Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
J Allergy Clin Immunol ; 147(4): 1420-1429.e7, 2021 04.
Article in English | MEDLINE | ID: mdl-32926878

ABSTRACT

BACKGROUND: Activation of the classical and lectin pathway of complement may contribute to tissue damage and organ dysfunction of antibody-mediated diseases and ischemia-reperfusion conditions. Complement factors are being considered as targets for therapeutic intervention. OBJECTIVE: We sought to characterize ARGX-117, a humanized inhibitory monoclonal antibody against complement C2. METHODS: The mode-of-action and binding characteristics of ARGX-117 were investigated in detail. Furthermore, its efficacy was analyzed in in vitro complement cytotoxicity assays. Finally, a pharmacokinetic/pharmacodynamic study was conducted in cynomolgus monkeys. RESULTS: Through binding to the Sushi-2 domain of C2, ARGX-117 prevents the formation of the C3 proconvertase and inhibits classical and lectin pathway activation upstream of C3 activation. As ARGX-117 does not inhibit the alternative pathway, it is expected not to affect the antimicrobial activity of this complement pathway. ARGX-117 prevents complement-mediated cytotoxicity in in vitro models for autoimmune hemolytic anemia and antibody-mediated rejection of organ transplants. ARGX-117 exhibits pH- and calcium-dependent target binding and is Fc-engineered to increase affinity at acidic pH to the neonatal Fc receptor, and to reduce effector functions. In cynomolgus monkeys, ARGX-117 dose-dependently reduces free C2 levels and classical pathway activity. A 2-dose regimen of 80 and 20 mg/kg separated by a week, resulted in profound reduction of classical pathway activity lasting for at least 7 weeks. CONCLUSIONS: ARGX-117 is a promising new complement inhibitor that is uniquely positioned to target both the classical and lectin pathways while leaving the alternative pathway intact.


Subject(s)
Antibodies, Monoclonal/pharmacology , Complement C2/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Calcium , Complement Activation/drug effects , Complement C2/analysis , Complement C2/metabolism , Complement Inactivating Agents/blood , Complement Inactivating Agents/pharmacokinetics , Epitope Mapping , Female , Humans , Hydrogen-Ion Concentration , Macaca fascicularis , Male
2.
Pharm Res ; 37(2): 17, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31879800

ABSTRACT

PURPOSE: Modulating sialylation of therapeutic glycoproteins may be used to influence their clearance and systemic exposure. We studied the effect of low and high sialylated IL4-10 fusion protein (IL4-10 FP) on in vitro and in vivo bioactivity and evaluated the effect of differential sialylation on pharmacokinetic parameters. METHODS: CHO cell lines producing low (IL4-10 FP lowSA) and high sialylated (IL4-10 FP highSA) fusion protein were generated. Bioactivity of the proteins was evaluated in an LPS-stimulated whole blood assay. Pharmacokinetics were studied in rats, analyzing plasma levels of IL4-10 FP upon intravenous injection. In vivo activity was assessed in an inflammatory pain mice model upon intrathecal injection. RESULTS: IL4-10 FP lowSA and IL4-10 FP highSA had similar potency in vitro. The pharmacokinetics study showed a 4-fold higher initial systemic clearance of IL4-10 FP lowSA, whereas the calculated half-life of both IL4-10 FP lowSA and IL4-10 FP highSA was 20.7 min. Finally, both IL4-10 FP glycoforms inhibited persistent inflammatory pain in mice to the same extent. CONCLUSIONS: Differential sialylation of IL4-10 fusion protein does not affect the in vitro and in vivo activity, but clearly results in a difference in systemic exposure. The rapid systemic clearance of low sialylated IL4-10 FP could be a favorable characteristic to minimize systemic exposure after administration in a local compartment.


Subject(s)
Anti-Inflammatory Agents/blood , Interleukin-10/blood , Interleukin-4/blood , N-Acetylneuraminic Acid/chemistry , Recombinant Fusion Proteins/blood , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , CHO Cells , Cricetulus , Disease Models, Animal , Glycosylation , HEK293 Cells , Humans , Interleukin-10/chemistry , Interleukin-10/pharmacology , Interleukin-4/chemistry , Interleukin-4/pharmacology , Metabolic Clearance Rate , Mice, Inbred C57BL , Pain/drug therapy , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
3.
Br J Haematol ; 180(6): 808-820, 2018 03.
Article in English | MEDLINE | ID: mdl-29468712

ABSTRACT

Based on their mechanisms-of-action, CD20 monoclonal antibodies (mAbs) are grouped into Type I [complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC)] and Type II [programmed cell death (PCD) and ADCC] mAbs. We generated 17 new hybridomas producing CD20 mAbs of different isotypes and determined unique heavy and light chain sequence pairs for 13 of them. We studied their epitope binding, binding kinetics and structural properties and investigated their predictive value for effector functions, i.e. PCD, CDC and ADCC. Peptide mapping and CD20 mutant screens revealed that 10 out of these 11 new mAbs have an overlapping epitope with the prototypic Type I mAb rituximab, albeit that distinct amino acids of the CD20 molecule contributed differently. Binding kinetics did not correlate with the striking differences in CDC activity among the mIgG2c mAbs. Interestingly, chimerization of mAb m1 resulted in a mAb displaying both Type I and II characteristics. PCD induction was lost upon introduction of a mutation in the framework of the heavy chain affecting the elbow angle, supporting that structural changes within this region can affect functional activities of CD20 mAbs. Together, these new CD20 mAbs provide further insights in the properties dictating the functional efficacy of CD20 mAbs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , Complement System Proteins/immunology , Epitopes/immunology , Antibodies, Monoclonal, Murine-Derived/genetics , Antibody-Dependent Cell Cytotoxicity/genetics , Cell Line , Epitope Mapping , Epitopes/genetics , Humans
4.
J Immunol ; 197(3): 807-13, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27316683

ABSTRACT

Emerging evidence suggests that FcγR-mediated cross-linking of tumor-bound mAbs may induce signaling in tumor cells that contributes to their therapeutic activity. In this study, we show that daratumumab (DARA), a therapeutic human CD38 mAb with a broad-spectrum killing activity, is able to induce programmed cell death (PCD) of CD38(+) multiple myeloma tumor cell lines when cross-linked in vitro by secondary Abs or via an FcγR. By comparing DARA efficacy in a syngeneic in vivo tumor model using FcRγ-chain knockout or NOTAM mice carrying a signaling-inactive FcRγ-chain, we found that the inhibitory FcγRIIb as well as activating FcγRs induce DARA cross-linking-mediated PCD. In conclusion, our in vitro and in vivo data show that FcγR-mediated cross-linking of DARA induces PCD of CD38-expressing multiple myeloma tumor cells, which potentially contributes to the depth of response observed in DARA-treated patients and the drug's multifaceted mechanisms of action.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Multiple Myeloma/immunology , Receptors, IgG/immunology , ADP-ribosyl Cyclase 1 , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/drug effects , Tumor Cells, Cultured
5.
Nanotechnology ; 27(31): 314002, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27334425

ABSTRACT

Gate control of donor electrons near interfaces is a generic ingredient of donor-based quantum computing. Here, we address the question: how is the phonon-assisted qubit relaxation time T 1 affected as the electron is shuttled between the donor and the interface? We focus on the example of the 'flip-flop qubit' (Tosi et al arXiv:1509.08538v1), defined as a combination of the nuclear and electronic states of a phosphorus donor in silicon, promising fast electrical control and long dephasing times when the electron is halfway between the donor and the interface. We theoretically describe orbital relaxation, flip-flop relaxation, and electron spin relaxation. We estimate that the flip-flop qubit relaxation time can be of the order of 100 µs, 8 orders of magnitude shorter than the value for an on-donor electron in bulk silicon, and a few orders of magnitude shorter (longer) than the predicted inhomogeneous dephasing time (gate times). All three relaxation processes are boosted by (i) the nontrivial valley structure of the electron-phonon interaction, and (ii) the different valley compositions of the involved electronic states.

6.
J Immunol ; 193(11): 5506-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25355925

ABSTRACT

The uptake of Ag-Ab immune complexes (IC) after the ligation of activating FcγR on dendritic cells (DC) leads to 100 times more efficient Ag presentation than the uptake of free Ags. FcγRs were reported to facilitate IC uptake and simultaneously induce cellular activation that drives DC maturation and mediates efficient T cell activation. Activating FcγRs elicit intracellular signaling via the ITAM domain of the associated FcRγ-chain. Studies with FcRγ-chain knockout (FcRγ(-/-)) mice reported FcRγ-chain ITAM signaling to be responsible for enhancing both IC uptake and DC maturation. However, FcRγ-chain is also required for surface expression of activating FcγRs, hampering the dissection of ITAM-dependent and independent FcγR functions in FcRγ(-/-) DCs. In this work, we studied the role of FcRγ-chain ITAM signaling using DCs from NOTAM mice that express normal surface levels of activating FcγR, but lack functional ITAM signaling. IC uptake by bone marrow-derived NOTAM DCs was reduced compared with wild-type DCs, but was not completely absent as in FcRγ(-/-) DCs. In NOTAM DCs, despite the uptake of ICs, both MHC class I and MHC class II Ag presentation was completely abrogated similar to FcRγ(-/-) DCs. Secretion of cytokines, upregulation of costimulatory molecules, and Ag degradation were abrogated in NOTAM DCs in response to FcγR ligation. Cross-presentation using splenic NOTAM DCs and prolonged incubation with OVA-IC was also abrogated. Interestingly, in this setup, proliferation of CD4(+) OT-II cells was induced by NOTAM DCs. We conclude that FcRγ-chain ITAM signaling facilitates IC uptake and is essentially required for cross-presentation, but not for MHC class II Ag presentation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Receptors, IgG/metabolism , Animals , Antigen-Antibody Complex/immunology , Antigens, CD/metabolism , Cell Differentiation/genetics , Cells, Cultured , Cross-Priming/genetics , Endocytosis/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Structure, Tertiary/genetics , Receptors, IgG/genetics , Signal Transduction
7.
J Immunol ; 192(12): 5540-7, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24842758

ABSTRACT

Extensive analysis of a variety of arthritis models in germline KO mice has revealed that all four receptors for the Fc part of IgG (FcγR) play a role in the disease process. However, their precise cell type-specific contribution is still unclear. In this study, we analyzed the specific role of the inhibiting FcγRIIb on B lymphocytes (using CD19Cre mice) and in the myeloid cell compartment (using C/EBPαCre mice) in the development of arthritis induced by immunization with either bovine or chicken collagen type II. Despite their comparable anti-mouse collagen autoantibody titers, full FcγRIIb knockout (KO), but not B cell-specific FcγRIIb KO, mice showed a significantly increased incidence and severity of disease compared with wild-type control mice when immunized with bovine collagen. When immunized with chicken collagen, disease incidence was significantly increased in pan-myeloid and full FcγRIIb KO mice, but not in B cell-specific KO mice, whereas disease severity was only significantly increased in full FcγRIIb KO mice compared with incidence and severity in wild-type control mice. We conclude that, although anti-mouse collagen autoantibodies are a prerequisite for the development of collagen-induced arthritis, their presence is insufficient for disease development. FcγRIIb on myeloid effector cells, as a modulator of the threshold for downstream Ab effector pathways, plays a dominant role in the susceptibility to collagen-induced arthritis, whereas FcγRIIb on B cells, as a regulator of Ab production, has a minor effect on disease susceptibility.


