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.
PLoS One ; 12(6): e0179239, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28617827

RESUMO

Immunodeficient mice transplanted with human peripheral blood mononuclear cells (PBMCs) are promising tools to evaluate human immune responses to vaccines. However, these mice usually develop severe graft-versus-host disease (GVHD), which makes estimation of antigen-specific IgG production after antigen immunization difficult. To evaluate antigen-specific IgG responses in PBMC-transplanted immunodeficient mice, we developed a novel NOD/Shi-scid-IL2rγnull (NOG) mouse strain that systemically expresses the human IL-4 gene (NOG-hIL-4-Tg). After human PBMC transplantation, GVHD symptoms were significantly suppressed in NOG-hIL-4-Tg compared to conventional NOG mice. In kinetic analyses of human leukocytes, long-term engraftment of human T cells has been observed in peripheral blood of NOG-hIL-4-Tg, followed by dominant CD4+ T rather than CD8+ T cell proliferation. Furthermore, these CD4+ T cells shifted to type 2 helper (Th2) cells, resulting in long-term suppression of GVHD. Most of the human B cells detected in the transplanted mice had a plasmablast phenotype. Vaccination with HER2 multiple antigen peptide (CH401MAP) or keyhole limpet hemocyanin (KLH) successfully induced antigen-specific IgG production in PBMC-transplanted NOG-hIL-4-Tg. The HLA haplotype of donor PBMCs might not be relevant to the antibody secretion ability after immunization. These results suggest that the human PBMC-transplanted NOG-hIL-4-Tg mouse is an effective tool to evaluate the production of antigen-specific IgG antibodies.


Assuntos
Anticorpos Antineoplásicos/imunologia , Formação de Anticorpos , Proteínas de Transporte/imunologia , Imunização , Imunoglobulina G/imunologia , Interleucina-4/imunologia , Peptídeos/farmacologia , Receptor ErbB-2/farmacologia , Animais , Anticorpos Antineoplásicos/genética , Linfócitos B/imunologia , Linfócitos B/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Proteínas de Transporte/genética , Xenoenxertos , Humanos , Imunoglobulina G/genética , Interleucina-4/genética , Camundongos , Camundongos SCID , Camundongos Transgênicos , Peptídeos/imunologia , Receptor ErbB-2/imunologia , Células Th2/imunologia , Células Th2/transplante
2.
Mol Biol Cell ; 28(12): 1612-1621, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28428257

RESUMO

Tumor development progresses through a complex path of biomechanical changes leading first to cell growth and contraction and then cell deadhesion, scattering, and invasion. Tumorigenic factors may act specifically on one of these steps or have a wider spectrum of actions, leading to a variety of effects and thus sometimes to apparent contradictory outcomes. Here we used micropatterned lines of collagen type I/fibronectin on deformable surfaces to standardize cell behavior and measure simultaneously cell size, speed of motion and magnitude of the associated traction forces at the level of a single cell. We analyzed and compared the normal human breast cell line MCF10A in control conditions and in response to various tumorigenic factors. In all conditions, a wide range of biomechanical properties was identified. Despite this heterogeneity, normal and transformed motile cells followed a common trend whereby size and contractile forces were negatively correlated with cell speed. Some tumorigenic factors, such as activation of ErbB2 or loss of the ßsubunit of casein kinase 2, shifted the whole population toward a faster speed and lower contractility state. Treatment with transforming growth factor ß induced some cells to adopt opposing behaviors such as extremely high versus extremely low contractility. Thus tumor transformation amplified preexisting population heterogeneity and led some cells to exhibit biomechanical properties that were more extreme than those observed with normal cells.


Assuntos
Fenômenos Biomecânicos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fenômenos Biomecânicos/fisiologia , Neoplasias da Mama , Carcinogênese , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células , Tamanho Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Colágeno Tipo I/efeitos dos fármacos , Proteínas Contráteis/efeitos dos fármacos , Feminino , Humanos , Receptor ErbB-2/farmacologia , Fator de Crescimento Transformador beta/farmacologia
3.
Future Oncol ; 12(11): 1321-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27044454

RESUMO

E75 (nelipepimut-S) is an immunogenic peptide derived from the HER2 protein. When combined with the immunoadjuvant granulocyte-macrophage colony-stimulating factor (GM-CSF), nelipepimut-S has been used as a vaccine that is capable of eliciting a robust anti-HER2 immune response. Early-phase clinical trials that enrolled women with node-positive or high-risk node-negative breast cancer who had been rendered disease free with standard of care therapy but were at risk for recurrence, demonstrated the vaccine to be safe with a suggestion of clinical benefit. Nelipepimut-S is currently being evaluated in a Phase III clinical trial. This article covers the preclinical and clinical development of nelipepimut-S.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor ErbB-2/imunologia , Animais , Ensaios Clínicos como Assunto , Desenho de Fármacos , Feminino , Humanos , Receptor ErbB-2/farmacologia , Vacinas de Subunidades Antigênicas/farmacologia
4.
J Natl Cancer Inst ; 107(6): djv090, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25888715

RESUMO

BACKGROUND: Several treatment strategies target the human epidermal growth factor receptor 2 (HER2) in breast carcinomas, including monoclonal antibodies directed against HER2's extracellular domain (ECD) and small molecule inhibitors of its tyrosine kinase activity. Yet, novel therapies are needed that prevent HER2 dimerization with other HER family members, because current treatments are only partially effective. METHODS: To test the hypothesis that HER2 activation requires a protein sequence in the HER2-ECD that mediates HER2 homo- and heterodimerization, we introduced a series of deletion mutations in the third subdomain of HER2-ECD. These deletion mutants were retrovirally expressed in breast cancer (BC) cells that naturally overexpress HER2 and in noncancerous, HER2-negative breast epithelial cells. One-factor analysis of variance or Student's t test were used to analyze differences. All statistical tests were two-sided. RESULTS: The smallest deletion in the ECD domain of HER2, which removed only 16 amino acids (HER2-ECDΔ451-466), completely disrupted the oncogenic potential of HER2. In contrast to wild-type HER2, the mutant-inhibited anchorage-independent growth (mean number of colonies: mutant, 70, 95% confidence interval [CI] = 55 to 85; wild-type, 400, 95% CI = 320 to 480, P < .001) increased sensitivity to paclitaxel treatment in both transformed and nontransformed cells. Overexpression of HER2Δ451-466 efficiently inhibited activation of HER1, HER2, and HER3 in all cell lines tested. CONCLUSIONS: These findings reveal that an essential "activating" sequence exists in the extracellular domain of HER2. Disruption of this sequence disables the HER2 dimerization loop, blocks subsequent activation of HER2-driven oncogenic signaling, and generates a dominant-negative form of HER2. Reagents specifically against this molecular activation switch may represent a novel targeted approach for the management of HER2-overexpressing carcinomas.


Assuntos
Neoplasias da Mama/metabolismo , Dimerização , Deleção de Genes , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologia , Mama/metabolismo , Neoplasias da Mama/virologia , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Paclitaxel/farmacologia , Receptor ErbB-2/farmacologia , Regulação para Cima
5.
Clin Cancer Res ; 21(12): 2737-44, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25370464

RESUMO

PURPOSE: The HER2 mAb, trastuzumab, is a standard therapy for patients with HER2-positive breast cancer before acquired resistance. Afatinib, an irreversible, oral, small-molecule ErbB family blocker, shows clinical activity in trastuzumab-refractory HER2-positive breast cancer. EXPERIMENTAL DESIGN: This phase I study used a 3+3 dose escalation to determine the MTD of oral once-daily afatinib in combination with the recommended dose of intravenous trastuzumab (4 mg/kg week 1; 2 mg/kg/wk thereafter). Adult women with confirmed advanced/metastatic HER2-positive breast cancer were eligible. RESULTS: Of 18 patients treated, 16 received daily afatinib 20 mg and two 30 mg. Overall, 4 of 13 and 2 of 2 patients receiving afatinib 20 mg and 30 mg, respectively, experienced dose-limiting toxicity (DLT; all CTCAE grade 3 diarrhea). Most frequent treatment-related adverse events were diarrhea (94%), rash (56%), and fatigue (56%). Overall, pharmacokinetic profiles of afatinib and trastuzumab in combination were consistent with the known characteristics of each alone. Overall, objective response and disease control rates were 11% and 39%, respectively, with median progression-free survival 111.0 days (95% confidence interval, 56.0-274.0). CONCLUSIONS: The MTD of afatinib was 20 mg daily combined with the recommended weekly dose of trastuzumab, with 1 of 6 patients showing DLTs in the dose escalation. However, additional DLTs occurred in the dose-expansion phase meaning that this MTD cannot be recommended for phase II development without strict diarrhea management. There was no evidence suggesting relevant pharmacokinetic drug-drug interactions. Signs of clinical activity were seen in trastuzumab-resistant HER2-positive breast cancer, suggesting further investigation with optimal diarrhea management is warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Receptor ErbB-2/metabolismo , Adulto , Afatinib , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais , Neoplasias da Mama/patologia , Terapia Combinada , Feminino , Humanos , Dose Máxima Tolerável , Pessoa de Meia-Idade , Metástase Neoplásica , Quinazolinas/administração & dosagem , Receptor ErbB-2/farmacologia , Retratamento , Trastuzumab/administração & dosagem
6.
Biochemistry ; 51(12): 2558-68, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22385253

RESUMO

Receptor tyrosine kinases bind ligands such as cytokines, hormones, and growth factors and regulate key cellular processes, including cell division. They are also implicated in the development of many types of cancer. One such example is the Neu receptor tyrosine kinase found in rats (homologous to the human ErbB2 protein), which can undergo a valine to glutamic acid (V(664)E) mutation at the center of its α-helical transmembrane domain. This substitution results in receptor activation and oncogenesis. The molecular basis of this dramatic change in behavior upon introduction of the V(664)E mutation has been difficult to pin down, with conflicting results reported in the literature. Here we report the first quantitative, thermodynamic analysis of dimerization and biophysical characterization of the rat Neu transmembrane domain and several mutants in a range of chemical environments. These data have allowed us to identify the effects of the V(664)E mutation in the isolated TM domain with respect to protein-protein and protein-lipid interactions, membrane insertion, and secondary structure. We also report the results from a 100 ns atomistic molecular dynamics simulation of the Neu transmembrane domain in a model membrane bilayer (dipalmitoylphosphatidylcholine). The results from simulation and experiment are in close agreement and suggest that, in the model systems investigated, the V(664)E mutation leads to a weakening of the TM dimer and a change in sequence-dependent interactions. These results are contrary to recent results obtained in mammalian membranes, and the implications of this are discussed.


Assuntos
Fenômenos Biofísicos , Membrana Celular/metabolismo , Micelas , Simulação de Dinâmica Molecular , Mutação Puntual , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/efeitos dos fármacos , Detergentes/química , Escherichia coli/citologia , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Dados de Sequência Molecular , Oncogenes/genética , Fosforilcolina/análogos & derivados , Fosforilcolina/química , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Receptor ErbB-2/genética , Receptor ErbB-2/farmacologia , Termodinâmica
7.
Mol Psychiatry ; 17(1): 1, 99-107, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21483438

RESUMO

Neuregulin 1 (NRG1) is a secreted trophic factor that activates the postsynaptic erbB4 receptor tyrosine kinase. Both NRG1 and erbB4 have been repeatedly associated with schizophrenia, but their downstream targets are not well characterized. ErbB4 is highly abundant in interneurons, and NRG1-mediated erbB4 activation has been shown to modulate interneuron function, but the role for NRG1-erbB4 signaling in regulating interneuron dendritic growth is not well understood. Here we show that NRG1/erbB4 promote the growth of dendrites in mature interneurons through kalirin, a major dendritic Rac1-GEF. Recent studies have shown associations of the KALRN gene with schizophrenia. Our data point to an essential role of phosphorylation in kalirin-7's C terminus as the critical site for these effects. As reduced interneuron dendrite length occurs in schizophrenia, understanding how NRG1-erbB4 signaling modulates interneuron dendritic morphogenesis might shed light on disease-related alterations in cortical circuits.


Assuntos
Dendritos/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Interneurônios/citologia , Neuregulina-1/metabolismo , Receptor ErbB-2/deficiência , Análise de Variância , Animais , Encéfalo/citologia , Células Cultivadas , Dendritos/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Guanilato Quinases/metabolismo , Humanos , Imunoprecipitação , Interneurônios/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação/fisiologia , Neuregulina-1/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-fyn/farmacologia , RNA Interferente Pequeno/metabolismo , Receptor ErbB-2/farmacologia , Transdução de Sinais/genética , Transfecção , Tirosina/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
8.
Future Cardiol ; 7(5): 693-704, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21929348

RESUMO

The HER family of tyrosine kinase receptors includes several members that are clinically important targets in cancer therapies, in particular HER1 (the EGF receptor) and HER2, other members include HER3 and HER4. Trastuzumab, a humanized monoclonal antibody and lapatinib, a tyrosine kinase inhibitor, are drugs that target HER2, which is highly expressed in 20-30% of breast cancers. Trastuzumab is recommended as an adjuvant therapy for lymph node positive, HER2-positive breast cancers, or node-negative cancer with high-risk of recurrence, as well as in stage IV cancers. One serious side effect of trastuzumab is cardiomyocyte dysfunction, resulting in reduced heart contractile efficiency. The incidence of collateral effects on the heart with trastuzumab therapy increases in people with cardiovascular risk factors, heart disease and when combined with other chemotherapeutics. When cardiotoxicity was observed with trastuzumab, several studies have addressed potential cardiac damage of trastuzumab itself and lapatinib. The differences in cardiovascular effects of these two compounds are somewhat unexpected and suggest distinct mechanisms of action, which have clear implications in clinical application and prevention of cardiotoxicity in cardio-oncological approaches.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Coração/efeitos dos fármacos , Receptor ErbB-2/farmacologia , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/sangue , Neoplasias da Mama/tratamento farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lapatinib , Neoplasias Ovarianas/tratamento farmacológico , Fosforilação , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/efeitos adversos , Quinazolinas/farmacologia , Razoxano/farmacologia , Receptor ErbB-2/efeitos adversos , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Receptor ErbB-2/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Trastuzumab
9.
J Cell Biochem ; 111(3): 643-52, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20589763

RESUMO

In several human cancers, ErbB2 over-expression facilitates the formation of constitutively active homodimers resistant to internalization which results in progressive signal amplification from the receptor, conducive to cell survival, proliferation, or metastasis. Here we report on studies of the influence of ErbB2 over-expression on localization and signaling in polarized Caco-2 and MDCK cells, two established models to study molecular trafficking. In these cells, ErbB2 is not over-expressed and shares basolateral localization with ErbB3. Over-expression of ErbB2 by transient transfection resulted in partial separation of the receptors by relocalization of ErbB2, but not ErbB3, to the apical surface, as shown by biotinylation of the apical or basolateral surfaces. These results were confirmed by immunofluorescence and confocal microscopy. Polarity controls indicated that the relocalization of ErbB2 is not the result of depolarization of the cells. Biotinylation and confocal microscopy also showed that apical, but not basolateral ErbB2 is activated at tyrosine 1139. This phosphotyrosine binds adaptor protein Grb2, as confirmed by immunoprecipitation. However, we found that it does not initiate the canonical Grb2-Ras-Raf-Erk pathway. Instead, our data supports the activation of a survival pathway via Bcl-2. The effects of ErbB2 over-expression were abrogated by the humanized anti-ErbB2 monoclonal antibody Herceptin added only from the apical side. The ability of apical ErbB2 to initiate an altered downstream cascade suggests that subcellular localization of the receptor plays an important role in regulating ErbB2 signaling in polarized epithelia.


Assuntos
Polaridade Celular , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais , Animais , Células CACO-2 , Linhagem Celular , Sobrevivência Celular , Cães , Expressão Gênica , Humanos , Transporte Proteico , Proteínas Proto-Oncogênicas c-bcl-2 , Receptor ErbB-2/farmacologia
10.
Clin Cancer Res ; 4(8): 2015-24, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9717833

RESUMO

Previous studies have characterized the reactivity of CD8+ CTLs with ovarian and breast cancer. There is little information about the antigens and epitopes recognized by CD4+ T cells in these patients. In this study, we analyzed the ability of T cells from peripheral blood mononuclear cells of breast cancer patients to recognize HER-2/neu (HER-2) peptides. We found that 13 of 18 patients responded by proliferation to at least one of the HER-2 peptides tested. Of these peptides, one designated G89 (HER-2: 777-789) was recognized by T cells from 10 patients. Seven of nine responding patients were HLA-DR4+, suggesting that this peptide is recognized preferentially in association with HLA-DR4. Analysis of the specificity and restriction of the cytokine responses to G89 by G89-stimulated T cells revealed that these cells secreted significantly higher levels of IFN-gamma than interleukin 4 and interleukin 10, suggesting priming for a Th0-T helper 1 response. The same pattern of cytokine responses was observed to the intracellular domain of HER-2 protein, suggesting that G89-stimulated T cells recognized epitopes of the HER-2 protein in association with HLA-DR4. Because HLA-DR4 is present in 25% of humans, characterization of MHC class II-restricted epitopes inducing Th0-T helper 1 responses may provide a basis for the development of multivalent HER-2-based vaccines against breast and ovarian cancer.


Assuntos
Neoplasias da Mama/sangue , Citocinas/biossíntese , Citocinas/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptor ErbB-2/farmacologia , Sequência de Aminoácidos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Interferon gama/biossíntese , Interferon gama/metabolismo , Dados de Sequência Molecular , Receptor ErbB-2/biossíntese , Homologia de Sequência de Aminoácidos , Estimulação Química , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
11.
Biochemistry ; 37(5): 1306-14, 1998 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-9477957

RESUMO

Membrane anchorage of Ras oncoproteins, required for transforming activity, depends on their carboxy-terminal farnesylcysteine. We previously showed that S-trans,trans-farnesylthiosalicylic acid (FTS), a synthetic farnesylcysteine mimetic, inhibits growth of ErbB2- and Ras-transformed cells, but not of v-Raf-transformed cells, suggesting that FTS interferes specifically with Ras functions. Here we demonstrate that FTS dislodges Ras from membranes of H-Ras-transformed (EJ) cells, facilitating its degradation and decreasing total cellular Ras. The dislodged Ras that was transiently present in the cytosol was degraded relatively rapidly, causing a decrease of up to 80% in total cellular Ras. The half-life of Ras was 10 +/- 4 h in FTS-treated EJ cells and 27 +/- 4 h in controls. The dislodgment of membrane Ras and decrease in total cellular Ras were dose-dependent: 50% of the effects occurred at 10-15 microM, comparable to concentrations (7-10 microM) required for 50% growth inhibition in EJ cells. Higher concentrations of FTS (25-50 microM) were required to dislodge Ras from Rat-1 cell membranes expressing normal Ras, suggesting some selectivity of FTS toward oncogenic Ras. Membrane localization of the prenylated G beta gamma of heterotrimeric G proteins was not affected by FTS in EJ cells. An FTS-related compound, N-acetyl-S-farnesyl-L-cysteine, which does not inhibit EJ cell growth, did not affect Ras. FTS did not inhibit growth of Rat-1 cells transformed by N-myristylated H-Ras and did not reduce the total amount of this Ras isoform. The results suggest that FTS affects docking of Ras in the cell membrane in a rather specific manner, rendering the protein susceptible to proteolytic degradation.


Assuntos
Proteínas ras/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Linhagem Celular Transformada , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacologia , Inibidores do Crescimento/farmacologia , Metilação/efeitos dos fármacos , Proteínas Oncogênicas v-raf , Ratos , Receptor ErbB-2/farmacologia , Proteínas Oncogênicas de Retroviridae/farmacologia , Salicilatos/farmacologia , Proteínas ras/antagonistas & inibidores , Proteínas ras/efeitos dos fármacos
12.
Eur J Immunol ; 27(5): 1115-23, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9174600

RESUMO

The protooncogene HER2/neu encodes a 185-kDa transmembrane protein with extensive homology to the epidermal growth factor receptor. It is overexpressed in several human cancers of epithelial origin, such as pancreatic cancer. Previously, we demonstrated that cytotoxic T lymphocytes (CTL) derived from breast, ovarian, and non-small cell lung cancer recognized a peptide derived from HER2/neu. To evaluate whether this HLA-A2-binding peptide is a tumor-associated antigen (TAA) in pancreatic cancer, the ability of HER2/neu-reactive CTL to lyse human pancreatic carcinoma cells was tested. CTL were generated from tumor-associated T lymphocytes from HLA-A2+ HER2/neu+ breast and ovarian cancer patients. All CTL recognized autologous and allogeneic HER2/ neu+ tumor cells in an HLA-A2-restricted fashion. Furthermore, all CTL recognized p654-662 (GP2) derived from HER2/neu. These CTL also recognized HER2/neu+ pancreatic cancer cells in an HLA-A2-restricted fashion. HER2/neu+ HLA-A2- pancreatic cancer were not or only poorly lysed. Repeated stimulation of HLA-A2+ PBL from pancreatic cancer patients using the HER2/neu-derived peptide resulted in specific recognition of this peptide and, more importantly, HER2/neu+ pancreatic tumors in an HLA-A2-restricted fashion. Autologous HLA-A2+ fibroblasts or HLA-A2+ malignant melanoma cells were not recognized. HLA-A2- peptide-stimulated T lymphocytes showed no significant cytotoxicity. These results demonstrate that this HER2/neu-derived peptide is a shared TAA among several adenocarcinomas including pancreatic carcinoma, suggesting a common mechanism of recognition of these human tumors by T lymphocytes. The identification of the HER2/neu-derived peptide GP2 as a TAA in pancreatic cancer provides an opportunity for the design of novel immunotherapy and vaccine strategies.


Assuntos
Adenocarcinoma/imunologia , Antígenos de Neoplasias/imunologia , Citotoxicidade Imunológica , Neoplasias Pancreáticas/imunologia , Fragmentos de Peptídeos/imunologia , Receptor ErbB-2/imunologia , Linfócitos T Citotóxicos/imunologia , Apresentação de Antígeno , Feminino , Antígeno HLA-A2/metabolismo , Humanos , Ativação Linfocitária , Fragmentos de Peptídeos/farmacologia , Receptor ErbB-2/farmacologia , Células Tumorais Cultivadas
13.
Pathobiology ; 65(2): 75-82, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9253031

RESUMO

The cultivation of cells from primary breast cancers is very unpredictable. The majority of breast-cancer-derived cell lines are of metastatic origin. To define the characteristics of tumor cells which govern their ability to grow in vitro as primary cultures as well as continuous or established culture cell lineages, human mammary epithelial cancer (HMEC) cells from 18 cases of unselected primary breast cancer were propagated in culture. Propagation of HMEC cells in vitro as monolayers in primary culture was successful in 10 out of 18 (55.5%) cases, which showed continous proliferation of tumor cells only up to 6-8 passages before they reached senescence. An investigation of the effects of phenotypic expression of estrogen receptors (ER), the progesterone receptors, c-erbB-2 oncoprotein and epidermal growth factor receptors (EGFR) on the capacity of HMEC cells to grow in vitro as monolayers showed that expression of ER and EGFR is required for controlling tumor proliferative activity in vitro. Expression of ER protein made the growth of HMEC cells more difficult, while expression of EGFR protein made their growth in vitro easier. Phenotypic characteristics of floating HMEC cells were found to be different from those grown on cover slips as adherent cultures, suggesting a selective growth of HMEC cells of a specific phenotype in culture. Cultured HMEC cells in subsequent passages showed a decrease in their proliferative capacity, alterations in phenotypic characteristics and development of morphologic features of terminal differentiation, resulting in senescence.


Assuntos
Neoplasias da Mama/patologia , Carcinoma/patologia , Receptores ErbB/fisiologia , Genes erbB-2/fisiologia , Receptor ErbB-2/fisiologia , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/fisiologia , Biomarcadores Tumorais/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Adesão Celular/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Vidro , Humanos , Fenótipo , Receptor ErbB-2/farmacologia , Células Tumorais Cultivadas
14.
Cancer Immunol Immunother ; 42(3): 179-84, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8640846

RESUMO

The neu proto-oncogene encodes a plasma membrane protein belonging to the epidermal growth factor receptor family. The cell line B104, derived from BDIX rat neuroblastoma, carries a point mutation in neu, and forms a tumor when injected into these rats. The human homologue of the neu oncogene (here called HER2) is overexpressed in certain types of cancer. Rats were immunized with HER2 protein (HER2) to investigate a possible cross-reaction between the homologous proteins which could protect them against subsequent inoculation with B104. Specific antibody in the serum was measured by cell-based enzyme-linked immunoabsorbent assay and fluorescence immunocytochemistry, and delayed-type hypersensitivity by an ear assay. Sera from animals immunized with the HER2 extracellular domain (HER2-ECD) reacted with both HER2- and neu-expressing cells. In the ear assay, a significant cellular response to both HER-ECD (P < 0.05) and neu protein (P < 0.001) was observed in HER2-ECD-immunized rats. However, the growth of B104 tumors in rats was not affected by preimmunization with HER2-ECD. The results indicate that an autoreactive immune response to neu was induced by immunization with HER2-ECD, but was too weak to affect the growth of the neu-bearing tumor.


Assuntos
Neuroblastoma/imunologia , Neuroblastoma/prevenção & controle , Mutação Puntual , Receptor ErbB-2/imunologia , Receptor ErbB-2/farmacologia , Células 3T3 , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Feminino , Genes erbB-2 , Humanos , Hipersensibilidade Tardia/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Imunização , Masculino , Camundongos , Neuroblastoma/genética , Proto-Oncogene Mas , Ratos , Ratos Endogâmicos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
15.
J Immunother Emphasis Tumor Immunol ; 18(4): 197-209, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8680648

RESUMO

The HER-2/neu protooncogene (HER-2) is overexpressed in a significant number of breast and ovarian tumors. Peptides of HER-2 sequence were recently found to reconstitute recognition of cytotoxic T lymphocytes (CTLs) from tumor-associated (TALs) and tumor-infiltrating (TILs) lymphocytes, indicating that they reconstitute natural epitopes recognized by CTLs on HLA-A2+ tumors. Because HER-2 is an important antigen (Ag) for tumor-specific CTL induction and the immunogenicity of peptides for CTL induction is dependent on their presentation as stable complexes with HLA-A2, we identified peptides of high and low stabilizing activity from the sequence of HER-2 and the folate-binding protein (FBP). Distinct sequence patterns in the region positions (P)3-P5 and P1 were found for peptides with high (HSA) and low (LSA) stabilizing ability. A low-HLA-A2-affinity HER-2 peptide, P1 of the CTL epitope, was found to be permissive to substitutions that enhanced HLA-A2-stabilizing ability and conserved CTL recognition. In contrast, the region P3-P5 was not permissive to sequence changes. We conclude that the selective permissivity of P1 and P9 in the tumor epitope sequence may have important implications for optimization of tumor Ag presentation, and "neoantigenicity" of self-antigens, aiming toward induction of tumor-reactive CTLs of defined affinity and specificity for target Ags.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Epitopos/química , Antígeno HLA-A2/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Conformação Proteica/efeitos dos fármacos , Receptor ErbB-2/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Regulação para Cima/imunologia , Sequência de Aminoácidos , Anticorpos Monoclonais/química , Citotoxicidade Imunológica/efeitos dos fármacos , Epitopos/efeitos dos fármacos , Epitopos/imunologia , Antígeno HLA-A2/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/imunologia , Oligopeptídeos/farmacologia , Receptor ErbB-2/química , Receptor ErbB-2/imunologia , Linfócitos T Citotóxicos/imunologia , Regulação para Cima/efeitos dos fármacos
16.
Cancer Res ; 55(5): 1099-104, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7532543

RESUMO

Dendritic antigen-presenting cells are considered to be the most effective stimulators of T cell immunity. The use of dendritic cells has been proposed to generate therapeutic T cell responses to tumor antigens in cancer patients. One limitation is that the number of dendritic cells in peripheral blood is exceedingly low. Dendritic cells originate from CD34+ hematopoietic progenitor cells (HPC) which are present in the bone marrow and in small numbers in peripheral blood. CD34+ HPC can be mobilized into the peripheral blood by in vivo administration of granulocyte-colony-stimulating factor. The aim of the current study was to determine whether functional dendritic cells could be elicited and grown in vitro from CD34+ HPC derived from bone marrow or granulocyte-colony-stimulating factor-mobilized peripheral blood. Culture of CD34+ HPC with granulocyte-macrophage-colony-stimulating factor and tumor necrosis factor alpha yielded a heterogeneous cell population containing cells with typical dendritic morphology. Phenotypic studies demonstrated a loss of the CD34 molecule over 1 week and an increase in cells expressing surface markers associated with dendritic cells, CD1a, CD80 (B7/BB1), CD4, CD14, HLA-DR, and CD64 (Fc gamma RI). Function was validated in experiments showing that cultured cells could stimulate proliferation of allogeneic CD4+ and CD8+ T lymphocytes. Antigen-presenting capacity was further confirmed in experiments showing that cultured cells could effectively stimulate tetanus toxoid-specific responses and HER-2/neu peptide-specific responses. The derivation and expansion of dendritic cells from cultured bone marrow or granulocyte-colony-stimulating factor-mobilized CD34+ HPC may provide adequate numbers for testing of dendritic cells in clinical studies, such as vaccine and T cell therapy trials.


Assuntos
Antígenos CD/análise , Células da Medula Óssea , Medula Óssea/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Leucócitos Mononucleares/citologia , Linfócitos T/imunologia , Apresentação de Antígeno , Antígenos CD34 , Antígeno B7-1/farmacologia , Medula Óssea/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Ativação Linfocitária/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Fragmentos de Peptídeos/farmacologia , Fenótipo , Receptor ErbB-2/farmacologia , Estimulação Química , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...