Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Immunol ; 197(12): 4829-4837, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27807190

ABSTRACT

Human IgG1 type I CD20 Abs, such as rituximab and ofatumumab (OFA), efficiently induce complement-dependent cytotoxicity (CDC) of CD20+ B cells by binding of C1 to hexamerized Fc domains. Unexpectedly, we found that type I CD20 Ab F(ab')2 fragments, as well as C1q-binding-deficient IgG mutants, retained an ability to induce CDC, albeit with lower efficiency than for whole or unmodified IgG. Experiments using human serum depleted of specific complement components demonstrated that the observed lytic activity, which we termed "accessory CDC," remained to be dependent on C1 and the classical pathway. We hypothesized that CD20 Ab-induced clustering of the IgM or IgG BCR was involved in accessory CDC. Indeed, accessory CDC was consistently observed in B cell lines expressing an IgM BCR and in some cell lines expressing an IgG BCR, but it was absent in BCR- B cell lines. A direct relationship between BCR expression and accessory CDC was established by transfecting the BCR into CD20+ cells: OFA-F(ab')2 fragments were able to induce CDC in the CD20+BCR+ cell population, but not in the CD20+BCR- population. Importantly, OFA-F(ab')2 fragments were able to induce CDC ex vivo in malignant B cells isolated from patients with mantle cell lymphoma and Waldenström macroglobulinemia. In summary, accessory CDC represents a novel effector mechanism that is dependent on type I CD20 Ab-induced BCR clustering. Accessory CDC may contribute to the excellent capacity of type I CD20 Abs to induce CDC, and thereby to the antitumor activity of such Abs in the clinic.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody-Dependent Cell Cytotoxicity , Antigens, CD20/metabolism , B-Lymphocytes/drug effects , Complement Pathway, Classical , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Rituximab/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, CD20/immunology , B-Lymphocytes/immunology , Cell Line, Tumor , Complement C1/metabolism , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Lymphoma, B-Cell/immunology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Rituximab/genetics , Rituximab/therapeutic use
2.
Methods ; 65(1): 5-10, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23872061

ABSTRACT

Transient expression systems in mammalian cells have become the method of choice for producing research quantities of antibodies. Both the speed and yield of the available transient systems and the natural posttranslational modifications favor these systems above expression in lower eukaryotes, prokaryotes or stable cell lines. We describe an optimized mammalian transient expression system, capable of producing up to 400mg/L of native secreted antibodies in less than a week. The system is composed of commercially available components and is based on expression in the fast growing suspension cell line, FreeStyle™ 293-F (HEK-293F). The method depends on an optimal combination of a gene transfer method, an expression vector and cotransfection with expression enhancing plasmids, encoding the large T antigen of the SV40 virus and the cell cycle inhibitors p21 and p27. Optimization of all components of the expression system, by experimental design techniques, yielded maximal expression levels (including antibody isotypes IgG1, 2, 3, 4 and Fab fragments of various species). Expression volumes were scalable from 0.1 ml up to 1.2L in a simple shaker flask system in animal component free, low protein medium, enabling consistent production of relatively high amounts of a large number of native antibodies.


Subject(s)
Immunoglobulin G/biosynthesis , Cloning, Molecular , Gene Expression , HEK293 Cells , Humans , Immunoglobulin G/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection
3.
J Immunol ; 189(7): 3430-8, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956577

ABSTRACT

Development of human therapeutic Abs has led to reduced immunogenicity and optimal interactions with the human immune system in patients. Humanization had as a consequence that efficacy studies performed in mouse models, which represent a crucial step in preclinical development, are more difficult to interpret because of gaps in our knowledge of the activation of murine effector cells by human IgG (hIgG) remain. We therefore developed full sets of human and mouse isotype variants of human Abs targeting epidermal growth factor receptor and CD20 to explore the crosstalk with mouse FcγRs (mFcγRs) and murine effector cells. Analysis of mFcγR binding demonstrated that hIgG1 and hIgG3 bound to all four mFcγRs, with hIgG3 having the highest affinity. hIgG1 nevertheless was more potent than hIgG3 in inducing Ab-dependent cellular cytotoxicity (ADCC) and Ab-dependent cellular phagocytosis with mouse NK cells, mouse polymorphonuclear leukocytes, and mouse macrophages. hIgG4 bound to all mFcγRs except mFcγRIV and showed comparable interactions with murine effector cells to hIgG3. hIgG4 is thus active in the murine immune system, in contrast with its inert phenotype in the human system. hIgG2 bound to mFcγRIIb and mFcγRIII, and induced potent ADCC with mouse NK cells and mouse polymorphonuclear leukocytes. hIgG2 induced weak ADCC and, remarkably, was unable to induce Ab-dependent cellular phagocytosis with mouse macrophages. Finally, the isotypes were studied in s.c. and i.v. tumor xenograft models, which confirmed hIgG1 to be the most potent human isotype in mouse models. These data enhance our understanding of the crosstalk between hIgGs and murine effector cells, permitting a better interpretation of human Ab efficacy studies in mouse models.


Subject(s)
Cytotoxicity Tests, Immunologic/methods , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antigens, CD20/immunology , Antigens, CD20/metabolism , Binding Sites, Antibody/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neutrophils/immunology , Neutrophils/metabolism
4.
J Immunol ; 187(6): 3383-90, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21832160

ABSTRACT

Ab-dependent cellular cytotoxicity (ADCC) is recognized as a prominent cytotoxic mechanism for therapeutic mAbs in vitro. However, the contribution of ADCC to in vivo efficacy, particularly for treatment of solid tumors, is still poorly understood. For zalutumumab, a therapeutic epidermal growth factor receptor (EGFR)-specific mAb currently in clinical development, previous studies have indicated signaling inhibition and ADCC induction as important therapeutic mechanisms of action. To investigate the in vivo role of ADCC, a panel of EGFR-specific mAbs lacking specific functionalities was generated. By comparing zalutumumab with mAb 018, an EGFR-specific mAb that induced ADCC with similar potency, but did not inhibit signaling, we observed that ADCC alone was insufficient for efficacy against established A431 xenografts. Interestingly, however, both zalutumumab and mAb 018 prevented tumor formation upon early treatment in this model. Zalutumumab and mAb 018 also completely prevented outgrowth of lung metastases, in A431 and MDA-MB-231-luc-D3H2LN experimental metastasis models, already when given at nonsaturating doses. Finally, tumor growth of mutant KRAS-expressing A431 tumor cells, which were resistant to EGFR signaling inhibition, was completely prevented by early treatment with zalutumumab and mAb 018, whereas ADCC-crippled N297Q-mutated variants of both mAbs did not show any inhibitory effects. In conclusion, ADCC induction by EGFR-specific mAbs represents an important mechanism of action in preventing tumor outgrowth or metastasis in vivo, even of cancers insensitive to EGFR signaling inhibition.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/immunology , ErbB Receptors/antagonists & inhibitors , Neoplasms, Experimental/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Separation , ErbB Receptors/immunology , Female , Flow Cytometry , Humans , Mice , Mice, SCID , Neoplasms, Experimental/metabolism , Xenograft Model Antitumor Assays
5.
J Immunol ; 187(6): 3238-46, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841137

ABSTRACT

A distinctive feature of human IgG4 is its ability to recombine half molecules (H chain and attached L chain) through a dynamic process termed Fab-arm exchange, which results in bispecific Abs. It is becoming evident that the process of Fab-arm exchange is conserved in several mammalian species, and thereby represents a mechanism that impacts humoral immunity more generally than previously thought. In humans, Fab-arm exchange has been attributed to the IgG4 core-hinge sequence (226-CPSCP-230) in combination with unknown determinants in the third constant H chain domain (CH3). In this study, we investigated the role of the CH3 domain in the mechanism of Fab-arm exchange, and thus identified amino acid position 409 as the critical CH3 determinant in human IgG, with R409 resulting in exchange and K409 resulting in stable IgG. Interestingly, studies with IgG from various species showed that Fab-arm exchange could not be assigned to a common CH3 domain amino acid motif. Accordingly, in rhesus monkeys (Macaca mulatta), aa 405 was identified as the CH3 determinant responsible (in combination with 226-CPACP-230). Using native mass spectrometry, we demonstrated that the ability to exchange Fab-arms correlated with the CH3-CH3 dissociation constant. Species-specific adaptations in the CH3 domain thus enable Fab-arm exchange by affecting the inter-CH3 domain interaction strength. The redistribution of Ag-binding domains between molecules may constitute a general immunological and evolutionary advantage. The current insights impact our view of humoral immunity and should furthermore be considered in the design and evaluation of Ab-based studies and therapeutics.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin G/chemistry , Immunoglobulin Heavy Chains/chemistry , Models, Molecular , Animals , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/chemistry , Macaca mulatta , Mass Spectrometry , Species Specificity
6.
J Immunol ; 186(3): 1840-8, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187443

ABSTRACT

CD38, a type II transmembrane glycoprotein highly expressed in hematological malignancies including multiple myeloma (MM), represents a promising target for mAb-based immunotherapy. In this study, we describe the cytotoxic mechanisms of action of daratumumab, a novel, high-affinity, therapeutic human mAb against a unique CD38 epitope. Daratumumab induced potent Ab-dependent cellular cytotoxicity in CD38-expressing lymphoma- and MM-derived cell lines as well as in patient MM cells, both with autologous and allogeneic effector cells. Daratumumab stood out from other CD38 mAbs in its strong ability to induce complement-dependent cytotoxicity in patient MM cells. Importantly, daratumumab-induced Ab-dependent cellular cytotoxicity and complement-dependent cytotoxicity were not affected by the presence of bone marrow stromal cells, indicating that daratumumab can effectively kill MM tumor cells in a tumor-preserving bone marrow microenvironment. In vivo, daratumumab was highly active and interrupted xenograft tumor growth at low dosing. Collectively, our results show the versatility of daratumumab to effectively kill CD38-expressing tumor cells, including patient MM cells, via diverse cytotoxic mechanisms. These findings support clinical development of daratumumab for the treatment of CD38-positive MM tumors.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/therapeutic use , Cytotoxicity, Immunologic , Hematologic Neoplasms/therapy , Membrane Glycoproteins/immunology , Multiple Myeloma/therapy , ADP-ribosyl Cyclase 1/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/metabolism , Binding Sites, Antibody , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , HEK293 Cells , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, SCID , Mice, Transgenic , Molecular Sequence Data , Multiple Myeloma/immunology , Multiple Myeloma/pathology , NIH 3T3 Cells , Xenograft Model Antitumor Assays
7.
Blood ; 112(6): 2390-9, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18566325

ABSTRACT

Glycosylation of the antibody Fc fragment is essential for Fc receptor-mediated activity. Carbohydrate heterogeneity is known to modulate the activity of effector cells in the blood, in which fucosylation particularly affects NK cell-mediated killing. Here, we investigated how the glycosylation profile of 2F8, a human IgG(1) monoclonal antibody against epidermal growth factor receptor in clinical development, impacted effector function. Various 2F8 batches differing in fucosylation, galactosylation, and sialylation of the complex-type oligosaccharides in the Fc fragment were investigated. Our results confirmed that low fucose levels enhance mononuclear cell-mediated antibody-mediated cellular cytotoxicity (ADCC). In contrast, polymorphonuclear cells were found to preferentially kill via high-fucosylated antibody. Whole blood ADCC assays, containing both types of effector cells, revealed little differences in tumor cell killing between both batches. Significantly, however, high-fucose antibody induced superior ADCC in blood from granulocyte colony-stimulating factor-primed donors containing higher numbers of activated polymorphonuclear cells. In conclusion, our data demonstrated for the first time that lack of fucose does not generally increase the ADCC activity of therapeutic antibodies and that the impact of Fc glycosylation on ADCC is critically dependent on the recruited effector cell type.


Subject(s)
Antibodies/metabolism , Antibody-Dependent Cell Cytotoxicity , Fucose/immunology , Killer Cells, Natural/immunology , Neutrophils/immunology , Antibodies/immunology , Cell Line , Fucose/metabolism , Glycosylation , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism
8.
Biotechnol Bioeng ; 105(2): 350-7, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19739094

ABSTRACT

Rapid production of recombinant human IgG with improved antibody dependent cell-mediated cytotoxicity (ADCC) effector function is presented. The technique employs transient expression of IgG in suspension growing HEK-293F cells in the presence of the glycosidase inhibitor kifunensine. The procedure takes approximately 7 days, provided that expression plasmids encoding the IgG of interest are available. Kifunensine inhibits the N-linked glycosylation pathway of HEK-293F cells in the endoplasmatic reticulum, resulting in IgG with oligomannose type glycans lacking core-fucose. IgG1 transiently produced in kifunensine- treated HEK-293F cells has improved affinity for the FcgammaRIIIA molecule as measured in an ELISA based assay, and almost eightfold enhanced ADCC using primary peripheral blood mononuclear effector cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Biotechnology/methods , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Alkaloids/metabolism , Biotechnology/economics , Cell Line , Gene Expression , Glycoside Hydrolases/antagonists & inhibitors , Humans , Immunoglobulin G/genetics , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Plasmids/genetics , Receptors, IgG/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Time Factors
9.
Sci Rep ; 7(1): 992, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28428630

ABSTRACT

Autoantibodies against ion channels are the cause of numerous neurologic autoimmune disorders. Frequently, such pathogenic autoantibodies have a restricted epitope-specificity. In such cases, competing antibody formats devoid of pathogenic effector functions (blocker antibodies) have the potential to treat disease by displacing autoantibodies from their target. Here, we have used a model of the neuromuscular autoimmune disease myasthenia gravis in rhesus monkeys (Macaca mulatta) to test the therapeutic potential of a new blocker antibody: MG was induced by passive transfer of pathogenic acetylcholine receptor-specific monoclonal antibody IgG1-637. The effect of the blocker antibody (IgG4Δhinge-637, the hinge-deleted IgG4 version of IgG1-637) was assessed using decrement measurements and single-fiber electromyography. Three daily doses of 1.7 mg/kg IgG1-637 (cumulative dose 5 mg/kg) induced impairment of neuromuscular transmission, as demonstrated by significantly increased jitter, synaptic transmission failures (blockings) and a decrease in the amplitude of the compound muscle action potentials during repeated stimulations (decrement), without showing overt symptoms of muscle weakness. Treatment with three daily doses of 10 mg/kg IgG4Δhinge-637 significantly reduced the IgG1-637-induced increase in jitter, blockings and decrement. Together, these results represent proof-of principle data for therapy of acetylcholine receptor-myasthenia gravis with a monovalent antibody format that blocks binding of pathogenic autoantibodies.


Subject(s)
Autoantibodies/metabolism , Immunoglobulin G/administration & dosage , Myasthenia Gravis/drug therapy , Receptors, Cholinergic/metabolism , Animals , CHO Cells , Cholinergic Antagonists , Cricetulus , Disease Models, Animal , Gene Expression Regulation/drug effects , HEK293 Cells , Hinge Exons , Humans , Immunoglobulin G/pharmacology , Macaca mulatta , Myasthenia Gravis/immunology , Myasthenia Gravis/metabolism , Synaptic Transmission/drug effects , Treatment Outcome
10.
Mol Cancer Ther ; 15(11): 2688-2697, 2016 11.
Article in English | MEDLINE | ID: mdl-27559142

ABSTRACT

Antibody-drug conjugates (ADC) are designed to be stable in circulation and to release potent cytotoxic drugs intracellularly following antigen-specific binding, uptake, and degradation in tumor cells. Efficient internalization and routing to lysosomes where proteolysis can take place is therefore essential. For many cell surface proteins and carbohydrate structures on tumor cells, however, the magnitude of these processes is insufficient to allow for an effective ADC approach. We hypothesized that we could overcome this limitation by enhancing lysosomal ADC delivery via a bispecific antibody (bsAb) approach, in which one binding domain would provide tumor specificity, whereas the other binding domain would facilitate targeting to the lysosomal compartment. We therefore designed a bsAb in which one binding arm specifically targeted CD63, a protein that is described to shuttle between the plasma membrane and intracellular compartments, and combined it in a bsAb with a HER2 binding arm, which was selected as model antigen for tumor-specific binding. The resulting bsHER2xCD63his demonstrated strong binding, internalization and lysosomal accumulation in HER2-positive tumor cells, and minimal internalization into HER2-negative cells. By conjugating bsHER2xCD63his to the microtubule-disrupting agent duostatin-3, we were able to demonstrate potent cytotoxicity of bsHER2xCD63his-ADC against HER2-positive tumors, which was not observed with monovalent HER2- and CD63-specific ADCs. Our data demonstrate, for the first time, that intracellular trafficking of ADCs can be improved using a bsAb approach that targets the lysosomal membrane protein CD63 and provide a rationale for the development of novel bsADCs that combine tumor-specific targeting with targeting of rapidly internalizing antigens. Mol Cancer Ther; 15(11); 2688-97. ©2016 AACR.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents/administration & dosage , Immunoconjugates/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Tetraspanin 30/antagonists & inhibitors , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacokinetics , Antibody Affinity/immunology , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Liberation , Female , Humans , Immunoconjugates/pharmacokinetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Lysosomes/metabolism , Mice , Molecular Targeted Therapy , Protein Binding , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Ann N Y Acad Sci ; 994: 267-74, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12851325

ABSTRACT

The melanocortin (MC) system is involved in the regulation of energy balance and in the development of obesity. Here we briefly review why we became interested in investigating whether the MC system - more particularly, the increased activity of the MC system - is also involved in disorders of negative energy balance. We provide evidence that suppression of increased MC receptor activity by treatment with the inverse agonist agouti-related peptide (AgRP) (83-132) rescues rats exposed to an animal model known as activity-based anorexia. Furthermore, we found a polymorphism, Ala67Thr AgRP, that was observed more frequently in anorexia nervosa.


Subject(s)
Feeding and Eating Disorders/metabolism , Pro-Opiomelanocortin/metabolism , Proteins/metabolism , Receptors, Corticotropin/metabolism , alpha-MSH/metabolism , Agouti-Related Protein , Animals , Behavior, Animal/drug effects , Binding Sites , Hypothalamo-Hypophyseal System/physiology , Intercellular Signaling Peptides and Proteins , Melanocyte-Stimulating Hormones/pharmacology , Peptides/metabolism , Peptides/therapeutic use , Pituitary-Adrenal System/physiology , Polymorphism, Genetic , Pro-Opiomelanocortin/genetics , Proteins/genetics , Proteins/therapeutic use , Rats , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Melanocortin , Starvation/drug therapy
13.
MAbs ; 6(2): 392-402, 2014.
Article in English | MEDLINE | ID: mdl-24492309

ABSTRACT

The human epidermal growth factor receptor (HER)2 provides an excellent target for selective delivery of cytotoxic drugs to tumor cells by antibody-drug conjugates (ADC) as has been clinically validated by ado-trastuzumab emtansine (Kadcyla(TM)). While selecting a suitable antibody for an ADC approach often takes specificity and efficient antibody-target complex internalization into account, the characteristics of the optimal antibody candidate remain poorly understood. We studied a large panel of human HER2 antibodies to identify the characteristics that make them most suitable for an ADC approach. As a model toxin, amenable to in vitro high-throughput screening, we employed Pseudomonas exotoxin A (ETA') fused to an anti-kappa light chain domain antibody. Cytotoxicity induced by HER2 antibodies, which were thus non-covalently linked to ETA', was assessed for high and low HER2 expressing tumor cell lines and correlated with internalization and downmodulation of HER2 antibody-target complexes. Our results demonstrate that HER2 antibodies that do not inhibit heterodimerization of HER2 with related ErbB receptors internalize more efficiently and show greater ETA'-mediated cytotoxicity than antibodies that do inhibit such heterodimerization. Moreover, stimulation with ErbB ligand significantly enhanced ADC-mediated tumor kill by antibodies that do not inhibit HER2 heterodimerization. This suggests that the formation of HER2/ErbB-heterodimers enhances ADC internalization and subsequent killing of tumor cells. Our study indicates that selecting HER2 ADCs that allow piggybacking of HER2 onto other ErbB receptors provides an attractive strategy for increasing ADC delivery and tumor cell killing capacity to both high and low HER2 expressing tumor cells.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/therapy , Drug Design , Immunotherapy/methods , Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/immunology , Cell Line, Tumor , Dimerization , Endocytosis , Epidermal Growth Factor/metabolism , Female , Humans , Immunotoxins/therapeutic use , Receptor Aggregation/drug effects , Receptor, ErbB-2/immunology , Signal Transduction/drug effects , Trastuzumab
14.
MAbs ; 5(2): 219-28, 2013.
Article in English | MEDLINE | ID: mdl-23406897

ABSTRACT

Antibody engineering is increasingly being used to influence the properties of monoclonal antibodies to improve their biotherapeutic potential. One important aspect of this is the modulation of glycosylation as a strategy to improve efficacy. Here, we describe mutations of Y407 in the CH3 domain of IgG1 and IgG4 that significantly increase sialylation, galactosylation, and branching of the N-linked glycans in the CH2 domain. These mutations also promote the formation of monomeric assemblies (one heavy-light chain pair). Hydrogen-deuterium exchange mass spectrometry was used to probe conformational changes in IgG1-Y407E, revealing, as expected, a more exposed CH3-CH3 dimerization interface. Additionally, allosteric structural effects in the CH2 domain and in the CH2-CH3 interface were identified, providing a possible explanation for the dramatic change in glycosylation. Thus, the mutation of Y407 in the CH3 domain remarkably affects both antibody conformation and glycosylation, which not only alters our understanding of antibody structure, but also reveals possibilities for obtaining recombinant IgG with glycosylation tailored for clinical applications.


Subject(s)
Antibodies, Monoclonal/genetics , Immunoglobulin G/genetics , Mutation , Protein Engineering/methods , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Deuterium Exchange Measurement , Glycosylation , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Mass Spectrometry , Models, Molecular , N-Acetylneuraminic Acid , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Structure-Activity Relationship
15.
Biotechnol Prog ; 25(1): 244-51, 2009.
Article in English | MEDLINE | ID: mdl-19224598

ABSTRACT

We studied the variations in N-linked glycosylation of human IgG molecules derived from 105 different stable cell lines each expressing one of the six different antibodies. Antibody expression was based on glutamine synthetase selection technology in suspension growing CHO-K1SV cells. The glycans detected on the Fc fragment were mainly of the core-fucosylated complex type containing zero or one galactose and little to no sialic acid. The glycosylation was highly consistent for the same cell line when grown multiple times, indicating the robustness of the production and glycan analysis procedure. However, a twofold to threefold difference was observed in the level of galactosylation and/or non-core-fucosylation between the 105 different cell lines, suggesting clone-to-clone variation. These differences may change the Fc-mediated effector functions by such antibodies. Large variation was also observed in the oligomannose-5 glycan content, which, when present, may lead to undesired rapid clearance of the antibody in vivo. Statistically significant differences were noticed between the various glycan parameters for the six different antibodies, indicating that the variable domains and/or light chain isotype influence Fc glycosylation. The glycosylation altered when batch production in shaker was changed to fed-batch production in bioreactor, but was consistent again when the process was scaled from 400 to 5,000 L. Taken together, the observed clone-to-clone glycosylation variation but batch-to-batch consistency provides a rationale for selection of optimal production cell lines for large-scale manufacturing of biopharmaceutical human IgG.


Subject(s)
Cell Line/metabolism , Glycosylation , Immunoglobulin G/biosynthesis , Animals , CHO Cells , Cell Line/cytology , Cricetinae , Cricetulus , Humans , Polysaccharides/metabolism
16.
Science ; 317(5844): 1554-7, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17872445

ABSTRACT

Antibodies play a central role in immunity by forming an interface with the innate immune system and, typically, mediate proinflammatory activity. We describe a novel posttranslational modification that leads to anti-inflammatory activity of antibodies of immunoglobulin G, isotype 4 (IgG4). IgG4 antibodies are dynamic molecules that exchange Fab arms by swapping a heavy chain and attached light chain (half-molecule) with a heavy-light chain pair from another molecule, which results in bispecific antibodies. Mutagenesis studies revealed that the third constant domain is critical for this activity. The impact of IgG4 Fab arm exchange was confirmed in vivo in a rhesus monkey model with experimental autoimmune myasthenia gravis. IgG4 Fab arm exchange is suggested to be an important biological mechanism that provides the basis for the anti-inflammatory activity attributed to IgG4 antibodies.


Subject(s)
Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Allergens/immunology , Animals , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antigens, CD20/immunology , Antigens, Plant , Autoantibodies/immunology , ErbB Receptors/immunology , Glycoproteins/immunology , Humans , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains , Macaca mulatta , Mice , Mutation , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/prevention & control , Protein Processing, Post-Translational , Receptors, Cholinergic/immunology
17.
J Immunol ; 177(1): 362-71, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785532

ABSTRACT

We have previously defined a panel of fully human CD20 mAb. Most of these were unexpectedly efficient in their ability to recruit C1q to the surface of CD20-positive cells and mediate tumor lysis via activation of the classical pathway of complement. This complement-dependent cytotoxicity (CDC) potency appeared to relate to the unusually slow off-rate of these human Abs. However, we now present epitope-mapping data, which indicates that all human mAb bind a novel region of CD20 that may influence CDC potency. Epitope mapping, using both mutagenesis studies and overlapping 15-mer peptides of the extracellular loops of CD20, defined the amino acids required for binding by an extensive panel of mouse and human mAb. Binding by rituximab and mouse CD20 mAb, had an absolute requirement for alanine and proline at positions 170 and 172, respectively, within the large extracellular loop of CD20. Surprisingly, however, all of the human CD20 mAb recognize a completely novel epitope located N-terminally of this motif, also including the small extracellular loop of CD20. Thus, although off-rate may influence biological activity of mAb, another critical factor for determining CDC potency by CD20 mAb appears to be the region of the target molecule they recognize. We conclude that recognition of the novel epitope cooperates with slow off-rate in determining the activity of CD20 Ab in activation of complement and induction of tumor cell lysis.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigens, CD20/immunology , Antigens, CD20/metabolism , Binding Sites, Antibody , Epitopes/immunology , Epitopes/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antigens, CD20/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/metabolism , Binding Sites, Antibody/genetics , Cell Line , Cell Line, Tumor , Complement Pathway, Classical/genetics , Complement Pathway, Classical/immunology , Cytotoxicity, Immunologic/genetics , Epitope Mapping , Epitopes/genetics , Humans , Immunoglobulin G/metabolism , Lymphoma/immunology , Lymphoma/pathology , Lymphoma/therapy , Mice , Mice, Transgenic , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Peptide Fragments/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL