Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Biol Chem ; 293(24): 9326-9334, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29678878

ABSTRACT

Despite the success of current biological therapeutics for rheumatoid arthritis, these therapies, targeting individual cytokines or pathways, produce beneficial responses in only about half of patients. Therefore, better therapeutics are needed. IL-6 and IL-17A are proinflammatory cytokines in many autoimmune and inflammatory diseases, and several therapeutics have been developed to specifically inhibit them. However, targeting both of these cytokines with a bispecific therapeutic agent could account for their nonoverlapping proinflammatory functions and for the fact that IL-6 and IL-17A act in a positive feedback loop. Here, we present the development of MT-6194, a bispecific antibody targeting both IL-6R and IL-17A that was developed with the FynomAb technology. We also present data from mouse inflammatory disease experiments, indicating that simultaneous inhibition of both IL-6 and IL-17A yields enhanced efficacy compared with inhibition of each cytokine alone.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Inflammation/drug therapy , Interleukin-17/immunology , Receptors, Interleukin-6/immunology , Animals , Antibodies, Bispecific/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Line , Humans , Inflammation/immunology , Interleukin-17/antagonists & inhibitors , Macaca fascicularis , Receptors, Interleukin-6/antagonists & inhibitors
2.
J Biol Chem ; 289(20): 14392-8, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24692552

ABSTRACT

Fynomers are small binding proteins derived from the human Fyn SH3 domain. Using phage display technology, Fynomers were generated inhibiting the activity of the proinflammatory cytokine interleukin-17A (IL-17A). One specific Fynomer called 2C1 inhibited human IL-17A in vitro with an IC50 value of 2.2 nm. Interestingly, when 2C1 was genetically fused to the Fc part of a human antibody via four different amino acid linkers to yield bivalent IL-17A binding proteins (each linker differed in length), the 2C1-Fc fusion protein with the longest linker displayed the most potent inhibitory activity. It blocked homodimeric IL-17A with an IC50 value of 21 pm, which corresponds to a hundredfold improved IC50 value as compared to the value obtained with monovalent Fynomer 2C1. In contrast, the 2C1-Fc fusion with the shortest linker showed only an ∼8-fold improved IC50 value of 260 pm. Furthermore, in a mouse model of acute inflammation, we have shown that the most potent 2C1-Fc fusion protein is able to efficiently inhibit IL-17A in vivo. With their suitable biophysical properties, Fynomer-Fc fusion proteins represent new drug candidates for the treatment of IL-17A mediated inflammatory conditions such as psoriasis, psoriatic arthritis, or rheumatoid arthritis.


Subject(s)
Immunoglobulin Fc Fragments/genetics , Interleukin-17/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , src Homology Domains , Amino Acid Sequence , Animals , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics
3.
J Nucl Med ; 48(4): 582-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17401095

ABSTRACT

UNLABELLED: Targeting proteins that are overexpressed in atherosclerotic plaques may open novel diagnostic applications. The C domain of tenascin-C is absent from normal adult tissues but can be inserted during tumor progression or tissue repair into the molecule by alternative splicing. We tested the ability of the human antibody G11, specific to this antigen, to reveal murine atherosclerotic plaques ex vivo. The antibody directed against the extra domain B of fibronectin (L19) was used as a reference. METHODS: We intravenously injected (125)I-labeled G11 or L19 antibodies into apolipoprotein E-deficient (ApoE(-/-)) mice and harvested the aortae 4 or 24 h later. En face analyses of distal aortae and longitudinal sections of the aortic arch were performed to compare antibody uptake using autoradiography with plaque staining using oil red O. Plaque macrophages were detected by immunohistochemistry (anti-CD68 staining). Biodistribution of injected antibodies was investigated in aortae and blood at 4 and 24 h. RESULTS: En face analyses revealed a significant correlation between radiolabeled G11 and fat-stained areas, increasing from 4 to 24 h, with a correlation coefficient of 0.92 (P < 0.0001) and an average signal-to-noise ratio of 104:1 at 24 h. Plaque imaging using L19 showed similar results (r = 0.86; P < 0.0001; signal-to-noise ratio, 72:1 at 24 h). Uptake of radiolabeled antibodies in histologic sections colocalized with fat staining and activated macrophages in aortic plaques. Biodistribution analyses confirmed specific accumulation in aortic plaques as well as rapid blood pool clearance of the antibodies 24 h after injection. Immunofluorescence analyses revealed increased expression of tenascin and fibronectin isoforms in macrophage-rich plaques. CONCLUSION: The antibody G11, specific to the C domain of tenascin-C, visualizes murine atherosclerotic plaques ex vivo. In conjunction with the increased expression of the C domain of tenascin-C in macrophage-rich plaques, the colocalization of G11 uptake with activated macrophages, and the favorable target-to-blood ratio at 24 h, this antibody may be useful for molecular imaging of advanced atherosclerotic plaques in the intact organism.


Subject(s)
Atherosclerosis/metabolism , Tenascin/chemistry , Alternative Splicing , Animals , Antibodies/chemistry , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/diagnosis , Atherosclerosis/pathology , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Time Factors , Tissue Distribution
4.
Clin Cancer Res ; 12(10): 3200-8, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707621

ABSTRACT

BACKGROUND: The targeted delivery of bioactive molecules with antibodies specific to tumor-associated antigens represents a promising strategy for improving the efficacy of tumor therapy. The large isoform of tenascin-C, an abundant glycoprotein of the tumor extracellular matrix, is strongly overexpressed in adult tissue undergoing tissue remodeling, including wound healing and neoplasia, and has been implicated in a variety of different cancers while being virtually undetectable in most normal adult tissues. EXPERIMENTAL DESIGN: We have used antibody phage technology to generate good-quality human recombinant antibodies (F16 and P12) specific to the alternatively spliced domains A1 and D of the large isoform of tenascin-C. The tumor-targeting properties of F16 and P12 were assessed by biodistribution studies in tumor xenografts using the antibodies in small immunoprotein (SIP) format. RESULTS: SIP(F16) selectively accumulated at the tumor site with 4.5%ID/g at 24 hours in the U87 glioblastoma model but was rapidly cleared from other organs (tumor-to-organ ratios, approximately 10:1). The accumulation of SIP(P12) in the tumor was lower compared with SIP(F16) and persistent levels of radioactivity were observed in the intestine. CONCLUSIONS: These data suggest that the F16 antibody, specific to domain A1 of tenascin-C, is a promising building block for the development of antibody-based pharmaceuticals in view of its excellent tumor-targeting performance and the strong expression of the antigen in a variety of primary and metastatic tumors.


Subject(s)
Antibodies/analysis , Antibodies/immunology , Immunoglobulin Variable Region , Tenascin/immunology , Alternative Splicing , Animals , Antibodies/therapeutic use , Antibody Formation , Brain Neoplasms/immunology , Extracellular Matrix , Glioblastoma/immunology , Immunoglobulin Fragments , Immunohistochemistry , Melanoma/immunology , Mice , Mice, Nude , Protein Isoforms , Skin Neoplasms/immunology , Tenascin/chemistry , Tissue Distribution , Transplantation, Heterologous
5.
Protein Eng Des Sel ; 19(10): 471-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16928692

ABSTRACT

We had previously reported that splice isoforms of tenascin-C containing the extra-domain C are virtually absent in normal adult tissues but are highly abundant in high-grade astrocytomas, with a prominent peri-vascular pattern of expression. We now report that the extra-domain C of tenascin-C is strongly expressed in the majority of lung cancers, with a vascular and stromal pattern of expression. Using antibody phage technology, we have generated a human monoclonal antibody (G11), with a dissociation constant K(D) = 4.2 nM for the human domain C. The G11 antibody, expressed in scFv and in mini-antibody (SIP) format, as well as a scFv-interleukin-2 fusion protein, was then characterized in quantitative biodistribution studies using mice grafted subcutaneously with U87 gliomas, revealing a selective tumor uptake, with tumor/blood ratios up to 11.8:1 at 24 h. A radioiodinated preparation of SIP(G11) was also investigated in a double tracer study using an orthotopic rat glioma model, confirming the antibody's ability to preferentially localize at the tumor site, with tumor/brain ratios superior to the ones observed with (18)F-fluorodeoxyglucose. These tumor-targeting properties, together with the strong immunohistochemical staining of human tumor sections, indicate that the G11 antibody may be used as a portable targeting moiety for the selective delivery of imaging and therapeutic agents to gliomas and lung tumors.


Subject(s)
Antibodies, Monoclonal/chemistry , Neoplasms/therapy , Tenascin/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Glioma/therapy , Humans , Immunotherapy/methods , Kinetics , Molecular Sequence Data , Neoplasms/immunology , Peptide Library , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry
6.
J Cancer Res Clin Oncol ; 132(8): 537-46, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16788848

ABSTRACT

PURPOSE: Through alternative splicing of the extracellular matrix protein tenascin-C (Tn-C) primary transcript nine type III homology repeats can be independently included or omitted. Large, low spliced Tn-C variants (Tn-C(L)) are preferentially expressed during tissue remodelling processes like tumour invasion to modulate cell migration. The study was aimed to evaluate the differential expression of Tn-C splicing domains in urinary bladder carcinoma with respect to the invasive behaviour. METHODS: The deposition and synthesis of the Tn-C splicing domains A1-D was analysed in 34 urinary bladder carcinomas by semiquantitative immunohistochemistry using domain specific antibodies and by RT-PCR. Results were correlated to tumour stage and grade. RESULTS: There is a significant increase of Tn-C(L) with higher tumour stage and grade. Immunohistochemistry revealed a more restricted distribution pattern of A1, B, and/or D domain containing Tn-C variants to invasive tumours, tumour vessels, and to destructed muscle. The mRNA expression patterns of the domains A1-A3 are similar among the different carcinomas. Stronger differences exist in the region from the B to D domain. In general, the domains AD1/C are rarely expressed. AD1 domain expression seems to be connected with compact invasion pattern. CONCLUSION: In urinary bladder carcinoma a differential expression of Tn-C splicing variants exists in dependence of tumour type, vascularization, and invasive behaviour. Therefore, the detection of different Tn-C splicing domains could be useful for assessment of muscle invasion, tumour surveillance, as well as target structures for antibody based tumour detection and therapy.


Subject(s)
Carcinoma, Transitional Cell/chemistry , Protein Splicing , Tenascin/analysis , Urinary Bladder Neoplasms/chemistry , Aged , Aged, 80 and over , Carcinoma, Papillary/chemistry , Carcinoma, Transitional Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/chemistry
7.
MAbs ; 8(1): 141-9, 2016.
Article in English | MEDLINE | ID: mdl-26390837

ABSTRACT

Biologic treatment options such as tumor necrosis factor (TNF) inhibitors have revolutionized the treatment of inflammatory diseases, including rheumatoid arthritis. Recent data suggest, however, that full and long-lasting responses to TNF inhibitors are limited because of the activation of the pro-inflammatory TH17/interleukin (IL)-17 pathway in patients. Therefore, dual TNF/IL-17A inhibition is an attractive avenue to achieve superior efficacy levels in such diseases. Based on the marketed anti-TNF antibody adalimumab, we generated the bispecific TNF/IL-17A-binding FynomAb COVA322. FynomAbs are fusion proteins of an antibody and a Fyn SH3-derived binding protein. COVA322 was characterized in detail and showed a remarkable ability to inhibit TNF and IL-17A in vitro and in vivo. Through its unique mode-of-action of inhibiting simultaneously TNF and the IL-17A homodimer, COVA322 represents a promising drug candidate for the treatment of inflammatory diseases. COVA322 is currently being tested in a Phase 1b/2a study in psoriasis ( ClinicalTrials.gov Identifier: NCT02243787).


Subject(s)
Interleukin-17/antagonists & inhibitors , Psoriasis/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Female , Humans , Interleukin-17/immunology , Male , Mice , Psoriasis/immunology , Tumor Necrosis Factor-alpha/immunology
8.
Trends Biotechnol ; 20(12 Suppl): S19-22, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12570155

ABSTRACT

The simultaneous identification and quantitative measurement of the production levels of thousands of different proteins in a biological specimen remains an unachieved goal of modern proteomic research. Advances in the development of microarray-based platforms for highly parallel detection of proteins have therefore received a considerable impulse during the last few years. Here, we review the existing reagents for affinity capture of protein targets, as well as the techniques used for their immobilization on solid supports and methods for the detection of binding events, underlining the problems and the opportunities in this continuously evolving research field.


Subject(s)
Peptides/chemistry , Protein Array Analysis , Proteins/chemistry , Biotechnology/trends , Enzyme-Linked Immunosorbent Assay , Kinetics , Peptide Library , Protein Binding , Proteome
9.
Arthritis Res Ther ; 9(1): R9, 2007.
Article in English | MEDLINE | ID: mdl-17261171

ABSTRACT

The antibody-mediated targeted delivery of cytokines to sites of disease is a promising avenue for cancer therapy, but it is largely unexplored for the treatment of chronic inflammatory conditions. Using both radioactive and fluorescent techniques, the human monoclonal antibodies L19 and G11 (specific to two markers of angiogenesis that are virtually undetectable in normal adult tissues) were found to selectively localize at arthritic sites in the murine collagen-induced model of rheumatoid arthritis following intravenous (i.v.) administration. The same animal model was used to study the therapeutic action of the L19 antibody fused to the cytokines IL-2, tumour necrosis factor (TNF) and IL-10. Whereas L19-IL-2 and L19-TNF treatment led to increased arthritic scores and paw swellings, the fusion protein L19-IL-10 displayed a therapeutic activity, which was superior to the activity of IL-10 fused to an antibody of irrelevant specificity in the mouse. The anti-inflammatory cytokine IL-10 has been investigated for the treatment of patients with rheumatoid arthritis, but clinical development plans have been discontinued because of a lack of efficacy. Because the antigen recognised by L19 is strongly expressed at sites of arthritis in humans and identical in both mice and humans, it suggests that the fusion protein L19-IL-10 might help overcome some of the clinical limitations of IL-10 and provide a therapeutic benefit to patients with chronic inflammatory disorders, including arthritis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/prevention & control , Drug Delivery Systems/methods , Interleukin-10/administration & dosage , Animals , Antibodies, Monoclonal/immunology , Arthritis, Experimental/immunology , Cell Line , Disease Progression , Humans , Interleukin-10/immunology , Male , Mice , Mice, Inbred DBA
10.
Proteomics ; 6(16): 4496-505, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16858729

ABSTRACT

The identification of biomarkers from serum or plasma is often hindered by a few proteins present at high concentrations, which may obscure less abundant proteins. Ideal serum depletion strategies would be flexible as regards the proteins to be removed, and would feature the use of reagents with long shelf-lives. In this article, we describe a novel protein depletion methodology based on the incubation of serum samples with phage-derived recombinant antibody fragments, which are able to bind to staphylococcal Protein A, and which carry a C-terminal peptide tag capable of streptavidin binding. The resulting protein-antibody complexes can be removed by simultaneous capture on Protein A and/or streptavidin resin. The depletion methodology was exemplified by the isolation of recombinant human mAb fragments specific to abundant human serum Ags and by the simultaneous depletion of albumin, immunoglobulins, alpha2-macroglobulin, hemoglobin, transferrin and haptoglobin. The depleted serum samples were analyzed by 2-DE and by gel-free MS-based methodologies, confirming the efficiency and selectivity of the depletion process. The methodology presented is modular in nature, since several recombinant antibodies can be combined in a single depletion experiment. Furthermore, antibodies do not have to be covalently coupled to a solid support facilitating long-term storage.


Subject(s)
Antibodies/immunology , Blood Proteins/analysis , Proteomics , Antibodies/chemistry , Electrophoresis, Gel, Two-Dimensional , Humans , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/immunology
11.
Proteomics ; 5(9): 2340-50, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15880779

ABSTRACT

Advances in proteomic research allow the identification of several hundred protein components in complex biological specimens. Structural information is typically lost during proteomic investigations. For this reason, the rapid isolation of monoclonal antibodies specific to proteins of interest would allow the study of structurally intact biological specimens, thus providing complementary proteomic information. Here, we describe the design, construction, characterization, and use of a large synthetic human antibody phage display library (ETH-2-Gold) containing three billion individual antibody clones. A large repertoire of antibodies with similar biochemical properties was produced by appending short variable complementarity-determining region 3 (CDR3) onto three antibody germline segments (DP47, DPK22, and DPL16), which are frequently found in human antibodies. The ETH-2-Gold library exhibits efficient display of antibody fragments on filamentous phage, as assessed by immunoblot. Furthermore, the library is highly functional, since >90% of clones express soluble antibodies in bacteria and since good quality monoclonal antibodies have been isolated against 16 different antigens. The usefulness of the library as a tool for generating monoclonal antibodies for biomedical applications was tested using the C-domain of tenascin-C (a marker of angiogenesis) as antigen and showing that specific antibodies to this target were able to stain vascular structures in tumor sections.


Subject(s)
Antibodies, Monoclonal , Peptide Library , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibody Specificity , Bacteria/metabolism , Capsid Proteins , Cloning, Molecular , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Glioblastoma/blood supply , Humans , Immunoglobulin Variable Region/genetics , Inovirus/metabolism , Neovascularization, Pathologic/metabolism , Protein Structure, Tertiary , Tenascin/analysis , Tenascin/immunology , Viral Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL