Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cancer Sci ; 106(1): 102-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25421609

RESUMO

Malignant pleural mesothelioma (MPM) is a rare and highly aggressive neoplasm that arises from the pleural, pericardial, or peritoneal lining. Although surgery, chemotherapy, radiotherapy, and combinations of these therapies are used to treat MPM, the median survival of such patients is dismal. Therefore, there is a compelling need to develop novel therapeutics with different modes of action. Ganglioside GM2 is a glycolipid that has been shown to be overexpressed in various types of cancer. However, there are no published reports regarding the use of GM2 as a potential therapeutic target in cases of MPM. In this study, we evaluated the efficacy of the anti-GM2 antibody BIW-8962 as an anti-MPM therapeutic using in vitro and in vivo assays. Consequently, the GM2 expression in the MPM cell lines was confirmed using flow cytometry. In addition, eight of 11 cell lines were GM2-positive (73%), although the GM2 expression was variable. BIW-8962 showed a significant antibody-dependent cellular cytotoxicity activity against the GM2-expressing MPM cell line MSTO-211H, the effect of which depended on the antibody concentration and effector/target ratio. In an in vivo orthotropic mouse model using MSTO-211H cells, BIW-8962 significantly decreased the incidence and size of tumors. Additionally, the GM2 expression was confirmed in the MPM clinical specimens. Fifty-eight percent of the MPM tumors were positive for GM2, with individual variation in the intensity and frequency of staining. These data suggest that anti-GM2 antibodies may become a therapeutic option for MPM patients.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Gangliosídeo G(M2)/imunologia , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Neoplasias Pleurais/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Gangliosídeo G(M2)/metabolismo , Humanos , Masculino , Mesotelioma Maligno , Camundongos SCID , Pessoa de Meia-Idade , Engenharia de Proteínas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Biochem Biophys Res Commun ; 458(4): 877-82, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25701778

RESUMO

TROP-2 is a type Ⅰ transmembrane glycoprotein that is highly expressed in various epithelial cancer cells, and its increased expression correlates with poor prognosis. Although several anti-TROP-2 antibodies have been described, they were found unsuitable for antitumor therapy use in vivo as naked antibodies. In this study, we established a novel anti-TROP-2 antibody, designated Pr1E11, from mice immunized with primary prostate cancer cells. Antibody screening was based on the infection activity of Adv-LacZ-FZ33, which displays an immunoglobulin G binding domain in the adenoviral fiber protein. We found that Pr1E11 specifically binds to TROP-2 with high affinity and recognizes diverse epithelial cancer cell lines and primary pancreatic cancer tissues. Epitope analysis using TROP-2 deletion mutants revealed that binding site of Pr1E11 is a cysteine-rich domain, a unique epitope compared with other available anti-TROP-2 antibodies. In addition, Pr1E11 exhibited low internalization activity, which may make it suitable for naked antibody therapeutics. Our results suggest that Pr1E11 may stimulate different biological activities from other anti-TROP-2 antibodies based on its unique binding epitope, and is a potential candidate for naked antibody therapeutics for various epithelial cancer treatments.


Assuntos
Adenoviridae/imunologia , Antígenos de Neoplasias/imunologia , Moléculas de Adesão Celular/imunologia , Epitopos/imunologia , Neoplasias da Próstata/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Humanos , Masculino , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Próstata/imunologia , Deleção de Sequência , Células Tumorais Cultivadas
3.
Biosci Biotechnol Biochem ; 79(8): 1272-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25744656

RESUMO

Human claudin-3 (CLDN3) is a tetraspanin transmembrane protein of tight junction structures and is known to be over-expressed in some malignant tumors. Although a specific monoclonal antibody (MAb) against the extracellular domains of CLDN3 would be a valuable tool, generation of such MAbs has been regarded as difficult using traditional hybridoma techniques, because of the conserved sequence homology of CLDN3s among various species. In addition, high sequence similarity is shared among claudin family members, and potential cross-reactivity of MAb should be evaluated carefully. To overcome these difficulties, we generated CLDN3-expressing Chinese hamster ovary and Sf9 cells to use an immunogens and performed cell-based screening to eliminate cross-reactive antibodies. As a result, we generated MAbs that recognized the extracellular loops of CLDN3 but not those of CLDN4, 5, 6, or 9. Further in vitro studies suggested that the isolated MAbs possessed the desired binding properties for the detection or targeting of CLDN3.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Claudina-3/imunologia , Estrutura Terciária de Proteína , Animais , Anticorpos Monoclonais/química , Células CHO , Claudina-3/química , Cricetinae , Cricetulus , Humanos , Camundongos
4.
Biochem Biophys Res Commun ; 394(3): 498-502, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20197059

RESUMO

LGR5 is an orphan G-protein-coupled receptor (GPCR) that is expressed on the cell surface membrane. LGR5 is reported to be overexpressed in colon, liver, and ovary tumor compared to normal tissue. However, a specific ligand for LGR5 has not yet been determined, and the function is still not clear. An LGR5-specific monoclonal antibody (mAb) is needed as a tool for detection and analysis of LGR5 biological function and cancer therapy. To date, no mAb against LGR5 that retains high affinity and specificity has been reported. Here, we report successful establishment and characterization of a mAb (KM4056) that specifically recognizes the extracellular N-terminal domain of human LGR5, but not LGR4 or LGR6. This mAb has potent complement-dependent cytotoxicity (CDC) activity in vitro and shows strong anti-tumor activity in vivo against xenograft model by transplanting LGR5 expressing CHO transfectants into SCID mice. Thus, KM4056 can be a useful tool for detection of LGR5 positive cells and analysis of LGR5 biological function.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Imunológica , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/análise , Animais , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos SCID , Estrutura Terciária de Proteína , Receptores Acoplados a Proteínas G/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Sci ; 100(9): 1623-30, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19555390

RESUMO

Claudin-4 (CLDN4) is a tetraspanin transmembrane protein of tight junction structure and is highly expressed in pancreatic and ovarian cancers. In this study, we aimed to generate an anti-Claudin-4 monoclonal antibody (mAb) and evaluate its antitumor efficacy in vitro and in vivo. To isolate specific mAb, we generated CLDN3, 4, 5, 6, and 9, expressing Chinese hamster ovary (CHO) cells, and then used them as positive and negative targets through cell-based screening. As a result, we succeeded in isolating KM3900 (IgG2a), which specifically bound to CLDN4, from BXSB mice immunized with pancreatic cancer cells. Immunoprecipitation and flow cytometry analysis revealed that KM3900 recognized the conformational structure and bound to extracellular loop 2 of CLDN4. Furthermore, binding of KM3900 was detected on CLDN4-expressing pancreatic and ovarian cancer cells, but not on negative cells. Next, we made the mouse-human chimeric IgG1 (KM3934) and evaluated its antitumor efficacy. KM3934 induced dose-dependent antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro, and significantly inhibited tumor growth in MCAS or CFPAC-1 xenograft SCID mice in vivo (P < 0.05). These results suggest that mAb therapy against CLDN4 is promising for pancreatic and ovarian cancers.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Proteínas do Sistema Complemento/imunologia , Citotoxicidade Imunológica/imunologia , Proteínas de Membrana/imunologia , Neoplasias Ovarianas/terapia , Neoplasias Pancreáticas/terapia , Animais , Western Blotting , Células CHO , Células Cultivadas , Claudina-4 , Cricetinae , Cricetulus , Feminino , Citometria de Fluxo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoprecipitação , Camundongos , Camundongos SCID , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Hum Antibodies ; 18(1-2): 17-27, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19478395

RESUMO

Human interleukin-5 is the key cytokine involved in regulating the production and function of human eosinophils. IL-5 binds to its specific receptor composed of two heterogeneous alpha and beta polypeptide chains (hIL-5Ralpha and betac) that are expressed on the cell surface. The hIL-5Ralpha specifically binds IL-5 without involvement of the betac. It has been suggested that neutralizing antibodies to hIL-5Ralpha could serve as a therapeutic agent in eosinophil-associated diseases. We describe here the creation and biologic activities of a mouse monoclonal antibody against hIL-5Ralpha that blocks the following IL-5 dependent activities (a) binding of the IL-5 ligand to its receptor, (b) IL-5 dependent growth of hIL-5R expressing cells, and (c) IL-5-induced adhesion of human eosinophils. We also describe the process for humanization of the mouse Mab towards development of a therapeutic MAb. The humanized version of the monoclonal antibody also displayed potent neutralizing activity against IL-5 dependent activities.


Assuntos
Anticorpos Monoclonais/biossíntese , Subunidade alfa de Receptor de Interleucina-5/antagonistas & inibidores , Subunidade alfa de Receptor de Interleucina-5/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Eosinófilos/imunologia , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Engenharia de Proteínas , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/imunologia
7.
Mol Immunol ; 44(12): 3122-31, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17379311

RESUMO

Antibody-dependent cellular cytotoxicity (ADCC) is considered to be an important therapeutic function for clinical efficacy of monoclonal antibodies. Recent studies have revealed two methods to increase binding affinity for FcgammaRIIIa and enhance ADCC more efficiently for antibodies: (i) fucose removal from antibody N-linked complex oligosaccharides and (ii) amino acid mutations in the antibody Fc region. In this study, we compare the biological activities of the methods of generating high ADCC antibodies. We used a fucose-negative antibody and two antibodies with sets of mutations, demonstrated previously to optimally enhance ADCC using the chimeric anti-CD20 antibody, rituximab, as the model. Both amino acid mutant antibodies showed a significantly higher affinity for recombinant FcgammaRIIIa than fucose-negative antibody when compared using biosensor analysis. The removal of fucose from the antibodies bearing amino acid mutations exhibited a further enhancement of binding to recombinant FcgammaRIIIa and significantly increased binding to natural killer (NK) cells. Despite the differences manifested in binding for the FcgammaR, ADCCs were indistinguishable between methods and even when the methods were combined. These results indicate that the affinity of binding to FcgammaRIIIa does not predict ADCC beyond a certain threshold and that each method alone is sufficient to induce maximal ADCC of the antibody.


Assuntos
Anticorpos/metabolismo , Citotoxicidade Celular Dependente de Anticorpos , Receptores de IgG/metabolismo , Anticorpos/química , Anticorpos/genética , Fucose , Humanos , Células Matadoras Naturais/metabolismo , Métodos , Mutação , Ligação Proteica
8.
Transl Oncol ; 10(5): 707-718, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28710915

RESUMO

Many ovarian cancer patients often show peritoneal metastasis with malignant ascites. However, unmet medical needs remain regarding controlling these symptoms after tumors become resistant to chemotherapies. We developed KHK2805, a novel anti-folate receptor α (FOLR1) humanized antibody with enhanced antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). The primary aim of the present study was to evaluate whether the anti-tumor activity of KHK2805 was sufficient for therapeutic application against peritoneal dissemination and malignant ascites of platinum-resistant ovarian cancer in preclinical models. Here, both the ADCC and CDC of KHK2805 were evaluated in ovarian cancer cell lines and patient-derived samples. The anti-tumor activity of KHK2805 was evaluated in a SCID mouse model of platinum-resistant peritoneal dissemination. As results, KHK2805 showed specific binding to FOLR1 with high affinity at a novel epitope. KHK2805 exerted potent ADCC and CDC against ovarian cancer cell lines. Furthermore, primary platinum-resistant malignant ascites cells were susceptible to autologous ADCC with KHK2805. Patient-derived sera and malignant ascites induced CDC of KHK2805. KHK2805 significantly reduced the total tumor burden and amount of ascites in SCID mice with peritoneal dissemination and significantly prolonged their survival. In addition, the parental rat antibody strongly stained serous and clear cell-type ovarian tumors by immunohistochemistry. Overall, KHK2805 showed cytotoxicity against both ovarian cancer cell lines and patient-derived cells. These translational study findings suggest that KHK2805 may be promising as a novel therapeutic agent for platinum-resistant ovarian cancer with peritoneal dissemination and malignant ascites.

9.
J Biochem ; 140(6): 777-83, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17038352

RESUMO

Fucose removal from complex-type oligosaccharide of human IgGs results in a major enhancement of Fc-dependent cellular cytotoxicity. The aim of this study was to determine the effect of fucose removal on the effector function of another class of clinically important molecules that can effect cellular cytotoxicity, Fc fusion proteins. The receptors chosen for study were TNF receptor II and LFA-3, both of which have therapeutic significance. The fucosylated versions of these fusion proteins were produced in unmodified CHO cells, whereas the nonfucosylated counterparts were produced in CHO cells with alpha-1,6-fucosyltransferase, an enzyme required for fucosylation, knocked-out. Whilst binding activity of TNFRII-Fc and LFA-3-Fc were unchanged by fucose-removal, nonfucosylated Fc fusion proteins exhibited significantly higher Fc receptor gammaIIIa-binding and increased Fc-mediated cytotoxicity on target cells compared to fucosylated counterparts. Notably, in case of TNFRII-Fc, only the nonfucosylated protein exhibited potent Fc dependent cytotoxicity to transmembrane TNF-alpha expressing cells. These results prove that enhancement of Fc dependent cellular cytotoxicity by fucose-removal is effective in not only whole IgG but also Fc fusion proteins, and thus widens the potential of Fc-fusion proteins as therapeutic candidates.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Fucose/química , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Oligossacarídeos/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/imunologia , Alefacept , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos CD58/imunologia , Células CHO , Cricetinae , Cricetulus , Etanercepte , Humanos , Oligossacarídeos/química , Receptores de IgG/imunologia
10.
Clin Cancer Res ; 11(6): 2327-36, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15788684

RESUMO

PURPOSE: Recent studies have revealed that fucose removal from the oligosaccharides of human IgG1 antibodies results in a significant enhancement of antibody-dependent cellular cytotoxicity (ADCC) via improved IgG1 binding to FcgammaRIIIa. In this report, we investigated the relationship between enhanced ADCC and antigen density on target cells using IgG1 antibodies with reduced fucose. EXPERIMENTAL DESIGN: Using EL4 cell-derived transfectants with differential expression levels of exogenous human CC chemokine receptor 4 or human CD20 as target cells, ADCC of fucose variants of chimeric IgG1 antibodies specific for these antigens were measured. We further investigated IgG1 binding to natural killer (NK) cells and NK cell activation during ADCC induction to elucidate the mechanism by which low-fucose IgG1 induces ADCC upon target cells with low antigen expression. RESULTS: Low-fucose IgG1s showed potent ADCC at low antigen densities at which their corresponding high-fucose counterparts could not induce measurable ADCC. The quantitative analysis revealed that fucose depletion could reduce the antigen amount on target cells required for constant degrees of ADCC induction by 10-fold for CC chemokine receptor 4 and 3-fold for CD20. IgG1 binding to NK cells was increased by ligating IgG1 with clustered antigen, especially for low-fucose IgG1. Up-regulation of an activation marker, CD69, on NK cells, particularly the CD56(dim) subset, in the presence of both the antibody and target cells was much greater for the low-fucose antibodies. CONCLUSIONS: Our data showed that fucose removal from IgG1 could reduce the antigen amount required for ADCC induction via efficient recruitment and activation of NK cells.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD20/metabolismo , Antígeno CD56/metabolismo , Fucose/imunologia , Imunoglobulina G/metabolismo , Células Matadoras Naturais/imunologia , Receptores de Quimiocinas/metabolismo , Humanos , Linfoma/metabolismo , Linfoma/patologia , Ligação Proteica , Receptores CCR4 , Receptores de Quimiocinas/genética , Células Tumorais Cultivadas
11.
Clin Cancer Res ; 11(9): 3494-502, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15867252

RESUMO

Environmental stimuli, such as organ-specific growth factors, can influence the metastatic potential of a tumor. The liver is the main source of insulin-like growth factors (IGFs). The importance of IGF signal in hepatic metastasis has been clarified mainly by IGF-I receptor targeting strategies. This study aims to confirm these precedent reports by novel tool, neutralizing antibodies against IGFs and to show that IGFs are promising therapeutic targets for hepatic metastasis in vivo. Hepatic metastasis was induced by intrasplenic injection of human colorectal cancer cell line, HT29. The antimetastatic effects of three antibodies (anti-mouse IGF-I, anti-mouse IGF-II, and anti-human/mouse IGF-II designated KM1468) were tested singly or in combination in the early phase of metastasis. The dose escalation effect of KM1468 and its survival benefit were examined in the early and late phases of metastasis. The mechanism of IGF neutralization was investigated with immunohistochemistry. Dual neutralization of paracrine IGF-I and IGF-II showed modest additive antimetastatic effects than single neutralization of IGF-I or IGF-II. In any phase of metastasis, neutralization led to significant tumor growth inhibition and longer survival. Dose escalation of KM1468 influenced survival only in the late phase of metastasis. Apoptosis increased significantly in the antibody-treated group compared with the control group (P = 0.0025) In conclusion, IGFs are promising therapeutic targets for hepatic metastases of colorectal cancers. However, the IGF dependency is probably variable in the metastatic process.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Hepáticas/prevenção & controle , Somatomedinas/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antígeno Carcinoembrionário/sangue , Neoplasias Colorretais/patologia , Células HT29 , Humanos , Fator de Crescimento Insulin-Like I/imunologia , Fator de Crescimento Insulin-Like II/imunologia , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos SCID , Testes de Neutralização , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Res ; 64(6): 2127-33, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15026353

RESUMO

Human IgG1 antibodies with low fucose contents in their asparagine-linked oligosaccharides have been shown recently to exhibit potent antibody-dependent cellular cytotoxicity (ADCC) in vitro. To additionally investigate the efficacy of the human IgG1 with enhanced ADCC, we generated the defucosylated chimeric anti-CC chemokine receptor 4 (CCR4) IgG1 antibody KM2760. KM2760 exhibited much higher ADCC using human peripheral blood mononuclear cells (PBMCs) as effector cells compared with the highly fucosylated, but otherwise identical IgG1, KM3060. In addition, KM2760 also exhibited potent ADCC in the presence of lower concentrations of human PBMCs than KM3060. Because CCR4 is a selective marker of T-cell leukemia/lymphoma, the effectiveness of KM2760 for T-cell malignancy was evaluated in several mouse models. First, to compare the antitumor activity of KM2760 and KM3060, we constructed a human PBMC-engrafted mouse model to determine ADCC efficacy with human effector cells. In this model, KM2760 showed significantly higher antitumor efficacy than KM3060, indicating that KM2760 retains its high potency in vivo. Second, KM2760 suppressed tumor growth in both syngeneic and xenograft mouse models in which human PBMCs were not engrafted. Although murine effector cells exhibited marginal ADCC mediated by KM2760 and KM3060, KM2760 unexpectedly showed higher efficacy than KM3060 in a syngeneic mouse model, suggesting that KM2760 functions in murine effector system in vivo via an unknown mechanism that differs from that in human. These results indicate that defucosylated antibodies with enhanced ADCC as well as potent antitumor activity in vivo are promising candidates for the novel antibody-based therapy.


Assuntos
Anticorpos Antineoplásicos/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Imunoglobulina G/imunologia , Leucemia de Células T/terapia , Linfoma de Células T/terapia , Receptores de Quimiocinas/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Sequência de Carboidratos , Sobrevivência Celular , Modelos Animais de Doenças , Citometria de Fluxo , Fucose , Humanos , Leucemia de Células T/imunologia , Linfoma de Células T/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Nus , Camundongos SCID , Dados de Sequência Molecular , Receptores CCR4 , Células Tumorais Cultivadas
13.
Cancer Res ; 64(17): 6252-8, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342412

RESUMO

Advanced prostate cancer frequently involves the bone that has the largest content of insulin-like growth factors (IGFs). However, the role of bone-derived IGFs in bone metastasis of prostate cancer has not been studied extensively because of the lack of a reliable animal model. Therefore, we investigated whether a novel antibody directed against human IGF-I and IGF-II (KM1468) could inhibit the development of new bone tumors and the progression of established bone tumors in nonobese diabetic/severe combined immunodeficient mice implanted with human adult bone. We first confirmed that KM1468 bound specifically to human IGF-I, human IGF-II, and mouse IGF-II but not to insulin. It also blocked autophosphorylation of the type I IGF receptor induced by the binding of IGFs in human-type I IGF receptor-overexpressing BALB/c 3T3 cells, and it inhibited the IGF-stimulated growth of MDA PCa 2b cells in vitro. Then mice were injected intraperitoneally with KM1468 once weekly for 4 weeks either immediately or 4 weeks after inoculation of MDA PCa 2b cells. KM1468 markedly and dose-dependently suppressed the development of new bone tumors and the progression of established tumor foci, as determined by histomorphometry, and it also decreased serum prostate-specific antigen levels, compared with the control. This is the first report of an IGF ligand-specific inhibitory antibody that suppresses the growth of human prostate cancer cells in human adult bone. These results indicate that the IGF signaling axis is a potential target for prevention and treatment of bone metastases arising from prostate cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Fator de Crescimento Insulin-Like II/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Neoplasias da Próstata/patologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Especificidade de Anticorpos , Células 3T3 BALB , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/terapia , Divisão Celular/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/imunologia , Fator de Crescimento Insulin-Like II/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Fosforilação , Antígeno Prostático Específico/biossíntese , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/terapia , Receptor IGF Tipo 1/biossíntese , Receptor IGF Tipo 1/metabolismo
14.
Anticancer Res ; 36(11): 5937-5944, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27793919

