Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 121(9): 1563-73, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23293080

RESUMO

Receptors for the Fc portion of IgG (FcγRs) are mandatory for the induction of various IgG-dependent models of autoimmunity, inflammation, anaphylaxis, and cancer immunotherapy. A few FcγRs have the ability to bind monomeric IgG: high-affinity mouse mFcγRI, mFcγRIV, and human hFcγRI. All others bind IgG only when aggregated in complexes or bound to cells or surfaces: low-affinity mouse mFcγRIIB and mFcγRIII and human hFcγRIIA/B/C and hFcγRIIIA/B. Although it has been proposed that high-affinity FcγRs are occupied by circulating IgG, multiple roles for mFcγRI and mFcγRIV have been reported in vivo. However, the potential roles of hFcγRI that is expressed on monocytes, macrophages, and neutrophils have not been reported. In the present study, we therefore investigated the role of hFcγRI in antibody-mediated models of disease and therapy by generating hFcγRI-transgenic mice deficient for multiple endogenous FcRs. hFcγRI was sufficient to trigger autoimmune arthritis and thrombocytopenia, immune complex-induced airway inflammation, and active and passive systemic anaphylaxis. We found monocyte/macrophages to be responsible for thrombocytopenia, neutrophils to be responsible for systemic anaphylaxis, and both cell types to be responsible for arthritis induction. Finally, hFcγRI was capable of mediating antibody-induced immunotherapy of metastatic melanoma. Our results unravel novel capabilities of human FcγRI that confirm the role of high-affinity IgG receptors in vivo.


Assuntos
Anafilaxia/genética , Imunoglobulina G/fisiologia , Imunoterapia , Inflamação/genética , Neoplasias/terapia , Receptores de IgG/fisiologia , Anafilaxia/imunologia , Animais , Antineoplásicos/farmacologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/farmacologia , Imunoterapia/métodos , Inflamação/induzido quimicamente , Inflamação/imunologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Oncol ; 34(1): 25-32, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19082474

RESUMO

Human carcinomas frequently express one or more members of the epidermal growth factor receptor family. Two family members, epidermal growth factor receptor (EGFR) and c-erbB2/neu (HER2), homodimerize or heterodimerize upon activation with ligand and trigger potent mechanisms of cellular proliferation, differentiation and migration. In this study, we examined the effect of the anti-EGFR monoclonal antibody Erbitux (cetuximab) on human tumor cells expressing both EGFR and HER2. Investigation of the effect of cetuximab on the activation of EGFR-EGFR, EGFR-HER2 and HER2-HER2 homodimers and heterodimers was conducted using the NCI-N87 human gastric carcinoma cell line. Treatment of NCI-N87 cells with cetuximab completely inhibited formation of EGFR-EGFR homodimers and EGFR-HER2 heterodimers. Activation of HER2-HER2 homodimers was not appreciably stimulated by exogenous ligand and was not inhibited by cetuximab treatment. Furthermore, cetuximab inhibited EGF-induced EGFR and HER2 phosphorylation in CAL27, NCI-H226 and NCI-N87 cells. The activation of downstream signaling molecules such as AKT, MAPK and STAT-3 were also inhibited by cetuximab in these cells. To examine the effect of cetuximab on the growth of tumors in vivo, athymic mice bearing established NCI-N87 or CAL27 xenografts were treated with cetuximab (1 mg, i.p., q3d). The growth of NCI-N87 and CAL27 tumors was significantly inhibited with cetuximab therapy compared to the control groups (p<0.0001 in both cases). In the CAL27 xenograft model, tumor growth inhibition by cetuximab treatment was similar to that by cetuximab and trastuzumab combination treatment. Immunohistological analysis of cetuximab-treated tumors showed a decrease in EGFR-HER2 signaling and reduced tumor cell proliferation. These results suggest that cetuximab may be useful in the treatment of carcinomas co-expressing EGFR and HER2.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Dimerização , Receptores ErbB/imunologia , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cetuximab , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Receptor ErbB-2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/metabolismo , Transplante Heterólogo , Ensaio Tumoral de Célula-Tronco
3.
MAbs ; 11(6): 1139-1148, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31242068

RESUMO

Lymphocyte activation gene 3 (LAG-3) is expressed on activated T cells, natural killer cells or B cells, and functions to negatively regulate homeostasis of these cells. Anti-LAG-3 antibodies might be useful for antitumor immunotherapy. In this study, we characterized a novel anti-LAG-3 antibody, LBL-007, which was isolated from a human antibody phage display library. LBL-007 was found to specifically bind to human LAG-3 antigen, but not to human CD4 or mouse LAG-3. LBL-007 bound activated T cells and promoted interleukin-2 secretion. LBL-007 internalization efficacy by endocytosis into different cells was better than that of another anti-LAG-3 antibody, relatlimab analog. Moreover, LBL-007 was able to block LAG-3 binding to MHC class II molecules and liver sinusoidal endothelial cell lectin, and block LAG-3-induced downstream signaling. In mice transplanted with colorectal cancer cells, treatment with either anti-PD-1 antibody or LBL-007 (10 mg/kg per mouse twice a week for three weeks) resulted in a significant delay in tumor growth compared with control IgG treatment, and their combination was even more effective. Serum LBL-007 levels were highly stable in monkeys after a single intravenous injection of LBL-007 at 3, 10, or 30 mg/kg. This study demonstrated that the combination of LBL-007 with an anti-PD-1 antibody is a promising antitumor regimen for immunotherapy of solid tumors in future that deserves further study.


Assuntos
Anticorpos Monoclonais Murinos , Antígenos CD/imunologia , Antineoplásicos Imunológicos , Neoplasias Colorretais , Imunoterapia , Proteínas de Neoplasias/imunologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Feminino , Células HEK293 , Haplorrinos , Humanos , Camundongos , Camundongos Transgênicos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína do Gene 3 de Ativação de Linfócitos
4.
Anticancer Res ; 28(5A): 2679-86, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19035294

RESUMO

Targeted immunotherapy against tumors or angiogenesis has shown promise as an alternative approach for the treatment of malignant disease. Whether or not combining these two treatment modalities would enhance the antitumor effect was tested in mouse models of malignant melanoma. C57BL/6 mice bearing established subcutaneous B16 tumors were treated with anti-vascular endothelial growth factor receptor (anti-VEGFR) fetal liver kinase-1 (Flk-1) monoclonal antibody (mAb) DC101 and/or anti-TYRP-1/gp75 (tyrosinase-related protein-1) mAb TA99. The growth of subcutaneous B16 tumors was significantly suppressed by the mAb DC101 (63%, p<0.001) and by mAb TA99 (75%, p<0.001) treatment alone. The combined antibody (TA99+DC101) treatment resulted in a significant enhancement (93%, p<0.001) of tumor growth suppression. In a B16 pulmonary metastasis model, combined therapy with mAb DC101 and mAb TA99 resulted in a significant reduction of lung metastases compared to the control (p<0.001) and the single agent treatment groups (p<0.05). A combined modality approach that provides passive immunity to melanoma differentiation antigens as well as inhibiting tumor neovascularization may be valuable for the treatment of malignant melanoma.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Melanoma Experimental/terapia , Glicoproteínas de Membrana/imunologia , Oxirredutases/imunologia , Receptores de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica
5.
Anticancer Res ; 27(5A): 3355-66, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17970081

RESUMO

BACKGROUND: The epidermal growth factor receptor (EGFR) plays an important role in the growth and survival of many human tumors of epithelial origin. EGFR variant III (EGFRvIII) is a truncated form of EGFR that does not bind ligand, is constitutively active, and is reported to be coexpressed with EGFR in some human tumors including breast, glioblastoma, lung, and prostate. MATERIALS AND METHODS: Here we have tested the anti-EGFR monoclonal antibody cetuximab for its interaction with EGFRvIII. Chinese hamster ovary (CHO), 32D (non-tumorigenic murine hematopoetic cells), and U87-MG stable transfectants were generated to express EGFRvIII. RESULTS: Analysis of receptor phosphorylation showed that the EGFRvIII was constitutively phosphorylated in transfected cells. Flow cytometry, direct binding, and immunoprecipitation analysis of EGFRvIII transfectants showed specific binding of cetuximab to EGFRvIII. Cetuximab bound to EGFRvIII with a KD of 0.38 nM determined by Scatchard analysis and 1.1 nM determined by Biacore analysis respectively. In internalization studies, binding of cetuximab to the EGFRvIII on the cell surface led to at least 50% of the cetuximab-EGFRvIII complex internalized from cell surface of CHO-EGFRvIII after 3 hours. This internalization led to a reduction in phosphorylated EGFRvIII in transfected cells. Furthermore, incubation of cells expressing EGFRvIII with cetuximab resulted in 40-50% inhibition of cell proliferation. CONCLUSION: These data suggest that cetuximab may be a potential candidate for the treatment of tumors that also express EGFRvIII.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Receptores ErbB/metabolismo , Animais , Anticorpos Monoclonais Humanizados , Células CHO , Linhagem Celular Tumoral , Cetuximab , Cricetinae , Cricetulus , Regulação para Baixo , Fator de Crescimento Epidérmico/antagonistas & inibidores , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Citometria de Fluxo , Humanos , Imunoprecipitação , Fosforilação , Transdução de Sinais , Especificidade por Substrato , Transfecção
6.
Hum Antibodies ; 16(3-4): 127-36, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18334748

RESUMO

TYRP1 (tyrosinase-related protein 1) is a melanoma antigen expressed in melanosomes and on the surface of melanoma cells. Previous studies have shown that mouse antibodies to TYRP1 localized to melanomas in vivo and inhibited tumor growth and metastasis. Here, we describe the characterization of a novel fully human anti-TYRP1 MAb (20D7) generated by immunizing HuMAb mice (Medarex). 20D7 recognized recombinant and native human TYRP1 by Western blotting and ELISA, and native TYRP1 in melanoma cells as determined by flow cytometry analysis. 20D7 cross-reacted with mouse TYRP1. The binding affinity to human TYRP1 for the human MAb was in the low nM range as determined by surface plasmon resonance kinetics. 20D7 can bind to human and mouse Fc receptor and induce a strong ADCC response against human melanoma cells in vitro. The antitumor activity of 20D7 was tested in human melanoma xenografts and mouse metastatic melanoma models in athymic nude mice. Growth of s.c. human melanoma tumors and metastatic nodules of murine B16 tumor were significantly suppressed by 20D7 compared to human IgG control. These results suggest that human anti-TYRP1 MAb may be a potent therapeutic for the treatment of malignant melanoma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Melanoma Experimental/terapia , Glicoproteínas de Membrana/imunologia , Oxirredutases/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Linhagem Celular Tumoral , Proteínas do Sistema Complemento/imunologia , Feminino , Humanos , Camundongos
7.
Cancer Res ; 62(18): 5144-7, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12234976

RESUMO

CD4(+) T cells can recognize "self" tumor antigens, but the impact of tumor cell expression of self-antigens on CD4(+) T-cell function in humans is unknown. Here, we identify a new epitope (ISPNSVFSQWRVVCDSLEDYD) derived from tyrosinase-related protein-1 (TRP-1) using a predictive algorithm and mice transgenic for a chimeric HLA-DRB1*0401 molecule. We then compared the functions of TRP-1-epitope-specific, CD4(+) T-cell responses in normal healthy individuals to those found in patients with metastatic malignant melanoma. Surprisingly, we found that tumor-bearing patients had significantly higher levels of TRP-1-specific, CD4(+) T-cell function than healthy volunteers as measured ex vivo. Thus, the net effect of "self" antigen expression by tumor cells was the enhancement of tumor antigen-specific CD4(+) T-cell function, rather than immunosuppression. These findings indicate that antigens expressed by malignant melanoma cells can partially activate CD4(+) T lymphocytes.


Assuntos
Antígenos de Neoplasias/imunologia , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Melanoma/imunologia , Glicoproteínas de Membrana , Oxirredutases , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/biossíntese , Autoantígenos/biossíntese , Antígenos HLA-DR/imunologia , Cadeias HLA-DRB1 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas/imunologia
8.
Hum Antibodies ; 13(3): 81-90, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15598988

RESUMO

Recombinant protein production in plants such as corn is a promising means to generate high product yields at low comparable production cost. The anti-EGFR monoclonal antibody C225, cetuximab, is a well-characterized receptor antagonist antibody recently approved for the treatment of refractory colorectal cancer. We initiated a study to test and compare the functional activity of glycosylated and aglycosylated C225 produced in stable transgenic corn seed. Both corn antibodies were shown to be functionally indistinguishable from mammalian-derived C225 in demonstrating high-affinity binding to the EGF receptor, blocking of ligand-dependent signaling, and inhibiting cell proliferation. In addition, consistent with cetuximab, both corn antibodies possessed strong anti-tumor activity in vivo. Acute dose primate pharmacokinetic studies, however, revealed a marked increase in clearance for the glycosylated corn antibody, while the aglycosylated antibody possessed in vivo kinetics similar to cetuximab. This experimentation established that corn-derived receptor blocking monoclonal antibodies possess comparable efficacy to mammalian cell culture-derived antibody, and offer a cost effective alternative to large-scale mammalian cell culture production.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/farmacologia , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Zea mays/genética , Zea mays/imunologia , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/farmacocinética , Cetuximab , Feminino , Humanos , Técnicas In Vitro , Cinética , Macaca fascicularis , Masculino , Camundongos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Plantas Geneticamente Modificadas , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Transplante Heterólogo
9.
Hum Antibodies ; 19(4): 89-99, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21178280

RESUMO

PURPOSE: To evaluate the antibody-dependent cellular cytotoxicity (ADCC) of cetuximab, an anti-epidermal growth factor receptor (EGFR) IgG1 antibody, in vitro. METHODS: Binding to human Fc receptors was measured by ELISA. ADCC against a panel of tumor cell lines was evaluated using peripheral blood mononuclear cells or NK cells as effectors and lactate dehydrogenase release as a marker of cell killing. Cetuximab was compared with two glycan variants of cetuximab and with panitumumab, an anti-EGFR IgG2. RESULTS: Cetuximab bound with high affinity to FcγRI (EC50 = 0.13 nM) and FcγRIIIa (EC50 = 6 nM) and effectively induced ADCC across multiple tumor cell lines. Panitumumab and aglycosylated cetuximab did not bind to FcγRI or FcγRIIIa nor have ADCC activity even at high effector-target cell ratios, even though the EGFR-binding affinity of cetuximab and panitumumab were shown to be comparable (KD = 87 pM and 83 pM, respectively). The extent of cetuximab-elicited ADCC was associated with the level of EGFR expression on tumor cells. CONCLUSIONS: Cetuximab elicits effective ADCC activity against a wide range of tumor cells in vitro. This activity is dependent on antibody glycosylation and IgG1 isotype as well as tumor-cell EGFR expression. These findings suggest that ADCC may contribute to the antitumor activity of cetuximab.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/imunologia , Receptores ErbB/metabolismo , Imunoglobulina G/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cetuximab , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/imunologia , Glicosilação , Humanos , Isotipos de Imunoglobulinas , Células Matadoras Naturais/imunologia , L-Lactato Desidrogenase/metabolismo , Leucócitos Mononucleares/imunologia , Panitumumabe
10.
J Biol Chem ; 280(20): 19665-72, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15757893

RESUMO

Both the epidermal growth factor receptor (EGFR) and the insulin-like growth factor receptor (IGFR) have been implicated in the tumorigenesis of a variety of cancers. Here we propose that simultaneous targeting of both receptors with a bispecific antibody would lead to enhanced antitumor activity. To this end, we produced a recombinant human IgG-like bispecific antibody, a Di-diabody, using the variable regions from two antagonistic antibodies: IMC-11F8 to EGFR and IMC-A12 to IGFR. The Di-diabody binds to both EGFR and IGFR and effectively blocked both EGF- and IGF-stimulated receptor activation and tumor cell proliferation. The Di-diabody also inherited the biological properties from both of its parent antibodies; it triggers rapid and significant IGFR internalization and degradation and mediates effective antibody-dependent cellular cytotoxicity in a variety of tumor cells. Finally, the Di-diabody strongly inhibited the growth of two different human tumor xenografts in vivo. Our results underscore the benefits of simultaneous targeting of two tumor targets with bispecific antibodies.


Assuntos
Anticorpos Biespecíficos/biossíntese , Anticorpos Biespecíficos/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/imunologia , Receptores de Somatomedina/imunologia , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/genética , Antineoplásicos/química , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/química , Imunoglobulina G/genética , Imunoglobulina G/farmacologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa