Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
Prostate ; 72(10): 1071-9, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22095553

RESUMO

BACKGROUND: Endocrine resistance is a critical issue in managing patients with prostate cancer. This study is undertaken to search for a potential molecular target connected with this process using a model system of androgen-dependent and androgen-unresponsive SC-3 and SC-4 cells. METHODS: Expression profiles, actin stress fiber organization, and the levels of activated Rho GTPases were compared between SC-4 and SC-3 cells using an oligonucleotide microarray, phalloidin staining, and a Rho activation assay. The cell viability was analyzed with a Rho inhibitor or by stable transfection with either a dominant-negative (DN) form of RhoC or a mutant form of NET1 (mutNET1). The expressions of RhoC, NET1, and epithelial-mesenchymal transition (EMT) markers were immunohistochemically analyzed in human prostate cancer specimens after short-term endocrine therapy and in an untreated condition. RESULTS: SC-4 cells exhibited mesenchymal phenotypes with activation of Rho signals. Treatment with a Rho inhibitor suppressed the cell viability in SC-4 cells, but not in SC-3 cells. The cell viability of SC-4 cells stably expressing DN-RhoC and mutNET1 was also attenuated. In the immunohistochemical analysis, NET1 and the EMT marker of N-cadherin were expressed at higher levels in prostate cancers after short-term endocrine therapy than in untreated tumors, and RhoC expression was maintained after short-term endocrine therapy. CONCLUSIONS: Rho signaling is involved in the cell survival of SC-4 cells. The higher expressions of RhoC and NET1 in human prostate cancers after short-term endocrine therapy suggest that RhoC and NET1 may become therapeutic targets during endocrine therapy.


Assuntos
Antagonistas de Androgênios/administração & dosagem , Androgênios/metabolismo , Neoplasias da Mama/metabolismo , Proteínas Oncogênicas/biossíntese , Neoplasias da Próstata/metabolismo , Proteínas rho de Ligação ao GTP/biossíntese , Idoso , Androgênios/uso terapêutico , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Humanos , Masculino , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Pessoa de Meia-Idade , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Fatores de Tempo , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteína de Ligação a GTP rhoC
3.
Clin Cancer Res ; 17(21): 6733-41, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21918176

RESUMO

PURPOSE: Heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF) is a member of the EGF family and plays a pivotal role in tumor progression in ovarian cancer. We developed an anti-HB-EGF monoclonal antibody (mAb) and investigated its antitumor activities in vitro and in vivo to evaluate its potential as a therapeutic antibody against ovarian cancer. EXPERIMENTAL DESIGN: We prepared mAbs from HB-EGF null mice immunized with recombinant human soluble HB-EGF and evaluated their binding and neutralizing activity against HB-EGF. Next, we generated a mouse-human chimeric antibody and examined its in vitro and in vivo antitumor activities. RESULTS: Two murine anti-HB-EGF mAbs were developed, and one of them, KM3566, was revealed to have a high binding reactivity for membrane-anchored HB-EGF (pro-HB-EGF) expressed on the cell surface, as well as neutralizing activity against growth promoting activity of soluble HB-EGF. The mouse-human chimeric counterpart for KM3566 (cKM3566) induced dose-dependent antibody-dependent cellular cytotoxicity (ADCC) against cancer cells expressing HB-EGF in vitro, and significantly inhibited tumor growth in severe combined immunodeficient mice inoculated with MCAS or ES-2 human ovarian cancer cells. CONCLUSIONS: A novel anti-HB-EGF chimeric antibody, cKM3566, with two antitumor mechanisms, neutralization and ADCC, exhibits potent in vivo antitumor activity. These results indicate that cKM3566 is a promising antiovarian cancer therapeutic antibody.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Neoplasias Ovarianas/terapia , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Especificidade de Anticorpos , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Feminino , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Imunoglobulina G/imunologia , Camundongos , Camundongos SCID , Neoplasias Ovarianas/imunologia , Especificidade por Substrato , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Cancer Res ; 16(5): 1520-31, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20160057

RESUMO

PURPOSE: Adult T-cell leukemia/lymphoma (ATLL) has a very poor prognosis. We have developed the humanized defucosylated anti-CC chemokine receptor 4 (CCR4) monoclonal antibody KW-0761 as a next generation immunotherapeutic agent. The first aim of the present study was to evaluate whether the antitumor activity of KW-0761 would likely be sufficient for therapeutic clinical application against ATLL. The second aim was to fully elucidate the mechanism of antibody-dependent cellular cytotoxicity (ADCC) mediated by this defucosylated monoclonal antibody. EXPERIMENTAL DESIGN: The antitumor activity of KW-0761 against ATLL cell lines was evaluated in vitro using human cells and in mice in vivo. Primary ATLL cells from 23 patients were evaluated for susceptibility to autologous ADCC with KW-0761 by two independent methods. RESULTS: KW-0761 showed potent antitumor activity against ATLL cell lines both in vitro and in the ATLL mouse model in vivo. In addition, KW-0761 showed potent antitumor activity mediated by highly enhanced ADCC against primary ATLL cells both in vitro and ex vivo in an autologous setting. The degree of KW-0761 ADCC against primary ATLL cells in an autologous setting was mainly determined by the amount of effector natural killer cells present, but not the amount of the target molecule CCR4 on the ATLL cell surface. CONCLUSION: KW-0761 should be sufficiently active for therapeutic clinical application for ATLL. In addition, combination treatment strategies that augment natural killer cell activity should be promising for amplifying the effect of KW-0761. In the near future, the actual efficacy of KW-0761 will be established in pivotal clinical trials.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Imunoterapia/métodos , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Adulto , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Técnicas Biossensoriais , Separação Celular , Citometria de Fluxo , Humanos , Leucemia-Linfoma de Células T do Adulto/imunologia , Masculino , Camundongos , Camundongos SCID , Receptores CCR4/imunologia
5.
Mol Cancer Ther ; 9(2): 419-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20103605

RESUMO

Insulin-like growth factors (IGF-I and IGF-II) play important roles in intestinal tumorigenesis. To investigate the effectiveness of IGF-targeting strategies, we conducted an in vivo study using anti-mouse neutralizing antibodies IGF-I (KM3168) and IGF-II (KM1468). Six- and 10-week-old Apc(+/-) mice were given KM3168 and/or KM1468 i.p. at two doses (0.01 or 0.1 microg/g weight) once or twice weekly for 4 weeks. To clarify the source of IGFs in vivo, we evaluated the expression levels of IGFs in the liver, normal small intestine, and polyps of the small intestine of Apc(+/-) mice. The phosphorylation status of IGF signal-related molecules was examined using immunostaining to understand the mechanism underlying the effects of IGF-neutralizing antibody. The plasma half-life was 168 for KM3168 and 85 hours for KM1468. In two lineages of Apc(+/-) mice (Apc(1309) and Apc(Min/+)), a low dose (0.01 microg/g weight) of KM3168 and KM1468 significantly reduced the number of polyps when given once and twice weekly, respectively. Combined administration of the effective dose of each antibody had an additive effect. The liver was the main source of IGF-I, whereas the polyps of the small intestine and normal small intestine were the main source of IGF-II. IGF-neutralizing antibodies decreased the phosphorylation of IGF type 1 receptor and inhibited the signal transduction of the Akt pathway. These results suggest that IGF-I and IGF-II play important roles in polyp formation in Apc(+/-) mice and that specific antibodies to IGF-I and IGF-II may be promising antitumor agents.


Assuntos
Pólipos Intestinais/patologia , Somatomedinas/química , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Fibroblastos/metabolismo , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Transdução de Sinais
6.
Cancer Sci ; 100(12): 2411-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19758394

RESUMO

One of the major issues in current antibody therapy is insufficient efficacy. Various biological factors relating to the host's immune system or tumor cells have been suggested to reduce the efficacy of anti-CD20 therapy in B-cell malignancies. In this study, we characterized the in vitro anti-lymphoma activity of anti-CD20 antibodies having a novel engineered heavy chain with enhanced complement-dependent cytotoxicity (CDC). Anti-CD20 antibodies having a variant heavy constant region of mixed IgG1/IgG3 isotype, which have previously been found to enhance CDC, were investigated for their in vitro CDC against lymphoma cells and whole blood B-cell depletion activity. Use of the variant constant region greatly increased the CDC of an anti-CD20 antibody having variable regions identical to those of rituximab to the level shown by an IgG1 antibody of ofatumumab. Although the whole blood assay showed different cytotoxicity patterns among individual blood donors, the CDC-enhancing variant of rituximab showed higher activity than the parent IgG1 and consistently showed maximized activity when further combined with antibody-dependent cellular cytotoxicity (ADCC)-enhancing modification by fucose removal from Fc-linked oligosaccharides. In addition, the rituximab variant showed potent CDC against transfectant cells with lower CD20 expression and chronic lymphocytic leukemia-derived cell lines with higher complement regulatory proteins. These findings suggest that CDC enhancement, both alone and in combination with ADCC enhancement, increases the anti-lymphoma activity of anti-CD20 antibodies irrespective of individual differences in effector functions, and renders current anti-CD20 therapy capable of overcoming the potential resistance mechanisms.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Linfoma/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Citotoxicidade Celular Dependente de Anticorpos , Células CHO , Proteínas do Sistema Complemento/imunologia , Cricetinae , Cricetulus , Humanos , Engenharia de Proteínas , Rituximab
7.
Cancer Sci ; 100(9): 1566-72, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19538497

RESUMO

In the past decade, more than 20 therapeutic antibodies have been approved for clinical use and many others are now at the clinical and preclinical stage of development. Fragment crystallizable (Fc)-dependent antibody functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and a long half-life, have been suggested as important clinical mechanisms of therapeutic antibodies. These functions are primarily triggered through direct interaction of the Fc domain with its corresponding receptors: FcgammaRIIIa for ADCC, C1q for CDC, and neonatal Fc receptor for prolongation of the clearance rate. However, current antibody therapy still faces the critical issues of insufficient efficacy and the high cost of the therapeutic agents. A possible solution to these issues could be to engineer antibody molecules to enhance their antitumor activity, leading to improved therapeutic outcomes and reduced doses. Here, we review advanced Fc engineering approaches for the enhancement of effector functions, some of which are now ready for evaluation of their effectiveness in clinical trials.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Neoplasias/terapia , Engenharia de Proteínas , Animais , Humanos
8.
Leuk Res ; 33(12): 1650-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19342098

RESUMO

By using neutralizing monoclonal antibodies to vascular endothelial growth factor receptor type 1 (VEGFR1) and VEGFR2, we have shown that acute myelogenous leukemia (AML) cells with specific chromosome abnormalities are dependent on VEGF/VEGFR system. AML with t(8;21) is the most dependent subtype on VEGF with both VEGFR1 and VEGFR2. t(15;17)AML cells depend on VEGF with VEGFR1. AML cells with 11q23 abnormalities showed variable dependence on VEGF. The growth of t(11;19)AML cells are most extensively inhibited by anti-VEGFR1 antibody. Then, the growth of Kasumi-1, a t(8;21) cell line was suppressed by either anti-VEGFR1 antibody (p=0.0022) or anti-VEGFR2 antibody (p=0.0029) in a dose-dependent manner. The growth of NB4, a t(15;17) cell line was more potently suppressed by anti-VEGFR1 antibody (p=0.0111) than by anti-VEGFR2 antibody (p=0.0477). These results are quite concordant with the results of clinical samples with t(8;21) or t(15;17). In addition, anti-VEGFR2 monoclonal antibody significantly potentiated the growth inhibitory effect of idarubicin for Kasumi-1. As for downstream signals, we have shown that VEGFR2 transduce growth and survival signals through phosphorylation of Akt and MEK in leukemia cells (Kasumi-1). However, VEGFR1 transduce growth and survival signals through pathways other than MEK and Akt (NB4), although Akt phosphorylation may account for some of the VEGFR1 signals (Kasumi-1). Finally, our data suggested that autocrine pathway of VEGF and VEGFRs observed in AML cells with specific chromosomal translocations have contributed to leukemogenesis as activated signaling of receptor tyrosine kinase.


Assuntos
Anticorpos Monoclonais/imunologia , Divisão Celular/imunologia , Aberrações Cromossômicas , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Receptores de Fatores de Crescimento do Endotélio Vascular/imunologia , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Fosforilação
9.
Exp Hematol ; 37(3): 309-21, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19218011

RESUMO

OBJECTIVE: Antibody-dependent cellular cytotoxicity mediated by natural killer cells via leukocyte receptor IIIa (FcgammaRIIIa) is greatly enhanced by the absence of the core fucose of Fc oligosaccharides, and is closely related to the clinical efficacy of anticancer processes in humans in vivo. Here, we focused on the physiological functions of nonfucosylated anti-CD20 IgG1 rituximab, in particular those functions mediated by human neutrophils, which highly express FcgammaRIIIb, a highly homologous FcgammaR to FcgammaRIIIa. MATERIALS AND METHODS: After treatment with anti-CD20, the response of neutrophils to fluorescently labeled CD20(+) B-cell lymphoma in human whole blood was quantitatively analyzed by measuring their activities of antibody-dependent phagocytosis and major histocompatibility complex (MHC) class II expression on the phagocytotic neutrophils using flow cytometry. RESULTS: In human whole blood, most of the added CD20(+) B-cell lymphoma died shortly, within 4 hours, irrespective of the presence or absence of anti-CD20. Neutrophils were not directly concerned in the death because depletion of neutrophils from human whole blood did not affect the phenomenon. However, neutrophils aggressively phagocytosed newly dead lymphoma cells, and the nonfucosylated anti-CD20 effectively enhanced neutrophil phagocytosis solely by enhancing binding for the phagocytosis coreceptor FcgammaRIIIb. Noteworthy, more increased expression of MHC class II was also observed on the phagocytotic neutrophils than those observed on spontaneous and fucosylated anti-CD20 stimulated phagocytotic neutrophils. CONCLUSIONS: Our data showed that antibody therapy composed of nonfucosylated rituximab can activate human neutrophil functions involving phagocytosis and MHC class II expression, which may favorably potentiate the adaptive immune response in cancer patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Histocompatibilidade Classe II/genética , Linfoma de Células B/tratamento farmacológico , Neutrófilos/imunologia , Fagocitose/efeitos dos fármacos , Receptores de IgG/metabolismo , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Murinos , Células Sanguíneas , Morte Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Proteínas Ligadas por GPI , Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma de Células B/patologia , Camundongos , Ligação Proteica , Rituximab
10.
BMC Cancer ; 9: 58, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19226457

RESUMO

BACKGROUND: Antibody-dependent cellular cytotoxicity (ADCC) has recently been identified as one of the critical mechanisms underlying the clinical efficacy of therapeutic antibodies, especially anticancer antibodies. Therapeutic antibodies fully lacking the core fucose of the Fc oligosaccharides have been found to exhibit much higher ADCC in humans than their fucosylated counterparts. However, data which show how fully non-fucosylated antibodies achieve such a high ADCC in human whole blood have not yet been disclosed. The precise mechanisms responsible for the high ADCC mediated by fully non-fucosylated therapeutic antibodies, even in the presence of human plasma, should be explained based on direct evidence of non-fucosylated antibody action in human blood. METHODS: Using a human ex vivo B-cell depletion assay with non-fucosylated and fucosylated anti-CD20 IgG1s rituximab, we monitored the binding of the therapeutic agents both to antigens on target cells (target side interaction) and to leukocyte receptors (FcgammaR) on effector cells (effector side interaction), comparing the intensities of ADCC in human blood. RESULTS: In the target side interaction, down-modulation of CD20 on B cells mediated by anti-CD20 was not observed. Simple competition for binding to the antigens on target B cells between fucosylated and non-fucosylated anti-CD20s was detected in human blood to cause inhibition of the enhanced ADCC of non-fucosylated anti-CD20 by fucosylated anti-CD20. In the effector side interaction, non-fucosylated anti-CD20 showed sufficiently high FcgammaRIIIa binding activity to overcome competition from plasma IgG for binding to FcgammaRIIIa on natural killer (NK) cells, whereas the binding of fucosylated anti-CD20 to FcgammaRIIIa was almost abolished in the presence of human plasma and failed to recruit NK cells effectively. The core fucosylation levels of individual serum IgG1 from healthy donors was found to be so slightly different that it did not affect the inhibitory effect on the ADCC of fucosylated anti-CD20. CONCLUSION: Our results demonstrate that removal of fucosylated antibody ingredients from antibody therapeutics elicits high ADCC in human blood by two mechanisms: namely, by evading the inhibitory effects both of plasma IgG on FcgammaRIIIa binding (effector side interaction) and of fucosylated antibodies on antigen binding (target side interaction).


Assuntos
Anticorpos/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Sangue/imunologia , Fucose/imunologia , Adulto , Anticorpos/genética , Anticorpos/imunologia , Reações Antígeno-Anticorpo , Antígenos CD20/genética , Antígenos CD20/imunologia , Linfócitos B/imunologia , Células Cultivadas , Feminino , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/uso terapêutico , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Yakugaku Zasshi ; 129(1): 3-9, 2009 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-19122430

RESUMO

Antibody-dependent cellular cytotoxicity (ADCC), a lytic attack on antibody-targeted cells, is triggered upon binding of lymphocyte receptors (FcgammaRs) to the antibody constant region. ADCC is considered to be a major therapeutic function of antibodies. ADCC requires the presence of oligosaccharides in the Fc region and is sensitive to change in the oligosaccharide structure. We have demonstrated that fucose is the most critical IgG1 oligosaccharide component, and the removal of fucose from IgG1 oligosaccharides results in a very significant enhancement of ADCC and anti-tumor activity in vivo. Many therapeutic antibodies approved or clinical development are produced using Chinese hamster ovary (CHO) cells that express high level of alpha1,6-fucosyltransferase and consequently produce highly fucosylated antibodies. We have established the fucosyltransferase knockout CHO cells which could stably produce non-fucosylated antibodies, designated as Potelligent antibodies. Potelligent antibodies show potent ADCC upon target cells through the effective and antigen-specific activation of NK cells due to augmented binding to FcgammaRIIIa. Moreover, Potelligent antibodies can evade the inhibitory effect of plasma IgG on ADCC through its high FcgammaRIIIa binding. Thus, the application of Potelligent antibodies is expected to be a promising approach as next-generation therapeutic antibodies with improved efficacy, even when administered at low doses in humans in vivo.


Assuntos
Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Desenho de Fármacos , Animais , Anticorpos/uso terapêutico , Células CHO , Cricetinae , Cricetulus , Fucose , Humanos , Imunoglobulina G , Oligossacarídeos , Receptores de IgG
12.
Glycobiology ; 19(2): 126-34, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18952826

RESUMO

Human leukocyte receptor IIIa (Fc gamma RIIIa) plays an important role in mediating therapeutic antibodies' antibody-dependent cellular cytotoxicity (ADCC), which is closely related to the clinical efficacy of anticancer processes in humans in vivo. The removal of the core fucose from oligosaccharides attached to the Fc region of antibodies improves Fc gamma RIIIa binding, allowing the antibodies to enhance dramatically the antibody effector functions of ADCC. In this study, the contribution of Fc gamma RIIIa oligosaccharides to the strength of the Fc gamma RIIIa/antibody complex was analyzed using a serial set of soluble human recombinant Fc gamma RIIIa lacking the oligosaccharides. A nonfucosylated antibody IgG1 appeared to have a significantly higher affinity to the wild-type Fc gamma RIIIa fully glycosylated at its five N-linked oligosaccharide sites than did the fucosylated IgG1, and this increased binding was almost abolished once all of the Fc gamma RIIIa glycosylation was removed. Our gain-of-function analysis in the Fc gamma RIIIa oligosaccharide at Asn-162 (N-162) confirmed that N-162 is the element required for the high binding affinity to nonfucosylated antibodies, as previously revealed by loss-of-function analyses. Interestingly, beyond our expectation, the Fc gamma RIIIa modified by N-162 alone showed a significantly higher binding affinity to nonfucosylated IgG1 than did the wild-type Fc gamma RIIIa. Attachment of the other four oligosaccharides, especially the Fc gamma RIIIa oligosaccharide at Asn-45 (N-45), hindered the high binding affinity of Fc gamma RIIIa to nonfucosylated IgG1. Our data clearly demonstrated that N-45 is an inhibitory element for the high Fc gamma RIIIa binding affinity mediated by N-162 to nonfucosylated antibodies. This information can be exploited for the structural-based functional study of Fc gamma RIIIa.


Assuntos
Fucose/metabolismo , Imunoglobulina G/metabolismo , Receptores de IgG/química , Receptores de IgG/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Antígenos CD20/imunologia , Antígenos CD20/metabolismo , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/química , Imunoglobulina G/imunologia , Modelos Biológicos , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Receptores de IgG/imunologia , Ressonância de Plasmônio de Superfície
13.
Biol Pharm Bull ; 31(9): 1739-44, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18758069

RESUMO

Tn-antigen (alpha-N-acetyl-galactosamine(GalNAc)-Ser/Thr) is a cancer-associated carbohydrate antigen expressed in various epithelial and hematological cancers, and although a number of anti-Tn IgG and IgM antibodies have been generated, they have not been fully validated for cancer immunotherapy. In this study, we generated a novel murine anti-Tn IgG1 monoclonal antibody, KM3413, by immunization of mucins purified from a culture supernatant of LS180: a human colon cancer cell line. The binding of KM3413 was detected against consecutive Tn-antigens (Tn3 and Tn2), but not against monovalent antigens (Tn1). The affinity (K(D)) of KM3413 was determined to be about 10(-7) M with BIAcore. Cross-reactivity against type-A blood antigen, which shares a sugar residue, alpha-linked GalNAc, with Tn-antigen, was not detected. Next, we generated mouse-human chimeric IgG1 of KM3413 (cKM3413) and evaluated its anti-tumor activities against Jurkat: a human T-lymphoid leukemia cell line. In vitro assay revealed that cKM3413 induced antibody-dependent cellular cytotoxicity (ADCC) and direct killing activity with cross-link antibody. Furthermore, treatment of cKM3413 (1 or 10 mg/kg) showed significantly better survival of Jurkat-inoculated C.B-17/lcr-scid Jcl mice compared with controls using PBS treatment (p<0.001). These results suggest that humanized antibody against clustered Tn-antigens is a promising therapeutic antibody against Tn-positive cancers.


Assuntos
Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Antígenos Glicosídicos Associados a Tumores/efeitos dos fármacos , Imunoglobulina G/farmacologia , Proteínas Mutantes Quiméricas/farmacologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antígenos Glicosídicos Associados a Tumores/imunologia , Western Blotting , Células CHO , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Células Jurkat , Camundongos , Camundongos SCID , Mucinas/isolamento & purificação , Mucinas/farmacologia
14.
Cancer Res ; 68(10): 3863-72, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483271

RESUMO

Enhancement of multiple effector functions of an antibody may be a promising approach for antibody therapy. We have previously reported that fucose removal from Fc-linked oligosaccharides greatly enhances antibody-dependent cellular cytotoxicity (ADCC) of therapeutic antibodies. Here, we report a unique approach to enhance complement-dependent cytotoxicity (CDC), another important effector function of antitumor antibodies, by using engineered constant region of human IgG1/IgG3 chimeric isotypes. We systematically shuffled constant domains of IgG1 and IgG3 to generate a comprehensive set of mixed chimeric isotypes of anti-CD20 antibodies. Among these, the variant 1133, consisting of the CH1 and the hinge each from IgG1 and the Fc from IgG3, was unexpectedly found to exhibit markedly enhanced CDC that exceeded wild-type levels. However, it lacked protein A-binding capacity, an important feature for the industrial production. To eliminate this deficiency, a portion in COOH-terminal CH3 domain of 1133 was substituted with IgG1, resulting in full recovery of protein A binding without compromising the enhanced CDC and ADCC activities. The CDC-enhancing effect using a chimeric isotype was also shown in CD52 antigen/antibody system. The ADCC activity of the variants was also maximized by the absence of fucose from its carbohydrate structure, a phenomenon that has previously been observed for wild-type antibodies. Enhanced cytotoxicity of a variant was confirmed in a cynomolgus monkey model. These findings suggest that the variant antibodies with IgG1/IgG3 chimeric constant regions and nonfucosylated oligosaccharides that possess dual-enhanced cytotoxic functions may be an improvement for the next generation of therapeutic antitumor antibodies.


Assuntos
Antígenos CD20/química , Imunoglobulina G/química , Engenharia de Proteínas/métodos , Animais , Antígenos CD/química , Antígenos de Neoplasias/química , Antígeno CD52 , Células CHO , Carboidratos/química , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Glicoproteínas/química , Humanos , Isotipos de Imunoglobulinas , Leucócitos Mononucleares/citologia , Estrutura Terciária de Proteína
15.
Prostate ; 68(6): 640-50, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18213631

RESUMO

BACKGROUND: Fibroblast growth factor 8-isoform b (FGF8b) has been detected in human clinical sex-organ related cancers including hormone-refractory prostate cancer. There are, however, few relevant experimental models. A murine monoclonal anti-FGF8 antibody, KM1334, has been shown to neutralize FGF8b and inhibit the growth of androgen-dependent mouse mammary SC-3 cells in vitro and in vivo. In the present study, we evaluated the anti-tumor activity of KM1334 against androgen-dependent and -independent progression of FGF8b-expressing human prostate cancer xenografts. METHODS: FGF8b cDNA was transfected into androgen-dependent human prostate cancer cell line LNCaP, and its xenograft tumors were established subcutaneously in SCID mice with or without castration. KM1334 at the dose of 400 microg/head was injected twice weekly. RESULTS: FGF8b-expressing LNCaP cells secreted FGF8b, showed enhanced level of Erk1/2 phosphorylation, and showed more potent growth properties than mock-expressing cells in vitro and in vivo. KM1334 reduced these properties in vitro, inhibited tumorigenecity in vivo (T/C=0.33), and showed anti-tumor activity against established tumors (T/C=0.47) of FGF8b-expressing cells. FGF8b-expressing LNCaP tumors were androgen-dependent. However, they recurred as androgen-independent FGF8b positive tumors after castration. KM1334 also inhibited the growth of established FGF8b-expressing tumors in the androgen-independent states (T/C=0.47). CONCLUSIONS: These results indicate that humanized monoclonal antibodies, conserving the paratope of KM1334, are a promising candidate for therapy of FGF8b-expressing clinical prostate cancers. Follow-up studies using xenograft models with clinical FGF8b-expressing tumors are required to validate these early findings.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Fator 8 de Crescimento de Fibroblasto/imunologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Antineoplásicos/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta Imunológica , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos SCID , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
16.
BMC Biotechnol ; 7: 84, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18047682

RESUMO

BACKGROUND: Antibody-dependent cellular cytotoxicity (ADCC) is greatly enhanced by the absence of the core fucose of oligosaccharides attached to the Fc, and is closely related to the clinical efficacy of anticancer activity in humans in vivo. Unfortunately, all licensed therapeutic antibodies and almost all currently-developed therapeutic antibodies are heavily fucosylated and fail to optimize ADCC, which leads to a large dose requirement at a very high cost for the administration of antibody therapy to cancer patients. In this study, we explored the possibility of converting already-established antibody-producing cells to cells that produce antibodies fully lacking core fucosylation in order to facilitate the rapid development of next-generation therapeutic antibodies. RESULTS: Firstly, loss-of-function analyses using small interfering RNAs (siRNAs) against the three key genes involved in oligosaccharide fucose modification, i.e. alpha1,6-fucosyltransferase (FUT8), GDP-mannose 4,6-dehydratase (GMD), and GDP-fucose transporter (GFT), revealed that single-gene knockdown of each target was insufficient to completely defucosylate the products in antibody-producing cells, even though the most effective siRNA (>90% depression of the target mRNA) was employed. Interestingly, beyond our expectations, synergistic effects of FUT8 and GMD siRNAs on the reduction in fucosylation were observed, but not when these were used in combination with GFT siRNA. Secondly, we successfully developed an effective short hairpin siRNA tandem expression vector that facilitated the double knockdown of FUT8 and GMD, and we converted antibody-producing Chinese hamster ovary (CHO) cells to fully non-fucosylated antibody producers within two months, and with high converting frequency. Finally, the stable manufacture of fully non-fucosylated antibodies with enhanced ADCC was confirmed using the converted cells in serum-free fed-batch culture. CONCLUSION: Our results suggest that FUT8 and GMD collaborate synergistically in the process of intracellular oligosaccharide fucosylation. We also demonstrated that double knockdown of FUT8 and GMD in antibody-producing cells could serve as a new strategy for producing next-generation therapeutic antibodies fully lacking core fucosylation and with enhanced ADCC. This approach offers tremendous cost- and time-sparing advantages for the development of next-generation therapeutic antibodies.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Fucosiltransferases/genética , Inativação Gênica/imunologia , Melhoramento Genético/métodos , Hidroliases/genética , Animais , Células CHO , Cricetinae , Cricetulus , Fucosiltransferases/imunologia , Hidroliases/imunologia , Engenharia de Proteínas/métodos
17.
Clin Cancer Res ; 13(6): 1875-82, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363544

RESUMO

PURPOSE: Removal of fucose residues from the oligosaccharides of human antibody is a powerful approach to enhance antibody-dependent cellular cytotoxicity (ADCC), a potential important antitumor mechanism of therapeutic antibodies. To provide clinically relevant evidence of this mechanism, we investigated ADCC of a fucose-negative version of trastuzumab [anti-human epidermal growth factor receptor 2 (HER2) humanized antibody] using peripheral blood mononuclear cells (PBMC) from breast cancer patients as effector cells. EXPERIMENTAL DESIGN: Thirty volunteers, including 20 breast cancer patients and 10 normal healthy control donors, were recruited randomly, and aliquots of peripheral blood were collected. ADCC of commercial trastuzumab (fucosylated) and its fucose-negative version were measured using PBMCs drawn from the volunteers as effector cells and two breast cancer cell lines with different HER2 expression levels as target cells. Relationships between cytotoxicity and characteristics of the patients, such as content of natural killer cells in PBMCs, type of therapy, FCGR3A genotypes, etc. were also analyzed. RESULTS: ADCC was significantly enhanced with the fucose-negative antibody compared with the fucose-positive antibody using PBMCs from either normal donors or breast cancer patients. Enhancement of ADCC was observed irrespective of the various clinical backgrounds of the patients, even in the chemotherapy cohort that presented with a reduced number of natural killer cells and weaker ADCC. CONCLUSIONS: This preliminary study suggests that the use of fucose-negative antibodies may improve the therapeutic effects of anti-HER2 therapy for patients independent of clinical backgrounds.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Neoplasias da Mama/imunologia , Fucose/química , Receptor ErbB-2/imunologia , Adulto , Idoso , Animais , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Humanos , Imunoterapia , Leucócitos/efeitos dos fármacos , Pessoa de Meia-Idade , Receptores de IgG/genética , Trastuzumab
18.
Glycobiology ; 17(1): 104-18, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17012310

RESUMO

The structure of asparagine-linked oligosaccharides attached to the antibody constant region (Fc) of human immunoglobulin G1 (IgG1) has been shown to affect the pharmacokinetics and antibody effector functions of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). However, it is still unclear how differences in the N-linked oligosaccharide structures impact the biological activities of antibodies, especially those lacking core fucose. Here, we succeeded in generating core fucose-lacking human IgG1 antibodies with three different N-linked Fc oligosaccharides, namely, a high-mannose, hybrid, and complex type, using the same producing clone, and compared their activities. Cultivation of an alpha-1,6-fucosyltransferase (FUT8) knockout Chinese hamster ovary cell line in the presence or absence of a glycosidase inhibitor (either swainsonine or kifunensine) yielded antibody production of each of the three types without contamination by the others. Two of three types of nonnaturally occurring atypical oligosaccharide IgG1, except the complex type, reduced the affinity for both human lymphocyte receptor IIIa (FcgammaRIIIa) and the C1q component of the complement, resulting in reduction of ADCC and CDC. The bulky structure of the nonreducing end of N-linked Fc oligosaccharides is considered to contribute the CDC change, whereas the structural change in the reducing end, i.e. the removal of core fucose, causes ADCC enhancement through improved FcgammaRIIIa binding. In the pharmacokinetic profile, although no significant difference of human neonatal Fc receptor (FcRn)-binding affinity was observed among the three types, the complex type showed longer serum half-lives than the other types irrespective of core fucosylation in mice, which also suggests the contribution of the nonreducing end structure. The present study provides basic information on the effects of core fucose-lacking N-linked Fc oligosaccharides on antibody biological activities.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Fatores Imunológicos/química , Mananas/química , Oligossacarídeos/química , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Citotoxicidade Celular Dependente de Anticorpos/fisiologia , Células CHO , Sequência de Carboidratos , Complemento C1q/metabolismo , Proteínas do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/fisiologia , Cricetinae , Cricetulus , Citotoxicidade Imunológica/fisiologia , Feminino , Fucosiltransferases/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Imunoglobulina G/uso terapêutico , Fatores Imunológicos/uso terapêutico , Taxa de Depuração Metabólica , Camundongos , Dados de Sequência Molecular , Mieloma Múltiplo/tratamento farmacológico , Organismos Geneticamente Modificados , Ligação Proteica , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Rituximab , Relação Estrutura-Atividade
19.
Expert Opin Biol Ther ; 6(11): 1161-73, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17049014

RESUMO

Most of the existing therapeutic antibodies that have been licensed and developed as medical agents are of the human IgG1 isotype, the molecular weight of which is approximately 150 kDa. Human IgG1 is a glycoprotein bearing two N-linked biantennary complex-type oligosaccharides bound to the antibody constant region (Fc), in which the majority of the oligosaccharides are core fucosylated, and it exercises the effector functions of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity through the interaction of the Fc with either leukocyte receptors (FcgammaRs) or complement. Recently, therapeutic antibodies have been shown to improve overall survival as well as time to disease progression in a variety of human malignancies, such as breast, colon and haematological cancers, and genetic analysis of FcgammaR polymorphisms of cancer patients has demonstrated that ADCC is a major antineoplasm mechanism responsible for clinical efficacy. However, the ADCC of existing licensed therapeutic antibodies has been found to be strongly inhibited by serum due to nonnpecific IgG competing for binding of the therapeutics to FcgammaRIIIa on natural killer cells, which leads to the requirement of a significant amount of drug and very high costs associated with such therapies. Moreover, enhanced ADCC of non-fucosylated forms of therapeutic antibodies through improved FcgammaRIIIa binding is shown to be inhibited by the fucosylated counterparts. In fact, non-fucosylated therapeutic antibodies, not including the fucosylated forms, exhibit the strongest and most saturable in vitro and ex vivo ADCC among such antibody variants with improved FcgammaRIIIa binding as those bearing naturally occurring oligosaccharide heterogeneities and artificial amino acid mutations, even in the presence of plasma IgG. Robust stable production of completely non-fucosylated therapeutic antibodies in a fixed quality has been achieved by the generation of a unique host cell line, in which the endogenous alpha-1,6-fucosyltransferase (FUT8) gene is knocked out. Thus, the application of non-fucosylated antibodies is expected to be a promising approach as next-generation therapeutic antibodies with improved efficacy, even when administrated at low doses in humans in vivo. Clinical trials using non-fucosylated antibody therapeutics are underway at present.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Fucose/uso terapêutico , Animais , Anticorpos/genética , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/genética , Fucose/metabolismo , Humanos , Neoplasias/imunologia , Neoplasias/terapia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA