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











Base de datos
Intervalo de año de publicación
1.
Fertil Steril ; 105(2): 529-38, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26616441

RESUMEN

OBJECTIVE: To assess the antiproliferative, proapoptotic, and antiangiogenic effects of the double-stranded RNA mimic polyinosine-polycytidylic acid (pIC) complexed with polyethylenimine [pIC(PEI)] in xenografted human leiomyomas. DESIGN: Heterologous leiomyoma mouse model. SETTING: University-affiliated infertility center. ANIMAL(S): Ovariectomized and hormone-replaced nude mice (n = 16) who received human leiomyoma fragment transplantation. INTERVENTION(S): Leiomyoma fragments placed in the peritoneum of 5-week-old nude female mice and treated with the vehicle (n = 8) or 0.6 mg/kg [pIC(PEI)] (n = 8) for 4 weeks. MAIN OUTCOME MEASURE(S): The size of the leiomyoma implants, and cellular proliferation (Ki67), vascularization (PECAM), and apoptosis (OH-ends) assessed by quantitative immunohistochemical/immunofluorescent analysis of the recovered implants. RESULT(S): No significant differences were observed in the size of the leiomyoma implants between groups. Vascularization and proliferation were significantly decreased, and apoptosis was increased in the [pIC(PEI)]-treated group versus control. CONCLUSION(S): We hypothesize that the antiangiogenic and apoptotic effects exerted by [pIC(PEI)] might lead to a decrease in lesion size in this animal model if the compound is administered for longer periods of time. This study provides promising data on [pIC(PEI)] as a potential novel therapeutic agent against human leiomyoma.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Leiomioma/tratamiento farmacológico , Neovascularización Patológica , Poli I-C/farmacología , Polietileneimina/farmacología , Neoplasias Uterinas/tratamiento farmacológico , Adulto , Animales , Modelos Animales de Enfermedad , Terapia de Reemplazo de Estrógeno , Femenino , Humanos , Antígeno Ki-67/metabolismo , Leiomioma/irrigación sanguínea , Leiomioma/genética , Leiomioma/metabolismo , Leiomioma/patología , Ratones Desnudos , Persona de Mediana Edad , Ovariectomía , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Neoplasias Uterinas/irrigación sanguínea , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Fertil Steril ; 104(5): 1310-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26297642

RESUMEN

OBJECTIVE: To assess the therapeutic potential of polyinosine-polycytidylic acid, a double-stranded RNA molecule with selective proapoptotic and antiangiogenic activity, complexed with polyethyleneimine (pIC(PEI)) in treating endometriosis. DESIGN: A heterologous mouse model of endometriosis was created by injecting human endometrial fragments into the peritoneum. Endometrial fragments were engineered to express the fluorescent protein mCherry as a reporter to monitor status over the course of the 4-week study. SETTING: University-affiliated infertility center. ANIMAL(S): Ovariectomized and hormone-replaced nude mice (n = 30) injected with fluorescent-labeled human endometrial fragments at 4-6 weeks of age. INTERVENTION(S): Animals (n = 10 per group) were injected with vehicle (control), the anti-VEGF compound CBO-P11 (0.6 mg/kg), or pIC(PEI) (0.6 mg/kg) twice weekly over the course of 4 weeks. MAIN OUTCOME MEASURE(S): Variations in the size of endometriotic implants were estimated by quantifying the expression of mCherry throughout the course of the experiment. Neovascularization, cellular proliferation, and apoptosis were estimated by quantitative immunofluorescence detection of PECAM, α-SMA, Ki67, and TUNEL. RESULT(S): pIC(PEI) promoted a significant increase in apoptosis and a decrease in neovascularization in human fragments, but did not reduce the size of endometriotic implants. CONCLUSION(S): While pIC(PEI) treatment had significant antiangiogenic and pro-apoptotic effects in this setting, longer periods of exposure than the ones supported by our heterologous model and/or assays in homologous mouse models of endometriosis may be necessary to detect an effect of this compound on lesion size.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endometriosis/tratamiento farmacológico , Endometrio/efectos de los fármacos , Poli I-C/farmacología , Polietileneimina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endometriosis/genética , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/metabolismo , Endometrio/patología , Endometrio/trasplante , Factores de Crecimiento Endotelial/farmacología , Terapia de Reemplazo de Estrógeno , Femenino , Genes Reporteros , Xenoinjertos , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Ratones Desnudos , Neovascularización Patológica , Ovariectomía , Péptidos Cíclicos/farmacología , Polietileneimina/análogos & derivados , Factores de Tiempo , Proteína Fluorescente Roja
3.
Cancer Cell ; 26(1): 61-76, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24981740

RESUMEN

Although common cancer hallmarks are well established, lineage-restricted oncogenes remain less understood. Here, we report an inherent dependency of melanoma cells on the small GTPase RAB7, identified within a lysosomal gene cluster that distinguishes this malignancy from over 35 tumor types. Analyses in human cells, clinical specimens, and mouse models demonstrated that RAB7 is an early-induced melanoma driver whose levels can be tuned to favor tumor invasion, ultimately defining metastatic risk. Importantly, RAB7 levels and function were independent of MITF, the best-characterized melanocyte lineage-specific transcription factor. Instead, we describe the neuroectodermal master modulator SOX10 and the oncogene MYC as RAB7 regulators. These results reveal a unique wiring of the lysosomal pathway that melanomas exploit to foster tumor progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linaje de la Célula , Lisosomas/enzimología , Melanoma/enzimología , Neoplasias Cutáneas/enzimología , Proteínas de Unión al GTP rab/metabolismo , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/mortalidad , Melanoma/secundario , Melanoma/terapia , Ratones , Invasividad Neoplásica , Estadificación de Neoplasias , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Factores de Tiempo , Transfección , Proteínas de Unión al GTP rab/genética , Proteínas de Unión a GTP rab7
4.
Cancer Cell ; 16(2): 103-14, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19647221

RESUMEN

Inappropriate drug delivery, secondary toxicities, and persistent chemo- and immunoresistance have traditionally compromised treatment response in melanoma. Using cellular systems and genetically engineered mouse models, we show that melanoma cells retain an innate ability to recognize cytosolic double-stranded RNA (dsRNA) and mount persistent stress response programs able to block tumor growth, even in highly immunosuppressed backgrounds. The dsRNA mimic polyinosine-polycytidylic acid, coadministered with polyethyleneimine as carrier, was identified as an unanticipated inducer of autophagy downstream of an exacerbated endosomal maturation program. A concurrent activity of the dsRNA helicase MDA-5 driving the proapoptotic protein NOXA resulted in an efficient autodigestion of melanoma cells. These results reveal tractable links for therapeutic intervention among dsRNA helicases, endo/lysosomes, and apoptotic factors.


Asunto(s)
Apoptosis/inmunología , Autofagia/inmunología , Inmunidad Innata , Melanoma/inmunología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Proteína 5 Relacionada con la Autofagia , Línea Celular Tumoral , Células Cultivadas , ARN Helicasas DEAD-box/metabolismo , Endosomas/efectos de los fármacos , Endosomas/genética , Endosomas/metabolismo , Humanos , Helicasa Inducida por Interferón IFIH1 , Lisosomas/efectos de los fármacos , Lisosomas/genética , Lisosomas/metabolismo , Melanoma/patología , Melanoma/terapia , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Fagosomas/efectos de los fármacos , Fagosomas/genética , Fagosomas/metabolismo , Poli C/farmacología , Polietileneimina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Bicatenario
5.
Nat Med ; 14(11): 1256-63, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18978796

RESUMEN

Genetic and epigenetic plasticity allows tumors to evade single-targeted treatments. Here we direct Bcl2-specific short interfering RNA (siRNA) with 5'-triphosphate ends (3p-siRNA) against melanoma. Recognition of 5'-triphosphate by the cytosolic antiviral helicase retinoic acid-induced protein I (Rig-I, encoded by Ddx58) activated innate immune cells such as dendritic cells and directly induced expression of interferons (IFNs) and apoptosis in tumor cells. These Rig-I-mediated activities synergized with siRNA-mediated Bcl2 silencing to provoke massive apoptosis of tumor cells in lung metastases in vivo. The therapeutic activity required natural killer cells and IFN, as well as silencing of Bcl2, as evidenced by rescue with a mutated Bcl2 target, by site-specific cleavage of Bcl2 messenger RNA in lung metastases and downregulation of Bcl-2 protein in tumor cells in vivo. Together, 3p-siRNA represents a single molecule-based approach in which Rig-I activation on both the immune- and tumor cell level corrects immune ignorance and in which gene silencing corrects key molecular events that govern tumor cell survival.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Silenciador del Gen , Melanoma/genética , Melanoma/metabolismo , Fosfatos/metabolismo , ARN Interferente Pequeño/genética , Animales , Antineoplásicos/uso terapéutico , Apoptosis/inmunología , Línea Celular Tumoral , Trasplante de Células , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata/inmunología , Melanoma/inmunología , Melanoma/patología , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Receptores Inmunológicos
6.
Handb Exp Pharmacol ; (183): 201-20, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18071661

