Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 8: 884, 2017.
Article in English | MEDLINE | ID: mdl-28871249

ABSTRACT

The heterodimeric cytokine interleukin (IL) 23 comprises the IL12-shared p40 subunit and an IL23-specific subunit, p19. Together with IL12 and IL27, IL23 sits at the apex of the regulatory mechanisms shaping adaptive immune responses. IL23, together with IL17, plays an important role in the development of chronic inflammation and autoimmune inflammatory diseases. In this context, we generated monovalent antihuman IL23 variable heavy chain domain of llama heavy chain antibody (VHH) domains (Nanobodies®) with low nanomolar affinity for human interleukin (hIL) 23. The crystal structure of a quaternary complex assembling hIL23 and several nanobodies against p19 and p40 subunits allowed identification of distinct epitopes and enabled rational design of a multivalent IL23-specific blocking nanobody. Taking advantage of the ease of nanobody formatting, multivalent IL23 nanobodies were assembled with properly designed linkers flanking an antihuman serum albumin nanobody, with improved hIL23 neutralization capacity in vitro and in vivo, as compared to the monovalent nanobodies. These constructs with long exposure time are excellent candidates for further developments targeting Crohn's disease, rheumatoid arthritis, and psoriasis.

2.
J Nucl Med ; 58(8): 1210-1215, 2017 08.
Article in English | MEDLINE | ID: mdl-28360206

ABSTRACT

The human epidermal growth factor receptor 3 (HER3) is an interesting target for antitumor therapy. For optimal HER3 signaling inhibition, a biparatopic Nanobody construct (MSB0010853) was developed that binds 2 different HER3 epitopes. In addition, MSB0010853 contains a third HER3 epitope that binds albumin to extend its circulation time. MSB0010853 is cross-reactive with HER3 and albumin of mouse origin. We aimed to gain insight into MSB0010853 biodistribution and tumor uptake by radiolabeling the Nanobody construct with 89Zr. Methods: MSB0010853 was radiolabeled with 89Zr. Dose- and time-dependent tumor uptake was studied in nude BALB/c mice bearing a subcutaneous HER3 overexpressing H441 non-small cell lung cancer xenograft. Dose-dependent biodistribution of 89Zr-MSB0010853 was assessed ex vivo at 24 h after intravenous injection. Protein doses of 5, 10, 25, 100, and 1,000 µg were used. Time-dependent biodistribution of MSB0010853 was analyzed ex vivo at 3, 6, 24, and 96 h after intravenous administration of 25 µg of 89Zr-MSB0010853. PET imaging and biodistribution were performed 24 h after administration of 25 µg of 89Zr-MSB0010853 to mice bearing human H441, FaDu (high HER3 expression), or Calu-1 (no HER3 expression) tumor xenografts. Results: Radiolabeling of MSB0010853 with 89Zr was performed with a radiochemical purity of greater than 95%. Ex vivo biodistribution showed protein dose- and time-dependent distribution of 89Zr-MSB0010853 in all organs. Uptake of 89Zr-MSB0010853 in H441 tumors was only time-dependent. Tumor could be visualized up to at least 96 h after injection. The highest mean SUV of 0.6 ± 0.2 was observed at 24 h after injection of 25 µg of 89Zr-MSB0010853. 89Zr-MSB0010853 tumor uptake correlated with HER3 expression and was highest in H441 (6.2 ± 1.1 percentage injected dose per gram [%ID/g]) and lowest in Calu-1 (2.3 ± 0.3 %ID/g) xenografts. Conclusion:89Zr-MSB0010853 organ distribution and tumor uptake in mice are time-dependent, and tumor uptake correlates with HER3 expression. In contrast to tumor uptake except for kidney uptake, organ distribution of 89Zr-MSB0010853 is protein dose-dependent for the tested doses. 89Zr-MSB0010853 PET imaging gives insight into the in vivo behavior of MSB0010853.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Positron-Emission Tomography , Receptor, ErbB-3/metabolism , Single-Chain Antibodies/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Image Processing, Computer-Assisted , Lung Neoplasms/pathology , Male , Mice , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution
3.
Arthritis Res Ther ; 17: 135, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25994180

