellular adhesion and its downregulation is a hallmark of epithelial-mesenchymal transition (EMT), which is associated with invasion, metastasis, and poor prognosis. Epithelial-mesenchymal transition is the major mechanism responsible for mediating invasiveness and metastasis of epithelial cancers. Recently, we reported the role of E-cadherin transcriptional repressors in the inflammation-induced promotion of EMT in HNSCC, which is mediated by COX-2. These findings suggest that therapies targeting the cyclooxygenase pathway may diminish the propensity for tumour metastasis in HNSCC by blocking the PGE2-mediated induction of E-cadherin transcriptional repressors. METHODS: Herein, we evaluate the efficacy of the COX-2 inhibitor, apricoxib, in HNSCC cell lines. Apricoxib is effective in preventing tumour cell growth in three-dimensional, and anchorage-independent growth assays, as well as decreasing the capacity for tumour cell migration. RESULTS: Herein, we evaluate the efficacy of the COX-2 inhibitor, apricoxib, in HNSCC cell lines. Apricoxib is effective in preventing tumour cell growth in three-dimensional, and anchorage-independent growth assays, as well as decreasing the capacity for tumour cell migration. Treatment of HNSCC cells with apricoxib also causes greater upregulation of E-cadherin and Muc1 expression and downregulation of vimentin, as compared with celecoxib treatment. This has significant implications for targeted chemoprevention and anti-cancer therapy because E-cadherin expression has been implicated as a marker of sensitivity to epidermal growth factor receptor tyrosine kinase inhibitor and other therapies. We show for the first time the molecular mechanisms underlying the efficacy of apricoxib in HNSCC cells. CONCLUSION: In addition to reversing EMT via inhibition of COX-2, apricoxib upregulates 15-prostaglandin dehydrogenase and the prostaglandin transporter, thereby reducing the levels of active PGE2 by both suppressing its synthesis and increasing its catabolism. These findings have significant implications for metastasis and tumour progression in HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Transportadores de Anión Orgánico/metabolismo , Pirroles/farmacología , Sulfonamidas/farmacología , Cadherinas/metabolismo , Carcinoma de Células Escamosas/etiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/etiología , Humanos , Hidroxiprostaglandina Deshidrogenasas , Fumar/efectos adversos , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba , Vimentina/metabolismo
2.
Oncogene ; 26(29): 4253-60, 2007 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-17237818

RESUMEN

Cyclooxygenase-2 (COX-2) is frequently overexpressed in human cancers and contributes to the malignant phenotype. Our data indicate unphosphorylated signal transducers and activators of transcription 6 (STAT6) may transcriptionally upregulate COX-2 expression and protect against apoptosis in NSCLC cells. In A427 and H2122, NSCLC cell lines that constitutively express COX-2, only unphosphorylated STAT6 was detectable by western blot, thus, all of the following STAT6-dependent effects are attributed to the unphosphorylated protein. In both cell lines, small-interfering RNA-mediated knockdown of STAT6 or stable expression of dominant-negative STAT6 decreased COX-2 expression. In contrast, transfection with a phosphorylation-deficient mutant STAT6 increased COX-2 levels. Immunofluorescent staining revealed the presence of STAT6 in H2122 nuclei, suggesting a direct role in gene regulation for the unphosphorylated protein. Consistent with this hypothesis, unphosphorylated STAT6 increased luciferase expression from a COX-2 promoter reporter construct. STAT6 co-immunoprecipitated with the transcriptional co-activator, p300, and chromatin immunoprecipitation assays demonstrated that these proteins bind a consensus STAT6 binding site located within the COX-2 promoter. STAT6 DNA-binding specificity was confirmed by electrophoretic mobility shift assay. As COX-2 over-expression has been clearly linked to apoptosis resistance and other hallmarks of malignancy, these findings suggest a novel role of unphosphorylated STAT6 in the pathogenesis of non-small cell lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Ciclooxigenasa 2/genética , Neoplasias Pulmonares/enzimología , Factor de Transcripción STAT6/metabolismo , Regulación hacia Arriba/fisiología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular , Línea Celular Tumoral , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Factor de Transcripción STAT6/antagonistas & inhibidores , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/fisiología , Regulación hacia Arriba/genética
3.
Br J Cancer ; 94(7): 1029-34, 2006 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-16598185

RESUMEN

Epstein-Barr virus-induced molecule 1 ligand chemokine (CCL19) is a CC chemokine that chemoattracts both dendritic cells (DC) and T lymphocytes. We evaluated the antitumour efficacy of CCL19 in a murine model of spontaneous bronchoalveolar cell carcinoma. These transgenic mice (CC-10 TAg) express the SV40 large T antigen under the Clara Cell promoter, develop bilateral, multifocal, pulmonary carcinomas and die at 4 months owing to progressive pulmonary tumour burden. To mimic therapy in late-stage disease, 3-month-old transgenic mice were treated with recombinant CCL19 (0.5 microg dose(-1)) by intranodal (axillary lymph node region) injection three times per week for 4 weeks. CCL19 treatment led to a marked reduction in tumour burden with extensive mononuclear infiltration of the tumours compared to diluent treated controls. Flow cytometric analyses showed significant increases in CD4 and CD8T cell subsets as well as DC in the lungs of CCL19-treated mice. Lung tissue cytokine profiles showed a shift towards immune stimulatory molecules with a decrease in the immunosuppressive cytokine TGF-beta. Our findings show that CCL19 may serve as a potential immune stimulator and provide a strong rationale for the evaluation of CCL19 in cancer immunotherapy.


Asunto(s)
Adenocarcinoma Bronquioloalveolar/tratamiento farmacológico , Quimiocinas CC/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma Bronquioloalveolar/inmunología , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Quimiocina CCL19 , Quimiocinas CC/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Inmunoterapia , Neoplasias Pulmonares/inmunología , Ratones , Ratones Transgénicos
4.
Cancer Gene Ther ; 13(1): 74-81, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16052231

RESUMEN

While external ionizing radiation has been used for treating non-small cell lung cancer (NSCLC), improved efficacy of this modality would be an important advance. Ectopic expression of the sodium iodide symporter (NIS) and thyroperoxidase (TPO) genes in NSCLC cells facilitated concentration of iodide in NSCLC cells, which markedly induced apoptosis in vitro and in vivo. Pre-incubation of the NIS/TPO-modified NSCLC cells in iodide followed by ionizing radiation generates bystander tumoricidal effects and potently enhances tumor cell killing. This iodide-induced bystander effect is associated with enhanced gap junction intercellular communication (GJIC) activity and increased connexin-43 (Cx43) expression. Thus, iodide may serve as an enhancer to markedly improve the efficacy of radiation therapy in combined therapeutic modalities.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Muerte Celular/efectos de la radiación , Yoduros/farmacología , Neoplasias Pulmonares/metabolismo , Apoptosis/efectos de los fármacos , Autoantígenos/metabolismo , Autoantígenos/farmacología , Efecto Espectador , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Conexina 43/metabolismo , Terapia Genética , Humanos , Yoduro Peroxidasa/metabolismo , Yoduro Peroxidasa/farmacología , Yoduros/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas de Unión a Hierro/farmacología , Neoplasias Pulmonares/genética , Radiación Ionizante , Sensibilidad y Especificidad , Simportadores/metabolismo , Transfección
6.
Cancer Gene Ther ; 8(8): 612-8, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11571539