Subject(s)
Arthritis, Experimental/immunology , Autoantibodies/immunology , B-Lymphocytes/immunology , Myeloid Cells/immunology , Receptors, IgG/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Autoantibodies/genetics , B-Lymphocytes/pathology , Cattle , Chickens , Collagen Type II/immunology , Mice , Mice, Knockout , Myeloid Cells/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Receptors, IgG/genetics
8.
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
9.
J Immunol ; 191(8): 4130-40, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24026082

ABSTRACT

Genetic deficiency of the inhibitory Fc receptor, FcγRIIB (CD32b), has been shown to augment the activity of activatory FcγR and promote mAb immunotherapy. To investigate whether mAbs capable of blocking FcγRIIB have similar capacity, we recently generated a panel of specific anti-mouse FcγRIIB mAbs that do not cross-react with other FcRs, allowing us to study the potential of FcγRIIB as a therapeutic target. Previous work revealed a number of these mAbs capable of eliciting programmed cell death of targets, and in the present study we demonstrated their ability to promote target cell phagocytosis. However, in a variety of murine tumor models, anti-FcγRIIB mAbs demonstrated limited therapeutic activity despite optimized treatment regimens. Unexpectedly, we observed that the anti-FcγRIIB mAbs are rapidly and extensively consumed in vivo, both by the tumor and host cells, including B cells, leading to a precipitous loss from the circulation. Closer analysis revealed that the anti-FcγRIIB mAbs become extensively internalized from the cell surface within 24 h in vivo, likely explaining their suboptimal efficacy. Subsequent studies revealed that anti-FcγRIIB mAb immunotherapy was effective when used against FcγRIIB(+) tumors in FcγRIIB(-/-) recipients, indicating that consumption of the mAb by nontumor cells is the primary limitation of these reagents. Importantly, similar rates of internalization were not seen on human target cells, at least in vitro. These studies further highlight the need to determine the propensity of mAb therapeutics to internalize target receptors and also identify potential key differences between human and mouse cells in this respect.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphoma, B-Cell/immunology , Macrophages/immunology , Multiple Myeloma/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/metabolism , Apoptosis/immunology , Cells, Cultured , Female , Humans , Immunotherapy , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Multiple Myeloma/therapy , Receptors, IgG/genetics
10.
J Immunol ; 190(1): 340-8, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23203925

ABSTRACT

FcγRIIb is the sole inhibitory FcR for IgG in humans and mice, where it is involved in the negative regulation of Ab production and cellular activation. FcγRIIb-deficient mice show exacerbated disease following the induction of nephrotoxic nephritis (NTN). In this study, we determined the cellular origin of the FcγRIIb-knockout phenotype by inducing NTN in mice with a deficiency of FcγRIIb on either B cells alone (FcγRIIB(fl/fl)/CD19Cre(+)) or myeloid cells (FcγRIIB(fl/fl)/CEBPαCre(+)). Deletion of FcγRIIb from B cells did not increase susceptibility to NTN, compared with wild-type (WT) mice, despite higher Ab titers in the FcγRIIB(fl/fl)/CD19Cre(+) mice compared with the WT littermate controls. In contrast, mice lacking FcγRIIb on myeloid cells had exacerbated disease as measured by increased glomerular thrombosis, glomerular crescents, albuminuria, serum urea, and glomerular neutrophil infiltration when compared with WT littermate controls. The role for FcγRIIb expression on radioresistant intrinsic renal cells in the protection from NTN was then investigated using bone marrow chimeric mice. FcγRIIb(-/-) mice transplanted with FcγRIIb(-/-) bone marrow were more susceptible to NTN than WT mice transplanted with FcγRIIb(-/-) bone marrow, indicating that the presence of WT intrinsic renal cells protects from NTN. These results demonstrate that FcγRIIb on myeloid cells plays a major role in protection from NTN, and therefore, augmentation of FcγRIIb on these cells could be a therapeutic target in human Ab-mediated glomerulonephritis. Where there was a lack of FcγRIIb on circulating myeloid cells, expression of FcγRIIb on intrinsic renal cells provided an additional level of protection from Ab-mediated glomerulonephritis.


