Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
J Immunother ; 23(3): 344-52, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10838663

RESUMO

Peptide vaccination of homozygous mice against syngeneic tumors using single major histocompatibility complex (MHC) class I-restricted cytotoxic T lymphocyte (CTL) epitopes elicits effective immune responses against metastatic growth. So far, single-peptide vaccination of patients against their autologous tumors seems to elicit less satisfactory results. In this study, the authors tried to determine whether effective anti-metastatic immunity requires the presentation of peptides restricted by the two parental class I major histocompatibility complex alleles in heterozygous hosts. The immune response against the H-2b-derived 3LL Lewis lung carcinoma was evaluated in heterozygous recombinant congenic F1 mice (Kk x K(b)) and (Kd x K(b)). Vaccination of such heterozygous animals with dendritic cells expressing the two parental H-2K alleles, pulsed with total tumor extract, elicited a potent anti-metastatic response. A comparable response was obtained after vaccination with tumor cells genetically modified to express the two class I alleles. In contrast, vaccination of the heterozygous mice with dendritic cells expressing only one of the parental F1 H-2K alleles or with tumors expressing only one H-2K allele failed to elicit effective immunity against tumor metastasis in recombinant congenic F1 mice. It appears, therefore, that to achieve effective anti-metastatic immunotherapy in heterozygous organisms, presentation of cytotoxic T lymphocyte epitopes restricted by the two parental class I alleles is required.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Neoplasias Pulmonares/terapia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Alelos , Animais , Testes Imunológicos de Citotoxicidade , Citometria de Fluxo , Genes MHC Classe I , Heterozigoto , Antígenos de Histocompatibilidade Classe I/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Camundongos , Camundongos Congênicos , Metástase Neoplásica/prevenção & controle , Células Tumorais Cultivadas
2.
Int J Cancer ; 85(3): 391-7, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10652432

RESUMO

The MUC1 protein was found to be up-regulated in a spectrum of malignant tumors. T-cell responses to the MUC1 extracellular tandem repeat array (TRA) were observed in murine models as well as in breast-carcinoma patients. In the present study, we evaluated the anti-tumor potential of HLA-A2.1-motif-selected peptides from non-TRA domains of the molecule. Peptide immunogenicity was examined in the Db-/- x beta2 microglobulin (beta2m) null mice transgenic for a modified HLA-A2.1/Db-beta2 microglobulin single chain (HHD mice). Our results show the existence of 3 novel HLA-A2.1-restricted MUC1-derived cytotoxic T-lymphocyte (CTL) epitopes. These peptides are processed and presented by the HHD-transfected breast-tumor cell line MDA-MB-157. Moreover, CTL induced by these 3 peptides show higher lysis of target cells pulsed with breast-carcinoma-derived peptides than of targets pulsed with normal breast-tissue-derived peptides. These data suggest an important role for non-TRA MUC1-derived peptides as inducers of a MHC-restricted CTL reaction to a breast-carcinoma cell line and patient-derived tumor extracts.


Assuntos
Neoplasias da Mama/imunologia , Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Mucina-1/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Microglobulina beta-2/imunologia , Animais , Apresentação de Antígeno , Neoplasias da Mama/metabolismo , Vacinas Anticâncer/química , Vacinas Anticâncer/genética , Citotoxicidade Imunológica , Epitopos de Linfócito T/química , Feminino , Fluorescência , Regulação Neoplásica da Expressão Gênica , Antígenos H-2/genética , Antígenos H-2/imunologia , Antígeno de Histocompatibilidade H-2D , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucina-1/química , Mucina-1/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Peptídeos/imunologia , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Células Tumorais Cultivadas , Microglobulina beta-2/química , Microglobulina beta-2/genética
3.
Int J Cancer ; 85(2): 236-42, 2000 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10629083

RESUMO

CTLs specific for tumor antigens play a major role in immunity against cancer. Improved binding affinity of putative TAA peptides could enhance the in vivo immunogenicity of these self-altered self- tumor antigens. We examined here the efficacy of tumor vaccines composed of an altered peptide ligand of MUT-1, designated MUT-D, which exhibited significantly higher class-I allele K(b) binding affinity than its native counterpart MUT-1. The peptide was loaded on antigen presenting cells composed of the C57BL/6-syngeneic fibroblast cell line BLK.CL4. These cells were treated with proteasome inhibitor in order to shut off the degradation of proteins and the subsequent loading of endogenous peptides onto MHC class-I molecules, thus allowing for the pulsing of these cells with the modified peptide MUT-D. Proteasome-inhibited and modified peptide-loaded fibroblasts induced a peptide-specific CTL that significantly delayed primary tumor progression and protected the pre-immunized mice against the development of lung metastasis following the surgical removal of the primary tumor. Genetic modification of the fibroblasts to express the immunostimulatory cytokine IL-2 did not improve the APC function of the modified cells, nor did it result in augmentation of the potency of the vaccine. Our results suggest that the proteasome-inhibited fibroblasts pulsed with modified, high binder tumor-associated antigen peptide are good antigen-presenting cells and represent an effective form of tumor vaccine.


Assuntos
Vacinas Anticâncer/imunologia , Fibroblastos/imunologia , Neoplasias Pulmonares/terapia , Oligopeptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Neoplasias/imunologia , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Epitopos de Linfócito T/imunologia , Fibroblastos/enzimologia , Imunoterapia , Interleucina-2/biossíntese , Interleucina-2/genética , Leupeptinas/farmacologia , Neoplasias Pulmonares/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multienzimáticos/metabolismo , Metástase Neoplásica/prevenção & controle , Transplante de Neoplasias , Complexo de Endopeptidases do Proteassoma
4.
J Interferon Cytokine Res ; 19(12): 1391-401, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10638708

RESUMO

Development of cytokine gene-modified autologous tumor vaccines must take into account the strictly paracrine physiology of cytokines whose expression at the tumor microenvironment is important for the successful induction of tumor-specific immunity. In this study, we investigated the efficacy of a tumor vaccine composed of inactivated autologous cells transfected with two plasmid vectors encoding a mutant membrane-bound murine granulocyte-macrophage colony-stimulating factor (MuGM-CSF) and murine interferon-gamma (MuIFN-gamma). Expression of both cytokines as cell surface ligands on the highly metastatic D122 clone of Lewis lung carcinoma led to abrogation of their tumorigenicity and metastatic phenotype. More importantly, vaccination with irradiated tumor cells expressing the membrane-bound GM-CSF and IFN-gamma induced a cytotoxic T lymphocyte (CTL) response that protected syngeneic mice against a subsequent challenge with D122 cells as a primary tumor in preimmunized mice as well as against lung metastasis developing after surgical removal of the primary tumor in naive mice. Autologous cells expressing the membrane-bound GM-CSF and IFN-gamma exhibited comparable efficacy as an antimetastatic vaccine to a vaccine composed of transfectants expressing wild-type secreted cytokine molecules. These results indicate that membrane-bound cytokines can cause enhanced immunogenicity when transfected into tumor cells for the induction of antitumor immunity.


Assuntos
Vacinas Anticâncer/farmacologia , Citocinas/genética , Animais , Sequência de Bases , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Membrana Celular/imunologia , Primers do DNA/genética , Feminino , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interferon gama/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proteínas Recombinantes , Linfócitos T Citotóxicos/imunologia , Transfecção , Transplante Autólogo
5.
Clin Exp Metastasis ; 16(7): 623-32, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9932609

RESUMO

Interleukin 12 (IL-12) is a disulfide-linked heterodimer molecule produced predominantly by professional antigen presenting cells. It promotes the induction of sundry biological effects with significant relevance to antitumor immunity, such as enhancing a T(H)1 helper response, an in vivo antiangiogenic effect, induction of adhesion molecules that assist in lymphocyte homing to sites of tumor growth, and a direct stimulatory effect on both T-cells and NK cells. We tested the efficacy of an antimetastatic vaccine composed of autologous murine D122 cells transfected with both subunits of IL-12 cDNA to express biologically-active IL-12 molecule. Expression of IL-12 by D122 cells significantly reduced their tumorigenicity and metastatic potential in immunocompetent syngeneic hosts. Furthermore, vaccination of mice with 2 x 10(6) irradiated IL-12-transfected D122 cells engendered a protective CTL response which rejected a subsequent challenge with parental D122 cells and eradicated lung micrometastasis in animals whose primary tumors have been surgically removed. The antitumor effects of IL-12 were mediated primarily by its ability to induce gammaIFN expression in vivo. CD8+ T-cells as well as NK cells were crucial in the execution of the antitumor effects of IL-12. These results suggest that autologous tumor cells expressing IL-12 by gene transfer are a potent antitumor vaccine able to induce a systemic immune response against poorly immunogenic and spontaneously metastatic tumors.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Pulmonar de Lewis/prevenção & controle , Terapia Genética/métodos , Interferon gama/genética , Interleucina-12/genética , Metástase Neoplásica/prevenção & controle , Animais , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/secundário , Divisão Celular , Células Cultivadas , Imunoterapia/métodos , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Plasmídeos/genética , Reação em Cadeia da Polimerase , Células Th1/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...