RESUMEN

Radioiodide is an effective therapy for thyroid cancer. This treatment modality exploits the thyroid-specific expression of the sodium iodide symporter (NIS) gene, which allows rapid internalization of iodide into thyroid cells. To test whether a similar treatment strategy could be exploited in nonthyroid malignancies, we transfected non-small cell lung cancer (NSCLC) cell lines with the NIS gene. Although the expression of NIS allowed significant radioiodide uptake in the transfected NSCLC cell lines, rapid radioiodide efflux limited tumor cell killing. Because thyroperoxidase (TPO) catalyzes iodination of proteins and subsequently causes iodide retention within thyroid cells, we hypothesized that coexpression of both NIS and TPO genes would overcome this deficiency. Our results show that transfection of NSCLC cells with both human NIS and TPO genes resulted in an increase in radioiodide uptake and retention and enhanced tumor cell apoptosis. These findings suggest that single gene therapy with only the NIS gene may have limited efficacy because of rapid efflux of radioiodide. In contrast, the combination of NIS and TPO gene transfer, with resulting TPO-mediated organification and intracellular retention of radioiodide, may lead to more effective tumor cell death. Thus, TPO could be used as a therapeutic strategy to enhance the NIS-based radioiodide concentrator gene therapy for locally advanced lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Yoduro Peroxidasa/genética , Radioisótopos de Yodo/metabolismo , Neoplasias Pulmonares/radioterapia , Simportadores/genética , Apoptosis , Southern Blotting , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Cartilla de ADN/química , Inhibidores Enzimáticos/farmacología , Terapia Genética , Humanos , Yoduro Peroxidasa/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Matriz Nuclear/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Simportadores/metabolismo , Factores de Tiempo , Transfección , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/efectos de la radiación
7.
Cancer Res ; 61(17): 6406-12, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11522634

RESUMEN

The antitumor efficiency of secondary lymphoid organ chemokine (SLC), a CC chemokine that chemoattracts both dendritic cells (DCs) and T lymphocytes,was evaluated in SV40 large T-antigen transgenic mice that develop bilateral multifocal pulmonary adenocarcinomas. Injection of recombinant SLC in the axillary lymph node region led to a marked reduction in tumor burden with extensive lymphocytic and DC infiltration of the tumors and enhanced survival. SLC injection led to significant increases in CD4 and CD8 lymphocytes as well as DC at the tumor sites, lymph nodes, and spleen. The cellular infiltrates were accompanied by the enhanced elaboration of Type 1 cytokines and the antiangiogenic chemokines IFN-gamma inducible protein 10, and monokine induced by IFN-gamma (MIG). In contrast, lymph node and tumor site production of the immunosuppressive cytokine transforming growth factor beta was decreased in response to SLC treatment. In vitro, after stimulation with irradiated autologous tumor, splenocytes from SLC-treated mice secreted significantly more IFN-gamma and granulocyte macrophage colony-stimulating factor, but reduced levels of interleukin 10. Significant reduction in tumor burden in a model in which tumors develop in an organ-specific manner provides a strong rationale for additional evaluation of SLC in regulation of tumor immunity and its use in lung cancer immunotherapy.


Asunto(s)
Adenocarcinoma Bronquioloalveolar/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Quimiocinas CC/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma Bronquioloalveolar/irrigación sanguínea , Adenocarcinoma Bronquioloalveolar/genética , Adenocarcinoma Bronquioloalveolar/inmunología , Inhibidores de la Angiogénesis/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/inmunología , Quimiocina CCL21 , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Factores de Crecimiento Endotelial/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Linfocinas/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Proteínas Recombinantes/farmacología , Bazo/citología , Bazo/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
8.
J Biol Chem ; 276(24): 20809-12, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11320076