ABSTRACT

INTRODUCTION: The pleiotropic cytokine interleukin-6 (IL-6) plays an important role in the pathogenesis of different diseases, including rheumatoid arthritis (RA). ALX-0061 is a bispecific Nanobody® with a high affinity and potency for IL-6 receptor (IL-6R), combined with an extended half-life by targeting human serum albumin. We describe here the relevant aspects of its in vitro and in vivo pharmacology. METHODS: ALX-0061 is composed of an affinity-matured IL-6R-targeting domain fused to an albumin-binding domain representing a minimized two-domain structure. A panel of different in vitro assays was used to characterize the biological activities of ALX-0061. The pharmacological properties of ALX-0061 were examined in cynomolgus monkeys, using plasma levels of total soluble (s)IL-6R as pharmacodynamic marker. Therapeutic effect was evaluated in a human IL-6-induced acute phase response model in the same species, and in a collagen-induced arthritis (CIA) model in rhesus monkeys, using tocilizumab as positive control. RESULTS: ALX-0061 was designed to confer the desired pharmacological properties. A 200-fold increase of target affinity was obtained through affinity maturation of the parental domain. The high affinity for sIL-6R (0.19 pM) translated to a concentration-dependent and complete neutralization of sIL-6R in vitro. In cynomolgus monkeys, ALX-0061 showed a dose-dependent and complete inhibition of hIL-6-induced inflammatory parameters, including plasma levels of C-reactive protein (CRP), fibrinogen and platelets. An apparent plasma half-life of 6.6 days was observed after a single intravenous administration of 10 mg/kg ALX-0061 in cynomolgus monkeys, similar to the estimated expected half-life of serum albumin. ALX-0061 and tocilizumab demonstrated a marked decrease in serum CRP levels in a non-human primate CIA model. Clinical effect was confirmed in animals with active drug exposure throughout the study duration. CONCLUSIONS: ALX-0061 represents a minimized bispecific biotherapeutic of 26 kDa, nearly six times smaller than monoclonal antibodies. High in vitro affinity and potency was demonstrated. Albumin binding as a half-life extension technology resulted in describable and expected pharmacokinetics. Strong IL-6R engagement was shown to translate to in vivo effect in non-human primates, demonstrated via biomarker deregulation as well as clinical effect. Presented results on preclinical pharmacological properties of ALX-0061 are supportive of clinical development in RA.


Subject(s)
Antibodies, Bispecific/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Receptors, Interleukin-6/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Animals , Arthritis, Rheumatoid/drug therapy , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Half-Life , Humans , Immunoglobulin Heavy Chains/immunology , Interleukin-6/immunology , Macaca fascicularis , Macaca mulatta , Serum Albumin/immunology
4.
J Biol Chem ; 288(41): 29562-72, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23979133

ABSTRACT

The chemokine receptor CXCR7, belonging to the membrane-bound G protein-coupled receptor superfamily, is expressed in several tumor types. Inhibition of CXCR7 with either small molecules or small interference (si)RNA has shown promising therapeutic benefits in several tumor models. With the increased interest and effectiveness of biologicals inhibiting membrane-bound receptors we made use of the "Nanobody platform" to target CXCR7. Previously we showed that Nanobodies, i.e. immunoglobulin single variable domains derived from naturally occurring heavy chain-only camelids antibodies, represent new biological tools to efficiently tackle difficult drug targets such as G protein-coupled receptors. In this study we developed and characterized highly selective and potent Nanobodies against CXCR7. Interestingly, the CXCR7-targeting Nanobodies displayed antagonistic properties in contrast with previously reported CXCR7-targeting agents. Several high affinity CXCR7-specific Nanobodies potently inhibited CXCL12-induced ß-arrestin2 recruitment in vitro. A wide variety of tumor biopsies was profiled, showing for the first time high expression of CXCR7 in head and neck cancer. Using a patient-derived CXCR7-expressing head and neck cancer xenograft model in nude mice, tumor growth was inhibited by CXCR7-targeting Nanobody therapy. Mechanistically, CXCR7-targeting Nanobodies did not inhibit cell cycle progression but instead reduced secretion of the angiogenic chemokine CXCL1 from head and neck cancer cells in vitro, thus acting here as inverse agonists, and subsequent angiogenesis in vivo. Hence, with this novel class of CXCR7 inhibitors, we further substantiate the therapeutic relevance of targeting CXCR7 in head and neck cancer.


Subject(s)
Head and Neck Neoplasms/immunology , Receptors, CXCR/immunology , Single-Domain Antibodies/immunology , Xenograft Model Antitumor Assays , Animals , Arrestins/immunology , Arrestins/metabolism , Binding, Competitive/immunology , Camelids, New World/immunology , Cell Line, Tumor , Chemokine CXCL12/pharmacology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/prevention & control , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Radioligand Assay , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Single-Domain Antibodies/pharmacology , Tumor Burden/drug effects , Tumor Burden/immunology , beta-Arrestins
5.
J Clin Invest ; 123(6): 2590-603, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676465

ABSTRACT

TNF has remarkable antitumor activities; however, therapeutic applications have not been possible because of the systemic and lethal proinflammatory effects induced by TNF. Both the antitumor and inflammatory effects of TNF are mediated by the TNF receptor p55 (p55TNFR) (encoded by the Tnfrsf1a gene). The antitumor effect stems from an induction of cell death in tumor endothelium, but the cell type that initiates the lethal inflammatory cascade has been unclear. Using conditional Tnfrsf1a knockout or reactivation mice, we found that the expression level of p55TNFR in intestinal epithelial cells (IECs) is a crucial determinant in TNF-induced lethal inflammation. Remarkably, tumor endothelium and IECs exhibited differential sensitivities to TNF when p55TNFR levels were reduced. Tumor-bearing Tnfrsf1a⁺⁺/⁻ or IEC-specific p55TNFR-deficient mice showed resistance to TNF-induced lethality, while the tumor endothelium remained fully responsive to TNF-induced apoptosis and tumors regressed. We demonstrate proof of principle for clinical application of this approach using neutralizing anti-human p55TNFR antibodies in human TNFRSF1A knockin mice. Our results uncover an important cellular basis of TNF toxicity and reveal that IEC-specific or systemic reduction of p55TNFR mitigates TNF toxicity without loss of antitumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Intestinal Mucosa/drug effects , Melanoma, Experimental/drug therapy , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis , Cytokines/genetics , Cytokines/metabolism , Drug Screening Assays, Antitumor , Endothelium/pathology , Gene Expression , Gene Knock-In Techniques , Humans , Inflammation/chemically induced , Interferon-gamma/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/therapeutic use , Tumor Necrosis Factor-alpha/toxicity
6.
Blood ; 121(25): 5088-97, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23589671

ABSTRACT

Thrombolytic therapy is the cornerstone of treatment of acute atherothrombotic ischemic stroke but is associated with brain hemorrhage; antiplatelet therapy has limited efficacy and is still associated with intracranial bleeding. Therefore, new antithrombotic approaches with a better efficacy/safety ratio are required. We have assessed the effect of ALX-0081, a Nanobody against the A1 domain of von Willebrand factor (VWF) that blocks VWF binding to GPIb, of the thrombolytic agent recombinant tissue plasminogen activator (rtPA), and of the GPIIb/IIIa antagonist tirofiban, in a middle cerebral artery (MCA) thrombosis model in guinea pigs. Drugs were administered before, immediately after, or 15 or 60 minutes after the total occlusion of the MCA. ALX-0081 prevented MCA thrombosis and induced reperfusion when given immediately after and 15 minutes after complete occlusion and reduced brain damage without inducing hemorrhage, whereas tirofiban prevented thrombosis but did not induce reperfusion and induced striking brain hemorrhage. rtPA also induced reperfusion when given 60 minutes after occlusion but provoked brain hemorrhage. Skin bleeding time was not modified or was moderately prolonged by ALX-0081, whereas tirofiban and rtPA prolonged it. The inhibition of the GPIb-VWF axis in guinea pigs prevents cerebral artery thrombosis and induces early reperfusion without provoking intracerebral bleeding thus reducing brain infarct area.


Subject(s)
Brain Infarction/prevention & control , Fibrinolytic Agents/pharmacology , Intracranial Thrombosis/drug therapy , Single-Domain Antibodies/pharmacology , Thrombolytic Therapy/methods , Animals , Cerebral Arteries/pathology , Disease Models, Animal , Guinea Pigs , Male , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Reperfusion/methods , von Willebrand Factor/antagonists & inhibitors
7.
Mol Cancer Ther ; 6(9): 2563-71, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17876053