RESUMO

BACKGROUND: Multiple factors affect the in vivo antitumor activity of antibody-based therapeutics; however, the influence of cell surface retention on antibody-dependent cellular cytotoxicity (ADCC) is not fully understood. Here we evaluated the importance of cell surface antibody retention in antitumor activity mediated by ADCC in vivo. MATERIALS AND METHODS: Two mAbs against tumor-associated calcium signal transducer 2 (TACSTD2/TROP2), AR47A6.4.2 and Pr1E11, were used. Antitumor activities against BxPC3 and Colo205 cells were investigated through in vitro and in vivo assays. RESULTS: Pr1E11 showed better cell surface retention than AR47A6.4.2 in vitro although Pr1E11 and AR47A6.4.2 showed equivalent ADCC activity. Complement-dependent cytotoxicity and antiproliferative activity were not observed for either antibody. Pr1E11 exhibited higher antitumor activity than AR47A6.4.2 in vivo. CONCLUSION: Our results suggest that high cell surface retention can result in potent ADCC activity in vivo. This observation could provide novel insight into how effectively screen for antibodies with strong in vivo antitumor activity.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos de Neoplasias/imunologia , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Humanos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Mol Biol ; 336(5): 1239-49, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-15037082

RESUMO

Depletion of fucose from human IgG1 oligosaccharide improves its affinity for Fcgamma receptor IIIa (FcgammaRIIIa). This is the first case where a glycoform modification is shown to improve glycoprotein affinity for the receptors without carbohydrate-binding capacity, suggesting a novel glyco-engineering strategy to improve ligand-receptor binding. To address the mechanisms of affinity improvement by the fucose depletion, we used isothermal titration calorimetry (ITC) and biosensor analysis with surface plasmon resonance. ITC demonstrated that IgG1-FcgammaRIIIa binding was driven by favorable binding enthalpy (DeltaH) but opposed by unfavorable binding entropy change (DeltaS). Fucose depletion from IgG1 enhanced the favorable DeltaH, leading to the increase in the binding constant of IgG1 for the receptor by a factor of 20-30. The increase in the affinity was mainly attributed to an enhanced association rate. A triple amino acid substitution in IgG1, S298A/E333A/K334A, is also known to improve IgG1 affinity for FcgammaRIIIa. ITC demonstrated that the amino acid substitution attenuated the unfavorable DeltaS resulting in a three- to fourfold increase in the binding constant. The affinity enhancement by the amino acid substitution was due to a reduced dissociation rate. These results indicate that the mechanism of affinity improvement by the fucose depletion is quite distinct from that by the amino acid substitution. Defucosylated IgG1 exhibited higher antibody-dependent cellular cytotoxicity (ADCC) than S298A/E333A/K334A-IgG1, showing a correlation between IgG1 affinity for FcgammaRIIIa and ADCC. We also examined the effect of FcgammaRIIIa polymorphism (Val158/Phe158) on IgG1-FcgammaRIIIa binding. The Phe to Val substitution increased FcgammaRIIIa affinity for IgG1 in an enthalpy-driven manner with the reduced dissociation rate. These results together highlight the distinctive functional improvement of affinity by IgG1 defucosylation and suggest that engineering of non-interfacial monosaccharides can improve glycoprotein affinity for receptors via an enthalpy-driven and association rate-assisted mechanism.


Assuntos
Fucose/química , Imunoglobulina G/química , Oligossacarídeos/química , Receptores de IgG/química , Substituição de Aminoácidos , Citotoxicidade Celular Dependente de Anticorpos , Calorimetria , Linhagem Celular Tumoral , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Cinética , Ligação Proteica , Engenharia de Proteínas/métodos , Receptores de IgG/imunologia , Ressonância de Plasmônio de Superfície , Termodinâmica
16.
Clin Cancer Res ; 10(18 Pt 1): 6248-55, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15448014

RESUMO

PURPOSE: The most common polymorphic variant of Fcgamma receptor type IIIa (FcgammaRIIIa), FcgammaRIIIa-158F, has been associated with inferior clinical responses to anti-CD20 chimeric IgG1 rituximab compared with FcgammaRIIIa-158V. As we previously found that removal of fucose residues from the oligosaccharides of human IgG1 results in enhanced antibody-dependent cellular cytotoxicity, we compared the effects of the FcgammaRIIIa gene (FCGR3A) polymorphism on normal and low-fucose versions of rituximab on antibody-dependent cellular cytotoxicity. EXPERIMENTAL DESIGN: The polymorphism at position 158 of FcgammaRIIIa was determined for the peripheral blood mononuclear cells (PBMCs) of 20 healthy donors. The PBMCs were then used as effector cells to compare the antibody-dependent cellular cytotoxicity of rituximab and a low-fucose version, KM3065. The contributions of the different cell types within the PBMC to antibody-dependent cellular cytotoxicity were examined. RESULTS: We found KM3065-mediated antibody-dependent cellular cytotoxicity was increased 10 to 100-fold compared with rituximab for each of the 20 donors. In contrast to rituximab, KM3065 antibody-dependent cellular cytotoxicity enhancement was similar for both FCGR3A alleles and thus independent of genotype. In addition, antibody-dependent cellular cytotoxicity of both KM3065 and rituximab requires natural killer cells but not monocytes nor polymorphonuclear cells. The antibody-dependent cellular cytotoxicity (ADCC) of each of the 20 donors correlated with the natural killer cell numbers present in the PBMCs. Importantly, using KM3065, the ADCC mediated by effector cells bearing the lower affinity variant FcgammaRIIIa-158F was significantly increased compared with rituximab-mediated ADCC using effector cells bearing the higher affinity FcgammaRIIIa-158V receptors. CONCLUSIONS: The use of low-fucose antibodies might improve the therapeutic effects of anti-CD20 therapy for all patients independent of FcgammaRIIIa phenotype beyond that currently seen with even the most responsive patients.


Assuntos
Fucose/química , Imunoglobulina G/química , Polimorfismo Genético , Receptores de IgG/química , Alelos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Anticorpos Antineoplásicos/química , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD20/biossíntese , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Fucose/metabolismo , Genótipo , Humanos , Células Matadoras Naturais/citologia , Leucócitos Mononucleares/metabolismo , Monócitos/metabolismo , Neutrófilos/citologia , Fenótipo , Ligação Proteica , Proteínas Recombinantes/química , Rituximab , Fatores de Tempo
17.
Anticancer Res ; 34(1): 89-97, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24403448

RESUMO

Prostate-specific membrane antigen (PSMA) is an attractive target for treatment of prostate cancer. Using the PSMA-recognizing mouse monoclonal antibody 2C9 obtained in our previous study, the biological activities of PSMA antibody were evaluated. Mouse-human chimeric IgG1 of 2C9 (KM2777) showed antibody-dependent cellular cytotoxicity activity against PSMA-expressing prostate cancer cells in the presence of human peripheral blood mononuclear cells (PBMCs). To increase lymphocyte-mediated cytotoxicity of KM2777, C-terminus interleukin-2 (IL-2)-fused KM2777 (KM2812) was constructed. KM2812 retained binding activity to PSMA and exhibited growth-stimulating activity equivalent to IL-2 on the IL-2-dependent T-cell line CTLL-2. Moreover, KM2812 exhibited enhanced cytotoxic activity against PSMA-expressing prostate cancer cells in the presence of PBMCs compared with KM2777. In a xenograft tumor model using PSMA-expressing prostate cancer cells, KM2812 exhibited marked antitumor activity, accompanied by complete regression of tumor in some of the KM2812-treated mice. These results suggest that KM2812 has a therapeutic potential for prostate cancer by stimulating lymphocyte-mediated antitumor cytotoxicity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos de Superfície/imunologia , Citotoxicidade Imunológica/imunologia , Glutamato Carboxipeptidase II/imunologia , Interleucina-2/imunologia , Glicoproteínas de Membrana/imunologia , Neoplasias da Próstata/terapia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Citometria de Fluxo , Glutamato Carboxipeptidase II/antagonistas & inibidores , Humanos , Interleucina-2/metabolismo , Leucócitos Mononucleares , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos SCID , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Linfócitos T , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Anticancer Res ; 30(9): 3397-405, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20944114

RESUMO

Tn[GalNAc(α1-3)-Ser/Thr] antigen, a tumor-associated carbohydrate antigen, is highly expressed in various tumors and an attractive candidate for cancer immunotherapy. The generation of an anti-Tn antibody is a first step toward the construction of new anticancer molecules. However, because of the simple and small conformation of the Tn molecule, it is difficult to generate an anti-Tn antibody for therapeutic use by conventional hybridoma technology. The purpose of this study was to isolate anti-Tn single-chain antibody fragments (scFv) by phage display technology from a novel immunised library, to attach an antibody constant region (Fc) and to convert them to scFv-Fc fusion proteins. The scFv-Fcs obtained here showed strict specificity against the Tn antigen and also showed antibody-dependent cellular cytotoxicity. These results suggest a potential use of this antibody generating method by phage display and indicate the potential of Fc-fusion proteins as therapeutic candidates.


Assuntos
Antígenos Glicosídicos Associados a Tumores/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/síntese química , Anticorpos de Cadeia Única/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Separação Celular , Feminino , Citometria de Fluxo , Humanos , Imunização , Células Jurkat , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Anticancer Res ; 30(11): 4555-62, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21115905

RESUMO

BACKGROUND: Because human claudin-3 and claudin-4 (CLDN3 and CLDN4) are overexpressed in a variety of carcinomas, they are promising targets for cancer therapy. The aim of the present study was to generate a dual-targeting monoclonal antibody against CLDN3 and CLDN4 and evaluate its antitumour activity. MATERIALS AND METHODS: BALB/c mice were immunised with CLDN4-expressing Chinese hamster ovary cells and cell-based screening was performed. The antibody-binding epitope of CLDN3 and CLDN4 and the antitumour activity of the antibody were evaluated. RESULTS: A monoclonal antibody, KM3907 (IgG2a), which recognised CLDN3 and CLDN4, but not CLDN5, CLDN6 and CLDN9, was successfully isolated. The binding assay of KM3907 revealed that KM3907 recognised the extracellular loop 1 of CLDN3 and CLDN4. Mouse human chimeric IgG1 induced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro, and treatment with murine KM3907 significantly inhibited tumour formation in SCID mice in vivo. CONCLUSION: A dual-targeting monoclonal antibody against CLDN3 and CLDN4 is a promising strategy for cancer immunotherapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/terapia , Citotoxicidade Imunológica/imunologia , Proteínas de Membrana/imunologia , Neoplasias Ovarianas/terapia , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Células CHO , Linhagem Celular , Proliferação de Células , Claudina-3 , Claudina-4 , Cricetinae , Cricetulus , Feminino , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos SCID , Dados de Sequência Molecular , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Homologia de Sequência de Aminoácidos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Proc Natl Acad Sci U S A ; 102(22): 7946-51, 2005 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-15905329

RESUMO

Because CD30 is highly expressed on Hodgkin's lymphoma and anaplastic large cell lymphoma, it is a promising target for immunotherapy. Soluble CD30, the extracellular domain of CD30 that is shed from the cells, can reduce the effects of CD30-targeting agents by competitive binding. In this study, we identified two epitopes on membrane-associated CD30 that are missing on soluble CD30 probably because of a conformational change upon shedding. These epitopes are potentially superior targets for immunotherapy because targeting them should be free from the competitive effects of soluble CD30. We studied 27 anti-native CD30 mAbs that were assigned to 8 different topographical epitopes. Soluble CD30 was prepared from culture supernatants of L540 cells or Karpas 299 cells. In an ELISA, the mAbs to two epitopes, Ep2 (amino acids 107-153) and Ep7 (amino acids 282-338), showed less than a 2% average cross-reactivity to soluble CD30 compared with a CD30-Fc fusion protein. In addition, these mAbs bound to CD30 on cells in the presence of an excess of soluble CD30. These epitopes (Ep2 and Ep7) are, therefore, more efficiently presented on cell-associated CD30 than on soluble CD30 (membrane-specific epitopes). Also, soluble CD30 in the sera of mice bearing L540 tumors did not form immune complexes with the membrane-specific mAbs analyzed by size-exclusion chromatography. In contrast, mAbs to the other epitopes reacted with both soluble CD30 and membrane CD30. Our results suggest that it may be possible to find membrane-specific epitopes on other immunotherapy target molecules.


Assuntos
Anticorpos Monoclonais/metabolismo , Epitopos/metabolismo , Imunoterapia/métodos , Antígeno Ki-1/imunologia , Antígeno Ki-1/metabolismo , Linfoma/terapia , Animais , Western Blotting , Linhagem Celular Tumoral , Cromatografia em Gel , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA