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










Base de dados
Intervalo de ano de publicação
1.
Fertil Steril ; 105(2): 529-38, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26616441

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Leiomioma/tratamento farmacológico , Neovascularização Patológica , Poli I-C/farmacologia , Polietilenoimina/farmacologia , Neoplasias Uterinas/tratamento farmacológico , Adulto , Animais , Modelos Animais de Doenças , Terapia de Reposição de Estrogênios , Feminino , Humanos , Antígeno Ki-67/metabolismo , Leiomioma/irrigação sanguínea , Leiomioma/genética , Leiomioma/metabolismo , Leiomioma/patologia , Camundongos Nus , Pessoa de Meia-Idade , Ovariectomia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Neoplasias Uterinas/irrigação sanguínea , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Fertil Steril ; 104(5): 1310-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26297642

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Endometriose/tratamento farmacológico , Endométrio/efeitos dos fármacos , Poli I-C/farmacologia , Polietilenoimina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Endometriose/genética , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Endométrio/transplante , Fatores de Crescimento Endotelial/farmacologia , Terapia de Reposição de Estrogênios , Feminino , Genes Reporter , Xenoenxertos , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos Nus , Neovascularização Patológica , Ovariectomia , Peptídeos Cíclicos/farmacologia , Polietilenoimina/análogos & derivados , Fatores de Tempo , Proteína Vermelha Fluorescente
3.
Cancer Cell ; 26(1): 61-76, 2014 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-24981740

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Linhagem da Célula , Lisossomos/enzimologia , Melanoma/enzimologia , Neoplasias Cutâneas/enzimologia , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/mortalidade , Melanoma/secundário , Melanoma/terapia , Camundongos , Invasividade Neoplásica , Estadiamento de Neoplasias , Transporte Proteico , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Fatores de Transcrição SOXE/genética , Fatores de Transcrição SOXE/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Fatores de Tempo , Transfecção , Proteínas rab de Ligação ao GTP/genética , proteínas de unión al GTP Rab7
4.
Cancer Cell ; 16(2): 103-14, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19647221

RESUMO

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.


Assuntos
Apoptose/imunologia , Autofagia/imunologia , Imunidade Inata , Melanoma/imunologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Linhagem Celular Tumoral , Células Cultivadas , RNA Helicases DEAD-box/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/genética , Endossomos/metabolismo , Humanos , Helicase IFIH1 Induzida por Interferon , Lisossomos/efeitos dos fármacos , Lisossomos/genética , Lisossomos/metabolismo , Melanoma/patologia , Melanoma/terapia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Fagossomos/efeitos dos fármacos , Fagossomos/genética , Fagossomos/metabolismo , Poli C/farmacologia , Polietilenoimina/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA de Cadeia Dupla
5.
Nat Med ; 14(11): 1256-63, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18978796

RESUMO

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.


Assuntos
RNA Helicases DEAD-box/metabolismo , Inativação Gênica , Melanoma/genética , Melanoma/metabolismo , Fosfatos/metabolismo , RNA Interferente Pequeno/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose/imunologia , Linhagem Celular Tumoral , Transplante de Células , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Melanoma/imunologia , Melanoma/patologia , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptores Imunológicos
6.
Handb Exp Pharmacol ; (183): 201-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18071661

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Receptores Toll-Like/agonistas , Adjuvantes Imunológicos/uso terapêutico , Animais , Antineoplásicos/farmacologia , DNA/uso terapêutico , Humanos , Imunoterapia , Oligonucleotídeos/uso terapêutico , RNA/uso terapêutico , Pele/citologia , Pele/crescimento & desenvolvimento , Neoplasias Cutâneas/imunologia , Fenômenos Fisiológicos da Pele , Receptores Toll-Like/imunologia
7.
Eur J Cell Biol ; 86(11-12): 817-26, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16928407

RESUMO

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.


Assuntos
Antígenos/administração & dosagem , Biolística/métodos , Células Dendríticas/imunologia , Imunização/métodos , Pele/imunologia , Linfócitos T Citotóxicos/imunologia , Transdução Genética , Transferência Adotiva , Animais , Proliferação de Células , Células Dendríticas/citologia , Injeções Intradérmicas , Luciferases/metabolismo , Proteínas Luminescentes/metabolismo , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Citotóxicos/citologia , Vacinas de DNA/imunologia , Imagem Corporal Total
8.
J Gene Med ; 8(11): 1300-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16952196

RESUMO

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.


Assuntos
Adenoviridae/genética , Adenoviridae/imunologia , Células Dendríticas/imunologia , Interferon-alfa/biossíntese , Receptor Toll-Like 9/fisiologia , Transdução Genética , Animais , Diferenciação Celular , Células Dendríticas/citologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodesoxirribonucleotídeos/genética , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética
9.
Am J Pathol ; 169(2): 665-72, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16877364

RESUMO

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.


Assuntos
Quinase 4 Dependente de Ciclina/genética , Fator de Crescimento de Hepatócito/genética , Melanoma Experimental/induzido quimicamente , Mutação/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Neoplasias Cutâneas/patologia , Animais , Animais Recém-Nascidos , Carcinógenos , Modelos Animais de Doenças , Expressão Gênica , Humanos , Pulmão/patologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mutantes/genética
10.
J Gene Med ; 8(10): 1243-50, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16892456

RESUMO

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.


Assuntos
Adenoviridae/genética , Células Dendríticas/transplante , Microscopia de Fluorescência/métodos , Vacinas de DNA/administração & dosagem , Animais , Antígenos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Clonagem Molecular , Células Dendríticas/diagnóstico por imagem , Células Dendríticas/imunologia , Diagnóstico por Imagem , Vias de Administração de Medicamentos , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Luciferases/genética , Luciferases/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Radiografia , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/metabolismo
11.
Cancer Res ; 66(10): 5427-35, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16707471

RESUMO

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.


Assuntos
Vacinas Anticâncer/farmacologia , Oxirredutases Intramoleculares/imunologia , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Receptores Toll-Like/imunologia , Vacinas de DNA/farmacologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Vacinas Anticâncer/imunologia , Ilhas de CpG/genética , Ilhas de CpG/imunologia , Quinase 4 Dependente de Ciclina/genética , Epitopos , Feminino , Fator de Crescimento de Hepatócito/genética , Humanos , Oxirredutases Intramoleculares/genética , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poli I-C/farmacologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Vacinas de DNA/imunologia
12.
Int J Cancer ; 118(2): 373-80, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16049960

RESUMO

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.


Assuntos
Vacinas Anticâncer/imunologia , Tolerância Imunológica , Melanoma/imunologia , Melanoma/prevenção & controle , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/prevenção & controle , Animais , Carcinógenos , Ciclo Celular/genética , Quinase 4 Dependente de Ciclina , Modelos Animais de Doenças , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Oxirredutases Intramoleculares/imunologia , Melanoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais , Neoplasias Cutâneas/genética , Raios Ultravioleta/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...