RESUMEN

This review will briefly cover some important aspects of skin structure and function before touching upon fundamental principles of neoplastic cell growth in the skin and some of the important molecular pathways involved. After presenting evidence for a role of the immune system in shaping the development of skin cancer, concepts for tumor immunotherapy with TLR-agonists are introduced from a historical point of view. Subsequently, the use of synthetic DNA, synthetic RNA and synthetic small immunostimulatory molecules for immunotherapy of early forms of epithelial carcinoma (actinic keratoses) and melanoma (lentigo maligna), as well as for advanced metastatic melanoma, is comprehensively presented. Finally, current developments and future prospects for immunotherapy of occult or unresectable melanoma metastastases, the most important clinical problem today, are discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Receptores Toll-Like/agonistas , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antineoplásicos/farmacología , ADN/uso terapéutico , Humanos , Inmunoterapia , Oligonucleótidos/uso terapéutico , ARN/uso terapéutico , Piel/citología , Piel/crecimiento & desarrollo , Neoplasias Cutáneas/inmunología , Fenómenos Fisiológicos de la Piel , Receptores Toll-Like/inmunología
7.
Eur J Cell Biol ; 86(11-12): 817-26, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16928407

RESUMEN

The skin is an attractive target for antigen-specific vaccination. Particle bombardment of the epidermis with plasmid DNA using the gene gun results in antigen expression in keratinocytes of the epidermis leading to antigen presentation in the draining lymph nodes by migratory dendritic cells (DC). In order to better understand the role of the skin in stimulating antigen-specific CD8+cytotoxic T cells (CTL), we compared gene gun immunization with intracutaneous injections of antigen-transduced DC. A single intracutaneous injection of antigen-transduced DC was able to induce in vivo expansion of CD8+CTL specific for the model antigen chicken ovalbumin while four simultaneous shots with the gene gun were not effective. Antigen-transduced DC were much more efficient than particle bombardment of the epidermis in stimulating adoptively transferred TCR-transgenic CD8+CTL in the draining lymph nodes. Employing the novel technique of in vivo bioluminescence imaging, we demonstrated efficient gene transfer to the skin following gene gun bombardment and confirmed that a similar amount of antigen reached the lymph node when compared with injection of antigen-transduced DC. Our results suggest that direct transfection of the skin does not optimally reach and activate appropriate antigen-presenting DC. We believe that this reflects the immunological function of the epidermis which must balance immunity and tolerance to foreign antigens. Further investigations will have to address the role of Langerhans cells for the activation of cellular immunity in the skin.


Asunto(s)
Antígenos/administración & dosificación , Biolística/métodos , Células Dendríticas/inmunología , Inmunización/métodos , Piel/inmunología , Linfocitos T Citotóxicos/inmunología , Transducción Genética , Traslado Adoptivo , Animales , Proliferación Celular , Células Dendríticas/citología , Inyecciones Intradérmicas , Luciferasas/metabolismo , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/citología , Vacunas de ADN/inmunología , Imagen de Cuerpo Entero
8.
J Gene Med ; 8(11): 1300-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16952196

RESUMEN

BACKGROUND: Recombinant replication-deficient adenoviral vectors (recAd) are attractive candidates for DNA vaccination approaches because they are able to activate the innate and adaptive immune systems. Here we explore the ability of recAd to transduce and activate subsets of dendritic cells, namely plasmacytoid dendritic cells (pDC) and conventional dendritic cells (cDC). METHODS: DC were derived from bone marrow precursors in vitro with the help of FLT3-ligand. Sorted populations of pDC and cDC were infected with recAd at various multiplicities of infection. Transduction efficiency, phenotypic maturation and production of IFN-alpha as well as IL-6 were assessed. Additionally, activation of DC and induction of cytotoxic T lymphocytes (CTL) were determined in vivo. The role of Toll-like receptor (TLR) 9 in recAd recognition was investigated as it has previously been shown that DNA viruses are recognized via this receptor. RESULTS: RecAd can efficiently transduce pDC as well as cDC in vitro. Both DC subsets mature and produce IFN-alpha upon interaction with recAd. In the absence of TLR9, activation and cytokine production was only detected in cDC but not in pDC. Importantly, induction of CD8+ CTL following in vivo injection of recAd was similar in TRL9-deficient mice when compared with wildtype controls. CONCLUSIONS: RecAd can efficiently transduce and activate both pDC and cDC. pDC required TLR9 to detect the presence of recAd whereas cDC also recognized recAd independently of TLR9. These unique immunostimulatory properties support the future development of recombinant Ad as a vector for DNA vaccine approaches.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Células Dendríticas/inmunología , Interferón-alfa/biosíntesis , Receptor Toll-Like 9/fisiología , Transducción Genética , Animales , Diferenciación Celular , Células Dendríticas/citología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos/genética , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética
9.
J Gene Med ; 8(10): 1243-50, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16892456

RESUMEN

Recombinant DNA vaccines are able to induce strong CD8+ T cell mediated immunity and have become increasingly attractive for the prevention and treatment of infectious diseases and cancer. Dendritic cells (DC), which critically control cellular immune responses, have been transduced with antigen ex vivo and used as 'nature's adjuvant' to enhance vaccine efficacy. The impact of the application route on the in vivo distribution of antigen and the stimulation of CD8+ T cells have been subjects of considerable debate. Here we report the construction of vectors expressing a fusion protein between EGFP, the H2-K(b)-binding peptide OVA(aa257-264) and green click beetle luciferase as a model antigen which allows for simultaneous quantitative assessment of antigen expression using fluorescence and bioluminescence imaging in correlation with CD8+ T cell stimulation in vivo. We applied this construct to evaluate DNA vaccination with recombinant adenoviral vectors, assess the impact of using cultured DC for vaccine delivery and investigate different application routes. Antigen expression was non-invasively followed in vivo by visualizing bioluminescence with an ultrasensitive CCD camera. CD8+ T cell stimulation was detected with H2-K(b)-OVA(aa257-264) tetramers. We found that intravenous injection of adenovirus-transduced DC stimulated the strongest OVA(aa257-264)-specific cytotoxic T-lymphocyte (CTL) responses although it delivered two orders of magnitude less antigen in vivo when compared to direct injection of recombinant adenovirus. We believe that our experimental approach has the potential to facilitate translational development of improved genetic immunization strategies targeting DC directly in vivo.


Asunto(s)
Adenoviridae/genética , Células Dendríticas/trasplante , Microscopía Fluorescente/métodos , Vacunas de ADN/administración & dosificación , Animales , Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Clonación Molecular , Células Dendríticas/diagnóstico por imagen , Células Dendríticas/inmunología , Diagnóstico por Imagen , Vías de Administración de Medicamentos , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Luciferasas/genética , Luciferasas/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Radiografía , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/metabolismo
10.
Am J Pathol ; 169(2): 665-72, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16877364

RESUMEN

Currently, novel mouse models of melanoma are being generated that recapitulate the histopathology and molecular pathogenesis observed in human disease. Impaired cell-cycle control, which is a hallmark of both familial and sporadic melanoma, promotes slowly growing carcinogen-induced melanomas in the skin of mice carrying a mutated cyclin-dependent kinase 4 (CDK4(R24C)). Deregulated receptor tyrosine kinase signaling, which is another important feature of human melanoma, leads to spontaneous development of metastatic melanoma after a long latency period in mice overexpressing hepatocyte growth factor/scatter factor (HGF/SF mice). Here we report that treatment with 7,12-dimethylbenz[a]anthracene and 12-O-tetradecanoylphorbol-13-acetate induced metastatic melanomas in all HGF/SF mice on the C57BL/6 background, which histologically resemble human melanoma. Importantly, mutant CDK4 dramatically increased the number and the growth kinetics of carcinogen-induced primary melanomas in the skin and promoted the growth of spontaneous metastases in lymph nodes and lungs in all HGF/SF mice within the first 3 months of life. Apart from very few skin papillomas, we did not observe tumors of other histology in carcinogen-treated HGF/SF x CDK4(R24C) mice. This new experimental mouse model can now be exploited to study further the biology of melanoma and evaluate new treatment modalities.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Factor de Crecimiento de Hepatocito/genética , Melanoma Experimental/inducido químicamente , Mutación/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Neoplasias Cutáneas/patología , Animales , Animales Recién Nacidos , Carcinógenos , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Pulmón/patología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mutantes/genética
11.
Cancer Res ; 66(10): 5427-35, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707471

RESUMEN

Malignant melanoma is an attractive model disease for the development of antigen-specific immunotherapy because many antigens recognized by tumor-specific T cells have been identified. In C57BL/6 mice, genetic immunization with recombinant adenovirus encoding xenogeneic human tyrosinase-related protein 2 (Ad-hTRP2) induces protective but not therapeutic cellular immunity against growth of transplanted B16 melanoma cells. Here, we additionally applied CpG DNA and synthetic double-stranded RNA, which activate the innate immune system via Toll-like receptors (TLR). Both adenoviral vaccination and peritumoral injections of TLR ligands were required for rejection of established B16 melanoma in the skin. To more closely mimic the clinical situation in patients with melanoma, we evaluated this combined immunotherapeutic strategy in genetically modified mice, which overexpress hepatocyte growth factor (HGF) and carry an oncogenic mutation in the cyclin-dependent kinase 4 (CDK4)(R24C). HGF x CDK4(R24C) mice rapidly develop multiple invasive melanomas in the skin following neonatal carcinogen treatment, which spontaneously metastasize to lymph nodes and lungs. Vaccination with Ad-hTRP2 followed by injections of TLR ligands resulted in delayed growth of autochthonous primary melanomas in the skin and reduction in the number of spontaneous lung metastases but did not induce tumor regression. Carcinogen-treated HGF x CDK4(R24C) mice bearing multiple autochthonous melanomas did not reject transplanted B16 melanoma despite treatment with Ad-hTRP2 and TLR ligands, suggesting the development of tumor immunotolerance. Further investigations in our novel genetic melanoma model may help to better understand the role of the immune system in the pathogenesis and treatment of this life-threatening disease.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Oxidorreductasas Intramoleculares/inmunología , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Receptores Toll-Like/inmunología , Vacunas de ADN/farmacología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Islas de CpG/genética , Islas de CpG/inmunología , Quinasa 4 Dependiente de la Ciclina/genética , Epítopos , Femenino , Factor de Crecimiento de Hepatocito/genética , Humanos , Oxidorreductasas Intramoleculares/genética , Neoplasias Pulmonares/secundario , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Vacunas de ADN/inmunología
12.
Int J Cancer ; 118(2): 373-80, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16049960

RESUMEN

We evaluated the efficacy of a candidate melanoma vaccine approach in mice genetically prone to develop melanoma due to the introduction of an oncogenic mutation (R24C) in the germline sequence of the cyclin-dependent kinase 4 (cdk4), a protein critically involved in cell cycle regulation. Melanomas were induced in cdk4-mutant mice by chemical carcinogenesis and UVB irradiation. A genetic prime-boost strategy targeting the clinically relevant differentiation antigen tyrosinase-related protein 2 (TRP2) was performed which was able to stimulate a melanocyte-specific cellular immune response associated with localized autoimmune vitiligo-like depigmentation. However, significant destruction of carcinogen-induced autochthonous melanocytic neoplasms in the skin was not observed following immunization. We provide evidence that autochthonous melanomas expressed TRP2 but not the MHC molecule H2-Kb and are immunologically tolerated in the skin. Our results highlight the importance of assessing melanoma vaccines in genetic mouse models that more adequately represent the expected clinical situation in order to identify strategies, which eventually may be of benefit for melanoma patients.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Tolerancia Inmunológica , Melanoma/inmunología , Melanoma/prevención & control , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/prevención & control , Animales , Carcinógenos , Ciclo Celular/genética , Quinasa 4 Dependiente de la Ciclina , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Oxidorreductasas Intramoleculares/inmunología , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales , Neoplasias Cutáneas/genética , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA