Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
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.
Oncoimmunology ; 5(1): e1058459, 2016.
Article in English | MEDLINE | ID: mdl-26942070

ABSTRACT

Antibody-dependent cell-mediated cytotoxicity (ADCC) mediated through the IgG Fc receptor FcγRIIIa represents a major effector function of many therapeutic antibodies. In an attempt to further enhance natural killer (NK) cell-mediated ADCC, we combined therapeutic antibodies against CD20 and CD38 with recombinant immunoligands against the stimulatory NK cell receptors NKG2D or NKp30. These immunoligands, respectively designated as ULBP2:7D8 and B7-H6:7D8, contained the CD20 scFv 7D8 as a targeting moiety and a cognate ligand for either NKG2D or NKp30 (i.e. ULBP2 and B7-H6, respectively). Both the immunoligands synergistically augmented ADCC in combination with the CD20 antibody rituximab and the CD38 antibody daratumumab. Combinations with ULBP2:7D8 resulted in higher cytotoxicity compared to combinations with B7-H6:7D8, suggesting that coligation of FcγRIIIa with NKG2D triggered NK cells more efficiently than with NKp30. Addition of B7-H6:7D8 to ULBP2:7D8 and rituximab in a triple combination did not further increase the extent of tumor cell lysis. Importantly, immunoligand-mediated enhancement of ADCC was also observed for tumor cells and autologous NK cells from patients with hematologic malignancies, in which, again, ULBP2:7D8 was particularly active. In summary, co-targeting of NKG2D was more effective in promoting rituximab or daratumumab-mediated ADCC by NK cells than co-ligation of NKp30. The observed increase in the ADCC activity of these therapeutic antibodies suggests promise for a 'dual-dual-targeting' approach in which tumor cell surface antigens are targeted in concert with two distinct activating NK cell receptors (i.e. FcγRIIIa and NKG2D or B7-H6).

3.
Science ; 343(6176): 1260-3, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24626930

ABSTRACT

Complement activation by antibodies bound to pathogens, tumors, and self antigens is a critical feature of natural immune defense, a number of disease processes, and immunotherapies. How antibodies activate the complement cascade, however, is poorly understood. We found that specific noncovalent interactions between Fc segments of immunoglobulin G (IgG) antibodies resulted in the formation of ordered antibody hexamers after antigen binding on cells. These hexamers recruited and activated C1, the first component of complement, thereby triggering the complement cascade. The interactions between neighboring Fc segments could be manipulated to block, reconstitute, and enhance complement activation and killing of target cells, using all four human IgG subclasses. We offer a general model for understanding antibody-mediated complement activation and the design of antibody therapeutics with enhanced efficacy.


Subject(s)
Cell Membrane/immunology , Complement Activation , Complement C1/immunology , Immunoglobulin G/chemistry , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/immunology , Liposomes , Protein Conformation , Protein Multimerization
4.
EMBO Mol Med ; 5(8): 1213-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23918228

ABSTRACT

Currently all approved anti-cancer therapeutic monoclonal antibodies (mAbs) are of the IgG isotype, which rely on Fcgamma receptors (FcγRs) to recruit cellular effector functions. In vitro studies showed that targeting of FcαRI (CD89) by bispecific antibodies (bsAbs) or recombinant IgA resulted in more effective elimination of tumour cells by myeloid effector cells than targeting of FcγR. Here we studied the in vivo anti-tumour activity of IgA EGFR antibodies generated using the variable sequences of the chimeric EGFR antibody cetuximab. Using FcαRI transgenic mice, we demonstrated significant in vivo anti-tumour activity of IgA2 EGFR against A431 cells in peritoneal and lung xenograft models, as well as against B16F10-EGFR cells in a lung metastasis model in immunocompetent mice. IgA2 EGFR was more effective than cetuximab in a short-term syngeneic peritoneal model using EGFR-transfected Ba/F3 target cells. The in vivo cytotoxic activity of IgA2 EGFR was mediated by macrophages and was significantly decreased in the absence of FcαRI. These results support the potential of targeting FcαRI for effective antibody therapy of cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , ErbB Receptors/antagonists & inhibitors , Immunoglobulin A/pharmacology , Neoplasms/drug therapy , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD/immunology , Cell Line, Tumor , Cetuximab , ErbB Receptors/immunology , Flow Cytometry , Humans , Immunotherapy/methods , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/immunology , Receptors, Fc/immunology , Recombinant Proteins/pharmacology
5.
J Immunol ; 191(1): 353-62, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23740955

ABSTRACT

To evade opsonophagocytosis, Staphylococcus aureus secretes various immunomodulatory molecules that interfere with effective opsonization by complement and/or IgG. Immune-evasion molecules targeting the phagocyte receptors for these opsonins have not been described. In this study, we demonstrate that S. aureus escapes from FcγR-mediated immunity by secreting a potent FcγR antagonist, FLIPr, or its homolog FLIPr-like. Both proteins were previously reported to function as formyl peptide receptor inhibitors. Binding of FLIPr was mainly restricted to FcγRII receptors, whereas FLIPr-like bound to different FcγR subclasses, and both competitively blocked IgG-ligand binding. They fully inhibited FcγR-mediated effector functions, including opsonophagocytosis and subsequent intracellular killing of S. aureus by neutrophils and Ab-dependent cellular cytotoxicity of tumor cells by both neutrophils and NK cells. In vivo, treatment of mice with FLIPr-like prevented the development of an immune complex-mediated FcγR-dependent Arthus reaction. This study reveals a novel immune-escape function for S. aureus-secreted proteins that may lead to the development of new therapeutic agents in FcγR-mediated diseases.


Subject(s)
Bacterial Proteins/physiology , Receptors, IgG/antagonists & inhibitors , Staphylococcus aureus/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites, Antibody/immunology , Humans , Immune Evasion/immunology , Leukemia P388/immunology , Leukemia P388/microbiology , Mice , Mice, Inbred BALB C , Phagocytosis/immunology , Protein Binding/immunology , Receptors, IgG/chemistry , Receptors, IgG/physiology , Sequence Homology, Amino Acid , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity
6.
Proc Natl Acad Sci U S A ; 110(13): 5145-50, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479652

ABSTRACT

The promise of bispecific antibodies (bsAbs) to yield more effective therapeutics is well recognized; however, the generation of bsAbs in a practical and cost-effective manner has been a formidable challenge. Here we present a technology for the efficient generation of bsAbs with normal IgG structures that is amenable to both antibody drug discovery and development. The process involves separate expression of two parental antibodies, each containing single matched point mutations in the CH3 domains. The parental antibodies are mixed and subjected to controlled reducing conditions in vitro that separate the antibodies into HL half-molecules and allow reassembly and reoxidation to form highly pure bsAbs. The technology is compatible with standard large-scale antibody manufacturing and ensures bsAbs with Fc-mediated effector functions and in vivo stability typical of IgG1 antibodies. Proof-of-concept studies with HER2×CD3 (T-cell recruitment) and HER2×HER2 (dual epitope targeting) bsAbs demonstrate superior in vivo activity compared with parental antibody pairs.


Subject(s)
Antibodies, Bispecific/biosynthesis , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin G/biosynthesis , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Jurkat Cells , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
7.
J Immunol ; 189(10): 5037-46, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23066150

ABSTRACT

Induced self expression of the NKp30 ligand B7-H6 facilitates NK cell-mediated elimination of stressed cells. A fusion protein consisting of the ectodomain of B7-H6 and the CD20 single-chain fragment variable 7D8 was generated to mimic an induced self phenotype required for NK cell-mediated target cell elimination. B7-H6:7D8 had bifunctional properties as reflected by its ability to simultaneously bind to the CD20 Ag and to the NKp30 receptor. B7-H6:7D8 bound by CD20(+) lymphoma cells activated human NK cells and triggered degranulation. Consequently, the immunoligand B7-H6:7D8 induced killing of lymphoma-derived cell lines as well as fresh tumor cells from chronic lymphocytic leukemia or lymphoma patients. B7-H6:7D8 was active at nanomolar concentrations in a strictly Ag-specific manner and required interaction with both CD20 and NKp30. Remarkably, NK cell cytotoxicity was further augmented by concomitant activation of Fcγ receptor IIIa or NK group 2 member D. Thus, B7-H6:7D8 acted synergistically with the CD20 Ab rituximab and the immunoligand ULBP2:7D8, which was similarly designed as B7-H6:7D8 but engaging the NK group 2 member D receptor. In conclusion, to our knowledge, B7-H6:7D8 represents the first Ab-based molecule stimulating NKp30-mediated NK cell cytotoxicity for therapeutic purposes and provides proof of concept that Ag-specific NKp30 engagement may represent an innovative strategy to enhance antitumoral NK cell cytotoxicity.


Subject(s)
B7 Antigens/pharmacology , Cell Degranulation/drug effects , Immunity, Cellular/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphoma/therapy , Natural Cytotoxicity Triggering Receptor 3/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/genetics , Antigens, CD20/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , B7 Antigens/agonists , B7 Antigens/genetics , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Line, Tumor , Drug Synergism , Humans , Immunity, Cellular/genetics , Immunotherapy , Lymphocyte Activation/genetics , Lymphoma/genetics , Lymphoma/immunology , Natural Cytotoxicity Triggering Receptor 3/genetics , Receptors, IgG , Rituximab
8.
Autoimmunity ; 45(7): 556-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22559288

ABSTRACT

Patients with Systemic Lupus Erythematosus (SLE) carry an increased risk for the development of coronary artery disease (CAD). The R131 allele of the Fc gamma receptor IIa (FcγRIIa) is associated with SLE incidence and disease severity but also with CAD. Compared to stable angina pectoris (SAP) the unstable angina (UAP), as a manifestation of destabilizing CAD, is associated with increased risk of persistent instability, myocardial infarction, and death. Identification of clinically relevant determinants for unstable angina promises reduction of UAP-associated mortality in patients with SLE. We conducted a clinical study among 553 consecutive patients with stable angina pectoris (n = 330) and unstable angina pectoris (n = 223). All patients were genotyped for a frequent functional variant at position 131 of the mature FcγRIIa. UAP, but not SAP was significantly associated with the R/R131 genotype (P < 0.001). In troponin-negative patients with angina carrying the R/R131 genotype the odds ratio for suffering from UAP was 4.02 (95% confidence interval, 2.52-6.41) compared to those with non-R/R131 genotypes. In a multivariable analysis, the R/R131 genotype independently predicted the risk for development of UAP in a model adjusted for classical atherogenic risk factors. Our data imply that risk stratification of SLE- and other high risk patients with troponin-negative angina could be significantly improved by FcγRIIa genotyping.


Subject(s)
Alleles , Angina, Unstable/genetics , Polymorphism, Single Nucleotide , Receptors, IgG/genetics , Aged , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Risk Factors
9.
Neoplasia ; 14(3): 190-205, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22496619

ABSTRACT

Oncogenic KRAS mutations in colorectal cancer (CRC) are associated with lack of benefit from epidermal growth factor receptor (EGFR)-directed antibody (Ab) therapy. However, the mechanisms by which constitutively activated KRAS (KRAS(G12V)) impairs effector mechanisms of EGFR-Abs are incompletely understood. Here, we established isogenic cell line models to systematically investigate the impact of KRAS(G12V) on tumor growth in mouse A431 xenograft models as well as on various modes of action triggered by EGFR-Abs in vitro. KRAS(G12V) impaired EGFR-Ab-mediated growth inhibition by stimulating receptor-independent downstream signaling. KRAS(G12V) also rendered tumor cells less responsive to Fc-mediated effector mechanisms of EGFR-Abs-such as complement-dependent cytotoxicity (CDC) and Ab-dependent cell-mediated cytotoxicity (ADCC). Impaired CDC and ADCC activities could be linked to reduced EGFR expression in KRAS-mutated versus wild-type (wt) cells, which was restored by small interfering RNA (siRNA)-mediated knockdown of KRAS4b. Immunohistochemistry experiments also revealed lower EGFR expression in KRAS-mutated versus KRAS-wt harboring CRC samples. Analyses of potential mechanisms by which KRAS(G12V) downregulated EGFR expression demonstrated significantly decreased activity of six distinct transcription factors. Additional experiments suggested the CCAAT/enhancer-binding protein (C/EBP) family to be implicated in the regulation of EGFR promoter activity in KRAS-mutated tumor cells by suppressing EGFR transcription through up-regulation of the inhibitory family member C/EBPß-LIP. Thus, siRNA-mediated knockdown of C/EBPß led to enhanced EGFR expression and Ab-mediated cytotoxicity against KRAS-mutated cells. Together, these results demonstrate that KRAS(G12V) signaling induced C/EBPß-dependent suppression of EGFR expression, thereby impairing Fc-mediated effector mechanisms of EGFR-Abs and rendering KRAS-mutated tumor cells less sensitive to these therapeutic agents.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , ras Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity/genetics , Antineoplastic Agents/pharmacology , Base Sequence , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cetuximab , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Signal Transduction/drug effects , Transcription, Genetic , ras Proteins/genetics
10.
Eur J Immunol ; 42(7): 1815-21, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22535639

ABSTRACT

Neutrophils potently kill tumour cells in the presence of anti-tumour antibodies in vitro. However, for in vivo targeting, the neutrophils need to extravasate from the circulation by passing through endothelial barriers. To study neutrophil migration in the presence of endothelial cells in vitro, we established a three-dimensional collagen culture in which SK-BR-3 tumour colonies were grown in the presence or absence of an endothelial barrier. We demonstrated that - in contrast to targeting FcγR on neutrophils with mAbs - targeting the immunoglobulin A Fc receptor (FcαRI) instead triggered neutrophil migration and degranulation leading to tumour destruction, which coincided with release of the pro-inflammatory cytokines interleukin (IL)-1ß and tumour necrosis factor (TNF)-α. Interestingly, neutrophil migration was enhanced in the presence of endothelial cells, which coincided with production of significant levels of the neutrophil chemokine IL-8. This supports the idea that stimulation of neutrophil FcαRI, but not FcγR, initiates cross-talk between neutrophils and endothelial cells, leading to enhanced neutrophil migration towards tumour colonies and subsequent tumour killing.


Subject(s)
Antigens, CD/immunology , Breast Neoplasms/immunology , Chemotaxis, Leukocyte/immunology , Human Umbilical Vein Endothelial Cells/immunology , Neutrophils/immunology , Receptors, Fc/immunology , Breast Neoplasms/pathology , Cell Communication/immunology , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/cytology , Humans , Immunity, Innate/immunology , Immunohistochemistry , Interleukin-1beta/analysis , Interleukin-1beta/immunology , Neutrophils/cytology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
12.
Haematologica ; 96(12): 1822-30, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21880632

ABSTRACT

BACKGROUND: CD20 monoclonal antibodies are widely used in clinical practice. Antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity and direct cell death have been suggested to be important effector functions for CD20 antibodies. However, their specific contributions to the in vivo mechanism of action of CD20 immunotherapy have not been well defined. DESIGN AND METHODS: Here we studied the in vivo mechanism of action of type I (rituximab and ofatumumab) and type II (HuMab-11B8) CD20 antibodies in a peritoneal, syngeneic, mouse model with EL4-CD20 cells using low and high tumor burden. RESULTS: Interestingly, we observed striking differences in the in vivo mechanism of action of CD20 antibodies dependent on tumor load. In conditions of low tumor burden, complement was sufficient for tumor killing both for type I and type II CD20 antibodies. In contrast, in conditions of high tumor burden, activating FcγR (specifically FcγRIII), active complement and complement receptor 3 were all essential for tumor killing. Our data suggest that complement-enhanced antibody-dependent cellular cytotoxicity may critically affect tumor killing by CD20 antibodies in vivo. The type II CD20 antibody 11B8, which is a poor inducer of complement activation, was ineffective against high tumor burden. CONCLUSIONS: Tumor burden affects the in vivo mechanism of action of CD20 antibodies. Low tumor load can be eliminated by complement alone, whereas elimination of high tumor load requires multiple effector mechanisms.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD20 , Antineoplastic Agents/pharmacology , Lymphoma/drug therapy , Tumor Burden/drug effects , Animals , Antibodies, Monoclonal, Humanized , Humans , Lymphoma/pathology , Mice , Mice, Knockout , Neoplasm Transplantation , Rituximab
13.
Structure ; 19(9): 1274-82, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21893287

ABSTRACT

Native mass spectrometry (MS) is a powerful technique for studying noncovalent protein-protein interactions. Here, native MS was employed to examine the noncovalent interactions involved in homodimerization of antibody half molecules (HL) in hinge-deleted human IgG4 (IgG4Δhinge). By analyzing the concentration dependence of the relative distribution of monomer HL and dimer (HL)(2) species, the apparent dissociation constant (K(D)) for this interaction was determined. In combination with site-directed mutagenesis, the relative contributions of residues at the CH3-CH3 interface to this interaction could be characterized and corresponding K(D) values quantified over a range of 10(-10)-10(-4) M. The critical importance of this noncovalent interaction in maintaining the intact dimeric structure was also proven for the full-length IgG4 backbone. Using time-resolved MS, the kinetics of the interaction could be measured, reflecting the dynamics of IgG4 HL exchange. Hence, native MS has provided a quantitative view of local structural features that define biological properties of IgG4.


Subject(s)
Binding Sites, Antibody , Immunoglobulin Fragments/chemistry , Immunoglobulin G/chemistry , Spectrometry, Mass, Electrospray Ionization , Amino Acid Motifs , Computer Simulation , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin G/genetics , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense , Protein Binding , Protein Stability , Thermodynamics
14.
J Immunol ; 187(6): 3438-47, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841127

ABSTRACT

We previously reported that 1 h after infusion of CD20 mAb rituximab in patients with chronic lymphocytic leukemia (CLL), >80% of CD20 was removed from circulating B cells, and we replicated this finding, based on in vitro models. This reaction occurs via an endocytic process called shaving/trogocytosis, mediated by FcγR on acceptor cells including monocytes/macrophages, which remove and internalize rituximab-CD20 immune complexes from B cells. Beers et al. reported that CD20 mAb-induced antigenic modulation occurs as a result of internalization of B cell-bound mAb-CD20 complexes by the B cells themselves, with internalization of ∼40% observed after 2 h at 37°C. These findings raise fundamental questions regarding the relative importance of shaving versus internalization in promoting CD20 loss and have substantial implications for the design of mAb-based cancer therapies. Therefore, we performed direct comparisons, based on flow cytometry, to determine the relative rates and extent of shaving versus internalization. B cells, from cell lines, from patients with CLL, and from normal donors, were opsonized with CD20 mAbs rituximab or ofatumumab and incubated for varying times and then reacted with acceptor THP-1 monocytes to promote shaving. We find that shaving induces considerably greater loss of CD20 and bound mAb from opsonized B cells in much shorter time periods (75-90% in <45 min) than is observed for internalization. Both shaving/trogocytosis and internalization could contribute to CD20 loss when CLL patients receive rituximab therapy, but shaving should occur more rapidly and is most likely to be the key mechanism of CD20 loss.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/metabolism , Antineoplastic Agents/pharmacology , B-Lymphocytes/metabolism , Endocytosis/physiology , Monocytes/metabolism , Animals , B-Lymphocytes/drug effects , Cell Line , Cell Separation , Endocytosis/drug effects , Flow Cytometry , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Receptors, IgG/metabolism , Rituximab , Transfection
15.
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
16.
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
17.
J Immunol Methods ; 373(1-2): 67-78, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21855548

ABSTRACT

Protein- or glyco-engineering of antibody molecules can be used to enhance Fc-mediated effector functions. ScFv-Fc fusion proteins (scFv-Fc) represent interesting antibody derivatives due to their relatively simple design and increased tissue penetration. Here, the impact of protein- and glyco-engineering on ADCC potency of a panel of human IgG1-based scFv-Fc was tested. Three matched sets of scFv-Fc variants targeting CD7, CD20 or HLA class II and optimized for CD16a binding by mutagenesis, lack of core-fucose, or their combination, were generated and functionally tested in comparison to the corresponding wild type scFv-Fc. Antigen binding activity was not compromised by altered glycosylation or Fc mutagenesis, whereas Fc binding to CD16a was significantly enhanced in the order: non-core fucosylated/Fc-mutated double-engineered≫Fc-mutated≥non-core-fucosylated>wild-type IgG1-Fc. All engineered variants triggered potent ADCC with up to 100-fold reduced EC50 values compared to non-engineered variants. Interestingly, double-engineered variants were similarly effective in triggering ADCC compared to single-engineered variants irrespective of their 1 log greater CD16a binding affinity. Thus, these data demonstrate that protein- and glyco-engineering enhances NK-cell mediated ADCC of scFv-Fc similarly and show that enhancing CD16a affinity beyond a certain threshold does not result in a further increase of NK-cell mediated ADCC.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Immunoglobulin Fc Fragments/immunology , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Affinity/immunology , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, CD7/immunology , Antigens, CD7/metabolism , Blotting, Western , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Flow Cytometry , Fucose/metabolism , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , HLA-D Antigens/immunology , HLA-D Antigens/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/immunology , Killer Cells, Natural/metabolism , Mutagenesis , Protein Binding , Protein Engineering/methods , Receptors, Fc/immunology , Receptors, Fc/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
18.
Cancer Sci ; 102(10): 1761-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21718386

ABSTRACT

Monoclonal antibodies (mAb) against variant III of epidermal growth factor receptor (EGFRvIII) hold promise for improving tumor selectivity of EGFR-targeted therapy. Here, we compared Fc-mediated effector functions of three mAb against EGFRvIII (MR1-1, ch806, 13.1.2) with those of zalutumumab, a high affinity EGFR mAb in advanced clinical trials. MR1-1 and ch806 demonstrated preferential and 13.1.2 exclusive binding to EGFRvIII, in contrast to zalutumumab, which bound both wild-type and EGFRvIII. All four human IgG1κ mAb mediated antibody-dependent cellular cytotoxicity (ADCC) of EGFRvIII-expressing cells with mononuclear cells and isolated monocytes, while only zalutumumab in addition triggered ADCC by polymorphonuclear cells. Interestingly, combinations of zalutumumab and EGFRvIII mAb specifically mediated complement-dependent cytotoxicity (CDC) of EGFRvIII-transfected but not wild-type cells. Moreover, EGFRvIII-specific CDC was significantly enhanced when zalutumumab was combined with a Fc-engineered variant of MR1-1 (K326A/E333A). These observations confirm the immunotherapeutic potential of antibody combinations against EGFR, and demonstrate that tumor selectivity can be improved by combining therapeutic EGFR mAb with an antibody against EGFRvIII.


Subject(s)
Antibodies, Monoclonal/immunology , Complement System Proteins/immunology , Cytotoxicity, Immunologic , ErbB Receptors/immunology , ErbB Receptors/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line, Tumor , Cricetinae , Epitope Mapping , HEK293 Cells , Humans , Neutrophils/immunology
19.
BMC Biotechnol ; 11: 77, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21781305

ABSTRACT

BACKGROUND: Human immunoglobulin G (IgG) plays an important role in mediating protective immune responses to malaria. Although human serum immunoglobulin A (IgA) is the second most abundant class of antibody in the circulation, its contribution, if any, to protective responses against malaria is not clear. RESULTS: To explore the mechanism(s) by which IgA may mediate a protective effect, we generated fully human IgA specific for the C-terminal 19-kDa region of Plasmodium falciparum merozoite surface protein 1 (PfMSP1 19), a major target of protective immune responses. This novel human IgA bound antigen with an affinity comparable to that seen for an epitope-matched protective human IgG1. Furthermore, the human IgA induced significantly higher NADPH-mediated oxidative bursts and degranulation from human neutrophils than the epitope-matched human IgG1 from which it was derived. Despite showing efficacy in in vitro functional assays, the human IgA failed to protect against parasite challenge in vivo in mice transgenic for the human Fcα receptor (FcαRI/CD89). A minority of the animals treated with IgA, irrespective of FcαRI expression, showed elevated serum TNF-α levels and concomitant mouse anti-human antibody (MAHA) responses. CONCLUSIONS: The lack of protection afforded by MSP1 19-specific IgA against parasite challenge in mice transgenic for human FcαRI suggests that this antibody class does not play a major role in control of infection. However, we cannot exclude the possibility that protective capacity may have been compromised in this model due to rapid clearance and inappropriate bio-distribution of IgA, and differences in FcαRI expression profile between humans and transgenic mice.


Subject(s)
Antibodies, Protozoan/immunology , Immunoglobulin A/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Recombinant Proteins/immunology , Animals , Antigens, CD/genetics , Disease Models, Animal , Humans , Immunization, Passive , Immunoglobulin G/metabolism , Malaria/immunology , Malaria/prevention & control , Mice , Mice, Transgenic , NADPH Oxidases/metabolism , Plasmodium berghei , Receptors, Fc/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...