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.
J Mol Med (Berl) ; 98(4): 585-593, 2020 04.
Article in English | MEDLINE | ID: mdl-32108909

ABSTRACT

Blocking the proteolytic capacity of urokinase-type plasminogen activator (uPA) with a monoclonal antibody (mAb) reduces arthritis progression in the collagen-induced mouse arthritis model to an extent that is on par with the effect of blocking tumor necrosis factor-alpha by etanercept. Seeking to develop a novel therapy for rheumatoid arthritis, a humanized mAb, NNC0266-0043, was selected for its dual inhibition of both the zymogen activation and the proteolytic capacity of human uPA. The antibody revealed nonlinear elimination kinetics in cynomolgus monkeys consistent with binding to and turnover of endogenous uPA. At a dose level of 20.6 mg kg-1, the antibody had a plasma half-life of 210 h. Plasma uPA activity, a pharmacodynamic marker of anti-uPA therapy, was reduced to below the detection limit during treatment, indicating that an efficacious plasma concentration was reached. Pharmacokinetic modeling predicted that sufficient antibody levels can be sustained in arthritis patients dosed subcutaneously once weekly. The anti-uPA mAb was also well tolerated in cynomolgus monkeys at weekly doses up to 200 mg kg-1 over 4 weeks. The data from cynomolgus monkeys and from human material presented here indicates that anti-uPA mAb NNC0266-0043 is suitable for clinical testing as a novel therapeutic for rheumatic diseases. KEY MESSAGES: Background: Anti-uPA therapy is on par with etanercept in a mouse arthritis model. A new humanized antibody blocks activation and proteolytic activity of human uPA. The antibody represents a radically novel mode-of-action in anti-rheumatic therapy. The antibody has PK/PD properties in primates consistent with QW clinical dosing.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/etiology , Drug Development , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Immunohistochemistry , Macaca fascicularis , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Monocytes/metabolism
2.
J Immunol ; 200(3): 957-965, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29282305

ABSTRACT

Genetic absence of the urokinase-type plasminogen activator (uPA) reduces arthritis progression in the collagen-induced arthritis (CIA) mouse model to an extent just shy of disease abrogation, but this remarkable observation has not been translated into therapeutic intervention. Our aim was to test the potential in mice of an Ab that blocks the proteolytic capacity of uPA in the CIA model and the delayed-type hypersensitivity arthritis model. A second aim was to determine the cellular origins of uPA and the uPA receptor (uPAR) in joint tissue from patients with rheumatoid arthritis. A mAb that neutralizes mouse uPA significantly reduced arthritis progression in the CIA and delayed-type hypersensitivity arthritis models. In the CIA model, the impact of anti-uPA treatment was on par with the effect of blocking TNF-α by etanercept. A pharmacokinetics evaluation of the therapeutic Ab revealed target-mediated drug disposition consistent with a high turnover of endogenous uPA. The cellular expression patterns of uPA and uPAR were characterized by double immunofluorescence in the inflamed synovium from patients with rheumatoid arthritis and compared with synovium from healthy donors. The arthritic synovium showed expression of uPA and uPAR in neutrophils, macrophages, and a fraction of endothelial cells, whereas there was little or no expression in synovium from healthy donors. The data from animal models and human material provide preclinical proof-of-principle that validates uPA as a novel therapeutic target in rheumatic diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Receptors, Urokinase Plasminogen Activator/metabolism , Synovial Membrane/pathology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Disease Progression , Endothelial Cells/immunology , Etanercept/pharmacology , Female , Humans , Hypersensitivity, Delayed/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neutrophils/immunology , Synovial Membrane/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
3.
J Immunol ; 194(7): 3169-79, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25725106

ABSTRACT

Osteoclast-associated receptor (OSCAR) is widely expressed on human myeloid cells. Collagen types (Col)I, II, and III have been described as OSCAR ligands, and ColII peptides can induce costimulatory signaling in receptor activator for NF-κB-dependent osteoclastogenesis. In this study, we isolated collagen as an OSCAR-interacting protein from the membranes of murine osteoblasts. We have investigated a functional outcome of the OSCAR-collagen interaction in human monocyte-derived dendritic cells (DCs). OSCAR engagement by ColI/II-induced activation/maturation of DCs is characterized by upregulation of cell surface markers and secretion of cytokines. These collagen-matured DCs (Col-DCs) were efficient drivers of allogeneic and autologous naive T cell proliferation. The T cells expanded by Col-DCs secreted cytokines with no clear T cell polarization pattern. Global RNA profiling revealed that multiple proinflammatory mediators, including cytokines and cytokine receptors, components of the stable immune synapse (namely CD40, CD86, CD80, and ICAM-1), as well as components of TNF and TLR signaling, are transcriptional targets of OSCAR in DCs. Our findings indicate the existence of a novel pathway by which extracellular matrix proteins locally drive maturation of DCs during inflammatory conditions, for example, within synovial tissue of rheumatoid arthritis patients, where collagens become exposed during tissue remodeling and are thus accessible for interaction with infiltrating precursors of DCs.


Subject(s)
Cell Differentiation , Collagen/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Monocytes/cytology , Monocytes/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Antigens, Surface/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Survival/drug effects , Chemokines/biosynthesis , Coculture Techniques , Collagen/pharmacology , Cytokines/biosynthesis , Dendritic Cells/drug effects , Gene Expression Regulation , Humans , Immunophenotyping , Ligands , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Monocytes/drug effects , NF-kappa B/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Front Pharmacol ; 3: 122, 2012.
Article in English | MEDLINE | ID: mdl-22754528

ABSTRACT

Identification of targets for cancer therapy requires the understanding of the in vivo roles of proteins, which can be derived from studies using gene-targeted mice. An alternative strategy is the administration of inhibitory monoclonal antibodies (mAbs), causing acute disruption of the target protein function(s). This approach has the advantage of being a model for therapeutic targeting. mAbs for use in mouse models can be obtained through immunization of gene-deficient mice with the autologous protein. Such mAbs react with both species-specific epitopes and epitopes conserved between species. mAbs against proteins involved in extracellular proteolysis, including plasminogen activators urokinase plasminogen activator (uPA), tissue-type plasminogen activator (tPA), their inhibitor PAI-1, the uPA receptor (uPAR), two matrix metalloproteinases (MMP9 and MMP14), as well as the collagen internalization receptor uPARAP, have been developed. The inhibitory mAbs against uPA and uPAR block plasminogen activation and thereby hepatic fibrinolysis in vivo. Wound healing, another plasmin-dependent process, is delayed by an inhibitory mAb against uPA in the adult mouse. Thromboembolism can be inhibited by anti-PAI-1 mAbs in vivo. In conclusion, function-blocking mAbs are well-suited for targeted therapy in mouse models of different diseases, including cancer.

5.
J Immunol Methods ; 339(1): 55-65, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18761343

ABSTRACT

The urokinase plasminogen activator receptor (uPAR) is a versatile three-domain GPI-anchored protein, which binds urokinase plasminogen activator (uPA) and thereby focalises plasminogen activation on the cell surface. Generation of a proteolytic potential is essential in both normal physiological and pathological extracellular tissue remodelling processes. uPA can also cleave uPAR, resulting in liberation of the amino-terminal domain I, which encompasses binding sites for both uPA and the adhesion molecule, vitronectin. In order to localise the different uPAR forms on the plasma membrane of murine monocyte macrophage-like P388D.1 cells, we have now generated and characterised two high-affinity murine mAbs, mR3 and mR4, raised against murine uPAR. mR3 was found to recognise an epitope located in domain I of uPAR. Surface plasmon resonance analyses and cell binding studies revealed that this mAb was able to bind preformed complexes of murine pro-uPA and murine uPAR. In contrast, mR4 recognises domains II-III in uPAR and does not bind preformed pro-uPA-uPAR complexes in similar analyses. Immunofluorescence microscopy of P388D.1 cells revealed that mR3 stained the cells equally well in the presence or absence of saturation with the amino-terminal fragment of uPA, ATF. However, the signal intensity obtained using another uPAR domain I specific mAb, mR1, was significantly reduced upon ATF saturation. Furthermore, when adding ATF, mR4 selectively stained the cleaved receptor. Applying these newly generated mAbs, we additionally demonstrated that cleaved and intact uPAR was evenly distributed on the surface of these cells.


Subject(s)
Antibodies, Monoclonal/chemistry , Receptors, Urokinase Plasminogen Activator/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Binding Sites, Antibody/immunology , Cell Line , Cell Membrane/chemistry , Cell Membrane/immunology , Humans , Mice , Protein Structure, Tertiary , Receptors, Urokinase Plasminogen Activator/immunology , Surface Plasmon Resonance/methods , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/immunology
6.
Thromb Haemost ; 97(6): 1013-22, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17549305

ABSTRACT

Binding of urokinase plasminogen activator (uPA) to its cellular receptor, uPAR, potentiates plasminogen activation and localizes it to the cell surface. Focal plasminogen activation is involved in both normal and pathological tissue remodeling processes including cancer invasion. The interaction between uPA and uPAR therefore represents a potential target for anti-invasive cancer therapy. Inhibitors of the human uPA-uPAR interaction have no effect in the murine system. To enable in-vivo studies in murine cancer models we have now generated murine monoclonal antibodies (mAbs) against murine uPAR (muPAR) by immunizing uPAR-deficient mice with recombinant muPAR and screened for antibodies, which inhibit the muPA-muPAR interaction. Two of the twelve mAbs obtained, mR1 and mR2, interfered with the interaction between muPAR and the amino-terminal fragment of muPA (mATF) when analyzed by surface plasmon resonance. The epitope for mR1 is located on domain I of muPAR, while that of mR2 is on domains (II-III). In cell binding experiments using radiolabelled mATF, the maximal inhibition obtained with mR1 was 85% while that obtained with mR2 was 50%. The IC(50) value for mR1 was 0.67 nM compared to 0.14 nM for mATF. In an assay based on modified anthrax toxins, requiring cell-bound muPA activity for its cytotoxity, an approximately 50% rescue of the cells could be obtained by addition of mR1. Importantly, in-vivo efficacy of mR1 was demonstrated by the ability of mR1 to rescue mice treated with a lethal dose of uPA-activatable anthrax toxins.


Subject(s)
Antibodies, Monoclonal/pharmacology , Macrophages/drug effects , Plasminogen/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antigen-Antibody Reactions , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Binding Sites, Antibody , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epitope Mapping , Female , Humans , Hybridomas/metabolism , Immunization , Iodine Radioisotopes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Peptide Fragments/metabolism , Protein Structure, Tertiary , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Time Factors
7.
Biochemistry ; 43(40): 12877-86, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15461460

ABSTRACT

Human cysteine-rich secretory protein 3 (CRISP-3; also known as SGP28) belongs to a family of closely related proteins found in mammals and reptiles. Some mammalian CRISPs are known to be involved in the process of reproduction, whereas some of the CRISPs from reptiles are neurotoxin-like substances found in lizard saliva or snake venom. Human CRISP-3 is present in exocrine secretions and in secretory granules of neutrophilic granulocytes and is believed to play a role in innate immunity. On the basis of the relatively high content of CRISP-3 in human plasma and the small size of the protein (28 kDa), we hypothesized that CRISP-3 in plasma was bound to another component. This was supported by size-exclusion chromatography and immunoprecipitation of plasma proteins. The binding partner was identified by mass spectrometry as alpha(1)B-glycoprotein (A1BG), which is a known plasma protein of unknown function and a member of the immunoglobulin superfamily. We demonstrate that CRISP-3 is a specific and high-affinity ligand of A1BG with a dissociation constant in the nanomolar range as evidenced by surface plasmon resonance. The A1BG-CRISP-3 complex is noncovalent with a 1:1 stoichiometry and is held together by strong electrostatic forces. Similar complexes have been described between toxins from snake venom and A1BG-like plasma proteins from opossum species. In these cases, complex formation inhibits the toxic effect of snake venom metalloproteinases or myotoxins and protects the animal from envenomation. We suggest that the A1BG-CRISP-3 complex displays a similar function in protecting the circulation from a potentially harmful effect of free CRISP-3.


Subject(s)
Blood Proteins/metabolism , Glycoproteins/blood , Glycoproteins/metabolism , Immunoglobulins/blood , Immunoglobulins/metabolism , Salivary Proteins and Peptides/blood , Salivary Proteins and Peptides/metabolism , Seminal Plasma Proteins/blood , Seminal Plasma Proteins/metabolism , Blood Proteins/chemistry , Chromatography , Glycoproteins/chemistry , Glycosylation , Humans , Immunoglobulins/chemistry , Kinetics , Ligands , Molecular Weight , Protein Binding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Plasmon Resonance , Titrimetry
8.
Clin Chem ; 50(11): 2059-68, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15345662

ABSTRACT

BACKGROUND: The cell surface receptor (uPAR) for urokinase plasminogen activator (uPA) is a strong prognostic marker in several types of cancer. uPA cleaves the three-domain protein uPAR(I-III) into two fragments: uPAR(I), which contains domain I; and uPAR(II-III), which contains domains II and III. Established immunoassays measure a combination of uPAR forms. Our aim was to design immunoassays for specific quantification of the individual forms of uPAR. METHODS: Using appropriate combinations of epitope-mapped monoclonal antibodies (Mabs) for capture and europium-labeled detection Mabs, we designed two-site sandwich time-resolved fluorescence immunoassays (TR-FIAs): TR-FIA 1 to measure uPAR(I-III) alone; TR-FIA 2 to measure both uPAR(I-III) and uPAR(II-III); and TR-FIA 3 to measure uPAR(I). To avoid detection of uPAR(I-III) in TR-FIA 3, we used a combination of the peptide uPAR antagonist AE120 and a domain I antibody, R3. AE120 blocks the binding of R3 to uPAR(I-III). In contrast, AE120 does not interact with liberated domain I and therefore does not interfere with the binding of R3 to uPAR(I). RESULTS: The limits of quantification (CV <20%) determined by adding the proteins to uPAR-depleted plasma were <3 pmol/L in all three assays. The interassay CVs in plasma with added analytes were <11%, and recoveries were between 93% and 105%. Cross-reactivities of purified proteins in the three TR-FIAs were no more than 4%. Studies on chymotrypsin cleavage of uPAR and size-exclusion chromatography of plasma with and without added protein further supported the specificity of the assays. CONCLUSIONS: The three novel TR-FIAs accurately quantify uPAR(I-III) alone, uPAR(I-III) together with uPAR(II-III), and uPAR(I), respectively, in biological samples, including plasma, and thus are well suited for studies of the diagnostic and prognostic value of individual uPAR forms in cancer patients.


Subject(s)
Receptors, Cell Surface/analysis , Urokinase-Type Plasminogen Activator/metabolism , Biomarkers, Tumor/blood , Chymotrypsin/chemistry , Cross Reactions , Fluoroimmunoassay , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/urine , Male , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Receptors, Cell Surface/blood , Receptors, Cell Surface/chemistry , Receptors, Urokinase Plasminogen Activator , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...