Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Hum Vaccin Immunother ; 15(1): 179-188, 2019.
Article in English | MEDLINE | ID: mdl-30156957

ABSTRACT

The rational for designing dendritic cell (DC)-targeted immunotherapies is their central role in orchestrating immunity. Most studies addressing antigen-targeting to DCs for eliciting T cell responses have employed ex-vivo matured DCs derived from monocytes or myeloid DCs isolated from peripheral blood. More recently, also plasmacytoid DCs (pDCs) emerged as attractive targets that can be readily isolated and activated ex vivo. pDCs are known as key effectors of innate and adaptive immunity due to their exquisite ability to produce large amounts of type-1 interferons upon signaling via TLR7 or TLR9 intracellular receptor for viral RNA or bacterial DNA, respectively. In this study, we describe and characterize the immune modulating and targeting module of a composite human specific vaccine platform for active immunotherapy. This module, called warhead (WH), is composed of a single-chain variable fragment (scFv) and CpG-C type oligonucleotides (ODNs) that are covalently coupled. The scFv mediates specific binding to FcγRII/CD32 on APCs and internalization of the ODNs which stimulate TLR9-expressing B cells and pDCs. Furthermore, the scFv in the WH is extended with a five-time heptad repeat (EVSALEK) alpha helix which allows for a coiled-coil complex formation with any immunogen also extended with another five-time heptad (KVSALKE) repeat. WH elicits fast and robust pDC activation as evidenced by the release of interferon-α, TNF-α and IL-6. The WH thus takes advantage of the key features of human pDCs for immunostimulation and can be a versatile tool for antigen-specific vaccination with a variety of proteins or peptides.


Subject(s)
Dendritic Cells/immunology , Immunomodulation , Receptors, IgG/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Vaccines/immunology , B-Lymphocytes/immunology , Cytokines/immunology , Humans , Immunotherapy, Active , Oligodeoxyribonucleotides/immunology , Single-Chain Antibodies/immunology
2.
Mol Ther ; 23(11): 1722-1733, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26234505

ABSTRACT

FS102 is a HER2-specific Fcab (Fc fragment with antigen binding), which binds HER2 with high affinity and recognizes an epitope that does not overlap with those of trastuzumab or pertuzumab. In tumor cells that express high levels of HER2, FS102 caused profound HER2 internalization and degradation leading to tumor cell apoptosis. The antitumor effect of FS102 in patient-derived xenografts (PDXs) correlated strongly with the HER2 amplification status of the tumors. Superior activity of FS102 over trastuzumab or the combination of trastuzumab and pertuzumab was observed in vitro and in vivo when the gene copy number of HER2 was equal to or exceeded 10 per cell based on quantitative polymerase chain reaction (qPCR). Thus, FS102 induced complete and sustained tumor regression in a significant proportion of HER2-high PDX tumor models. We hypothesize that the unique structure and/or epitope of FS102 enables the Fcab to internalize and degrade cell surface HER2 more efficiently than standard of care antibodies. In turn, increased depletion of HER2 commits the cells to apoptosis as a result of oncogene shock. FS102 has the potential of a biomarker-driven therapeutic that derives superior antitumor effects from a unique mechanism-of-action in tumor cells which are oncogenically addicted to the HER2 pathway due to overexpression.


Subject(s)
Apoptosis/drug effects , Immunoglobulin Fc Fragments/pharmacology , Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation , Humans , Immunoglobulin Fc Fragments/therapeutic use , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Receptor, ErbB-2/immunology , Signal Transduction , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
3.
J Immunol ; 195(5): 2343-52, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26209628

ABSTRACT

Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine and counterregulator of glucocorticoids, is a potential therapeutic target. MIF is markedly different from other cytokines because it is constitutively expressed, stored in the cytoplasm, and present in the circulation of healthy subjects. Thus, the concept of targeting MIF for therapeutic intervention is challenging because of the need to neutralize a ubiquitous protein. In this article, we report that MIF occurs in two redox-dependent conformational isoforms. We show that one of the two isoforms of MIF, that is, oxidized MIF (oxMIF), is specifically recognized by three mAbs directed against MIF. Surprisingly, oxMIF is selectively expressed in the plasma and on the cell surface of immune cells of patients with different inflammatory diseases. In patients with acute infections or chronic inflammation, oxMIF expression correlated with inflammatory flare-ups. In addition, anti-oxMIF mAbs alleviated disease severity in mouse models of acute and chronic enterocolitis and improved, in synergy with glucocorticoids, renal function in a rat model of crescentic glomerulonephritis. We conclude that oxMIF represents the disease-related isoform of MIF; oxMIF is therefore a new diagnostic marker for inflammation and a relevant target for anti-inflammatory therapy.


Subject(s)
Inflammation/immunology , Inflammation/prevention & control , Macrophage Migration-Inhibitory Factors/immunology , Molecular Targeted Therapy/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Blotting, Western , Dexamethasone/immunology , Dexamethasone/therapeutic use , Disease Models, Animal , Enterocolitis/immunology , Enterocolitis/metabolism , Enterocolitis/prevention & control , Flow Cytometry , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/prevention & control , Glucocorticoids/immunology , Glucocorticoids/therapeutic use , Humans , Inflammation/metabolism , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rabbits , Rats, Inbred WKY
4.
Biotechnol J ; 9(6): 844-51, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24806546

ABSTRACT

Antigen-binding Fc fragments (Fcabs) are a new unique class of immunotherapeutics. They are small (50 kD) fully functional antibody alternatives that bind antigen and elicit effector functions such as antibody-dependent cytotoxicity (ADCC) and complement-dependent cytotoxicity. Since Fcabs carry the natural FcRn binding site of antibodies, they have very favorable pharmacokinetics. We showed recently that Fcab H10-03-6 is a high-affinity binder of Her-2/neu (ErbB2/neu) mediating killing of Her-2/neu-overexpressing tumor cells in the presence of immune effector cells, strongly suggesting that the mechanism of killing is due to ADCC. The present study further confirms ADCC as the mechanism by which H10-03-6 mediates tumor cell killing, since H10-03-6 was shown to interact simultaneously with Her-2/neu and the Fc receptor CD16a. The epitope recognized by H10-03-6 overlaps with that of the clinically used monoclonal antibody trastuzumab. However, unlike trastuzumab, Fcab H10-03-6 did not inhibit proliferation of human tumor cells in vitro even under conditions favoring Her-2/neu crosslinking. Treatment of mice harboring human BT-474 cell xenograft tumors with Fcab H10-03-6 led to statistically significant retardation of tumor growth. For the first time, in vivo properties of an Fcab are presented, supporting the view that Fcabs could become highly efficacious immunotherapeutics for human use.


Subject(s)
Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/metabolism , Binding Sites , Cell Line , Cell Proliferation/drug effects , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/metabolism , In Vitro Techniques , Mice , Neoplasm Transplantation , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Trastuzumab
5.
Immunobiology ; 217(10): 1017-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22349518

ABSTRACT

Atopic diseases are characterized by the presence of Th2 cells. Recent studies, in mice and man, demonstrated that allergen-specific Th2 responses can be shifted to Th0/Th1 responses. Plasmacytoid dendritic cells (pDCs) produce large amounts of type I interferons (IFNs) after stimulation of Toll Like Receptor 9 (TLR9) and are likely to play an important role in the reorientation of these Th2 cells. The expression of CD32a on the cell surface of pDCs makes this cell type attractive for targeted delivery of antigen and TLR agonists to revert Th2 responses. Therefore we sought to determine the efficacy of targeted delivery of CpG-C ODN to CD32a on the ability of human and monkey pDCs to secrete inflammatory cytokines. Here we demonstrate that targeted delivery of 3'-biotinylated CpG-C to CD32a on pDC induced phenotypical maturation as determined by CD80, CD83 and CD86 expression. Furthermore, targeting both monkey and human pDCs strongly augmented the secretion of IFNα compared to the delivery of CpG-C in an untargeted fashion (p<0.001). TLR9 induced activation hampers the ability of human pDCs to internalize CD32a. Therefore we opted for targeted delivery of CpG-ODNs to CD32a, which reduces the risk of undesired side effects of systemic TLR treatment and in addition delivers a superior signal for the activation of pDCs. This approach opens new treatment principles for allergic patients.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-alpha/biosynthesis , Oligodeoxyribonucleotides/metabolism , Receptors, IgG/metabolism , Animals , Biotinylation , Cells, Cultured , Cytokines/biosynthesis , Humans , Inflammation Mediators/metabolism , Macaca mulatta , Oligodeoxyribonucleotides/chemistry , Receptors, Fc/genetics , Receptors, Fc/metabolism , Toll-Like Receptor 9/metabolism
6.
EMBO J ; 30(13): 2705-18, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21623346

ABSTRACT

The quality and quantity of BCR signals impact on cell fate decisions of B lymphocytes. Here, we describe novel gene-targeted mice, which in the context of normal VDJ recombination show hypomorphic expression of immunoglobulin µ heavy chain (µHC) mRNA levels and hence lower pre-BCR and BCR levels. Hypomorphic expression of µHC leads to augmented selection processes at all stages of B-cell development, noticeably at the expansion of pre-B cells, the positive selection of immature B lymphocytes in the bone marrow and the selection of the follicular (FO), marginal zone (MZ) and B1 B-lymphocyte compartment in peripheral lymphoid organs. Immature as well as mature FO and MZ B lymphocytes in the peripheral lymphoid organs express lower levels of the receptor for B-cell activating factor (BAFF). In addition, hypomorphic expression of the BCR favours receptor editing. Together, our results highlight the critical importance of pre-BCR and BCR receptor levels for the normal development of B-lymphocyte subpopulations in the context of intact VDJ recombination and a diverse antibody repertoire.


Subject(s)
B-Lymphocyte Subsets/physiology , Cell Differentiation/immunology , Genes, Immunoglobulin Heavy Chain/physiology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Alleles , Animals , Antibody Formation/genetics , Antibody Formation/physiology , B-Cell Activation Factor Receptor/metabolism , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Differentiation/genetics , Cells, Cultured , Genes, Immunoglobulin Heavy Chain/genetics , Immunoglobulin mu-Chains/genetics , Immunoglobulin mu-Chains/metabolism , Lectins/genetics , Lectins/immunology , Lectins/metabolism , Mice , Mice, Transgenic , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/physiology , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins
7.
Biotechnol J ; 2(6): 700-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17427997

ABSTRACT

Recent studies have demonstrated that the reduction of the core fucosylation on N-glycans of human IgGs is responsible for a clearly enhanced antibody-dependent cellular cytotoxicity (ADCC). This finding might give access to improved active therapeutic antibodies. Here, the expression of the tumor antigen-specific antibody IGN311 was performed in a glyco-optimized strain of the moss Physcomitrella patens. Removal of plant specific N-glycan structures in this plant expression host was achieved by targeted knockout of corresponding genes and included quantitative elimination of core fucosylation. Antibodies transiently expressed and secreted by such genetically modified moss protoplasts assembled correctly, showed an unaltered antigen-binding affinity and, in extensive tests, revealed an up to 40-fold enhanced ADCC. Thus, the glyco-engineered moss-based transient expression platform combines a rapid technology with the subsequent analysis of glycooptimized therapeutics with regard to advanced properties.


Subject(s)
Antibodies, Monoclonal/immunology , Bryopsida/metabolism , Genetic Enhancement/methods , Polysaccharides/metabolism , Protein Engineering/methods , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Bryopsida/genetics , Humans
8.
Cancer Immunol Immunother ; 56(6): 863-73, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17009044

ABSTRACT

Tumor-associated antigens resulting from aberrant glycosylation, such as the SialylTn carbohydrate antigen, are frequently over-expressed on cancer cells and provide potential targets for cancer vaccination. Immunization of Rhesus monkeys with SialylTn coupled to a highly immunogenic carrier molecule and formulated on aluminum hydroxide induced a strong immune response against the carrier protein but only a moderate IgM immune response against the SialylTn carbohydrate antigen. Co-formulation with QS-21 adjuvant dramatically enhanced the anti-SialylTn immune response and resulted in a SialylTn-specific IgG switch. The kinetics of the carbohydrate-specific IgG response correlated with a temporary release of cytokines such as IFNgamma, IL-2, IL-1beta, TNFalpha and GM-CSF which was measurable in the immune serum by xMAP Multiplex technology. Furthermore, tumor cell killing by activated natural killer cells was induced. These data demonstrate that immunization with a tumor-associated carbohydrate antigen in a highly immunogenic formulation results in a temporary release of type 1 cytokines which may be required for the induction of a specific IgG immune response against the carbohydrate antigen as well as for activation of effector cells against tumor cells.


Subject(s)
Adjuvants, Immunologic , Antigens, Tumor-Associated, Carbohydrate/immunology , Cancer Vaccines/immunology , Cytokines/blood , Killer Cells, Natural/immunology , Saponins/immunology , Animals , Antibodies, Monoclonal/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , Macaca mulatta , Vaccines, Conjugate/immunology
9.
Blood ; 109(7): 2815-22, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17164349

ABSTRACT

To study both the pathophysiologic and the prognostic value of ADAMTS13 in thrombotic microangiopathies (TMAs), we enrolled a cohort of 35 adult patients combining a first acute episode of TMA, an undetectable (below 5%) ADAMTS13 activity in plasma, and no clinical background such as sepsis, cancer, HIV, and transplantation. All patients were treated by steroids and plasma exchange, and an 18-month follow-up was scheduled. Remission was obtained in 32 patients (91.4%), and 3 patients died (8.6%) after the first attack. At presentation, ADAMTS13 antigen was decreased in 32 patients (91.4%), an ADAMTS13 inhibitor was detectable in 31 patients (89%), and an anti-ADAMTS13 IgG/IgM/IgA was present in 33 patients (94%). The 3 decedent patients were characterized by the association of several anti-ADAMTS13 Ig isotypes, including very high IgA titers, while mortality was independent of the ADAMTS13 inhibitor titer. In survivors, ADAMTS13 activity in remission increased to levels above 15% in 19 patients (59%) but remained undetectable in 13 patients (41%). Six patients relapsed either once or twice (19%) during the follow-up. High levels of inhibitory anti-ADAMTS13 IgG at presentation were associated with the persistence of an undetectable ADAMTS13 activity in remission, the latter being predictive for relapses within an 18-month delay.


Subject(s)
ADAM Proteins/deficiency , ADAM Proteins/immunology , Autoantibodies/blood , Thrombosis/immunology , ADAM Proteins/blood , ADAMTS13 Protein , Adolescent , Adult , Anemia, Hemolytic/blood , Anemia, Hemolytic/immunology , Cohort Studies , Female , France , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/immunology , Humans , Immunoglobulin Isotypes/blood , Male , Middle Aged , Prognosis , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/immunology , Thrombosis/blood , von Willebrand Factor/metabolism
10.
J Pharmacol Exp Ther ; 319(3): 1459-66, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16963623

ABSTRACT

The blood group-related Lewis Y antigen is expressed on the majority of human cancers of epithelial origin with only limited expression on normal tissue. Therefore, the Lewis Y antigen represents an interesting candidate for antibody-based treatment strategies. Previous experiments showed that the humanized Lewis Y-specific monoclonal antibody, IGN311, reduced ErbB-receptor-mediated stimulation of mitogen-activated protein kinase by altering receptor recycling. Here, we tested whether binding of IGN311 to growth factor receptors is relevant also to inhibition of tumor growth in vivo. Prolonged incubation with IGN311 of human tumor cell lines, which express high levels of ErbB1 (A431) or ErbB2 (SK-BR-3), resulted in down-regulation of the receptors and inhibition of cell proliferation. IGN311 inhibited the growth of tumors derived from A431 cells xenografted in nude mice. Treatment with IGN311 was associated with a down-regulation of ErbB1 in the excised tumor tissue. Importantly, these effects of IGN311 were also mimicked by the Fab fragment of IGN311. These data indicate that tumor cell growth inhibition by IGN311 cannot solely be accounted for by invoking cellular and humoral immunological mechanisms. A direct effect on signaling via binding to Lewis Y glycosylated growth factor receptors on tumor cells is also likely to contribute to the therapeutic effect of IGN311 in vivo.


Subject(s)
Antibodies, Blocking/pharmacology , Genes, erbB-1/drug effects , Lewis Blood Group Antigens/immunology , Animals , Antibodies, Blocking/immunology , Cells, Cultured , Down-Regulation/drug effects , Female , Immunoglobulin Fab Fragments/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Signal Transduction/drug effects , Transplantation, Heterologous
11.
Vaccine ; 24(13): 2349-57, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16406172

ABSTRACT

Tumor-associated antigens resulting from aberrant glycosylation, such as the SialylTn carbohydrate antigen, are over-expressed on cancer cells and provide potential targets for cancer vaccination. However, as T-cell-independent antigens carbohydrates are poorly immunogenic, and fail to induce memory. In order to increase the immunogenicity we have coupled the SialylTn carbohydrate antigen to a highly immunogenic carrier molecule, the murine monoclonal antibody mAb17-1A. An immunogenic formulation of the SialylTn-mAb17-1A conjugate on alhydrogel, IGN402, with or without additional adjuvants was tested in Rhesus monkeys for tolerability and immunogenicity. A significant antibody response against mAb17-1A antibody was found. Importantly, also a specific immune response against SialylTn carbohydrate and binding to tumor cells was induced. Immunization in the presence of additional adjuvants, such as QS-21, strongly enhanced the immune response against the carbohydrate antigen, and resulted in induction of SialylTn-specific IgG antibodies. Noteworthy, also an induced temporary release of cytokines including IFNgamma and IL-2, indicative for T-cell activation, was measured. The data indicate that carrier-induced T-cell help together with strong adjuvant is sufficient for carbohydrate specific class switch induction.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Cancer Vaccines/immunology , Immunoglobulin G/biosynthesis , Animals , Cytokines/biosynthesis , Immunization , Lymphocyte Activation , Macaca mulatta , Rabbits , Saponins/administration & dosage , T-Lymphocytes/immunology , Vaccines, Conjugate/immunology
12.
Bioconjug Chem ; 16(6): 1519-28, 2005.
Article in English | MEDLINE | ID: mdl-16287250

ABSTRACT

Carbohydrate antigens resulting from aberrant glycosylation of tumor cells, such as SialylTn, represent attractive targets for cancer vaccination. However, T-cell-independent carbohydrate antigens are poorly immunogenic and fail to induce memory and IgG class switch. Clustered expression patterns of some carbohydrates on the cell surface add further complexity to the design of carbohydrate-based vaccines. We describe here a vaccine consisting of SialylTn carbohydrate epitopes coupled to a highly immunogenic carrier molecule, mAb17-1A, adsorbed on alhydrogel and coformulated with a strong adjuvant, QS-21. The SialylTn-mAb17-1A conjugate vaccine was administered in Rhesus monkeys, and the immune responses against mAb17-1A, SialylTn, ovine submaxillary mucin, and tumor cells were analyzed. The data demonstrate that the density of carbohydrate epitopes on the carrier is an essential parameter for induction of anti-carbohydrate specific memory IgG immune responses. Furthermore, the influence of different types of presentation of SialylTn (monomeric vs trimers vs clustered via a branched polyethylenimine linker) on antibody titers and specificity was studied. High-density coupling of SialylTn epitopes to mAb17-1A induced the strongest immune response against synthetic SialylTn and showed also the highest reactivity against natural targets, such as OSM and tumor cells.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/immunology , Immunoconjugates/administration & dosage , Vaccines, Conjugate/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibody Specificity , Antigens, Tumor-Associated, Carbohydrate/administration & dosage , Cancer Vaccines/immunology , Epitopes , Immunoglobulin G , Immunologic Memory , Macaca mulatta
13.
Cancer Res ; 65(17): 7934-41, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140965

ABSTRACT

The aim of the present study was to produce glycosylation variants of the therapeutic Lewis Y-specific humanized IgG1 antibody IGN311 to enhance cell-killing effector function. This was achieved via genetic engineering of the glycosylation machinery of the antibody-producing host. Antibody genes were transiently cotransfected with acetyl-glycosaminyltransferase-III genes into human embryonic kidney-EBV nuclear antigen cells. A control wild-type antibody, IGN311wt, was expressed in the same host using identical expression vectors, but without cotransfection of genes for acetyl-glycosaminyltransferase-III expression. Both expression products were purified to homogeneity and characterized. The glyco-engineered expression product (IGN312-Glyco-I) showed a remarkably homogenous N-linked glycosylation pattern consisting of one major hybrid-type, non-fucosylated and agalactosylated form carrying a bisecting GlcNAc-group. Wild-type expression product (IGN311wt) on the other hand was glycosylated by a multitude of different core-fucosylated complex-type structures of variable degrees of galactosylation. Target affinity of the glyco-engineered antibody as well as heavy and light chain assembly were not affected by acetyl-glycosaminyltransferase-III expression. In vitro experiments showed a approximately 10-fold increase of antibody-dependent cellular cytotoxicity of the glyco-engineered antibody using different Lewis Y-positive target cancer cell lines (SK-BR-3, SK-BR-5, OVCAR-3, and Kato-III). Complement-mediated cytotoxicity of IGN312-Glyco-I was 0.4-fold reduced using SK-BR-5 as target cell line. The reduction of complement activation could be prevented and even converted into a slight increase of activity by using a different molecular-biological approach directing the glycosylation towards increased levels of complex N-linked oligosaccharides of bisected, non-fucosylated type, as a result of cotransfection of mannosidase II together with acetyl-glycosaminyltransferase-III.


Subject(s)
Antibodies, Monoclonal/immunology , Lewis Blood Group Antigens/immunology , Oligosaccharides/metabolism , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibody Specificity , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Glycosylation , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mannosidases/genetics , Mannosidases/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neoplasms/immunology , Neoplasms/therapy , Oligosaccharides/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
14.
Immunology ; 115(3): 407-15, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946258

ABSTRACT

The inhibitory effect on antibody production by immune complexes has been shown to depend on co-ligation of the B-cell antigen receptor (BCR) with the low-affinity receptor for immunoglobulin G (IgG) (Fc gammaRIIb, CD32). Here we report that immunoglobulin E (IgE) synthesis, induced in a BCR-independent manner by interleukin-4 (IL-4) and anti-CD40 antibody, was inhibited by CD32 ligation. The observed effect was specific for CD32 as, first, antibodies directed against other B-cell surface structures had no inhibitory effect, and, second, treatment with anti-CD32 of cells that had been in culture for 2 days was ineffective owing to the down-regulation of CD32 expression. IgE inhibition was also observed in cells stimulated by IL-4/CD40 F(ab')(2) or IL-4 plus soluble CD40 ligand, demonstrating that co-cross-linking of CD32 and CD40 was not necessary to induce inhibition. Mechanistic studies into the IgE class switch process demonstrated that IL-4/anti-CD40-induced IgE germline gene transcription and B-cell proliferation were not affected by CD32 ligation. The data demonstrate that the negative regulatory role of the CD32 molecule is not restricted to BCR-induced B-cell activation, but is also functional on other B-cell activation pathways mediated by CD40 and IL-4.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin E/biosynthesis , Receptors, Antigen, B-Cell/immunology , Receptors, IgG/immunology , Antibody Specificity/immunology , CD40 Antigens/immunology , Cell Division/immunology , Cell Line , Cells, Cultured , Down-Regulation/immunology , Humans , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Immunoglobulin Isotypes/immunology , Immunoglobulin M/biosynthesis , Interleukin-4/immunology , Ligands , Palatine Tonsil/immunology , Transcription, Genetic/immunology
15.
Immunity ; 22(4): 451-65, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15845450

ABSTRACT

The identification of specific genetic loci that contribute to inflammatory and autoimmune diseases has proved difficult due to the contribution of multiple interacting genes, the inherent genetic heterogeneity present in human populations, and a lack of new mouse mutants. By using N-ethyl-N-nitrosourea (ENU) mutagenesis to discover new immune regulators, we identified a point mutation in the murine phospholipase Cg2 (Plcg2) gene that leads to severe spontaneous inflammation and autoimmunity. The disease is composed of an autoimmune component mediated by autoantibody immune complexes and B and T cell independent inflammation. The underlying mechanism is a gain-of-function mutation in Plcg2, which leads to hyperreactive external calcium entry in B cells and expansion of innate inflammatory cells. This mutant identifies Plcg2 as a key regulator in an autoimmune and inflammatory disease mediated by B cells and non-B, non-T haematopoietic cells and emphasizes that by distinct genetic modulation, a single point mutation can lead to a complex immunological phenotype.


Subject(s)
Autoimmunity , Calcium/metabolism , Inflammation/genetics , Point Mutation , Type C Phospholipases/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , B-Lymphocytes/metabolism , Base Sequence , Bone Marrow Cells/cytology , Dermatitis/genetics , Dermatitis/immunology , Male , Mice , Molecular Sequence Data , Phospholipase C gamma , Type C Phospholipases/metabolism , Up-Regulation
16.
Cancer Res ; 64(3): 1087-93, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871842

ABSTRACT

The majority of cancer cells derived from epithelial tissue express Lewis-Y (LeY) type difucosylated oligosaccharides on their plasma membrane. This results in the modification of cell surface receptors by the LeY antigen. We used the epidermal growth factor (EGF) receptor family members ErbB1 and ErbB2 as model systems to investigate whether the sugar moiety can be exploited to block signaling by growth factor receptors in human tumor cells (i.e., SKBR-3 and A431, derived from a breast cancer and a vulval carcinoma, respectively). The monoclonal anti-LeY antibody ABL364 and its humanized version IGN311 immunoprecipitated ErbB1 and ErbB2 from detergent lysates of A431 and SKBR-3, respectively. ABL364 and IGN311 blocked EGF- and heregulin-stimulated phosphorylation of mitogen-activated protein kinase [MAPK = extracellular signal-regulated kinase 1/2] in SKBR-3 and A431 cells. The effect was comparable in magnitude with that of trastuzumab (Herceptin) and apparently noncompetitive with respect to EGF. Stimulation of MAPK by ErbB was dynamin dependent and contingent on receptor internalization. ABL364 and IGN311 changed the intracellular localization of fluorescent EGF-containing endosomes and accelerated recycling of intracellular [(125)I]EGF to the plasma membrane. Taken together, these observations show that antibodies directed against carbohydrate side chains of ErbB receptors are capable of inhibiting ErbB-mediated signaling. The ability of these antibodies to reroute receptor trafficking provides a mechanistic explanation for their inhibitory action.


Subject(s)
Antibodies, Monoclonal/pharmacology , ErbB Receptors/antagonists & inhibitors , Lewis Blood Group Antigens/immunology , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Cell Line, Tumor , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/metabolism , ErbB Receptors/immunology , Female , Humans , Iodine Radioisotopes , Kinetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Precipitin Tests , Receptor, ErbB-2/immunology , Tunicamycin/pharmacology , Vulvar Neoplasms/immunology , Vulvar Neoplasms/metabolism , Vulvar Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...