Subject(s)
B-Lymphocyte Subsets/immunology , Glomerulonephritis/immunology , Glomerulonephritis/prevention & control , Kidney/immunology , Myeloid Cells/immunology , Receptors, IgG/physiology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Genetic Predisposition to Disease , Glomerulonephritis/pathology , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Radiation Chimera/genetics , Radiation Chimera/immunology , Receptors, IgG/deficiency , Receptors, IgG/genetics
11.
Clin Immunol ; 155(1): 108-117, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25242138

ABSTRACT

We identified a novel Q27W FcγRIIa variant that was found more frequently in common variable immunodeficiency (CVID) or CVID-like children. We analyzed the possible functional consequence of the Q27W FcγRIIa mutation in human cells. We used peripheral blood mononuclear cells from Q27W FcγRIIa patients and healthy controls, and cultured cells that overexpress the Q27W and common FcγRIIa variants. The Q27W FcγRIIa mutation does not disrupt FcγRIIa surface expression in peripheral blood mononuclear cells. Mononuclear cells express multiple FcγR, precluding careful analysis of Q27W FcγRIIa functional deviation. For functional analysis of FcγRIIa function, we therefore overexpressed the Q27W FcγRIIa and common FcγRIIa variant in IIA1.6 cells that are normally deficient in FcγR. We show that FcγRIIa triggering-induced signaling is obstructed, as measured by both decrease in calcium flux and defective MAPK phosphorylation. In conclusion, we here describe a novel Q27W FcγRIIa variant that causes delayed downstream signaling. This variant may contribute to CVID.


Subject(s)
Common Variable Immunodeficiency/genetics , Receptors, IgG/metabolism , Signal Transduction/genetics , Adolescent , Calcium/metabolism , Child , Gene Expression Regulation/immunology , Genetic Variation , Humans , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Receptors, IgG/genetics
12.
Blood ; 119(25): 5945-7, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22730521

ABSTRACT

In this issue of Blood, Elvington et al present a method to enhance complement activation during antibody therapy of cancer. Using a fusion protein of a complement receptor and an IgG Fc fragment, therapeutic outcome was improved in vivo.

13.
FASEB J ; 27(10): 4136-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23896727

ABSTRACT

The CD40/CD40L dyad is deemed to play a central role in several inflammatory processes, including atherosclerosis. As CD40 is overexpressed in atherosclerotic lesions, it constitutes a promising candidate for targeted imaging approaches. Here we describe the design of a novel, selective peptide ligand for CD40 by phage display. A synthetic peptide corresponding with the phage insert NP31 displayed nanomolar affinity for CD40. Affinity was further enhanced by mutimeric presentation of NP31. An essential 11-mer peptide motif was identified by truncation and alanine scan studies. Enriched phage selectively bound human CD40 and homed to inflammatory joints in a murine model of rheumatoid arthritis. NP31 ablated VEGF and IL-6 transcriptional activation and partially inhibited IL-6 production by CD40L-activated endothelial cells. Notably, NP31 did not only alter the biodistribution profile of a streptavidin scaffold but also markedly increased accumulation of the carrier in atherosclerotic aortic lesions of aged ApoE(-/-) mice in a CD40-dependent manner. This potent and selective peptide ligand has potential for targeted imaging and drug delivery approaches in CD40-dependent inflammatory disorders such as atherosclerosis.


Subject(s)
CD40 Ligand/metabolism , Peptide Library , Amino Acid Sequence , Animals , Aorta/pathology , Arthritis, Rheumatoid/diagnosis , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD40 Ligand/genetics , Cell Line , Humans , Inflammation , Mice , Protein Binding
14.
J Immunol ; 189(1): 92-101, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649202

ABSTRACT

FcγR ligation by Ag-Ab immune complexes (IC) not only mediates effective Ag uptake, but also strongly initiates dendritic cell (DC) maturation, a requirement for effective T cell activation. Besides the activating FcγRI, FcγRIII, and FcγRIV, the inhibitory FcγRIIb is expressed on DCs. It is unclear how the ratio between signals from the activating FcγR and the inhibitory FcγRIIb determines the outcome of FcγR ligation on DCs. By microarray analysis, we compared the transcriptomes of steady state and IC-activated bone marrow-derived wild-type (WT) DCs expressing all FcγR or DCs expressing only activating FcγR (FcγRIIb knockout [KO]) or only the inhibitory FcγRIIb (FcR γ-chain KO). In WT DCs, we observed a gene expression profile associated with effective T cell activation, which was absent in FcR γ-chain KO, but strikingly more pronounced in FcγRIIb KO bone marrow-derived DCs. These microarray results, confirmed at the protein level for many cytokines and other immunological relevant genes, demonstrate that the transcriptome of IC-activated DCs is dependent on the presence of the activating FcγR and that the modulation of the expression of the majority of the genes was strongly regulated by FcγRIIb. Our data suggest that FcγRIIb-deficient DCs have an improved capacity to activate naive T lymphocytes. This was confirmed by their enhanced FcγR-dependent Ag presentation and in vivo induction of CD8(+) T cell expansion compared with WT DCs. Our findings underscore the potency of FcγR ligation on DCs for the effective induction of T cell immunity by ICs and the strong regulatory role of FcγRIIb.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation/immunology , Receptors, IgG/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cells, Cultured , Coculture Techniques , Female , Gene Knockout Techniques , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Receptors, IgG/deficiency , Receptors, IgG/genetics , T-Lymphocyte Subsets/metabolism
15.
J Biol Chem ; 287(30): 25139-50, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22679018

ABSTRACT

IgA antibodies constitute an important part of the mucosal immune system, but their immunotherapeutic potential remains rather unexplored, in part due to biotechnological issues. For example, the IgA2m(1) allotype carries an unusual heavy and light chain pairing, which may confer production and stability concerns. Here, we report the generation and the biochemical and functional characterization of a P221R-mutated IgA2m(1) antibody against the epidermal growth factor receptor (EGFR). Compared with wild type, the mutated antibody demonstrated heavy chains covalently linked to light chains in monomeric as well as in joining (J)-chain containing dimeric IgA. Functional studies with wild type and mutated IgA2m(1) revealed similar binding to EGFR and direct effector functions such as EGFR down-modulation and growth inhibition. Furthermore, both IgA molecules triggered similar levels of indirect tumor cell killing such as antibody-dependent cell-mediated cytotoxicity (ADCC) by isolated monocytes, activated polymorphonuclear cells, and human whole blood. Interestingly, the dimeric IgA antibodies demonstrated higher efficiency in direct as well as in indirect effector mechanisms compared with their respective monomeric forms. Both wild type and mutated antibody triggered effective FcαRI-mediated tumor cell killing by macrophages already at low effector to target cell ratios. Interestingly, also polarized macrophages mediated significant IgA2-mediated ADCC. M2 macrophages, which have been described as promoting tumor growth and progression, may convert to ADCC-mediating effector cells in the presence of EGFR-directed antibodies. In conclusion, these results provide further insight into the immunotherapeutic potential of recombinant IgA antibodies for tumor immunotherapy and suggest macrophages as an additional effector cell population.


Subject(s)
Antibodies, Neoplasm/immunology , Immunity, Cellular , Immunoglobulin A/immunology , Immunoglobulin Allotypes/immunology , Immunotherapy , Macrophages/immunology , Monocytes/immunology , Mutation , Neoplasms/therapy , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Cell Line, Tumor , Cricetinae , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , ErbB Receptors/genetics , ErbB Receptors/immunology , Humans , Immunoglobulin A/genetics , Immunoglobulin A/pharmacology , Immunoglobulin Allotypes/genetics , Immunoglobulin Allotypes/pharmacology , Neoplasms/genetics , Neoplasms/immunology , Receptors, Fc/genetics , Receptors, Fc/immunology
16.
J Immunol ; 187(3): 1304-13, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21724994

ABSTRACT

FcγRIIB-deficient mice generated in 129 background (FcγRIIB(129)(-/-)) if back-crossed into C57BL/6 background exhibit a hyperactive phenotype and develop lethal lupus. Both in mice and humans, the Fcγr2b gene is located within a genomic interval on chromosome 1 associated with lupus susceptibility. In mice, the 129-derived haplotype of this interval, named Sle16, causes loss of self-tolerance in the context of the B6 genome, hampering the analysis of the specific contribution of FcγRIIB deficiency to the development of lupus in FcγRIIB(129)(-/-) mice. Moreover, in humans genetic linkage studies revealed contradictory results regarding the association of "loss of function" mutations in the Fcγr2b gene and susceptibility to systemic lupus erythematosis. In this study, we demonstrate that FcγRIIB(-/-) mice generated by gene targeting in B6-derived ES cells (FcγRIIB(B6)(-/-)), lacking the 129-derived flanking Sle16 region, exhibit a hyperactive phenotype but fail to develop lupus indicating that in FcγRIIB(129)(-/-) mice, not FcγRIIB deficiency but epistatic interactions between the C57BL/6 genome and the 129-derived Fcγr2b flanking region cause loss of tolerance. The contribution to the development of autoimmune disease by the resulting autoreactive B cells is amplified by the absence of FcγRIIB, culminating in lethal lupus. In the presence of the Yaa lupus-susceptibility locus, FcγRIIB(B6)(-/-) mice do develop lethal lupus, confirming that FcγRIIB deficiency only amplifies spontaneous autoimmunity determined by other loci.


Subject(s)
Genetic Predisposition to Disease/prevention & control , Immunoglobulin G/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Receptors, IgG/physiology , Animals , Cells, Cultured , Crosses, Genetic , Disease Models, Animal , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Female , Gene Targeting , Humans , Immunophenotyping , Lupus Nephritis/prevention & control , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, IgG/deficiency , Receptors, IgG/genetics
17.
EBioMedicine ; 93: 104663, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37379657

ABSTRACT

BACKGROUND: HexaBody®-CD38 (GEN3014) is a hexamerization-enhanced human IgG1 that binds CD38 with high affinity. The E430G mutation in its Fc domain facilitates the natural process of antibody hexamer formation upon binding to the cell surface, resulting in increased binding of C1q and potentiated complement-dependent cytotoxicity (CDC). METHODS: Co-crystallization studies were performed to identify the binding interface of HexaBody-CD38 and CD38. HexaBody-CD38-induced CDC, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), trogocytosis, and apoptosis were assessed using flow cytometry assays using tumour cell lines, and MM patient samples (CDC). CD38 enzymatic activity was measured using fluorescence spectroscopy. Anti-tumour activity of HexaBody-CD38 was assessed in patient-derived xenograft mouse models in vivo. FINDINGS: HexaBody-CD38 binds a unique epitope on CD38 and induced potent CDC in multiple myeloma (MM), acute myeloid leukaemia (AML), and B-cell non-Hodgkin lymphoma (B-NHL) cells. Anti-tumour activity was confirmed in patient-derived xenograft models in vivo. Sensitivity to HexaBody-CD38 correlated with CD38 expression level and was inversely correlated with expression of complement regulatory proteins. Compared to daratumumab, HexaBody-CD38 showed enhanced CDC in cell lines with lower levels of CD38 expression, without increasing lysis of healthy leukocytes. More effective CDC was also confirmed in primary MM cells. Furthermore, HexaBody-CD38 efficiently induced ADCC, ADCP, trogocytosis, and apoptosis after Fc-crosslinking. Moreover, HexaBody-CD38 strongly inhibited CD38 cyclase activity, which is hypothesized to relieve immune suppression in the tumour microenvironment. INTERPRETATION: Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of HexaBody-CD38 in patients with MM. FUNDING: Genmab.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Humans , Animals , Mice , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Complement System Proteins/metabolism , Tumor Microenvironment
18.
Blood ; 116(24): 5327-33, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20805361

ABSTRACT

FcγRI is the sole high-affinity immunoglobulin G (IgG) receptor on leukocytes. Its role in immunity and the clearance of opsonized particles has been challenged, as the receptor function may well be hindered by serum IgG. Here, we document immune complex binding by FcγRI to be readily enhanced by cytokine stimulation, whereas binding of monomeric IgG only modestly increased. Enhanced immune complex binding was independent of FcγRI surface expression levels. FcγRI, saturated with prebound IgG, was found capable of effective immune complex binding upon cytokine stimulation. Cytokine-enhanced binding was observed across a variety of immune complexes, including huIgG3- or mIgG2a-opsonized red blood cells, rituximab- or ofatumumab-opsonized B-cell lymphoma, and cetuximab-opsonized glioblastoma cells. This study contributes to our understanding of how FcγRI can participate in the clearance of opsonized particles despite saturation by monomeric IgG.


Subject(s)
Antigen-Antibody Complex/metabolism , Cytokines/pharmacology , Immunoglobulin G/metabolism , Receptors, IgG/metabolism , Animals , Cell Line , Erythrocytes/immunology , Glioblastoma/immunology , Humans , Lymphoma, B-Cell/immunology , Mice , Opsonin Proteins/metabolism , Protein Binding
19.
J Control Release ; 341: 475-486, 2022 01.
Article in English | MEDLINE | ID: mdl-34890719

ABSTRACT

PEGylation of lipid-based nanoparticles and other nanocarriers is widely used to increase their stability and plasma half-life. However, either pre-existing or de novo formed anti-PEG antibodies can induce hypersensitivity reactions and accelerated blood clearance through binding to the nanoparticle surfaces, leading to activation of the complement system. In this study, we investigated the consequences and mechanisms of complement activation by anti-PEG antibodies interacting with different types of PEGylated lipid-based nanoparticles. By using both liposomes loaded with different (model) drugs and LNPs loaded with mRNA, we demonstrate that complement activation triggered by anti-PEG antibodies can compromise the bilayer/surface integrity, leading to premature drug release or exposure of their mRNA contents to serum proteins. Anti-PEG antibodies also can induce deposition of complement fragments onto the surface of PEGylated lipid-based nanoparticles and induce the release of fluid phase complement activation products. The role of the different complement pathways activated by lipid-based nanoparticles was studied using deficient sera and/or inhibitory antibodies. We identified a major role for the classical complement pathway in the early activation events leading to the activation of C3. Our data also confirm the essential role of amplification of C3 activation by alternative pathway components in the lysis of liposomes. Finally, the levels of pre-existing anti-PEG IgM antibodies in plasma of healthy donors correlated with the degree of complement activation (fixation and lysis) induced upon exposure to PEGylated liposomes and mRNA-LNPs. Taken together, anti-PEG antibodies trigger complement activation by PEGylated lipid-based nanoparticles, which can potentially compromise their integrity, leading to premature drug release or cargo exposure to serum proteins.


Subject(s)
Liposomes , Nanoparticles , Complement System Proteins , Lipids , Liposomes/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry
20.
Life Sci Alliance ; 5(11)2022 Nov.
Article in English | MEDLINE | ID: mdl-36271507

ABSTRACT

CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4- but not of 5T4-IFNGR1- tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/pharmacology , CD8-Positive T-Lymphocytes , Granzymes/pharmacology , CD3 Complex/pharmacology , Cytotoxicity, Immunologic , Perforin/pharmacology , Programmed Cell Death 1 Receptor , Neoplasms/drug therapy , Cytokines
SELECTION OF CITATIONS
SEARCH DETAIL