Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Cancer ; 125(3): 630-8, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19455675

RESUMO

Regulatory T cells (Tregs) play an important role in maintaining immunological tolerance that is one of the main obstacles to overcome for improving antitumor immunity. Recently, the Treg has been shown to constitutively express OX40 (CD134), which is a member of the TNF-receptor family that is transiently expressed on effector T cells after TCR triggering, and through which the signal enhances effector T-cell proliferation and memory T-cell development. However, little is known about the role of OX40 costimulation to Tregs in tumor immunology. Here we show that OX40 signaling modulates the function of naturally occurring Tregs in vitro and in vivo. Foxp3 expression on Tregs was reduced by OX40 costimulation, but not by IL-2 stimulation. Tregs suppressed the proliferation of naïve CD4(+) CD25(-) T cells after TCR triggering, in contrast, OX40 costimulated Tregs that reduced Foxp3 expression reversed the suppressive function. In addition, Tregs inhibited the proliferation of TCR-stimulated (primed) CD4(+) T cells and naïve CD8(+) T cells after TCR-mediated activation, however, Tregs with OX40 costimulation lost their suppressive function. Interestingly, Tregs minimally suppressed the proliferation or the cytokine secretion of Ag-specific CD8(+) T cells after Ag-restimulation. Furthermore, Tregs suppressive function to the antitumor effect was reversed by OX40 costimulation in vivo. Our data indicate that, in addition to controlling effector T-cell function, OX40 costimulation directly controls Treg-mediated suppression in tumor immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/metabolismo , Glicoproteínas de Membrana/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Necrose Tumoral/imunologia , Transferência Adotiva , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Fluoresceínas , Corantes Fluorescentes , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Ligante OX40 , Receptor ErbB-2/genética , Succinimidas
2.
Clin Cancer Res ; 14(19): 6116-24, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829490

RESUMO

PURPOSE: Animal models of breast cancer metastases that recapitulate the pattern of metastatic progression seen in patients are lacking; metastatic breast cancer models do not currently exist for evaluation of immune-mediated therapies. We have developed and characterized a preclinical model for the evaluation of immune-mediated metastatic breast cancer therapies. EXPERIMENTAL DESIGN: The NT2.5 mammary tumor cell line was injected into the left cardiac ventricle of immunotolerant transgenic neu-N mice and athymic nu/nu mice. Metastatic progression was monitored by bioluminescent, small-animal magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography/computed tomography imaging, and also by histopathology. Antigen expression in normal organs and tumor metastases was evaluated by Western blot analysis and flow cytometry. RESULTS: Left cardiac ventricle injection of NT2.5 cells yielded widespread metastases in bones, liver, and spleen. Three to four weeks after injection, mice exhibited hind limb paralysis and occasional abdominal enlargement. Bioluminescence imaging of metastatic progression was successful in nude mice but the bioluminescent cells were rejected in immunocompetent mice. Other imaging modalities allowed successful imaging of nonbioluminescent cells. Small-animal positron emission tomography imaging allowed visualization of disease, in vivo, in the bones and liver. Magnetic resonance imaging revealed initial dissemination of the tumor cells to the bone marrow. Small-animal single-photon emission computed tomography/computed tomography imaging identified metastatic bone lesions targeted by a radiolabeled antibody. CONCLUSION: The model closely recapitulates the pattern of metastatic spread in breast cancer. This immunotolerant metastatic model is a novel addition to existing breast cancer models and coupling the model with in vivo imaging greatly facilitates the evaluation of targeted immunotherapies of metastasis.


Assuntos
Modelos Animais de Doenças , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Receptor ErbB-2/genética , Animais , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Metástase Neoplásica , Transplante de Neoplasias , Ratos , Receptor ErbB-2/metabolismo , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único/métodos
3.
Cancer Res ; 68(10): 3873-80, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483272

RESUMO

Treatment failure in breast cancer is largely the failure to control metastatic dissemination. In this study, we investigated the efficacy of an antibody against the rat variant of HER-2/neu, labeled with the alpha-particle emitter (213)Bi to treat widespread metastases in a rat/neu transgenic mouse model of metastatic mammary carcinoma. The model manifests wide-spread dissemination of tumor cells leading to osteolytic bone lesions and liver metastases, common sites of clinical metastases. The maximum tolerated dose was 120 muCi of (213)Bi-7.16.4. The kinetics of marrow suppression and subsequent recovery were determined. Three days after left cardiac ventricular injection of 10(5) rat HER-2/neu--expressing syngeneic tumor cells, neu-N mice were treated with (a) 120 muCi (213)Bi-7.16.4, (b) 90 muCi (213)Bi-7.16.4, (c) 120 muCi (213)Bi-Rituximab (unreactive control), and (d) unlabeled 7.16.4. Treatment with 120 muCi (213)Bi-7.16.4 increased median survival time to 41 days compared with 28 days for the untreated controls (P < 0.0001); corresponding median survival times for groups b, c, and d were 36 (P < 0.001), 31 (P < 0.01), and 33 (P = 0.05) days, respectively. Median survival relative to controls was not significantly improved in mice injected with 10-fold less cells or with multiple courses of treatment. We concluded that alpha-emitter (213)Bi-labeled monoclonal antibody targeting the HER-2/neu antigen was effective in treating early-stage HER-2/neu--expressing micrometastases. Analysis of the results suggests that further gains in efficacy may require higher specific activity constructs or target antigens that are more highly expressed on tumor cells.


Assuntos
Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/radioterapia , Radioimunoterapia/métodos , Receptor ErbB-2/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Cinética , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Transplante de Neoplasias , Radioimunoterapia/instrumentação , Ratos , Receptor ErbB-2/genética , Rituximab , Esferoides Celulares/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas/metabolismo
4.
J Clin Invest ; 118(5): 1700-11, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18398507

RESUMO

The pathogenic nature of cancer is attributed, at least in part, to the ability of tumors cells to induce systemic and local mechanisms of immune tolerance. However, we previously reported that tumor-free survival in up to 100% of tolerized HER-2/neu transgenic mice can be achieved by administration of neu-specific mAb concurrently with a HER-2/neu-expressing, GM-CSF-secreting whole cell vaccine. In this report, we show that one mechanism of improved antitumor activity induced by the combination of these 2 neu-targeted interventions was enhanced Fc-mediated activation of APCs. Specifically, in vivo studies demonstrated localization of radiolabeled neu-specific mAb at the vaccine site. Subsequently, increased accumulation of neu-specific mAb at the vaccine-draining lymph node correlated with increased vaccine cell uptake by DCs in vivo. This led to enhancement of CD8(+) neu-specific T cell function in terms of proliferation, cytokine production, and central memory development. Thus, the administration of a neu-specific mAb with a neu-targeted GM-CSF-secreting tumor vaccine enhanced induction of neu-specific CD8(+) T cells through Fc-mediated activation of DCs. This multimodality attack on the same tumor antigen may have the potential to overcome tolerance to self antigens and weaken the immunosuppressive networks within the tumor microenvironment.


Assuntos
Anticorpos Monoclonais/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Receptor ErbB-2/imunologia , Transferência Adotiva , Animais , Anticorpos Monoclonais/uso terapêutico , Antígeno CD11c/imunologia , Linfócitos T CD8-Positivos/citologia , Linhagem Celular , Células Dendríticas/citologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/patologia , Receptor ErbB-2/genética , Taxa de Sobrevida
5.
J Immunol ; 176(2): 974-83, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16393983

RESUMO

T cell costimulation via OX40 is known to increase CD4+ T cell expansion and effector function and enhances the development of T cell memory. OX40 costimulation can also prevent, and even reverse, CD4+ T cell anergy. However, the role of OX40 in CD8+ T cell function is less well defined, particularly in the setting of immune tolerance. To determine the effects of OX40 costimulation on the induction of the host CD8+ T cell repertoire to an endogenous tumor Ag, we examined the fate of CD8+ T cells specific for the immunodominant rat HER-2/neu epitope, RNEU420-429, in FVB MMTV-neu (neu-N) mice, which express rat HER-2/neu protein in a predominantly mammary-restricted fashion. We show that the RNEU420-429-specific T cell repertoire in neu-N mice expands transiently after vaccination with a neu-targeted GM-CSF-secreting whole-cell vaccine, but quickly declines to an undetectable level. However, OX40 costimulation, when combined with GM-CSF-secreting tumor-targeted vaccination, can break established CD8+ T cell tolerance in vivo by enhancing the expansion, and prolonging the survival and effector function of CD8+ T cells specific for RNEU420-429. Moreover, we demonstrate that OX40 expression is up-regulated on both CD4+ and CD8+ T cells shortly after administration of a GM-CSF expressing vaccine. These studies highlight the increased efficacy of OX40 costimulation when combined with a GM-CSF-secreting vaccine, and define a new role for OX40 costimulation of CD8+ T cells in overcoming tolerance and boosting antitumor immunity.


Assuntos
Antígenos de Neoplasias , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Glicoproteínas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Receptores do Fator de Necrose Tumoral/administração & dosagem , Animais , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Feminino , Tolerância Imunológica , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Fragmentos de Peptídeos/imunologia , Ratos , Receptor ErbB-2 , Receptores OX40 , Proteínas Recombinantes , Linfócitos T Auxiliares-Indutores/imunologia
6.
Cancer Res ; 66(2): 638-45, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16423990

RESUMO

In cancer patients and in those at high risk, systemic exposure to agents for therapy or prevention is accompanied by undesirable side effects. We hypothesized that it is possible to prevent and treat breast cancer by introducing anticancer agents into the mammary ductal network. Here, we show the efficacy of intraductally administered anticancer agents 4-hydroxytamoxifen and pegylated liposomal doxorubicin (PLD) in the prevention and treatment of breast cancer using the rat N-methyl-N'-nitrosourea-induced and spontaneous HER-2/neu transgenic mouse (neu-N) models of breast cancer. Intraductal administration of PLD to neu-N mice caused regression of established tumors and prevented tumor development more effectively than i.v. injection (P < 0.0001). Intraductal administration resulted in lower circulating levels of PLD compared with i.v. administration, with no evidence of systemic toxicity or long-term histopathologic changes in the mammary gland. Compared with systemic administration, intraductal injection provides direct access to breast lesions with higher local and lower systemic drug exposure. These studies suggest that this approach has potential for application to prevention and neoadjuvant therapy of early breast cancer.


Assuntos
Doxorrubicina/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Neoplasias Mamárias Animais/prevenção & controle , Polietilenoglicóis/farmacologia , Tamoxifeno/análogos & derivados , Animais , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Vias de Administração de Medicamentos , Antagonistas de Estrogênios/administração & dosagem , Feminino , Humanos , Glândulas Mamárias Animais , Camundongos , Camundongos Transgênicos , Terapia Neoadjuvante , Polietilenoglicóis/administração & dosagem , Ratos , Tamoxifeno/administração & dosagem , Tamoxifeno/farmacologia
8.
J Exp Med ; 201(10): 1591-602, 2005 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-15883172

RESUMO

A major barrier to successful antitumor vaccination is tolerance of high-avidity T cells specific to tumor antigens. In keeping with this notion, HER-2/neu (neu)-targeted vaccines, which raise strong CD8(+) T cell responses to a dominant peptide (RNEU(420-429)) in WT FVB/N mice and protect them from a neu-expressing tumor challenge, fail to do so in MMTV-neu (neu-N) transgenic mice. However, treatment of neu-N mice with vaccine and cyclophosphamide-containing chemotherapy resulted in tumor protection in a proportion of mice. This effect was specifically abrogated by the transfer of neu-N-derived CD4(+)CD25(+) T cells. RNEU(420-429)-specific CD8(+) T cells were identified only in neu-N mice given vaccine and cyclophosphamide chemotherapy which rejected tumor challenge. Tetramer-binding studies demonstrated that cyclophosphamide pretreatment allowed the activation of high-avidity RNEU(420-429)-specific CD8(+) T cells comparable to those generated from vaccinated FVB/N mice. Cyclophosphamide seemed to inhibit regulatory T (T reg) cells by selectively depleting the cycling population of CD4(+)CD25(+) T cells in neu-N mice. These findings demonstrate that neu-N mice possess latent pools of high-avidity neu-specific CD8(+) T cells that can be recruited to produce an effective antitumor response if T reg cells are blocked or removed by using approaches such as administration of cyclophosphamide before vaccination.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Epitopos de Linfócito T/imunologia , Ativação Linfocitária/imunologia , Neoplasias Mamárias Experimentais/imunologia , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/genética , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Ciclofosfamida/administração & dosagem , Citotoxicidade Imunológica , Epitopos de Linfócito T/genética , Feminino , Genes erbB-2/imunologia , Imunossupressores/administração & dosagem , Imunoterapia Adotiva , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Transgênicos , Receptores de Interleucina-2/imunologia , Vacinação
9.
J Biol Chem ; 280(1): 54-63, 2005 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-15507452

RESUMO

Cancer vaccines designed to elicit an antibody response that target antigenic sites on a tumor antigen must closely mimic the three-dimensional structure of the corresponding region on the antigen. We have designed a complex immunogen derived from the extracellular domain of human HER-2/neu-(626-649) that represents a three-dimensional epitope. We have successfully introduced two disulfide bonds into this sequence, thereby recapitulating the natural disulfide pairings observed in the native protein. To evaluate the immunogenicity of the doubly cyclized disulfide-linked peptide versus the free uncyclized peptide we examined the induction of antibody responses in both inbred and outbred mice strains, with both constructs eliciting high titered antibodies. The disulfide-paired specific antibodies exhibited enhanced cross-reactivity to HER-2/neu expressed on BT-474 cell line as determined by flow cytometry. The antitumor activities of the disulfidepaired specific antibodies did not improve the in vitro growth inhibition of human breast cancer cells overexpressing HER-2, but showed superior antitumor responses in the context of ADCC and interferon-gamma induction. Inbred mice (FVB/n) vaccinated with the disulfide-paired epitope exhibited a statistically significant reduction in the development of exogenously administered tumors in vivo compared with mice receiving either the free uncyclized or the promiscuous T-cell epitope (MVF) control peptide (p = 0.001). This study demonstrates the feasibility and importance of designing conformational epitopes that mimic the tertiary structure of the native protein for eliciting biologically relevant anti-tumor antibodies. Such approaches are a prerequisite to the design of effective peptide vaccines.


Assuntos
Neoplasias da Mama/imunologia , Vacinas Anticâncer , Epitopos de Linfócito B/imunologia , Receptor ErbB-2/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antineoplásicos/imunologia , Neoplasias da Mama/terapia , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Dissulfetos , Epitopos de Linfócito B/química , Epitopos de Linfócito B/metabolismo , Feminino , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/prevenção & controle , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Conformação Proteica , Receptor ErbB-2/química , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia
10.
Anticancer Res ; 24(5A): 2879-84, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15517891

RESUMO

Bamboo grass leaves of different Sasa species have been widely used in food and medicine in Eastern Asia for hundreds of years. Of special interest are Kumazasa (Sasa senanensis rehder) leaves used to prepare an alkaline extract known as Sasa Health. This extract was reported to inhibit both the development and growth of mammary tumors in a mammary tumor strain of virgin SHN mice (1). We found that Sasa Health exerts a significant protective effect on spontaneous mammary tumorigenesis in another mouse model of human breast cancer, the transgenic FVB-Her2/NeuN mouse model. Two cohorts of Her2/NeuN female mice of different age (eleven-week-old and twenty-four-week-old) chronically treated with Sasa Health in drinking water showed both a delay in the development of tumors and reduced tumor multiplicity. Sasa Health also induced inhibition of mammary duct branching and side bud development in association with reduced angiogenesis. Altogether these findings indicate that Sasa Health contains phytochemicals that can effectively retard spontaneous mammary tumorigenesis.


Assuntos
Neoplasias Mamárias Experimentais/prevenção & controle , Fitoterapia/métodos , Receptor ErbB-2/genética , Sasa , Animais , Feminino , Genes erbB-2 , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Extratos Vegetais/farmacologia
11.
IDrugs ; 7(1): 64-9, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14730469
12.
Breast Dis ; 20: 13-24, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15687703

RESUMO

Rapid progress in defining the molecular underpinnings of the antitumor immune response has laid the foundation for tumor immunotherapy, leading to multiple early clinical studies testing vaccines for the treatment of breast cancer. Together, these small trials have provided early evidence for the induction of clinically relevant vaccine-induced tumor-specific immunity in some patients. However, they have not convincingly demonstrated a significant impact on disease progression or overall survival in women with advanced breast cancer. These disappointing results are likely due to the negative impact of standard cancer treatments on vaccine-activated antitumor immunity, the limited potency of current tumor vaccine formulations against large burdens of established tumor, and the presence of pre-existing tumor-specific immune tolerance. It is increasingly clear that standard and novel breast cancer treatments can influence the antitumor immune response. Also, signaling pathways that regulate immune responses have emerged as novel targets for immune modulation. The use of preclinical models to elucidate the pharmacodynamic interactions of standard breast cancer treatment modalities and novel, targeted immunotherapeutics with breast cancer vaccines will facilitate the development of combinatorial immunotherapeutic strategies. Combined modality immunotherapies should maximize the potency of the antitumor immune response, thereby improving the outcome of breast cancer therapy.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Vacinas Anticâncer/uso terapêutico , Imunoterapia , Humanos
14.
J Immunol ; 171(4): 2161-9, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902523

RESUMO

HER-2/neu is overexpressed in several cancers including 30% of breast carcinomas, and correlates with a poor outcome. HER-2/neu-transgenic (neu-N) mice that overexpress the non-transforming rat neu develop spontaneous mammary carcinomas and demonstrate immunotolerance to the neu protein similar to that observed in patients with neu-expressing cancers. In neu-N mice, neu-targeted vaccination induces weak T cell and negligible Ab responses sufficient to delay but not eradicate transplanted neu-expressing tumor. Here we demonstrate that passive infusion of neu-specific mAbs in sequence with whole cell vaccination significantly improves tumor-free survival over either modality alone. Importantly, treatment of neu-N mice with vaccine in combination with two distinct neu-specific Abs is particularly efficacious, preventing tumor in 70% and eradicating established tumor in 30% of neu-N mice. In vivo lymphocyte subpopulation depletion experiments demonstrate that the efficacy of Ab, alone or combined with vaccine, is dependent on both CD4(+) and CD8(+) T cells. Furthermore, the in vivo antitumor effects of vaccine and Ab are associated with a significant increase in the number and function of neu-specific CD8(+) T cells. Collectively, these observations suggest that similarly increased efficacy could be obtained by combining neu-targeted vaccination and neu-specific Abs such as trastuzumab (Herceptin) in patients with neu-expressing cancers.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Epitopos de Linfócito T/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/genética , Células 3T3 , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/imunologia , Feminino , Esquemas de Imunização , Injeções Subcutâneas , Contagem de Linfócitos , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/mortalidade , Camundongos , Camundongos Transgênicos , Ratos , Receptor ErbB-2/biossíntese , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Análise de Sobrevida , Ubiquitinas/metabolismo , Regulação para Cima/genética , Regulação para Cima/imunologia
15.
J Immunol ; 170(8): 4273-80, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12682262

RESUMO

The HER-2/neu (neu-N)-transgenic mice are a clinically relevant model of breast cancer. They are derived from the parental FVB/N mouse strain and are transgenic for the rat form of the proto-oncogene HER-2/neu (neu). In this study, we report the identification of a MHC class I peptide in the neu protein that is recognized by CD8(+) T cells derived from vaccinated FVB/N mice. This 10-mer was recognized by all tumor-specific FVB/N T cells generated regardless of the TCR Vbeta region expressed by the T cell or the method of vaccination used, establishing it as the immunodominant MHC class I epitope in neu. T cells specific for this epitope were able to cure FVB/N mice of transplanted neu-expressing tumor cells, demonstrating that this is a naturally processed peptide. Altered peptide analogs of the epitope were analyzed for immunogenicity. Vaccination with dendritic cells pulsed with a heteroclitic peptide provided FVB/N and neu-N mice with increased protection against tumor challenge as compared with mice immunized with dendritic cells loaded with either wild-type or irrelevant peptide. Discovery of this epitope allows for better characterization of the CD8(+) T cell responses in the neu-N mouse model in which neu-specific tolerance must be overcome to produce effective antitumor immunity.


Assuntos
Apresentação de Antígeno , Genes erbB-2/imunologia , Antígenos H-2/metabolismo , Epitopos Imunodominantes/isolamento & purificação , Epitopos Imunodominantes/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Células 3T3 , Transferência Adotiva , Animais , Apresentação de Antígeno/genética , Linhagem Celular Transformada , Células Clonais , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/biossíntese , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/genética , Inibidores do Crescimento/imunologia , Antígenos H-2/genética , Antígenos H-2/imunologia , Antígenos H-2/isolamento & purificação , Antígeno de Histocompatibilidade H-2D , Humanos , Epitopos Imunodominantes/genética , Injeções Intravenosas , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Mapeamento de Peptídeos , Proto-Oncogene Mas , Ratos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...