Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 9(7): e103776, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25080123

RESUMO

BACKGROUND: CC chemokine receptor 4 (CCR4) represents a potentially important target for cancer immunotherapy due to its expression on tumor infiltrating immune cells including regulatory T cells (Tregs) and on tumor cells in several cancer types and its role in metastasis. METHODOLOGY: Using phage display, human antibody library, affinity maturation and a cell-based antibody selection strategy, the antibody variants against human CCR4 were generated. These antibodies effectively competed with ligand binding, were able to block ligand-induced signaling and cell migration, and demonstrated efficient killing of CCR4-positive tumor cells via ADCC and phagocytosis. In a mouse model of human T-cell lymphoma, significant survival benefit was demonstrated for animals treated with the newly selected anti-CCR4 antibodies. SIGNIFICANCE: For the first time, successful generation of anti- G-protein coupled chemokine receptor (GPCR) antibodies using human non-immune library and phage display on GPCR-expressing cells was demonstrated. The generated anti-CCR4 antibodies possess a dual mode of action (inhibition of ligand-induced signaling and antibody-directed tumor cell killing). The data demonstrate that the anti-tumor activity in vivo is mediated, at least in part, through Fc-receptor dependent effector mechanisms, such as ADCC and phagocytosis. Anti-CC chemokine receptor 4 antibodies inhibiting receptor signaling have potential as immunomodulatory antibodies for cancer.


Assuntos
Antineoplásicos/farmacologia , Quimiotaxia/efeitos dos fármacos , Receptores CCR4/antagonistas & inibidores , Anticorpos de Cadeia Única/farmacologia , Animais , Antineoplásicos/uso terapêutico , Sinalização do Cálcio , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CCL17/fisiologia , Células HEK293 , Humanos , Concentração Inibidora 50 , Macaca mulatta , Masculino , Camundongos Nus , Biblioteca de Peptídeos , Agregação Plaquetária/efeitos dos fármacos , Receptores CCR4/imunologia , Anticorpos de Cadeia Única/uso terapêutico , Especificidade da Espécie , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
MAbs ; 6(3): 728-39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24670809

RESUMO

To improve recruitment and activation of natural killer (NK) cells to lyse tumor cells, we isolated a human anti-CD16A antibody with similar affinity for the CD16A 158F/V allotypes, but no binding to the CD16B isoform. Using CD16A-targeting Fv domains, we constructed a tetravalent bispecific CD30/CD16A tandem diabody (TandAb®) consisting solely of Fv domains. This TandAb has two binding sites for CD16A and two for CD30, the antigen identifying Hodgkin lymphoma cells. The binding and cytotoxicity of the TandAb were compared with antibodies with identical anti-CD30 domains: (1) a native IgG, (2) an IgG optimized for binding to Fc receptors, and (3) a bivalent bispecific CD30/CD16A diabody. Due to its CD16A-bivalency and reduced koff, the TandAb was retained longer on the surface of NK cells than the IgGs or the diabody. This contributed to the higher potency and efficacy of the TandAb relative to those of the other anti-CD30 antibodies. TandAb cytotoxicity was independent of the CD16A allotype, whereas the anti-CD30 IgGs were substantially less cytotoxic when NK cells with low affinity CD16A allotype were employed. TandAb activation of NK cells was strictly dependent on the presence of CD30(+) target cells. Therefore, the CD30/CD16A TandAb may represent a promising therapeutic for the treatment of Hodgkin's lymphoma; further, anti-CD16A TandAbs may function as potent immunotherapeutics that specifically recruit NK cells to destroy cancer cells.


Assuntos
Anticorpos Biespecíficos/imunologia , Doença de Hodgkin/imunologia , Antígeno Ki-1/imunologia , Células Matadoras Naturais/imunologia , Receptores de IgG/imunologia , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/uso terapêutico , Especificidade de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Sítios de Ligação de Anticorpos , Linhagem Celular Tumoral , Doença de Hodgkin/terapia , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Imunoterapia , Biblioteca de Peptídeos
3.
MAbs ; 6(4): 1038-50, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24525727

RESUMO

The epithelial cell adhesion molecule (EpCAM) is expressed by a wide range of human carcinomas, making it an attractive diagnostic and therapeutic target in oncology. Its recent identification on cancer stem cells has raised further interest in its use for tumor targeting and therapy. Here, we present the characterization and therapeutic potential of 3-17I, a novel human EpCAM-targeting monoclonal antibody. Strong reaction of 3-17I was observed in all lung, colon, and breast human tumor biopsies evaluated. By flow cytometry and confocal fluorescence microscopy, we demonstrate that 3-17I specifically targets EpCAM-positive cell lines. We also show evidence for mAb-sequestration in endo-/lysosomes, suggesting internalization of 3-17I by receptor-mediated endocytosis. The ribosomal-inactivating toxin saporin was linked to 3-17I, creating the per se non-toxic immunotoxin 3-17I-saporin, a promising candidate for the drug delivery technology photochemical internalization (PCI). PCI is based on a light-controlled destruction of endolysosomal membranes and subsequent cytosolic release of the sequestered payload upon light exposure. EpCAM-positive human cancer cell lines MCF7 (breast), BxPC-3 (pancreas), WiDr (colon), and the EpCAM-negative COLO320DM (colon), were treated with 3-17I-saporin in combination with the clinically relevant photosensitizer TPCS2a (Amphinex), followed by exposure to light. No cytotoxicity was observed after treatment with 3-17I-saporin without light exposure. However, cell viability, proliferation and colony-forming capacity was strongly reduced in a light-dependent manner after PCI of 3-17I. Our results show that 3-17I is an excellent candidate for diagnosis of EpCAM-positive tumors and for development of clinically relevant antibody-drug conjugates, using PCI for the treatment of localized tumors.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Imunotoxinas/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antineoplásicos/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Imunotoxinas/imunologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Fotoquímica/métodos , Proteínas Inativadoras de Ribossomos Tipo 1/imunologia , Saporinas
4.
J Mol Biol ; 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22985968

RESUMO

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...