Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Mol Med ; 10(4): 419-29, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20455858

RESUMO

Chondroitin sulfate proteoglycan 4 (CSPG4), also known as High Molecular Weight-Melanoma Associated Antigen, is a cell surface proteoglycan which has been recently shown to be expressed not only by melanoma cells, but also by various types of human carcinoma and sarcoma. Furthermore, at least in squamous cell carcinoma of head and neck and in basal breast carcinoma, CSPG4 is expressed by cancer stem cells. CSPG4 plays an important role in tumor cell growth and survival. These CSPG4-associated functional properties of tumor cells are inhibited by CSPG4-specific monoclonal antibodies (mAb) in vitro. Moreover, CSPG4-specific mAb can also inhibit tumor growth and metastasis in vivo. The anti-tumor effects of CSPG4-specific mAb are likely to reflect the blocking of important migratory, mitogenic and survival signaling pathways in tumor cells. These results indicate that CSPG4 is a promising new target to implement mAb-based immunotherapy of various types of cancer.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/antagonistas & inibidores , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Imunoterapia , Técnicas In Vitro , Masculino , Melanoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Retinal Desidrogenase , Linfócitos T/imunologia
2.
J Neurooncol ; 55(2): 91-100, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11817706

RESUMO

Monoclonal antibodies raised to peptide sequences of vascular endothelial growth factor (VEGF) and the VEGF receptor, FLT-1, inhibited the growth of C6 tumors growing subcutaneously in nude mice. Immunohistochemical analysis demonstrated antibody targeting of blood vessels, tumor cells, and macrophages. A control antibody demonstrated no growth inhibition or tumor uptake. An antibody to FLT- I impaired microvascular maturation and diminished the accumulation of tumor infiltrating macrophages. The antibodies demonstrated affinity for microvasculature and tumor cells in immunohistochemistry of human glioblastoma multiforme. Targeting VEGF and its receptors has potential in the treatment of tumors of the central nervous system. FLT-1 presents an attractive target due to its presence on multiple cell types.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Fatores de Crescimento Endotelial/imunologia , Glioma/irrigação sanguínea , Linfocinas/imunologia , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Receptores Proteína Tirosina Quinases/imunologia , Receptores de Fatores de Crescimento/imunologia , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Feminino , Glioblastoma/patologia , Glioma/tratamento farmacológico , Humanos , Técnicas Imunoenzimáticas , Macrófagos/patologia , Camundongos , Camundongos Nus , Ratos , Receptores de Fatores de Crescimento do Endotélio Vascular , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
3.
J Immunol ; 165(10): 5938-44, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11067956

RESUMO

Alterations in the p53 gene occur frequently and can lead to accumulation of p53 protein in squamous cell carcinomas of the head and neck (SCCHN). Since accumulation of p53 is associated with enhanced presentation of wild-type sequence (wt) p53 peptides to immune cells, the development of pan vaccines against SCCHN has focused on wt p53 epitopes. We used the HLA-A2.1-restricted wt p53(264-272) epitope to generate CTL from circulating precursor T cells of HLA-A2.1(+) healthy donors and patients with SCCHN. Autologous peptide-pulsed dendritic cells were used for in vitro sensitization. CTL specific for the wt p53(264-272) peptide were generated from PBMC obtained from two of seven normal donors and three of seven patients with SCCHN. These CTL were HLA class I restricted and responded to T2 cells pulsed with p53(264-272) peptide as well as HLA-A2-matched SCCHN cell lines naturally presenting the epitope. Paradoxically, none of the tumors in the three patients who generated CTL could adequately present the epitope; two had a wt p53 genotype and no p53 protein accumulation, while the third tumor expressed a point mutation (R to H) in codon 273 that prevents presentation of the p53(264-272) epitope. In contrast, patients who did not generate CTL had tumors that accumulated altered p53 and potentially could present the p53(264-272) epitope. These findings suggest that in vivo, CTL specific for the wt p53(264-272) peptide might play a role in the elimination of tumor cells expressing this epitope and in immunoselection of epitope-loss tumor cells. Immunoselection of tumors that become resistant to anti-p53 immune responses has important implications for future p53-based vaccination strategies.


Assuntos
Carcinoma de Células Escamosas/imunologia , Epitopos de Linfócito T/metabolismo , Neoplasias de Cabeça e Pescoço/imunologia , Epitopos Imunodominantes/metabolismo , Ativação Linfocitária/genética , Fragmentos de Peptídeos/metabolismo , Subpopulações de Linfócitos T/imunologia , Proteína Supressora de Tumor p53/metabolismo , Autoanticorpos/sangue , Carcinoma de Células Escamosas/genética , Citotoxicidade Imunológica/genética , Análise Mutacional de DNA , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Citometria de Fluxo , Variação Genética/imunologia , Antígeno HLA-A2/imunologia , Antígeno HLA-A2/metabolismo , Neoplasias de Cabeça e Pescoço/genética , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Coloração e Rotulagem , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/química , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia
4.
Cytometry ; 41(4): 321-8, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11084618

RESUMO

BACKGROUND: Major histocompatibility complex (MHC)-peptide tetrameric complexes (tetramers) are valuable tools for detecting and characterizing peptide-specific T cells. Because the frequency of these cells is generally very low, it may be difficult to discriminate between nonspecific and specific tetramer binding. METHODS: A four-color flow cytometric assay that simultaneously measures tetramer, CD3, CD8, and CD14 was used to investigate the sensitivity and specificity of MHC class I tetramer staining. This was accomplished by using the influenza virus matrix protein peptide, GILGFVFTL (FLU), as a model recall antigen and the human immunodeficiency virus (HIV) reverse transcriptase peptide, ILKEPVHGV (HIV), as a model novel antigen. Peripheral blood mononuclear cells (PBMC) from 31 HLA-A2.1(+) and 10 HLA-A2.1(-) healthy individuals were stained with the tetramers. RESULTS: The lower limit of detection was established at approximately 1/8,000. In HLA-A2(+) PMBC, frequencies of tetramer-positive CD8(+) T cells were log normally distributed and were high for FLU (1/910) but low for HIV (1/6,067). A novel competition assay, in which tetramer binding was shown to diminish subsequent staining with anti-CD3 antibody, was used to confirm the specificity of tetramer binding to the T-cell receptor (TCR) complex. The competition assay was validated by evaluating several anti-CD3 antibodies and showing that in PBMC from HLA-A2(-) subjects, spurious tetramer-positive events (1/20,000) failed to compete with CD3 binding. For the "recall" FLU tetramer, the degree of competition was proportional to the frequency, suggesting a selection of high avidity cells. Although CD3 competition was also highly correlated with the intensity of tetramer staining, competition allowed the identification of false positive cases with relatively high tetramer staining intensity. CONCLUSION: The data indicate that competition of CD3 binding allows confirmation of the specificity of tetramer binding to the TCR, extending the usefulness of tetramers in the frequency analysis of peptide-specific T lymphocytes.


Assuntos
Complexo CD3/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais/imunologia , Antígenos Virais/química , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Ligação Competitiva , Complexo CD3/análise , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citometria de Fluxo/métodos , HIV/imunologia , Transcriptase Reversa do HIV/química , Transcriptase Reversa do HIV/imunologia , Transcriptase Reversa do HIV/metabolismo , Antígeno HLA-A2/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Antígenos Comuns de Leucócito/análise , Substâncias Macromoleculares , Orthomyxoviridae/imunologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Sensibilidade e Especificidade , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/metabolismo
5.
Clin Cancer Res ; 6(3): 979-86, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10741724

RESUMO

Evidence has accumulated indicating that HLA-A2-restricted CTLs specific for human wild-type sequence p53 epitopes lyse tumor cells expressing mutant p53. To explore the possibility that wild-type sequence p53 peptides could also be used in vaccines for patients expressing HLA-A24 antigen, another frequent HLA class I allele, we investigated the induction of HLA-A24-restricted p53-specific CTLs from the peripheral blood lymphocytes of normal donors. Of six p53-derived peptides possessing an HLA-A24 binding motif, the p53 peptide 125-134 (p53(125-134)) was found to have a high binding capacity and induced peptide-specific CTLs from peripheral blood mononuclear cells, using peptide-pulsed autologous dendritic cells and subsequent cultivation with cytokines interleukin 2 and interleukin 7. Bulk CTL populations lysed peptide-pulsed HLA-A24+ targets as well as HLA-A24+ squamous cell carcinoma of the head and neck (SCCHN) cell lines. However, IFN-gamma pretreatment of HLA-A24+ SCCHN cell lines was necessary for lysis, suggesting that a ligand density higher than that normally expressed by tumor cells is required for these CTLs to mediate lysis. Moreover, a cloned CTL, designated TH#99, isolated from the bulk population by limiting dilution, lysed HLA-A24+ SCCHN targets more efficiently than the bulk CTL population. Lysis was inhibited by anti-HLA class I monoclonal antibody but not by anti-HLA-DR monoclonal antibody. These results indicate that HLA-A24-restricted CTLs recognizing the wild-type sequence p53(125-134) can be generated using autologous dendritic cells from precursors present in peripheral blood lymphocytes obtained from normal HLA-A24+ donors. This finding suggests that vaccine strategies targeting wild-type sequence p53 epitopes can be extended to a wider range of cancer patients.


Assuntos
Carcinoma de Células Escamosas/imunologia , Antígenos HLA-A/metabolismo , Neoplasias de Cabeça e Pescoço/imunologia , Linfócitos T Citotóxicos/imunologia , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais/farmacologia , Ligação Competitiva , Carcinoma de Células Escamosas/patologia , Citotoxicidade Imunológica/efeitos dos fármacos , Antígenos HLA-A/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Células K562 , Mutação , Oligopeptídeos/imunologia , Oligopeptídeos/metabolismo , Ligação Proteica , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia
6.
Gene Ther ; 7(23): 2036-40, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11175316

RESUMO

Enhanced green fluorescent protein (EGFP) is a novel marker gene product, which is readily detectable using techniques of fluorescence microscopy, flow cytometry, or macroscopic imaging. In the present studies, we have examined the immunogenicity of EGFP in murine models. A stable transfectant of the transplantable CMS4 sarcoma of BALB/c origin expressing EGFP, CMS4-EGFP-Zeo, was generated. Splenocytes harvested from mice immunized with a recombinant adenovirus expressing EGFP (Ad-EGFP) were restimulated in vitro with CMS4-EGFP-Zeo. Effector lymphocytes displayed strong cytotoxicity against CMS4-EGFP-Zeo, but not against mock-transfected CMS4-Zeo tumor cells. A number of candidate H2-Kd-binding peptides derived from the EGFP protein were chosen according to an epitope prediction program and synthesized. These peptides were tested for their ability to bind to H2-Kd molecules and stimulate IFNgamma-production by splenocytes harvested from Ad-EGFP-immunized mice. Using this methodology, the peptide, HYLSTQSAL (corresponding to EGFP200-208) which strongly binds to H2-Kd molecules, was identified as a naturally occurring epitope of EGFP. These results should facilitate the use of EGFP as a model tumor antigen in BALB/c mice.


Assuntos
Antígenos de Neoplasias/análise , Vacinas Anticâncer , Antígenos H-2/imunologia , Proteínas Luminescentes/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Proteínas de Fluorescência Verde , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais
7.
Gene Ther ; 6(4): 629-36, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10476222

RESUMO

DNA-based immunization is currently being investigated as a new method for the induction of cellular and humoral immunity directed against viral disease and cancer. In the present study we characterized and compared the immune responses induced in mice following particle-bombardment of the skin ('gene gun' immunization) with those elicited by intracutaneous injection of a recombinant adenoviral vector. Using the well characterized beta-galactosidase (beta gal) model Ag system we find that both in vivo gene transfer systems elicit potent and long-lasting anti-beta gal-specific CD8+ and CD4+ T cell responses. However, gene gun immunization predominantly promotes the production of anti-beta gal antibodies of the gamma 1 isotype, indicative of a Th2-biased immune response, while intradermal injection of recombinant adenovirus primarily leads to the production of anti-beta gal gamma 2a antibodies, indicative of a Th1-biased immune response. Since viral infections are generally associated with the production of large amounts of IFN-alpha and IL-12, we investigated whether administration of expression plasmids encoding these Th1-associated cytokines along with antigen-encoding cDNA can influence the nature of the immune response resulting from gene gun immunization. We observed that co-delivery of IFN-alpha or IL-12 resulted in increased production of anti-beta gal gamma 2a antibodies. This suggests a shift towards a Th1 phenotype of the resulting immune response, thus mimicking a viral infection. Importantly, gene gun immunization of mice with a naturally occurring tumor antigen, the tumor-specific p53 mutant antigen expressed by the chemically induced BALB/c Meth A sarcoma, required co-delivery of IL-12 for the induction of effective antitumor immunity. These results have important implications for the design of clinically relevant gene gun immunization strategies for tumor immunotherapy.


Assuntos
Biolística , Citocinas/genética , Neoplasias/terapia , Linfócitos T/imunologia , Vacinas de DNA/administração & dosagem , Animais , Anticorpos Monoclonais/sangue , Antígenos de Neoplasias/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Genes p53 , Imunidade Celular , Interferon gama/genética , Interleucina-12/genética , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Pele/imunologia , Células Th1/imunologia , beta-Galactosidase/genética , beta-Galactosidase/imunologia
8.
Clin Cancer Res ; 5(6): 1281-8, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10389910

RESUMO

CTLs recognizing the HLA-A2.1-restricted, wild-type sequence p53 epitopes p53(149-157) and p53(264-272) were generated from CD8-enriched populations of nonadherent peripheral blood lymphocytes (PBLs) obtained from healthy donors. The PBLs were restimulated in vitro with peptide-pulsed granulocyte macrophage colony-stimulating factor- and interleukin (IL)-4-induced autologous dendritic cells in the presence of IL-6 and IL-12 and subsequently cultivated with IL-1alpha, IL-2, IL-4, IL-6, and IL-7. Bulk anti-p53(264-272) CTL populations were generated from PBLs obtained from two of five donors. Both CTL populations were cytotoxic against peptide-pulsed HLA-A2+ target cells, but not against untreated target cells. A CD8+ anti-p53 CTL clone designated p264#2 was isolated from one of the bulk populations. It was found to have an intermediate affinity of approximately 10(-9) M for the epitope and to mediate cytotoxicity against several human tumor cell lines, including the squamous cell carcinoma of the head and neck cell line SCC-9, which is known to present the wild-type sequence p53(264-272) epitope. In addition, CTLs reactive against p53(149-157)-pulsed targets as well as a HLA-A2+ tumor cell line were cloned from a bulk population of antitumor CTLs obtained from one of the five normal PBLs restimulated with this epitope. The results indicate that CTLs recognizing wild-type sequence epitopes can be generated from precursors present in PBLs obtained from some normal individuals using autologous dendritic cells as antigen-presenting cells and suggest that vaccine strategies targeting these epitopes can lead to antitumor CTL generation, thereby emphasizing the therapeutic potential of p53-based cancer vaccines.


Assuntos
Células Dendríticas/imunologia , Linfócitos T Citotóxicos/imunologia , Proteína Supressora de Tumor p53/imunologia , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/imunologia , Separação Celular , Células Cultivadas , Células Clonais/imunologia , Testes Imunológicos de Citotoxicidade , Epitopos/imunologia , Antígeno HLA-A2/imunologia , Humanos , Interleucinas/metabolismo , Subpopulações de Linfócitos/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
9.
J Gene Med ; 1(6): 400-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10753065

RESUMO

BACKGROUND: The induction of cellular immune responses to melanocyte-specific enzymes such as the tyrosinase family of proteins is the goal of various clinical studies for the immunotherapy of melanoma. Tyrosinase-related protein-2 (TRP2) is an attractive model antigen for preclinical studies in C57BL/6 mice because it is naturally expressed by the murine B16 melanoma and can be recognized by self-reactive cytolytic T lymphocytes (CTL). Here we describe efforts to develop genetic immunization with dendritic cells (DC) for the immunotherapy of melanoma in this clinically relevant system. METHODS: Recombinant adenoviruses encoding green fluorescent protein (Ad-EGFP) and murine TRP2 (Ad-mTRP2) were constructed using Cre-loxP-mediated recombination. DC were generated in vitro from precursors in bone marrow and transduced with Ad-EGFP or Ad-mTRP2. Mice were immunized by direct injection of adenovirus or by injection of Ad-transduced DC. Induction of tumor immunity was assessed by intravenous challenge with B16 melanoma cells and enumeration of experimentally induced lung metastases. RESULTS: Flowcytometric analysis of DC transduced with Ad-EGFP demonstrated endogenous fluorescence due to cytoplasmatic expression of EGFP in 30-60% of cells. Ad-EGFP-transduced DC simultaneously displayed the DC-specific marker NLDC145 and high levels of MHC and costimulatory molecules on their cell surface. Transduction of DC with Ad-mTRP2 resulted in strong intracellular expression of TRP2 which could be readily detected by immunostaining. Importantly, immunization of mice with cultured Ad-mTRP2-transduced DC completely prevented the development of lung metastases following an intravenous challenge with B16 melanoma cells. This striking protective effect was observed with both the intravenous and the subcutaneous route of DC immunization. In vivo depletion of T-cell subsets suggested that the protective effect of an immunization with Ad-mTRP2-transduced DC involved both CD8+ and CD4+ T-cells. CONCLUSIONS: Our results demonstrate that DC-based genetic immunization of mice with TRP2, a clinically relevant melanocyte-specific self-antigen, induces effective cellular immunity and prevents metastatic growth of B16 melanoma cells in vivo.


Assuntos
Adenoviridae/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Vetores Genéticos/genética , Imunização , Imunoterapia Ativa , Oxirredutases Intramoleculares/imunologia , Melanoma Experimental/imunologia , Animais , Antígenos de Neoplasias/genética , Células Cultivadas , Células Dendríticas/transplante , Células Dendríticas/virologia , Feminino , Citometria de Fluxo , Genes Reporter , Rejeição de Enxerto , Tolerância Imunológica , Oxirredutases Intramoleculares/genética , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias
10.
Crit Rev Immunol ; 18(1-2): 29-35, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9419445

RESUMO

Immunotherapy targeting p53 missense mutations, which occur in nearly half of all human tumors, is limited by several factors, including the constraints of antigen processing and presentation. Due to the accumulation of mutated p53 molecules in tumors expressing p53 mutations, an alternative approach would be to target wild-type sequence, CTL-defined p53 epitopes. Obviously, the possibility of an autoimmune response is a major potential drawback to this therapy. Immunization of BALB/c mice with bone marrow-derived dendritic cells (DC) generated in the presence of GM-CSF/IL-4 and prepulsed with the H-2Kd-binding wild-type p53(232-240) peptide has been shown to induce anti-peptide CTL. These effectors were cross-reactive against sarcomas expressing p53 missense mutations outside of the p53(232-240) epitope, but not within it. Mitogen-activated splenocytes, which express elevated levels of p53, were not sensitive to these CTL. The p53 peptide-pulsed DC-based vaccine was shown to be effective in inducing tumor rejection in immunization and therapy models in the absence of any observable deleterious effect on naive mice. The murine model has now been extended to include the use of genetically modified DC-based vaccines as well.


Assuntos
Imunoterapia , Neoplasias/terapia , Proteína Supressora de Tumor p53/uso terapêutico , Animais , Autoimunidade , Modelos Animais de Doenças , Previsões , Antígeno HLA-A2/imunologia , Humanos , Neoplasias/imunologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia
11.
Eur J Immunol ; 27(10): 2702-7, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9368629

RESUMO

The clinical application of synthetic tumor peptide-based vaccines is currently limited to patients with specified major histocompatibility complex (MHC) class I alleles. Such logistic limitations may be overcome using tumor gene-based approaches. Here we describe the effective generation of dendritic cells (DC) expressing tumor peptide-MHC complexes as a result of particle-mediated transfer of genes encoding tumor-associated antigens (TAA). Bone marrow-derived DC were transfected with plasmid DNA encoding the tumor-associated viral antigen E7 derived from human papilloma virus (HPV) 16. When applied as a vaccine, these genetically modified DC induced antigen-specific CD8+ cytotoxic T lymphocytes (CTL) in vivo and promoted the rejection of a subsequent, normally lethal challenge with an HPV 16-transformed tumor cell line. Of greatest interest, immunization of mice with syngeneic DC genetically modified to enhance their presentation of a constitutive "self" epitope derived from the tumor-suppressor gene product p53 caused a significant reduction in the in vivo growth of a chemically induced p53-positive sarcoma. These results suggest that cancer vaccines consisting of DC genetically modified to express TAA of viral or "self" origin effectively induce antitumor immunity in vivo.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Células da Medula Óssea/imunologia , Vacinas Anticâncer , Células Dendríticas/imunologia , Epitopos/imunologia , Imunização/métodos , Proteínas Oncogênicas Virais/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Vacinas Anticâncer/imunologia , DNA Recombinante/genética , Feminino , Técnicas de Transferência de Genes , Genes Reporter , Antígenos H-2/imunologia , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus , Transfecção
12.
Stem Cells ; 15(2): 94-103, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9090785

RESUMO

Dendritic cells (DCs) are considered the most effective antigen-presenting cells (APCs) for primary immune responses. Since presentation of antigens to the immune system by appropriate professional APCs is critical to elicit a strong immune reaction and DCs seem to be quantitatively and functionally defective in the tumor host, DCs hold great promise to improve cancer vaccines. Even though they are found in lymphoid organs, skin and mucosa, the difficulty of generating large numbers of DCs has been a major limitation for their use in vaccine studies. A simple method for obtaining DCs from mouse bone marrow cells cultured in the presence of GM-CSF + interleukin 4 is now available. In four different tumor models, mice injected with DCs grown in GM-CSF plus interleukin 4 and prepulsed with a cytotoxic T lymphocyte-recognized tumor peptide epitope developed a specific cytotoxic T lymphocyte response and were protected against a subsequent tumor challenge with tumor cells expressing the relevant tumor antigen. Moreover, treatment of day 5-14 tumors with peptide-pulsed DCs resulted in sustained tumor regression in five different tumor models. These results suggest that presentation of tumor antigens to the immune system by professional APCs is a promising method to circumvent tumor-mediated immunosuppression and is the basis for ongoing clinical trials of cancer immunotherapy with tumor peptide-pulsed DCs.


Assuntos
Medula Óssea/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Fragmentos de Peptídeos/imunologia , Animais , Células da Medula Óssea , Vacinas Anticâncer/metabolismo , Terapia Combinada , Células Dendríticas/citologia , Humanos , Imunoterapia Adotiva , Fragmentos de Peptídeos/metabolismo
13.
J Exp Med ; 183(4): 1357-65, 1996 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8666894

RESUMO

The BALB/c Meth A sarcoma carries a p53 missense mutation at codon 234, which occurs in a peptide, termed 234CM, capable of being presented to cytotoxic T lymphocytes (CTL) by H-2Kd molecules (Noguchi, Y., E.C. Richards, Y.-T. Chen, and L.J. Old. 1994. Proc. Natl. Acad. Sci. USA. 91:3171-3175). Immunization of BALB/c mice with bone marrow-derived dendritic cells (DC), generated in the presence of granulocyte macrophage colony-stimulating factor and interleukin 4, and prepulsed with the Meth A p53 mutant peptide, induced CTL that specifically recognized peptide-pulsed P815 cells, as well as Meth A cells naturally expressing this epitope. Immunization with this vaccine also protected naive mice from a subsequent tumor challenge, and it inhibited tumor growth in mice bearing day 7 subcutaneous Meth A tumors. We additionally determined that immunization of BALB/c mice with DC pulsed with the p53 peptide containing the wild-type residue at position 234, 234CW, induced peptide-specific CTL that reacted against several methylcholanthrene-induced BALB/c sarcomas, including CMS4 sarcoma, and rejection of CMS4 sarcoma in vaccination and therapy (day 7) protocols. These results support the efficacy of DC-based, p53-derived peptide vaccines for the immunotherapy of cancer. The translational potential of this strategy is enhanced by previous reports showing that DC can readily be generated from human peripheral blood lymphocytes.


Assuntos
Fragmentos de Peptídeos/uso terapêutico , Sarcoma Experimental/terapia , Proteína Supressora de Tumor p53/uso terapêutico , Vacinação , Sequência de Aminoácidos , Animais , Sequência de Bases , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
14.
J Exp Med ; 183(1): 87-97, 1996 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8551248

RESUMO

Antigen presentation by host dendritic cells (DC) is critical for the initiation of adaptive immune responses. We have previously demonstrated in immunogenic murine tumor models that bone marrow (BM)-derived DC pulsed ex vivo with synthetic tumor-associated peptides, naturally expressed by tumor cells, serve as effective antitumor vaccines, protecting animals against an otherwise lethal tumor challenge (Mayordomo, J.I., T. Zorina, W.J. Storkus, C. Celluzzi, L.D. Falo, C.J. Melief, T. Ildstad, W.M. Kast, A.B. DeLeo, and M.T. Lotze. 1995. Nature Med. 1:1297-1302). However, T cell-defined epitopes have not been identified for most human cancers. To explore the utility of this approach in the treatment of tumors expressing as yet uncharacterized epitopes, syngeneic granulocyte/macrophage colony-stimulating factor-stimulated and BM-derived DC, pulsed with unfractionated acid-eluted tumor peptides (Storkus, W.J., H.J. Zeh III, R.D. Salter, and M.T. Lotze. 1993. J. Immunother. 14:94-103) were used to treat mice bearing spontaneous, established tumors. The adoptive transfer of 5 x 10(5) tumor peptide-pulsed DC dramatically suppressed the growth of weakly immunogenic tumors in day 4 to day 8 established MCA205 (H-2b) and TS/A (H-2d) tumor models, when applied in three biweekly intravenous injections. Using the immunogenic C3 (H-2b) tumor model in B6 mice, tumor peptide-pulsed DC therapy resulted in the erradication of established d14 tumors and long-term survival in 100% of treated animals. The DC-driven antitumor immune response was primarily cell mediated since the transfer of spleen cells, but not sera, from immunized mice efficiently protected sublethally irradiated naive mice against a subsequent tumor challenge. Furthermore, depletion of either CD4+ or CD8+ T cells from tumor-bearing mice before therapy totally suppressed the therapeutic efficacy of DC pulsed with tumor-derived peptides. Costimulation of the host cell-mediated antitumor immunity was critical since inoculation of the chimeric fusion protein CTLA4-Ig virtually abrogated the therapeutic effects of peptide-pulsed DC in vivo. The analysis of the cytokine pattern in the draining lymph nodes and spleens of tumor-bearing mice immunized with DC pulsed with tumor-eluted peptides revealed a marked upregulation of interleukin (IL) 4 and interferon (IFN) gamma production, as compared with mice immunized with DC alone or DC pulsed with irrelevant peptides. DC-induced antitumor effects were completely blocked by coadministration of neutralizing monoclonal antibody directed against T helper cell 1-associated cytokines (such as IL-12, tumor necrosis factor alpha, IFN-gamma), and eventually, but not initially, blocked by anti-mIL-4 mAb. Based on these results, we believe that DC pulsed with acid-eluted peptides derived from autologous tumors represents a novel approach to the treatment of established, weakly immunogenic tumors, and serves as a basis for designing clinical trials in cancer patients.


Assuntos
Células Dendríticas/imunologia , Imunoterapia Adotiva , Proteínas de Neoplasias/imunologia , Neoplasias Experimentais/terapia , Peptídeos/imunologia , Animais , Apresentação de Antígeno , Antígenos CD/imunologia , Antígeno B7-2 , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Linfonodos/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/metabolismo , Peptídeos/metabolismo , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Th1/imunologia
15.
Nat Med ; 1(12): 1297-302, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7489412

RESUMO

Dendritic cells, the most potent 'professional' antigen-presenting cells, hold promise for improving the immunotherapy of cancer. In three different well-characterized tumour models, naive mice injected with bone marrow-derived dendritic cells prepulsed with tumour-associated peptides previously characterized as being recognized by class I major histocompatibility complex-restricted cytotoxic T lymphocytes, developed a specific T-lymphocyte response and were protected against a subsequent lethal tumour challenge. Moreover, in the C3 sarcoma and the 3LL lung carcinoma murine models, treatment of animals bearing established macroscopic tumours (up to 1 cm2 in size) with tumour peptide-pulsed dendritic cells resulted in sustained tumour regression and tumour-free status in more than 80% of cases. These results support the clinical use of tumour peptide-pulsed dendritic cells as components in developing effective cancer vaccines and therapies.


Assuntos
Células da Medula Óssea , Células Dendríticas/transplante , Neoplasias Pulmonares/terapia , Sarcoma Experimental/terapia , Vacinas Sintéticas/imunologia , Animais , Apresentação de Antígeno , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas
17.
Mol Immunol ; 32(11): 805-17, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7675042

RESUMO

We have previously shown that specific T cell receptor (TCR) gamma V regions genes (V gamma 4 and V gamma 6) are rearranged and expressed by murine fetal liver (FL) cells cultured with IL-7. The present studies determined that the sequences of the TCR V region gene transcripts expressed in response to IL-7 included diverse and functional sequences expressed by thymocyte and peripheral V gamma 4+ and V gamma 6+ T cells, indicating that the IL-7-induced expression of these genes is functionally relevant and mimics normal in vivo developmental events of gamma delta T cells. We found that more than 50% of these TCR transcripts had N region diversity. The presence of N region diversity indicates that these TCR rearrangements took place in vitro, presumably in response to IL-7, because fresh (uncultured) FL cells do not produce detectable terminal deoxynucleotidyl transferase (TdT) mRNA or protein. We also found that 100% of immunoglobulin (Ig) VH7183-JH4 transcripts from FL cells cultured with IL-7 had N region diversity at the V-DJ region, while only 40% of Ig VH7183-JH4 transcripts from FL cells cultured in the absence of IL-7 had N region diversity at this region. FL cell cultures supplemented for 7 days with IL-7 had increased TdT mRNA and protein levels. However, since 1-day culture of FL cells with or without IL-7 resulted in induction of expression of TdT, IL-7 probably does not directly stimulate TdT expression, but increases the development and expansion of TdT+ lymphoid cells. These findings implicate IL-7 as a regulator of the molecular signals involved in controlling TCR gamma rearrangement and diversity, and provide an in vitro system for studying the regulation of TdT and N region diversity in B and T lymphoid progenitors by environmental signals.


Assuntos
Cadeias Pesadas de Imunoglobulinas/biossíntese , Interleucina-7/farmacologia , Fígado/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/biossíntese , Animais , Sequência de Bases , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Cadeias Pesadas de Imunoglobulinas/genética , Fígado/embriologia , Camundongos , Dados de Sequência Molecular , Gravidez , Receptores de Antígenos de Linfócitos T gama-delta/genética , Transcrição Gênica
18.
Cancer Res ; 55(1): 124-8, 1995 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-7528638

RESUMO

The finding that class I major histocompatibility complex (MHC)-restricted cytotoxic T lymphocytes (CTL) recognize peptide antigens (epitopes) bound to class I MHC molecules has accelerated efforts to identify CTL-defined tumor peptides for the development of peptide-based cancer immunotherapy. The Meth A sarcoma is probably one of the best studied of all murine tumors. It is extremely lethal unless protective immunity is induced. We recently reported the characterization of a cloned H-2Kd-restricted, CD8+ anti-Meth A CTL line (CTLMA-9C; Frassanito et al., Cancer Res., 54: 4424-4429, 1994). The cytotoxic reactivity of this CTL was shown to be restricted to Meth A sarcoma, and the results of the analysis of the immunogenicity of the CTL-resistant variant of Meth A, designated Meth A4R, indicate that the CTL-defined epitope is functional in tumor rejection. Here we have isolated class I MHC-associated peptides from Meth A sarcoma by mild acid treatment and resolved them into sixty fractions by reverse phase-HPLC. These fractions were then tested for their ability to sensitize the DBA/2 mastocytoma P815 to cytolysis by the anti-Meth A CTL. A single fraction, fraction 27, has been repeatedly identified as containing the CTL-defined epitope. Peptides eluted from the CTL-resistant variant, Meth A4R, failed to sensitize P815 to cytolysis by the anti-Meth A CTL, while fraction 27 derived from Meth A sensitized Meth A4R to lysis by the CTL. These findings confirm the peptide nature of the epitope recognized by CTL on the surface of Meth A. Our future efforts will focus on the identification and sequence analysis of the tumor peptides and the development of a tumor peptide-based vaccine model for immunotherapy.


Assuntos
Antígenos de Neoplasias/análise , Antígenos de Histocompatibilidade Classe I/análise , Sarcoma Experimental/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular , Testes Imunológicos de Citotoxicidade , Epitopos , Sarcoma de Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Células Tumorais Cultivadas
19.
Nat Immun ; 13(6): 289-300, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7894200

RESUMO

In this report, we present data on the expression and function of Fc gamma RII (CD32) by natural killer (NK) cells. Highly enriched NK cell populations were isolated from peripheral blood lymphocytes by negative selection and consisted of > or = 95% CD3-/CD56+ cells. Flow cytometric analyses with anti-CD32 monoclonal antibodies (mAbs) demonstrated that a small proportion of NK cells were recognized by mAbs IV.3 and 41H16. Two-color flow cytometric analysis indicated coexpression of the epitope on NK cells recognized by both these mAbs. Verification of expression of CD32 on NK cells was obtained by demonstrating coexpression of CD32 on either CD16+ or CD56+ cells. The CD32+/CD16+ and CD32+/CD56+ cells represented approximately 7 and 3% of the total, respectively. CD32 transcripts were identified from highly purified NK cells using reverse transcription-polymerase chain reaction with CD32-specific primers, followed by Southern blotting. Enhanced chemiluminescence-Western blot (ECL-WB) analysis of lysates of purified NK cells indicated that mAb IV.3 recognized a molecule of approximately 40 kD. The Fc gamma RII on NK cells was able to transduce intracellular signals in several types of assay. Cross-linking of anti-CD32 resulted in a mobilization of intracellular Ca2+, although to a lesser extent than that induced by cross-linking CD16. Both mAbs IV.3 and 41H16 were found to be capable of inducing reverse antibody-dependent cellular cytotoxicity against FcR+ target cells (e.g. P815). These data represent the first direct description of the expression and function of Fc gamma RII on human NK cells.


Assuntos
Células Matadoras Naturais/imunologia , Receptores de IgG/biossíntese , Receptores de IgG/imunologia , Anticorpos Monoclonais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sequência de Bases , Western Blotting , Cálcio/metabolismo , Linhagem Celular , Citometria de Fluxo , Humanos , Medições Luminescentes , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , Transdução de Sinais/imunologia , Espectrometria de Fluorescência
20.
Nat Immun ; 13(6): 315-30, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7894202

RESUMO

Studies have shown that the administration of anti-Ly-6E.1 monoclonal antibody (mAb) to tumor-bearing mice augments their T-cell-dependent functional activities, splenic natural killer (NK) cell activity, and antitumor resistance. The effect of the mAb on splenic NK cell activity resembles that of biological-response modifiers, which involves enhanced large granular lymphocyte (LGL) development in the bone marrow (BM) from pre-NK cells, and their subsequent migration and localization in peripheral organs. We analyze here the effect of anti-Ly-6E.1 mAb on the IL-2-dependent generation of NK cell activity in short-term cultures of mouse BM cells. The results indicate that an increase in the population of LGLs paralleled the mAb-mediated augmentation of IL-2-induced generation of NK cell activity in the cultures. Although pre-NK and T cells present in the BM expressed Ly-6 antigens, the augmentation of NK cell generation appears to be due to the mAb interacting with the T cells, not the pre-NK cells, resulting in increased synthesis of tumor necrosis factor-alpha, which in turn enhanced the interleukin-2-dependent development of NK cell activity.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos Ly/imunologia , Diferenciação Celular/imunologia , Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Animais , Células da Medula Óssea , Testes Imunológicos de Citotoxicidade , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...