Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Mol Biol ; 368(3): 767-79, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17368483

RESUMO

Removal of the fucose residue from the oligosaccharides attached to Asn297 of human immunoglobulin G1 (IgG1) results in a significant enhancement of antibody-dependent cellular cytotoxicity (ADCC) via improved IgG1 binding to Fcgamma receptor IIIa. To provide structural insight into the mechanisms of affinity enhancement, we determined the crystal structure of the nonfucosylated Fc fragment and compared it with that of fucosylated Fc. The overall conformations of the fucosylated and nonfucosylated Fc fragments were similar except for hydration mode around Tyr296. Stable-isotope-assisted NMR analyses confirmed the similarity of the overall structures between fucosylated and nonfucosylated Fc fragments in solution. These data suggest that the glycoform-dependent ADCC enhancement is attributed to a subtle conformational alteration in a limited region of IgG1-Fc. Furthermore, the electron density maps revealed that the traces between Asp280 and Asn297 of our fucosylated and nonfucosylated Fc crystals were both different from that in previously reported isomorphous Fc crystals.


Assuntos
Aminoácidos/química , Fucose/química , Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Modelos Moleculares , Animais , Células CHO , Sequência de Carboidratos , Cricetinae , Cricetulus , Cristalografia por Raios X , Glicosilação , Humanos , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Conformação Proteica , Soluções
8.
Dev Growth Differ ; 50(2): 97-107, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18067585

RESUMO

Cleavage of the extracellular matrix (ECM) by proteolysis unmasks cryptic sites and generates novel fragments with biological activities functionally distinct from those of the intact ECM molecule. The laminin G-like (LG)4-5 fragment has been shown to be excised from the laminin alpha4 chain in various tissues. However, the functional role of this fragment has remained unknown to date. To investigate this, we prepared alpha4 LG1-3 and alpha4 LG4-5 fragments by elastase digestion of recombinant alpha4 LG1-5, and examined their effects on de novo adipogenesis in mice at the site of injection of basement membrane extract (Matrigel) and fibroblast growth factor (FGF)-2. Although the addition of whole alpha4 LG1-5 suppressed adipogenesis to some extent, the alpha4 LG4-5 fragment could strongly suppress adipogenesis at a concentration of less than 20 nm. Addition of the alpha4 LG4 module, which contains a heparin-binding region, had a suppressive effect, but this was lost in mutants with reduced heparin-binding activity. In addition, antibodies against the extracellular domain of syndecan-2 and -4, which are known receptors for the alpha4 LG4 module, suppressed adipogenesis. Thus, these results suggest that the cryptic alpha4 LG4-5 fragment derived from the laminin alpha4 chain inhibits de novo adipogenesis by modulating the effect of FGF-2 through syndecans.


Assuntos
Adipogenia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Laminina/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/metabolismo , Animais , Células CHO , Células Cultivadas , Cromatografia de Afinidade , Colágeno/metabolismo , Cricetinae , Cricetulus , Combinação de Medicamentos , Heparina/metabolismo , Técnicas In Vitro , Laminina/química , Laminina/genética , Camundongos , Camundongos Nus , Modelos Biológicos , Elastase Pancreática/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Suínos , Sindecana-2/química , Sindecana-2/metabolismo , Sindecana-4/química , Sindecana-4/metabolismo
9.
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
10.
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
11.
Biochim Biophys Acta ; 1760(4): 693-700, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16343775

RESUMO

The Fc portion of immunoglobulin G (IgG) expresses the biantennary complex type oligosaccharides at Asn297 of the C(H)2 domain of each heavy chain with microheterogeneities depending on physiological and pathological states. These N-glycans are known to be essential for promotion of proper effector functions of IgG such as complement activation and Fcgamma receptor (FcgammaR)-mediated activities. To gain a better understanding of the role of Fc glycosylation, we prepared a series of truncated glycoforms of human IgG1-Fc and analyzed their interactions with human soluble FcgammaRIIIa (sFcgammaRIIIa) and with staphylococcal protein A by surface plasmon resonance and nuclear magnetic resonance (NMR) methods. Progressive but less pronounced reductions in the affinity for sFcgammaRIIIa were observed as a result of the galactosidase and subsequent N-acetylhexosaminidase treatments of IgG1-Fc. The following endoglycosidase D treatment, giving rise to a disaccharide structure composed of a fucosylated GlcNAc, abrogated the affinity of IgG1-Fc for sFcgammaRIIIa. On the other hand, those glycosidase treatments did not significantly affect the affinity of IgG1-Fc for protein A. Inspection of stable-isotope-assisted NMR data of a series of Fc glycoforms indicates that the stepwise trimming out of the carbohydrate residues results in concomitant increase in the number of amino acid residues perturbed thereby in the C(H)2 domains. Furthermore, the cleavage at the GlcNAcbeta1-4GlcNAc glycosidic linkage induced the conformational alterations of part of the lower hinge region, which makes no direct contact with the carbohydrate moieties and forms the major FcgammaR-binding site, while the conformation of the C(H)2/C(H)3 interface was barely perturbed that is the protein A-binding site. These results indicate that the carbohydrate moieties are required for maintaining the structural integrity of the FcgammaR-binding site.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Ressonância Magnética Nuclear Biomolecular , Oligossacarídeos/análise , Sítios de Ligação , Glicosídeo Hidrolases/metabolismo , Glicosilação , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Ligação Proteica , Conformação Proteica , Receptores de IgG/metabolismo , Proteína Estafilocócica A/metabolismo
12.
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
13.
J Biotechnol ; 130(3): 300-10, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17559959

RESUMO

Currently, removal of core fucose from the Fc oligosaccharides of therapeutic antibodies is widely recognized as being of great importance for the effector function of antibody-dependent cellular cytotoxicity, and alpha-1,6-fucosyltransferase (FUT8) knockout cells have been generated as an ideal host cell line for manufacturing such therapeutics. Here, we attempted to identify genes other than FUT8 that could be targeted for the manufacture of non-fucosylated therapeutics. Loss-of-function analyses using siRNAs against three key genes involved in oligosaccharide fucosylation in Chinese hamster ovary (CHO) cells revealed that there was a positive correlation between the Fc oligosaccharide fucosylation and the mRNA expression through the origin in the cases of both GDP-fucose 4,6-dehydratase (GMD) and FUT8, but not for the GDP-fucose transporter, suggesting that there is no functional redundancy in GMD and FUT8. GMD knockout CHO/DG44 cells were successfully established, and were confirmed to be devoid of intracellular GDP-fucose and to produce completely non-fucosylated antibodies. GMD knockout cells recovered their fucosylation capability through the salvage pathway upon addition of l-fucose into the culture medium, and exhibited equable morphology, growth kinetics and recombinant protein productivity, demonstrating that loss of oligosaccharide fucosylation has no impact on these cellular phenotypes. Our results demonstrate that GMD knockout is a new strategy applicable to the manufacture of non-fucosylated therapeutic antibodies, and completely O-fucose-negative therapeutics as well.


Assuntos
Biotecnologia/métodos , Fucose/metabolismo , Hidroliases/deficiência , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Animais , Antígenos CD20/imunologia , Células CHO , Cricetinae , Cricetulus , Meios de Cultura Livres de Soro , DNA Complementar , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Hidroliases/genética , Hidroliases/metabolismo , Imunoglobulina G/imunologia , Camundongos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Oligossacarídeos/metabolismo , Lectinas de Plantas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
14.
Clin Cancer Res ; 12(9): 2879-87, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16675584

RESUMO

PURPOSE: Recent studies have revealed that fucosylated therapeutic IgG1s need high concentrations to compensate for FcgammaRIIIa-competitive inhibition of antibody-dependent cellular cytotoxicity (ADCC) by endogenous human plasma IgG. Here, we investigated whether ADCC of nonfucosylated therapeutic IgG1 is also influenced by plasma IgG in the same way as fucosylated IgG1s. EXPERIMENTAL DESIGN: Ex vivo ADCC upon CD20+ human B cells was induced by incubation of human whole blood with nonfucosylated and/or fucosylated anti-CD20 IgG1s rituximab, and quantified by measuring the remaining CD19+ human B cells using flow cytometry. RESULTS: Nonfucosylated anti-CD20 showed markedly higher (over 100-fold based on EC50) ex vivo B-cell depletion activity than its fucosylated counterpart in the presence of plasma IgG. The efficacy of fucosylated anti-CD20 was greatly diminished in plasma, resulting in the need for a high concentration (over 1.0 microg/mL) to achieve saturated efficacy. In contrast, nonfucosylated anti-CD20 reached saturated ADCC at lower concentrations (0.01-0.1 microg/mL) with much higher efficacy than fucosylated anti-CD20 in all nine donors through improved FcgammaRIIIa binding. Noteworthy, the high efficacy of nonfucosylated anti-CD20 was inhibited by addition of fucosylated anti-CD20. Thus, the efficacy of a 1:9 mixture (10 microg/mL) of nonfucosylated and fucosylated anti-CD20s was inferior to that of a 1,000-fold dilution (0.01 microg/mL) of nonfucosylated anti-CD20 alone. CONCLUSIONS: Our data showed that nonfucosylated IgG1, not including fucosylated counterparts, can evade the inhibitory effect of plasma IgG on ADCC through its high FcgammaRIIIa binding. Hence, nonfucosylated IgG1 exhibits strong therapeutic potential through dramatically enhanced ADCC at low doses in humans in vivo.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Imunoglobulina G/uso terapêutico , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Linfócitos B/imunologia , Doadores de Sangue , Células CHO , Cricetinae , Fucose/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/genética , Depleção Linfocítica , Camundongos , Receptor ErbB-2/imunologia , Rituximab
15.
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
16.
J Biochem ; 140(3): 359-68, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16861252

RESUMO

Bispecific antibodies (bsAbs) have the potential to extend binding selectivity, increase avidity and exert potent cytotoxicity due to the combination of dual specificities. scFv2-Fc type of single-gene-encoded bispecific antibody, composed of two different single-chain Fvs and an Fc, has been reported to be capable of binding to different antigens. The aim of this study was to determine the effect of fucose removal on effector functions of scFv2-Fc since fucose depletion from oligosaccharide of human IgG1 and scFv-Fc results in significant enhancement of ADCC. We generated novel single-gene-encoded bsAb with dual specificity against tumor associated glycoprotein (TAG)-72 and MUC1 mucin as fucose-negative scFv2-Fc from alpha-1,6-fucosyltransferase knock-out CHO cells and a highly fucosylated scFv2-Fc comparator from parental CHO cells. Expression, assembly and the antigen-binding activity of the scFv2-Fc were not influenced by removal of fucose. The fucose negative scFv2-Fc bound with higher avidity to FcgammaRIIIa and enhanced ADCC compared to the highly fucosylated scFv2-Fc. These results demonstrate that ADCC-enhancement by removal of fucose is effective in not only whole IgG1 and scFv-Fc, but also scFv2-Fc targeting two different antigens, and thus increases the potential of fucose-negative scFv2-Fcs as novel therapeutic candidates.


Assuntos
Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/imunologia , Especificidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Fucose/química , Oligossacarídeos/química , Animais , Antígenos de Neoplasias/imunologia , Células CHO , Cricetinae , Cricetulus , Vetores Genéticos/genética , Glicoproteínas/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Mucina-1/imunologia
17.
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
18.
Clin Cancer Res ; 11(10): 3897-904, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15897591

RESUMO

PURPOSE: Fibroblast growth factor 8b (FGF8b) has been implicated in oncogenesis of sex hormone-related malignancies. A murine monoclonal anti-FGF8 antibody, KM1334, has been raised against a FGF8b-derived peptide and shown to neutralize FGF8b activity in an androgen-dependent mouse mammary cell line (SC-3) in vitro growth. The purpose of this study was to evaluate KM1334 as a therapeutic agent for FGF8-dependent cancer. EXPERIMENTAL DESIGN: Specificity and neutralizing activity of KM1334 were examined in vitro. In vivo therapeutic studies were done in nude mice bearing SC-3 tumors s.c. RESULTS: KM1334 recognized FGF8b and FGF8f specifically out of four human FGF8 isoforms and showed little binding to other members of FGF family. Neutralizing activity of KM1334 was confirmed by both blocking of FGF8b binding to its three receptors (FGFR2IIIc, FGFR3IIIc, and FGFR4) and FGF8b-induced phosphorylation of FGFR substrate 2alpha and extracellular signal-regulated kinase 1/2 in SC-3 cells. The in vitro inhibitory effect could be extended to in vivo tumor models, where KM1334 caused rapid regression of established SC-3 tumors in nude mice. This rapid regression of tumors after KM1334 treatment was explained by two independent mechanisms: (a) decreased DNA synthesis, as evidenced by a decrease in uptake of 5-bromo-2'-deoxyuridine, and (b) induction of apoptosis as shown by the terminal deoxynucleotidyl transferase-mediated nick end labeling assay. CONCLUSIONS: KM1334 possesses strong blocking activity in vitro and antitumor activity in vivo and therefore may be an effective therapeutic candidate for the treatment of cancers that are dependent on FGF8b signaling for growth and survival.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos/imunologia , Fatores de Crescimento de Fibroblastos/imunologia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Animais , Proliferação de Células , DNA/biossíntese , Fator 8 de Crescimento de Fibroblasto , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Isoformas de Proteínas , Transdução de Sinais , Células Tumorais Cultivadas
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA