Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cytokine ; 98: 59-70, 2017 10.
Article in English | MEDLINE | ID: mdl-28863833

ABSTRACT

T helper 2 cells produce a number of cytokines including inteleukin (IL)-5, IL-4 and IL-13. Group 2 innate lymphoid cells (ILC2s) also produce IL-5 under sterile conditions. IL-5 is interdigitating homodimeric glycoprotein and a member of the four α helical bundle motifs conserved among hematopoietic cytokines. IL-5 exerts its effects on target cells via IL-5 receptor (IL-5R), composed of an IL-5R α and ßc subunit. The membrane proximal proline-rich motif of the cytoplasmic domain of both IL-5R α and ßc subunits is essential for IL-5 signal transduction. Although IL-5 was initially identified by its ability to support the growth and terminal differentiation of mouse B cells into antibody-secreting cells, recombinant IL-5 exerts pleiotropic activities on various target cells. For example, IL-5 is now recognized as the major maturation and differentiation factor for eosinophils in mice and humans. Overexpression of IL-5 in mouse significantly increases eosinophil numbers and antibody levels in vivo, while mice lacking a functional gene for IL-5 or IL-5R display developmental and functional impairments in B cell and eosinophil lineages. In mice, the role of the IL-5/IL-5R system in the production and secretion of Immunoglobulin (Ig) M and IgA in mucosal tissues has been reported. Although eosinophils protect against invading pathogens including virus, bacteria and helminthes, they are also involved in the pathogenesis of various diseases, such as food allergy, asthma, and inflammatory bowel diseases. The recent expansion in our understanding in the context of IL-5 and IL-5-producing ILC2s in eosinophil activation and the pathogenesis of eosinophil-dependent inflammatory diseases has led to advances in therapeutic options. A new therapy currently under invetigarion in clinical trials uses humanized monoclonal antibodies against IL-5 or the IL-5R. In this review, we summarize our current understanding of the functions of IL-5 and its receptor, the innate regulation of IL-5-producing cells, and therapeutic potential of anti-IL-5 and anti-eosinophil (IL-5R) antibodies.


Subject(s)
Hypersensitivity , Interleukin-5/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Clinical Trials as Topic , Eosinophils/immunology , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Interleukin-5/antagonists & inhibitors , Interleukin-5/genetics , Lymphocytes/immunology , Mice , Receptors, Interleukin-5/immunology , Receptors, Interleukin-5/metabolism , Signal Transduction/immunology , Th2 Cells/immunology
2.
Glycobiology ; 26(5): 482-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26747427

ABSTRACT

The structure of the N-linked oligosaccharides attached to antithrombin (AT) has been shown to affect its anticoagulant activity and pharmacokinetics. Human AT has biantennary complex-type oligosaccharides with the unique feature of lacking a core fucose, which affects its biological activities by changing its heparin-binding affinity. In human plasma, AT circulates as a mixture of the α-form bearing four oligosaccharides and the ß-form lacking an oligosaccharide at Asn135. However, it remains unclear how the immature high-mannose-type oligosaccharides produced by mammalian cells affect biological activities of AT. Here, we succeeded in directly comparing the activities between the high-mannose and complex types. Interestingly, although there were no substantial differences in thrombin inhibitory activity, the high-mannose type showed higher heparin-binding affinity. The anticoagulant activities were increased by heparin and correlated with the heparin-binding affinity, resulting in the strongest anticoagulant activity being displayed in the ß-form with the high-mannose type. In pharmacokinetic profiling, the high-mannose type showed a much shorter plasma half-life than the complex type. The ß-form was found to have a prolonged plasma half-life compared with the α-form for the high-mannose type; conversely, the α-form showed a longer half-life than the ß-form for the complex-type. The present study highlights that AT physiological activities are strictly controlled not only by a core fucose at the reducing end but also by the high-mannose-type structures at the nonreducing end. The ß-form with the immature high-mannose type appears to function as a more potent anticoagulant than the AT typically found in human plasma, once it emerges in the blood.


Subject(s)
Antithrombin Proteins/metabolism , Heparin/metabolism , Mannose/metabolism , Oligosaccharides/metabolism , Antithrombin Proteins/chemistry , Glycosylation , Heparin/chemistry , Humans , Mannose/chemistry , Oligosaccharides/chemistry , Protein Binding
3.
PLoS One ; 10(10): e0140120, 2015.
Article in English | MEDLINE | ID: mdl-26444434

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is an important effector function determining the clinical efficacy of therapeutic antibodies. Core fucose removal from N-glycans on the Fc portion of immunoglobulin G (IgG) improves the binding affinity for Fcγ receptor IIIa (FcγRIIIa) and dramatically enhances ADCC. Our previous structural analyses revealed that Tyr-296 of IgG1-Fc plays a critical role in the interaction with FcγRIIIa, particularly in the enhanced FcγRIIIa binding of nonfucosylated IgG1. However, the importance of the Tyr-296 residue in the antibody in the interaction with various Fcγ receptors has not yet been elucidated. To further clarify the biological importance of this residue, we established comprehensive Tyr-296 mutants as fucosylated and nonfucosylated anti-CD20 IgG1s rituximab variants and examined their binding to recombinant soluble human Fcγ receptors: shFcγRI, shFcγRIIa, shFcγRIIIa, and shFcγRIIIb. Some of the mutations affected the binding of antibody to not only shFcγRIIIa but also shFcγRIIa and shFcγRIIIb, suggesting that the Tyr-296 residue in the antibody was also involved in interactions with FcγRIIa and FcγRIIIb. For FcγRIIIa binding, almost all Tyr-296 variants showed lower binding affinities than the wild-type antibody, irrespective of their core fucosylation, particularly in Y296K and Y296P. Notably, only the Y296W mutant showed improved binding to FcγRIIIa. The 3.00 Å-resolution crystal structure of the nonfucosylated Y296W mutant in complex with shFcγRIIIa harboring two N-glycans revealed that the Tyr-to-Trp substitution increased the number of potential contact atoms in the complex, thus improving the binding of the antibody to shFcγRIIIa. The nonfucosylated Y296W mutant retained high ADCC activity, relative to the nonfucosylated wild-type IgG1, and showed greater binding affinity for FcγRIIa. Our data may improve our understanding of the biological importance of human IgG1-Fc Tyr-296 in interactions with various Fcγ receptors, and have applications in the modulation of the IgG1-Fc function of therapeutic antibodies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Fucose/immunology , Immunoglobulin Fc Fragments/immunology , Receptors, IgG/immunology , Animals , Antigens, CD20/immunology , CHO Cells , Cell Line , Cricetulus , Crystallography, X-Ray , Fucose/chemistry , Fucose/genetics , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Models, Molecular , Point Mutation , Rituximab/chemistry , Rituximab/genetics , Rituximab/immunology
4.
J Pharm Sci ; 104(3): 930-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25583555

ABSTRACT

Monoclonal antibodies have demonstrated enormous potential as new classes of drugs that confer great benefits to patients, and more than 40 therapeutic antibodies have already been approved for clinical use. In particular, the past 5 years might be recognized as the period guiding the new era for "engineered antibodies," with the successful approval of numerous antibody-drug conjugates, bispecific antibodies, and glyco-engineered antibodies for clinical applications. In this review, we summarize the development of antibody engineering technologies that are proving their concepts in the clinic, mainly focusing on the latest trends in defucosylated antibody technologies.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Monoclonal/biosynthesis , Immunoconjugates/metabolism , Protein Engineering , Technology, Pharmaceutical/methods , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Glycosylation , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Immunotherapy/methods , Protein Conformation , Protein Processing, Post-Translational , Structure-Activity Relationship
5.
Genes Cells ; 16(11): 1071-80, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22023369

ABSTRACT

Removal of the fucose residue from the N-glycans of the Fc portion of immunoglobulin G (IgG) results in a dramatic enhancement of antibody-dependent cellular cytotoxicity (ADCC) through improved affinity for Fcγ receptor IIIa (FcγRIIIa). Here, we present the 2.2-Šstructure of the complex formed between nonfucosylated IgG1-Fc and a soluble form of FcγRIIIa (sFcγRIIIa) with two N-glycosylation sites. The crystal structure shows that one of the two N-glycans of sFcγRIIIa mediates the interaction with nonfucosylated Fc, thereby stabilizing the complex. However, fucosylation of the Fc N-glycans inhibits this interaction, because of steric hindrance, and furthermore, negatively affects the dynamics of the receptor binding site. Our results offer a structural basis for improvement in ADCC of therapeutic antibodies by defucosylation.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Polysaccharides/chemistry , Receptors, IgG/chemistry , Receptors, IgG/immunology , Antibodies/chemistry , Antibodies/immunology , Antibodies/therapeutic use , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Polysaccharides/immunology , Protein Conformation
6.
Int J Oncol ; 39(1): 203-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21503570

ABSTRACT

Golgi phosphoprotein 2 (GP73) is a type II Golgi protein, which was found on examination of the fucosylated proteome as a potential tumor marker for hepatocellular carcinoma (HCC). The serum levels of both total and fucosylated GP73 were increased in the sera of patients with HCC. Fucosylation is one of the most important oligosaccharide modifications involved in cancer and is catalyzed by α1,6-fucosyltransferase (Fut8). In the present study, we investigated the effect of Fut8 overexpression on GP73 production in the human hepatoma cell line Hep3B. The Fut8 expression vector was transfected into Hep3B cells and the expression of GP73 was investigated by Western blotting and real-time PCR. Overexpression of Fut8 dramatically enhanced the expression of GP73 at the transcriptional level. Surprisingly, this effect was not dependent on cellular fucosylation. Overexpression of a mutant Fut8, which was unable to be localized to the Golgi, did not induce GP73 production, suggesting that the localization of Fut8 in the Golgi apparatus was important for the increase in GP73 expression. This is the first demonstration of GP73 regulation through overexpression of a glycosyltransferase, which may lead to Golgi stress.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Fucosyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/physiopathology , Membrane Proteins/metabolism , Animals , Base Sequence , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Cricetinae , Fucosyltransferases/genetics , Gene Expression , Genetic Vectors/genetics , Humans , Intracellular Space/enzymology , Intracellular Space/metabolism , Liver Neoplasms/enzymology , Membrane Proteins/genetics , Mutation/genetics , Protein Transport , Sequence Alignment , Transfection
7.
J Proteome Res ; 9(10): 5047-52, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20681733

ABSTRACT

Peptide hormones and neuropeptides constitute an important class of naturally occurring peptides that are generated from precursor proteins by limited proteolytic processing. An important but unaddressed issue in peptidomics is to pin down novel bioactive peptides in a bulk of peptide sequences provided by tandem mass spectrometry. Here, we describe an approach to simultaneously screen for bioactive peptides and their target tissues. The principle behind this approach is to identify intact secretory peptides that have the ability to raise intracellular calcium levels. In practice, we used nanoflow liquid chromatography-tandem mass spectrometry to analyze peptides released by exocytosis from cultured cells. Peptide sequence information was utilized to deduce intact peptide forms, among which those highly conserved between species are selected and tested on an ex vivo calcium assay using tissue pieces from transgenic mice that systemically express the calcium indicator apoaequorin. The calcium assay can be applied to various cell types, including those not amenable to in vitro culture. We used this approach to identify novel bioactive neuropeptides derived from the neurosecretory protein VGF, which evoke a calcium response in the pituitary and hypothalamus.


Subject(s)
Neuropeptides/metabolism , Organ Specificity , Peptides/metabolism , Proteomics/methods , Aequorin/genetics , Aequorin/metabolism , Amino Acid Sequence , Animals , Apoproteins/genetics , Apoproteins/metabolism , Cell Line, Tumor , Chromatography, Liquid , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Neuropeptides/genetics , Peptides/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tandem Mass Spectrometry
8.
Biochem Biophys Res Commun ; 394(3): 498-502, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20197059

ABSTRACT

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


Subject(s)
Antibodies, Monoclonal/immunology , Cytotoxicity, Immunologic , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/analysis , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Mice , Mice, SCID , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/immunology , Xenograft Model Antitumor Assays
9.
Leuk Res ; 34(5): 666-71, 2010 May.
Article in English | MEDLINE | ID: mdl-20022111

ABSTRACT

We demonstrate herein the augmentation of rituximab-mediated apoptosis in lymphoma cell lines by cross-linking with recombinant FcgammaRs, which is further enhanced by using a nonfucosylated variant of rituximab having strong FcgammaRIII-binding capacity. Furthermore, we show that neutrophils can serve as physiological cross-linkers that augment anti-CD20-mediated apoptosis, as evidenced by (i) the neutrophil-augmented apoptosis was more profound for the nonfucosylated variant of rituximab and (ii) the mechanism depended on FcgammaRIIIb but not on FcgammaRIIa. Taken together, we suggest a potential anti-tumour mechanism of nonfucosylated anti-CD20 antibody by which antibody molecules are cross-linked through enhanced interaction with FcgammaRIIIb in neutrophils.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphoma/immunology , Neutrophils/drug effects , Receptors, IgG/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/metabolism , Apoptosis/immunology , Cell Line, Tumor , Cross-Linking Reagents , Humans , Neutrophils/immunology , Neutrophils/metabolism , Receptors, IgG/immunology , Rituximab
10.
Drug Des Devel Ther ; 3: 7-16, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19920917

ABSTRACT

As platforms for therapeutic agents, monoclonal antibodies (MAbs) have already been approved, and several MAbs have demonstrated clinical effectiveness in a variety of malignancies. However, several issues have also been emerging in antibody therapy, such as high cost and insufficient drug action. Recently, to improve MAb activity in humans, effector functions have been subjects of focus, especially antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Extensive efforts have been made to enhance these effector functions of MAbs, and successful approaches have been reported by us and others, wherein the binding activity of MAbs to FcgammaRIIIa or C1q is increased by introducing amino acid mutations into heavy chain constant regions or through glyco-modification of Fc-linked oligosaccharides. In addition, one of the next approaches to optimizing therapeutic antibodies would be to combine multiple enhancing modifications into a single antibody platform to overcome the diverse mechanisms of clinical resistance of tumor cells. For this aim, we have recently developed a successful combination composed of ADCC-enhancing modification by the fucose depletion from Fc-linked oligosaccharides and CDC-enhancing modification by IgG1 and IgG3 isotype shuffling in heavy chains, which could be of great value for the development of third-generation antibody therapeutics.

11.
Cancer Sci ; 100(12): 2411-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19758394

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , Antineoplastic Agents/pharmacology , Complement Activation/drug effects , Lymphoma/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Complement System Proteins/immunology , Cricetinae , Cricetulus , Humans , Protein Engineering , Rituximab
12.
Cancer Sci ; 100(9): 1566-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19538497

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Neoplasms/therapy , Protein Engineering , Animals , Humans
13.
Exp Hematol ; 37(3): 309-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19218011

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Histocompatibility Antigens Class II/genetics , Lymphoma, B-Cell/drug therapy , Neutrophils/immunology , Phagocytosis/drug effects , Receptors, IgG/metabolism , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Murine-Derived , Blood Cells , Cell Death/drug effects , Cells, Cultured , Flow Cytometry , GPI-Linked Proteins , Gene Expression/drug effects , Humans , Lymphoma, B-Cell/pathology , Mice , Protein Binding , Rituximab
14.
BMC Cancer ; 9: 58, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19226457

ABSTRACT

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).


Subject(s)
Antibodies/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Blood/immunology , Fucose/immunology , Adult , Antibodies/genetics , Antibodies/immunology , Antigen-Antibody Reactions , Antigens, CD20/genetics , Antigens, CD20/immunology , B-Lymphocytes/immunology , Cells, Cultured , Female , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Young Adult
15.
Glycobiology ; 19(2): 126-34, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18952826

ABSTRACT

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.


Subject(s)
Fucose/metabolism , Immunoglobulin G/metabolism , Receptors, IgG/chemistry , Receptors, IgG/metabolism , Amino Acid Sequence , Animals , Antibodies/immunology , Antibodies/metabolism , Antigens, CD20/immunology , Antigens, CD20/metabolism , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Models, Biological , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Receptors, IgG/immunology , Surface Plasmon Resonance
16.
MAbs ; 1(3): 230-6, 2009.
Article in English | MEDLINE | ID: mdl-20065644

ABSTRACT

The clinical success of therapeutic antibodies is demonstrated by the number of antibody therapeutics that have been brought to market and the increasing number of therapeutic antibodies in development. Recombinant antibodies are molecular-targeted therapeutic agents and represent a major new class of drugs. However, it is still very important to optimize and maximize the clinical efficacy of therapeutic antibodies, in part to help lower the cost of therapeutic antibodies by potentially reducing the dose or the duration of treatment. Clinical trials using therapeutic antibodies fully lacking core fucose residue in the Fc oligosaccharides are currently underway, and their remarkable physiological activities in humans in vivo have attracted attention as next-generation therapeutic antibody approaches with improved efficacy. Thus, an industrially applicable antibody production process that provides consistent yields of fully non-fucosylated antibody therapeutics with fixed quality has become a key goal in the successful development of next-generation therapeutic agents. In this article, we review the current technologies for production of therapeutic antibodies with control of fucosylation of the Fc N-glycans.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Fucose/metabolism , Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Biotechnology , Drug Design , Fucose/chemistry , Fucose/genetics , Humans , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Constant Regions/metabolism , Neoplasms/immunology , Protein Binding/drug effects , Protein Binding/genetics , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
17.
Cancer Res ; 68(10): 3863-72, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483271

ABSTRACT

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.


Subject(s)
Antigens, CD20/chemistry , Immunoglobulin G/chemistry , Protein Engineering/methods , Animals , Antigens, CD/chemistry , Antigens, Neoplasm/chemistry , CD52 Antigen , CHO Cells , Carbohydrates/chemistry , Cell Line, Tumor , Cricetinae , Cricetulus , Glycoproteins/chemistry , Humans , Immunoglobulin Isotypes , Leukocytes, Mononuclear/cytology , Protein Structure, Tertiary
18.
Methods Mol Biol ; 435: 1-16, 2008.
Article in English | MEDLINE | ID: mdl-18370064

ABSTRACT

Chinese hamster ovary (CHO) cells are the most common host cells and are widely used in the manufacture of approved recombinant therapeutics. They represent a major new class of universal hosts in biopharmaceutical production. However, there remains room for improvement to create more ideal host cells that can add greater value to therapeutic recombinant proteins at reduced production cost. A promising approach to this goal is biallelic gene knockout in CHO cells, as it is the most reliable and effective means to permanent phenotypic change, owing to the complete removal of gene function. In this chapter, we describe a biallelic gene knockout process in CHO cells, as exemplified by the successful targeted disruption of both FUT8 alleles encoding alpha-1,6-fucosyltransferase gene in CHO/DG44 cells. Wild-type alleles are sequentially disrupted by homologous recombination using two targeting vectors to generate homozygous disruptants, and the drug-resistance gene cassettes remaining on the alleles are removed by a Cre/loxP recombination system so as not to leave the extraphenotype except for the functional loss of the gene of interest.


Subject(s)
CHO Cells/enzymology , Gene Targeting/methods , Alleles , Animals , Base Sequence , Blotting, Southern , CHO Cells/drug effects , Cricetinae , Cricetulus , DNA Primers/genetics , Drug Resistance/genetics , Fucosyltransferases/genetics , Gene Deletion , Genetic Vectors , Phenotype , Polymerase Chain Reaction , Recombination, Genetic , Transfection
20.
BMC Biotechnol ; 7: 84, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-18047682

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Fucosyltransferases/genetics , Gene Silencing/immunology , Genetic Enhancement/methods , Hydro-Lyases/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Fucosyltransferases/immunology , Hydro-Lyases/immunology , Protein Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...