RESUMEN

Elevated tumor cyclooxygenase (COX-2) expression is associated with increased angiogenesis, tumor invasion, and suppression of host immunity. We have previously shown that genetic inhibition of tumor COX-2 expression reverses the immunosuppression induced by non-small cell lung cancer (NSCLC). To assess the impact of COX-2 expression in lung cancer invasiveness, NSCLC cell lines were transduced with a retroviral vector expressing the human COX-2 cDNA in the sense (COX-2-S) and antisense (COX-2-AS) orientations. COX-2-S clones expressed significantly more COX-2 protein, produced 10-fold more prostaglandin E(2), and demonstrated an enhanced invasive capacity compared with control vector-transduced or parental cells. CD44, the cell surface receptor for hyaluronate, was overexpressed in COX-2-S cells, and specific blockade of CD44 significantly decreased tumor cell invasion. In contrast, COX-2-AS clones had a very limited capacity for invasion and showed diminished expression of CD44. These findings suggest that a COX-2-mediated, CD44-dependent pathway is operative in NSCLC invasion. Because tumor COX-2 expression appears to have a multifaceted role in conferring the malignant phenotype, COX-2 may be an important target for gene or pharmacologic therapy in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Receptores de Hialuranos/fisiología , Isoenzimas/metabolismo , Neoplasias Pulmonares/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Antígenos CD/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Ciclooxigenasa 2 , Dinoprostona/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Ácido Hialurónico/metabolismo , Isoenzimas/genética , Neoplasias Pulmonares/patología , Proteínas de la Membrana , Invasividad Neoplásica , Prostaglandina-Endoperóxido Sintasas/genética , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
9.
J Immunol ; 165(1): 373-80, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10861074

RESUMEN

In this study, we show that Delta-9-tetrahydrocannabinol (THC), the major psychoactive component of marijuana, suppresses host immune reactivity against lung cancer. In two different weakly immunogenic murine lung cancer models, intermittent administration of THC (5 mg/kg, four times/wk i.p. for 4 wk) led to accelerated growth of tumor implants compared with treatment with diluent alone. In contrast to our findings in immunocompetent mice, THC did not affect tumor growth in tumor-bearing SCID mice. The immune inhibitory cytokines, IL-10 and TGF-beta, were augmented, while IFN-gamma was down-regulated at both the tumor site and in the spleens of THC-treated mice. Administration of either anti-IL-10- or anti-TGF-beta-neutralizing Abs prevented the THC-induced enhancement in tumor growth. Both APC and T cells from THC-treated mice showed limited capacities to generate alloreactivity. Furthermore, lymphocytes from THC-treated mice transferred the effect to normal mice, resulting in accelerated tumor growth similar to that seen in the THC-treated mice. THC decreased tumor immunogenicity, as indicated by the limited capacity for tumor-immunized, THC-treated mice to withstand tumor rechallenge. In vivo administration of a specific antagonist of the CB2 cannabinoid receptor also blocked the effects of THC. Our findings suggest the THC promotes tumor growth by inhibiting antitumor immunity by a CB2 receptor-mediated, cytokine-dependent pathway.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Citocinas/fisiología , Dronabinol/farmacología , Inmunosupresores/farmacología , Receptores de Droga/fisiología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Células Presentadoras de Antígenos/inmunología , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/prevención & control , División Celular/efectos de los fármacos , División Celular/inmunología , Dronabinol/antagonistas & inhibidores , Dronabinol/metabolismo , Inhibidores de Crecimiento/administración & dosificación , Inmunidad Innata/efectos de los fármacos , Inmunosupresores/antagonistas & inhibidores , Inmunosupresores/metabolismo , Inyecciones Intraperitoneales , Interleucina-10/inmunología , Prueba de Cultivo Mixto de Linfocitos , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Subgrupos Linfocitarios/trasplante , Transfusión de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Trasplante de Neoplasias , Receptores de Cannabinoides , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Células Tumorales Cultivadas
11.
J Immunol ; 164(9): 4558-63, 2000 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10779757

RESUMEN

Secondary lymphoid tissue chemokine (SLC, also referred to as Exodus 2 or 6Ckine) is a recently identified high endothelial-derived CC chemokine. The ability of SLC to chemoattract both Th1 lymphocytes and dendritic cells formed the rationale to evaluate this chemokine in cancer immunotherapy. Intratumoral injection of recombinant SLC evidenced potent antitumor responses and led to complete tumor eradication in 40% of treated mice. SLC-mediated antitumor responses were lymphocyte dependent as evidenced by the fact that this therapy did not alter tumor growth in SCID mice. Studies performed in CD4 and CD8 knockout mice also revealed a requirement for both CD4 and CD8 lymphocyte subsets for SLC-mediated tumor regression. In immunocompetent mice, intratumoral SLC injection led to a significant increase in CD4 and CD8 T lymphocytes and dendritic cells, infiltrating both the tumor and the draining lymph nodes. These cell infiltrates were accompanied by the enhanced elaboration of Th1 cytokines and chemokines monokine induced by IFN-gamma and IFN-gamma-inducible protein 10 but a concomitant decrease in immunosuppressive cytokines at the tumor site. In response to irradiated autologous tumor, splenic and lymph node-derived cells from SLC-treated tumor-bearing mice secreted significantly more IFN-gamma, GM-CSF, and IL-12 and reduced levels of IL-10 than did diluent-treated tumor-bearing mice. After stimulation with irradiated autologous tumor, lymph node-derived lymphocytes from SLC-treated tumor-bearing mice demonstrated enhanced cytolytic capacity, suggesting the generation of systemic immune responses. These findings provide a strong rationale for further evaluation of SLC in tumor immunity and its use in cancer immunotherapy.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Quimiocinas CC/inmunología , Quimiocinas CC/uso terapéutico , Neoplasias Pulmonares/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/terapia , Quimiocina CCL21 , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CC/administración & dosificación , Quimiocinas CXC/biosíntesis , Citocinas/biosíntesis , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inyecciones Intralesiones , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucina-12/biosíntesis , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Subgrupos de Linfocitos T/metabolismo , Células Tumorales Cultivadas
12.
Am J Respir Cell Mol Biol ; 22(5): 613-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10783134

RESUMEN

Direct in vivo gene delivery is a prerequisite for many gene therapy strategies; however, efficacy has been limited by a lack of therapeutic gene transfer. In studying intrapleural malignancy as a model for the gene therapy of non-small cell lung cancer, we previously identified soluble chondroitin sulfate-proteoglycans/glycosaminoglycans (CS-PG/GAGs) in malignant pleural effusions (MPE) as factors that inhibit retroviral vector (RV) transduction. Similarly, we have observed inhibition to gene transfer in the fluid component of MPE using adenoviral (Ad) vectors. Analyses indicate that the factors responsible for the block are filterable, soluble, titrable, and heat stable (56 degrees C). Passage through microporous membranes fractionates the inhibitory factors into large (> 100 kD) components of the effusions. In contrast to RV transduction, hyaluronic acid or CS-PG/GAGs are not the inhibitors because the block is not reversed by pretreatment of the effusions with mammalian hyaluronidase, and exogenous addition of GAGs into the transduction media does not diminish Ad transduction. In considering the mechanism of action of the inhibitory factors, we observe that Ad entry, and specifically the binding of radiolabeled Ad to its target cell, is inhibited in the presence of MPE. Ad internalization may also be impaired; however, these studies exclude soluble fibronectin in MPE as a competitive inhibitor of Ad transduction. Lastly, sepharose A- mediated immunoglobulin depletion of MPE only partially reverses the block, and significant inhibition to Ad gene transfer persists at lower adenovirus:target cell ratios. Identifying the structural and functional basis for inhibition to Ad gene transfer may yield specific strategies to enable better in vivo translation of gene therapy approaches.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Derrame Pleural Maligno/química , Carcinoma de Pulmón de Células no Pequeñas , Proteínas de la Matriz Extracelular/química , Genes Reporteros , Terapia Genética , Glicosaminoglicanos/farmacología , Humanos , Hialuronoglucosaminidasa/farmacología , Neoplasias Pulmonares , Derrame Pleural Maligno/clasificación , Derrame Pleural Maligno/genética , Células Tumorales Cultivadas
13.
Hum Gene Ther ; 11(1): 53-65, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10646639

RESUMEN

In two murine lung cancer models adenoviral interleukin 7-transduced dendritic cells (DC-AdIL-7) were administered intratumorally, resulting in complete tumor regression. Intratumoral DC-AdIL-7 therapy was as effective as DCs pulsed with specific tumor peptide antigens. Comparison with other intratumoral therapies including recombinant IL-7, AdIL-7 vector alone, unmodified DCs, IL-7-transduced fibroblasts, or DCs pulsed with tumor lysates revealed DC-AdIL-7 therapy to be superior in achieving antitumor responses and augmenting immunogenicity. Mice with complete tumor eradication as a result of either DC-AdIL-7 or AdIL-7 therapy were rechallenged with parental tumor cells 30 days or more after complete tumor eradication. All the DC-AdIL-7-treated mice completely rejected a secondary rechallenge, whereas the AdIL-7-treated mice had sustained antitumor effects in only 20-25% of the mice. DC-AdIL-7 therapy was more effective than AdIL-7 in achieving systemic antitumor responses and enhancing immunogenicity. After complete tumor eradication, those mice treated with DC-AdIL-7 evidenced significantly greater release of splenocyte GM-CSF and IFN-gamma than did controls or AdIL-7-treated mice. After intratumoral injection, gene-modified DCs trafficked from the tumor to lymph node sites and spleen. DCs were detected in nodal tissues for up to 7 days after intratumoral injection. We report that intratumoral DC-AdIL-7 leads to significant systemic immune responses and potent antitumor effects in murine lung cancer models.


Asunto(s)
Adenoviridae/genética , Células Dendríticas/inmunología , Interleucina-7/genética , Neoplasias Pulmonares/terapia , Animales , Femenino , Terapia Genética , Inmunoterapia , Inyecciones Intralesiones , Interleucina-7/administración & dosificación , Neoplasias Pulmonares/inmunología , Ganglios Linfáticos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Inducción de Remisión , Bazo/inmunología
14.
J Immunol ; 164(3): 1269-76, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10640740

RESUMEN

Tumors produce a number of immunosuppressive factors that block the maturation of CD34+ stem cells into dendritic cells (DC). We hypothesized that tumors might also interfere with the maturation and/or function of human monocyte-derived DC. In contrast to stem cells, we found that CD14+ cells responded to tumor culture supernatant (TSN) by increasing expression of APC surface markers, up-regulating nuclear translocation of RelB, and developing allostimulatory activity. Although displaying these characteristics of mature DC, TSN-exposed DC lacked the capacity to produce IL-12, did not acquire full allostimulatory activity, and rapidly underwent apoptosis. The effects of TSN appeared to be specific for maturing DC, and were not reversed by Abs against known DC regulatory factors including IL-10, vascular endothelial growth factor, TGF-beta, or PGE2. Supernatants collected from nonmalignant cell sources had no effect on DC maturation. The altered maturation and early apoptosis of monocyte-derived DC may represent another mechanism by which tumors evade immune detection.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Monocitos/citología , Monocitos/inmunología , Células Tumorales Cultivadas/inmunología , Transporte Biológico/inmunología , Neoplasias de la Mama , Carcinoma de Células Renales , Diferenciación Celular/inmunología , Núcleo Celular/metabolismo , Sistema Libre de Células/inmunología , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/metabolismo , Humanos , Inmunofenotipificación , Interleucina-12/farmacología , Neoplasias Pulmonares , Melanoma , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción ReIB , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas/química
15.
Semin Respir Crit Care Med ; 21(5): 463-72, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-16088747

RESUMEN

Lung cancer is the leading cause of cancer-related mortality in men and women in the United States, in part, because of the poor treatment options available. New treatment strategies that specifically target discreet steps in the molecular and cellular pathogenesis of this disease are under development. This review highlights many of the basic defects that result in the cellular transformation and subsequent progression of lung cancer, and how the understanding of those fundamental defects lead to the formulation of rational gene-based or cell-based therapies.

