Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Immunol Immunother ; 64(10): 1215-27, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26076666

RESUMEN

The MYCN oncogene is a strong genetic marker associated with poor prognosis in neuroblastoma (NB). Therefore, MYCN gene amplification and subsequent overexpression provide a possible target for new treatment approaches in NB. We first identified an inverse correlation of MYCN expression with CD45 mRNA in 101 NB tumor samples. KEGG mapping further revealed that MYCN expression was associated with immune-suppressive pathways characterized by a down-regulation of T cell activation and up-regulation of T cell inhibitory gene transcripts. We then aimed to investigate whether DNA vaccination against MYCN is effective to induce an antigen-specific and T cell-mediated immune response. For this purpose, we generated a MYCN-expressing syngeneic mouse model by MYCN gene transfer to NXS2 cells. MYCN-DNA vaccines were engineered based on the pCMV-F3Ub plasmid backbone to drive ubiquitinated full-length MYCN-cDNA and minigene expression. Vaccines were delivered orally with attenuated S. typhimurium strain SL7207 as a carrier. Immunization with both MYCN-DNA vaccines significantly reduced primary tumor growth of MYCN-expressing NB cells in contrast to negative controls. The immune response was mediated by tumor-infiltrating T cells in vivo, which revealed MYCN-specific and MHC class I-restricted lysis of inducible MYCN-expressing NB target cells in vitro. Finally, these antigen-specific T cells also killed MYCN-negative mammary carcinoma cells pulsed with MYCN peptides in contrast to controls. In summary, we demonstrate proof of concept that MYCN can be targeted by DNA vaccination, which may provide an approach to overcoming MYCN immune-suppressive activities in patients with MYCN-amplified disease.


Asunto(s)
Carcinoma/inmunología , Epítopos de Linfocito B/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/inmunología , Neuroblastoma/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Vacunas contra la Salmonella/administración & dosificación , Salmonella typhimurium/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas de ADN/administración & dosificación , Administración Oral , Animales , Carcinoma/microbiología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Epítopos de Linfocito B/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Animales/microbiología , Ratones , Ratones Endogámicos , Proteína Proto-Oncogénica N-Myc , Trasplante de Neoplasias , Neoplasias Experimentales , Neuroblastoma/genética , Neuroblastoma/microbiología , Fragmentos de Péptidos , Proteínas Proto-Oncogénicas/genética , Transgenes/genética , Carga Tumoral , Vacunación
2.
Clin Dev Immunol ; 2010: 169484, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21197271

RESUMEN

Cancer is one of the most challenging diseases of today. Optimization of standard treatment protocols consisting of the main columns of chemo- and radiotherapy followed or preceded by surgical intervention is often limited by toxic side effects and induction of concomitant malignancies and/or development of resistant mechanisms. This requires the development of therapeutic strategies which are as effective as standard therapies but permit the patients a life without severe negative side effects. Along this line, the development of immunotherapy in general and the innovative concept of DNA vaccination in particular may provide a venue to achieve this goal. Using the patient's own immune system by activation of humoral and cellular immune responses to target the cancer cells has shown first promising results in clinical trials and may allow reduced toxicity standard therapy regimen in the future. The main challenge of this concept is to transfer the plethora of convincing preclinical and early clinical results to an effective treatment of patients.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , ADN de Neoplasias/inmunología , Neoplasias/terapia , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/genética , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Neoplasias/genética , Neoplasias/inmunología , Vacunación
3.
Int J Cancer ; 125(1): 104-14, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19291796

RESUMEN

The inhibitor of apoptosis protein survivin is highly expressed in neuroblastoma (NB) and survivin-specific T cells were identified in Stage 4 patients. Therefore, we generated a novel survivin minigene DNA vaccine (pUS-high) encoding exclusively for survivin-derived peptides with superior MHC class I (H2-K(k)) binding affinities and tested its efficacy to suppress tumor growth and metastases in a syngeneic NB mouse model. Vaccination was performed by oral gavage of attenuated Salmonella typhimurium SL7207 carrying pUS-high. Mice receiving the pUS-high in the prophylactic setting presented a 48-52% reduction in s.c. tumor volume, weight and liver metastasis level in contrast to empty vector controls. This response was as effective as a survivin full-length vaccine and was associated with an increased target cell lysis, increased presence of CD8(+) T-cells at the primary tumor site and enhanced production of proinflammatory cytokines by systemic CD8(+) T cells. Furthermore, depletion of CD8(+) but not CD4(+) T-cells completely abrogated the pUS-high mediated primary tumor growth suppression, demonstrating a CD8(+) T-cell mediated effect. Therapeutic vaccination with pUS-high led to complete NB eradication in over 50% of immunized mice and surviving mice showed an over 80% reduction in primary tumor growth upon rechallenge in contrast to controls. In summary, survivin-based DNA vaccination is effective against NB and the rational minigene design provides a promising approach to circumvent potentially hazardous effects of using full length antiapoptotic genes as DNA vaccines.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Neuroblastoma/prevención & control , Vacunas de ADN/inmunología , Animales , Apoptosis , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Diseño de Fármacos , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/inmunología , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Endogámicos A , Neuroblastoma/inmunología , Fragmentos de Péptidos/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Survivin , Vacunación
4.
J Mol Med (Berl) ; 91(4): 459-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23052481

RESUMEN

Neuroblastoma (NB) is the most common extracranial solid tumor in children. Combining passive immunotherapy with an antibody to the disialoganglioside GD2 (ch14.18/SP2/0) and cytokines with 13-cis-retinoic acid for post-myeloablative maintenance therapy increased survival in high-risk NB, but the overall prognosis for these children is still in need of improvement. Fenretinide (4-HPR) is a synthetic retinoid that has shown clinical activity in recurrent NB and is cytotoxic to a variety of cancer cells, in part via the accumulation of dihydroceramides, which are precursors of GD2. We investigated the effect of 4-HPR on CHO-derived, ch14.18-mediated anti-NB effector functions, complement-dependent cytotoxicity (CDC), and antibody-dependent and antibody-independent cellular cytotoxicity (ADCC and AICC, respectively). Here, we demonstrate for the first time that pretreatment of fenretinide-resistant NB cells with 4-HPR significantly enhanced ch14.18/CHO-mediated CDC and ADCC and AICC by both human natural killer cells and peripheral blood mononuclear cells. Treatment with 4-HPR increased GD2 and death receptor (DR) expression in resistant NB cells and induced an enhanced granzyme B and perforin production by effector cells. Blocking of ganglioside synthesis with a glucosylceramide synthase inhibitor abrogated the increased ADCC response but had no effect on the AICC, indicating that GD2 induced by 4-HPR mediates the sensitization of NB cells for ADCC. We also showed that 4-HPR induced increased GD2 and DR expression in a resistant NB xenograft model that was associated with an increased ADCC and AICC response using explanted tumor target cells from 4-HPR-treated mice. In summary, these findings provide an important baseline for the combination of 4-HPR and passive immunotherapy with ch14.18/CHO in future clinical trials for high-risk NB patients.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Fenretinida/farmacología , Células Asesinas Naturales/inmunología , Neuroblastoma/inmunología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Proteínas del Sistema Complemento/inmunología , Femenino , Gangliósidos/metabolismo , Humanos , Ratones , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Receptores de Muerte Celular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Lett ; 331(2): 167-73, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23337288

RESUMEN

Attenuated Salmonella is an approved oral life vaccine that is currently entering pre-clinical cancer vaccination studies as a promising DNA carrier. In a syngeneic mouse model for neuroblastoma, oral gavage of Salmonella typhimurium (SL7207) carrying recent generated survivin DNA vaccines induced a stronger cellular anti-NB immune response than gene gun application or injection of lentivirally transduced bone marrow-derived DCs. The level of Salmonella-associated side effects was not significant as indicated by unaffected survivin-mediated hematopoiesis and wound healing. We believe that our findings provide an important baseline to translate Salmonella-based DNA vaccination into a clinical application for neuroblastoma.


Asunto(s)
Modelos Animales de Enfermedad , Neuroblastoma/terapia , Salmonella/inmunología , Vacunas de ADN/administración & dosificación , Animales , Biolística , Línea Celular Tumoral , Femenino , Ratones , Neuroblastoma/patología , Plásmidos
6.
Mol Cancer Ther ; 8(8): 2392-401, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19671753

RESUMEN

Neuroblastoma (NB) is a challenging malignancy of the sympathetic nervous tissue characterized by a very poor prognosis. One important marker for NB is the expression of tyrosine hydroxylase (TH), the first-step enzyme of catecholamine biosynthesis. We could show stable and high TH gene expression in 67 NB samples independent of the clinical stage. Based on this observation, we addressed the question of whether xenogeneic TH DNA vaccination is effective in inducing an anti-NB immune response. For this purpose, we generated three DNA vaccines based on pCMV-F3Ub and pBUD-CE4.1 plasmids encoding for human (h)THcDNA (A), hTH minigene (B), and hTHcDNA in combination with the proinflammatory cytokine interleukin 12 (C), and tested prophylactic and therapeutic efficacy to suppress primary tumor growth and spontaneous metastasis. Here we report that xenogeneic TH DNA vaccination was effective in eradicating established primary tumors and inhibiting metastasis. Interestingly, this effect could not be enhanced by adding the Th1 cytokine interleukin 12. However, increased IFN-gamma production and NB cytotoxicity of effector cells harvested from vaccinated mice suggested the participation of tumor-specific CTLs in the immune response. The depletion of CD8(+)T cells completely abrogated the hTH vaccine-mediated anti-NB immune response. Furthermore, rechallenging of surviving mice resulted in reduced primary tumor growth, indicating the induction of a memory immune response. In conclusion, xenogeneic immunization with TH-derived DNA vaccines is effective against NB, and may open a new venue for a novel and effective immunotherapeutic strategy against this challenging childhood tumor.


Asunto(s)
Antígenos Heterófilos/administración & dosificación , Neuroblastoma/terapia , Tirosina 3-Monooxigenasa/genética , Vacunas de ADN/administración & dosificación , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Humanos , Inmunización , Interleucina-12/metabolismo , Ratones , Neuroblastoma/inmunología , Neuroblastoma/patología , Tirosina 3-Monooxigenasa/metabolismo , Vacunas de ADN/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA