Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Viruses ; 16(4)2024 04 12.
Article in English | MEDLINE | ID: mdl-38675937

ABSTRACT

Antibodies that specifically bind to individual human fragment crystallizable γ receptors (FcγRs) are of interest as research tools in studying immune cell functions, as well as components in bispecific antibodies for immune cell engagement in cancer therapy. Monoclonal antibodies for human low-affinity FcγRs have been successfully generated by hybridoma technology and are widely used in pre-clinical research. However, the generation of monoclonal antibodies by hybridoma technology that specifically bind to the high-affinity receptor FcγRI is challenging. Monomeric mouse IgG2a, IgG2b, and IgG3 bind human FcγRI with high affinity via the Fc part, leading to an Fc-mediated rather than a fragment for antigen binding (Fab)-mediated selection of monoclonal antibodies. Blocking the Fc-binding site of FcγRI with an excess of human IgG or Fc during screening decreases the risk of Fc-mediated interactions but can also block the potential epitopes of new antibody candidates. Therefore, we replaced hybridoma technology with phage display of a single-chain fragment variable (scFv) antibody library that was generated from mice immunized with FcγRI-positive cells and screened it with a cellular panning approach assisted by next-generation sequencing (NGS). Seven new FcγRI-specific antibody sequences were selected with this methodology, which were produced as Fc-silent antibodies showing FcγRI-restricted specificity.


Subject(s)
Antibodies, Monoclonal , Receptors, IgG , Receptors, IgG/immunology , Receptors, IgG/metabolism , Animals , Mice , Humans , Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Immunization , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Peptide Library , Cell Surface Display Techniques , Hybridomas , Antibody Specificity , Female , Mice, Inbred BALB C
2.
J Immunol ; 210(4): 389-397, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36637221

ABSTRACT

Signal inhibitory receptor on leukocytes-1 (SIRL-1) is an immune inhibitory receptor expressed on human granulocytes and monocytes that dampens antimicrobial functions. We previously showed that sputum neutrophils from infants with severe respiratory syncytial virus (RSV) bronchiolitis have decreased SIRL-1 surface expression compared with blood neutrophils and that SIRL-1 surface expression is rapidly lost from in vitro activated neutrophils. This led us to hypothesize that activated neutrophils lose SIRL-1 by ectodomain shedding. Here, we developed an ELISA and measured the concentration of soluble SIRL-1 (sSIRL-1) in patients with RSV bronchiolitis and hospitalized patients with COVID-19, which are both characterized by neutrophilic inflammation. In line with our hypothesis, sSIRL-1 concentration was increased in sputum compared with plasma of patients with RSV bronchiolitis and in serum of hospitalized patients with COVID-19 compared with control serum. In addition, we show that in vitro activated neutrophils release sSIRL-1 by proteolytic cleavage and that this diminishes the ability to inhibit neutrophilic reactive oxygen species production via SIRL-1. Finally, we found that SIRL-1 shedding is prevented by proteinase 3 inhibition and by extracellular adherence protein from Staphylococcus aureus. Notably, we recently showed that SIRL-1 is activated by PSMα3 from S. aureus, suggesting that S. aureus may counteract SIRL-1 shedding to benefit from preserved inhibitory function of SIRL-1. In conclusion, we report that SIRL-1 is released from activated neutrophils by proteinase 3 cleavage and that endogenous sSIRL-1 protein is present in vivo.


Subject(s)
Bronchiolitis , COVID-19 , Respiratory Syncytial Virus Infections , Humans , Infant , Bronchiolitis/metabolism , COVID-19/metabolism , Myeloblastin , Neutrophils , Receptors, Immunologic , Staphylococcus aureus , Leukocytes/metabolism
3.
MAbs ; 12(1): 1795505, 2020.
Article in English | MEDLINE | ID: mdl-32744145

ABSTRACT

Current combination therapies elicit high response rates in B cell malignancies, often using CD20 antibodies as the backbone of therapy. However, many patients eventually relapse or develop progressive disease. Therefore, novel CD20 antibodies combining multiple effector mechanisms were generated. To study whether neutrophil-mediated destruction of B cell malignancies can be added to the arsenal of effector mechanisms, we chimerized a panel of five previously described murine CD20 antibodies to the human IgG1, IgA1 and IgA2 isotype. Of this panel, we assessed in vitro antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and direct cell death induction capacity and studied the efficacy in two different in vivo mouse models. IgA antibodies outperformed IgG1 antibodies in neutrophil-mediated killing in vitro, both against CD20-expressing cell lines and primary patient material. In these assays, we observed loss of CD19 with both IgA and IgG antibodies. Therefore, we established a novel method to improve the assessment of B-cell depletion by CD20 antibodies by including CD24 as a stable cell marker. Subsequently, we demonstrated that only IgA antibodies were able to reduce B cell numbers in this context. Additionally, IgA antibodies showed efficacy in both an intraperitoneal tumor model with EL4 cells expressing huCD20 and in an adoptive transfer model with huCD20-expressing B cells. Taken together, we show that IgA, like IgG, can induce ADCC and CDC, but additionally triggers neutrophils to kill (malignant) B cells. We conclude that antibodies of the IgA isotype offer an attractive repertoire of effector mechanisms for the treatment of CD20-expressing malignancies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Antigens, CD20/immunology , B-Lymphocytes/immunology , Hematologic Neoplasms/immunology , Immunoglobulin A/pharmacology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Neutrophil Activation/drug effects , Neutrophils/immunology , Animals , B-Lymphocytes/pathology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Immunoglobulin A/immunology , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neutrophils/pathology , Xenograft Model Antitumor Assays
4.
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
5.
Blood ; 120(3): e9-e16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653974

ABSTRACT

Interactions within the hematopoietic niche in the BM microenvironment are essential for maintenance of the stem cell pool. In addition, this niche is thought to serve as a sanctuary site for malignant progenitors during chemotherapy. Therapy resistance induced by interactions with the BM microenvironment is a major drawback in the treatment of hematologic malignancies and bone-metastasizing solid tumors. To date, studying these interactions was hampered by the lack of adequate in vivo models that simulate the human situation. In the present study, we describe a unique human-mouse hybrid model that allows engraftment and outgrowth of normal and malignant hematopoietic progenitors by implementing a technology for generating a human bone environment. Using luciferase gene marking of patient-derived multiple myeloma cells and bioluminescent imaging, we were able to follow pMM cells outgrowth and to visualize the effect of treatment. Therapeutic interventions in this model resulted in equivalent drug responses as observed in the corresponding patients. This novel human-mouse hybrid model creates unprecedented opportunities to investigate species-specific microenvironmental influences on normal and malignant hematopoietic development, and to develop and personalize cancer treatment strategies.


Subject(s)
Hematopoietic Stem Cells/cytology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Stem Cell Niche/immunology , Transplantation Chimera/immunology , Tumor Microenvironment/immunology , Animals , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear Ossicles/cytology , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mice , Mice, Mutant Strains , Neoplasm Transplantation , Osteolysis/immunology , Tissue Scaffolds , Transplantation, Heterologous
6.
Cancer Cell ; 20(3): 370-83, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21907927

ABSTRACT

The development of resistance to chemotherapy is a major obstacle for lasting effective treatment of cancer. Here, we demonstrate that endogenous mesenchymal stem cells (MSCs) become activated during treatment with platinum analogs and secrete factors that protect tumor cells against a range of chemotherapeutics. Through a metabolomics approach, we identified two distinct platinum-induced polyunsaturated fatty acids (PIFAs), 12-oxo-5,8,10-heptadecatrienoic acid (KHT) and hexadeca-4,7,10,13-tetraenoic acid (16:4(n-3)), that in minute quantities induce resistance to a broad spectrum of chemotherapeutic agents. Interestingly, blocking central enzymes involved in the production of these PIFAs (cyclooxygenase-1 and thromboxane synthase) prevents MSC-induced resistance. Our findings show that MSCs are potent mediators of resistance to chemotherapy and reveal targets to enhance chemotherapy efficacy in patients.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclooxygenase 1/metabolism , Drug Resistance, Neoplasm , Fatty Acids, Unsaturated/metabolism , Fatty Acids/metabolism , Mesenchymal Stem Cells/metabolism , Platinum Compounds/pharmacology , Thromboxane-A Synthase/metabolism , Animals , Apoptosis/drug effects , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cyclooxygenase Inhibitors , Humans , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Oxaliplatin , Thromboxane-A Synthase/antagonists & inhibitors , Tumor Cells, Cultured
7.
Haematologica ; 95(12): 2063-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20851867

ABSTRACT

BACKGROUND: Incorporation of the chimeric CD20 monoclonal antibody rituximab in the treatment schedule of patients with non-Hodgkin's lymphoma has significantly improved outcome. Despite this success, about half of the patients do not respond to treatment or suffer from a relapse and additional therapy is required. A low CD20-expression level may in part be responsible for resistance against rituximab. We therefore investigated whether the CD20-expression level related resistance to rituximab could be overcome by a new group of CD20 mAbs (HuMab-7D8 and ofatumumab) targeting a unique membrane-proximal epitope on the CD20 molecule. DESIGN AND METHODS: By retroviral transduction of the CD20 gene into CD20-negative cells and clonal selection of transduced cells a system was developed in which the CD20-expression level is the only variable. These CD20 transduced cells were used to study the impact of rituximab and HuMab-7D8 mediated complement-dependent cytotoxicity. To study the in vivo efficacy of these mAbs an in vivo imaging system was generated by retroviral expression of the luciferase gene in the CD20-positive cells. RESULTS: We show that HuMab-7D8 efficiently killed CD20(low) cells that are not susceptible to rituximab-induced killing in vitro. In a mouse xenograft model, we observed a comparable increase in survival time between HuMab-7D8 and rituximab-treated mice. Most significantly, however, HuMab-7D8 eradicated all CD20-expressing cells both in the periphery as well as in the bone marrow whereas after rituximab treatment CD20(low) cells survived. CONCLUSIONS: Cells that are insensitive to in vitro and in vivo killing by rituximab as the result of their low CD20-expression profile may be efficiently killed by an antibody against the membrane-proximal epitope on CD20. Such antibodies should, therefore, be explored to overcome rituximab resistance in the clinic.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD20/metabolism , Cytotoxicity, Immunologic/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, CD20/genetics , Antigens, CD20/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Epitopes/immunology , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Membrane Microdomains/metabolism , Mice , Protein Transport , Rituximab , Xenograft Model Antitumor Assays
8.
Cell ; 111(2): 241-50, 2002 Oct 18.
Article in English | MEDLINE | ID: mdl-12408868

ABSTRACT

The transactivation of TCF target genes induced by Wnt pathway mutations constitutes the primary transforming event in colorectal cancer (CRC). We show that disruption of beta-catenin/TCF-4 activity in CRC cells induces a rapid G1 arrest and blocks a genetic program that is physiologically active in the proliferative compartment of colon crypts. Coincidently, an intestinal differentiation program is induced. The TCF-4 target gene c-MYC plays a central role in this switch by direct repression of the p21(CIP1/WAF1) promoter. Following disruption of beta-catenin/TCF-4 activity, the decreased expression of c-MYC releases p21(CIP1/WAF1) transcription, which in turn mediates G1 arrest and differentiation. Thus, the beta-catenin/TCF-4 complex constitutes the master switch that controls proliferation versus differentiation in healthy and malignant intestinal epithelial cells.


Subject(s)
Colorectal Neoplasms/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Cell Cycle , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Humans , Intestinal Mucosa/metabolism , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Tumor Cells, Cultured , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...