16.
J Immunol ; 164(1): 361-70, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10605031

RESUMEN

Cyclooxygenase-2 (COX-2), the enzyme at the rate-limiting step of prostanoid production, has been found to be overexpressed in human lung cancer. To evaluate lung tumor COX-2 modulation of antitumor immunity, we studied the antitumor effect of specific genetic or pharmacological inhibition of COX-2 in a murine Lewis lung carcinoma (3LL) model. Inhibition of COX-2 led to marked lymphocytic infiltration of the tumor and reduced tumor growth. Treatment of mice with anti-PGE2 mAb replicated the growth reduction seen in tumor-bearing mice treated with COX-2 inhibitors. COX-2 inhibition was accompanied by a significant decrement in IL-10 and a concomitant restoration of IL-12 production by APCs. Because the COX-2 metabolite PGE2 is a potent inducer of IL-10, it was hypothesized that COX-2 inhibition led to antitumor responses by down-regulating production of this potent immunosuppressive cytokine. In support of this concept, transfer of IL-10 transgenic T lymphocytes that overexpress IL-10 under control of the IL-2 promoter reversed the COX-2 inhibitor-induced antitumor response. We conclude that abrogation of COX-2 expression promotes antitumor reactivity by restoring the balance of IL-10 and IL-12 in vivo.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Isoenzimas/farmacología , Prostaglandina-Endoperóxido Sintasas/farmacología , Adyuvantes Inmunológicos/fisiología , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/prevención & control , Ciclooxigenasa 2 , Dinoprostona/antagonistas & inhibidores , Dinoprostona/biosíntesis , Dinoprostona/fisiología , Regulación hacia Abajo/inmunología , Inducción Enzimática/inmunología , Femenino , Interleucina-12/antagonistas & inhibidores , Isoenzimas/biosíntesis , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Bazo/citología , Bazo/metabolismo , Células Tumorales Cultivadas
17.
J Immunol ; 163(9): 5020-8, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10528207

RESUMEN

We have found previously that human lung cancers potently induce T lymphocyte IL-10 production in vitro. To assess the impact of enhanced T cell-derived IL-10 on antitumor immunity in vivo, we utilized transgenic mice expressing IL-10 under the control of the IL-2 promoter. We have shown previously that Lewis lung carcinoma cells (3LL) have more aggressive growth potential in IL-10 transgenic mice compared with control littermates. In this study, we show that transfer of T cells from IL-10 transgenic mice to control littermates transferred the IL-10 immunosuppressive effect and led to enhanced 3LL tumor growth. In addition to changes in T cell-mediated immunity, professional APC from IL-10 transgenic mice were found to have significantly suppressed capacity to induce MHC alloreactivity, CTL responses, and IL-12 production. Tumor Ag-pulsed dendritic cells from IL-10 transgenic mice also failed to generate antitumor reactivity. These results suggest that increased levels of T cell-derived IL-10 severely impair antitumor immunity in vivo, due to defects in both T cell and APC function.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Inmunosupresores/farmacología , Interleucina-10/fisiología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/metabolismo , Carcinoma Pulmonar de Lewis/etiología , Carcinoma Pulmonar de Lewis/genética , División Celular/inmunología , Citocinas/biosíntesis , Citocinas/deficiencia , Citotoxicidad Inmunológica/genética , Células Dendríticas/inmunología , Femenino , Humanos , Interleucina-10/biosíntesis , Interleucina-10/genética , Interleucina-12/biosíntesis , Interleucina-12/deficiencia , Interleucina-12/genética , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/metabolismo , Linfocitos T/trasplante , Linfocitos T Citotóxicos/inmunología , Células TH1/metabolismo , Células Tumorales Cultivadas
18.
Am J Respir Cell Mol Biol ; 21(2): 238-45, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10423407

RESUMEN

Current paradigms in cancer therapy suggest that activation of nuclear factor-kappaB (NF-kappaB) by a variety of stimuli, including some cytoreductive agents, may inhibit apoptosis. Thus, inhibiting NF-kappaB activation may sensitize cells to anticancer therapy, thereby providing a more effective treatment for certain cancers. E-1-deleted adenoviral (Ad) vectors encoding a "superrepressor" form of the NF-kappaB inhibitor IkappaBalpha (AdIkappaBalphaSR) or beta-galactosidase (AdLacZ) were tested alone and in combination with tumor necrosis factor-alpha (TNF-alpha) in lung cancer cells for sensitization of the cells to death. Following transduction with AdIkappaBalphaSR, lung cancer cells expressed IkappaBalphaSR in a dose-dependent manner. Probing nuclear extracts of lung cancer cells with NF-kappaB-sequence-specific oligonucleotides indicated that there was a minimal amount of NF-kappaB in the nucleus at baseline and an expected and dramatic increase in nuclear NF-kappaB following exposure of cells to TNF-alpha. Control E-1-deleted AdLacZ did not promote NF-kappaB activation. Importantly, AdIkappaBalphaSR-mediated gene transfer resulted in the complete block of nuclear translocation of NF-kappaB by specific binding of its p65/relA component with transgenic IkappaBalphaSR. At the cellular level, transduction with AdIkappaBalphaSR resulted in increased cytotoxicity in lung cancer cells as opposed to transduction with equivalent doses of AdLacZ. In addition, whereas the parental cells were resistant to TNF-alpha-mediated cytotoxicity, IkappaBalphaSR-transduced cells could be sensitized to TNF-alpha. Consequently, AdIkappaBalphaSR transduction followed by exposure to TNF-alpha uniformly resulted in the death of non-small-cell lung cancer cells. These data suggest that novel approaches incorporating IkappaBalpha gene therapy may have a role in the treatment of lung cancer.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Proteínas de Unión al ADN/genética , Técnicas de Transferencia de Gen , Proteínas I-kappa B , Factor de Necrosis Tumoral alfa/farmacología , Adenoviridae/genética , Carcinoma de Células Escamosas/metabolismo , Muerte Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Vectores Genéticos , Humanos , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Transducción Genética , Transgenes , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
19.
Novartis Found Symp ; 215: 54-66; discussion 66-72, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9760571

RESUMEN

The overall T cell response to a multideterminant antigen consists of the sum of responses to a limited number of different determinants on the protein. Antigen-presenting cells (APCs) are crucial in delimiting the determinants on the protein to which a response will be mounted. This influence is apparent at two levels. First, the determinants that are generated and displayed by APCs in the thymus are pivotal in shaping the T cell repertoire that will be available for responding to antigen determinants in the periphery. Second, antigen processing affects the selection of determinants that become displayed by the various peripheral APC populations that are involved in inducing and promoting a T cell response. We have studied the effect of the display hierarchy on tolerance induction to individual determinants in transgenic mice expressing different serum levels of hen egg lysozyme. We have also analysed aspects of the processing machinery that contribute to shaping the hierarchy of determinant display on MHC class II molecules: proteolysis and reduction of disulfide bonds.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Epítopos , Tolerancia Inmunológica , Linfocitos T/inmunología , Aminoácidos Diaminos/inmunología , Animales , Presentación de Antígeno , Disulfuros/metabolismo , Ratones , Fragmentos de Péptidos/inmunología
20.
Hematol Oncol Clin North Am ; 12(3): 569-94, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9684099

RESUMEN

Gene therapy has received considerable attention and some speculation as to its value. Although few patients have been treated, the preliminary results of the phase I lung cancer gene therapy clinical trials are very promising. Clinically relevant basic research in the molecular pathogenesis and immunology of lung cancer is progressing. As improved vector technologies are developed, new opportunities will be available to initiate lung cancer gene therapy trials that are based on a more detailed understanding of lung cancer biology. In conclusion, although important biologic and technical questions remain unanswered, recent research suggests that gene therapy will have a profound impact on lung cancer treatment.


Asunto(s)
Terapia Genética , Inmunoterapia , Neoplasias Pulmonares/terapia , Animales , Vacunas contra el Cáncer/uso terapéutico , Terapia Combinada , Terapia Genética/métodos , Vectores Genéticos , Humanos , Inmunoterapia/métodos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...