ABSTRACT

The potent antitumor activity of tumor necrosis factor (TNF) in combination with IFN-gamma can only be applied in local regimens due to their strong proinflammatory properties. It has been shown that the broad-spectrum matrix metalloproteinase (MMP) inhibitor BB-94 protects against TNF/IFNgamma-induced toxicity without blocking the antitumor effect. Here, we tried to explain this protective role of BB-94 and sought to assign roles to specific MMPs in TNF/IFNgamma-induced toxicity. By studying the expression of MMP genes in different organs and in the tumor, we observed that the expression levels of MMP-7, MMP-8, MMP-9, and MMP-12 and tissue inhibitor of metalloproteinase-4 are clearly up-regulated in the liver during therapy. MMP-8 and MMP-9 are also up-regulated in the lung and kidney, respectively. In the tumor, most MMP genes are expressed, but only MMP-3 is up-regulated during TNF/IFNgamma treatment. Using MMP-deficient or double-deficient mice, we have shown a mediating role for MMP-3 during TNF/IFNgamma treatment in tumor-free and B16BL6 melanoma-bearing mice. By contrast, MMP-12 seemed to have some protective role in both models. However, because most phenotypes were not extremely outspoken, we have to conclude, based on the set of MMP-deficient mice we have studied, that inhibition of a single MMP will probably not increase the therapeutic value of TNF/IFNgamma, but that rather, broad-spectrum MMP inhibitors will be required.


Subject(s)
Antiviral Agents/therapeutic use , Interferon-gamma/therapeutic use , Matrix Metalloproteinases/physiology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Phenylalanine/analogs & derivatives , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Disease Models, Animal , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/physiology , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/physiology , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/physiology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/physiology , Matrix Metalloproteinases/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylalanine/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinases/metabolism
8.
Cancer Res ; 67(15): 7301-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671199

ABSTRACT

Tumor necrosis factor (TNF)-induced inflammation prevents its broad application as an antitumor agent. We here report that addition of ZnSO(4) to the drinking water of mice induces expression of heat shock protein 70 (HSP70) in several organs, notably the gastrointestinal track. Zinc conferred dose-responsive protection against TNF-induced hypothermia, systemic induction of interleukin-6 and NO(x), as well as against TNF-induced bowel cell death and death of the organism. The protective effect of zinc was completely absent in mice deficient in the major HSP70-inducible gene, hsp70.1, whereas transgenic mice constitutively expressing the human HSP70.A gene, under control of a beta-actin promoter, was also protected against TNF, indicating that an increase in HSP70 is necessary and sufficient to confer protection. The therapeutic potential of the protection induced by ZnSO(4) was clearly shown in a TNF/IFNgamma-based antitumor therapy using three different tumor models. In hsp70.1 wild-type mice, but not in hsp70.1-deficient mice, zinc very significantly protected against lethality but left the antitumor effect intact. We conclude that zinc protects against TNF in a HSP70-dependent way and that protection by zinc could be helpful in developing a safer anticancer therapy with TNF/IFNgamma.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Inflammation/prevention & control , Tumor Necrosis Factor-alpha/toxicity , Zinc Sulfate/therapeutic use , Animals , Antiviral Agents/therapeutic use , Dose-Response Relationship, Drug , Female , HSP70 Heat-Shock Proteins/genetics , Humans , Hypothermia , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/therapeutic use , Interleukin-6/metabolism , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Necrosis , Neoplasms, Experimental/drug therapy , Survival Rate , Tumor Cells, Cultured
9.
J Immunol ; 178(8): 5069-75, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404289

ABSTRACT

In our search for genes that inhibit the inflammatory effects of TNF without diminishing its antitumor capacities we found that, compared with C57BL/6 mice, DBA/2 mice exhibit a dominant resistance to TNF-induced lethality. Tumor-bearing (C57BL/6 x DBA/2)(BXD)F(1) mice completely survived an otherwise lethal TNF/IFN-gamma-antitumor therapy with complete regression of the tumor. This was not the case for C57BL/6 mice. Genetic linkage analysis revealed that TNF resistance is linked to a major locus on distal chromosome 6 and a minor locus on chromosome 17. Compared with littermate controls, chromosome substitution mice carrying a DBA/2 chromosome 6 in a C57BL/6 background were significantly protected against TNF and TNF/IFN-gamma, albeit less so than DBA/2 mice. Definition of a critical region of 13 Mb on chromosome 6 was the highest mapping resolution obtained. Further analysis of candidate genes may provide a powerful tool to control TNF-induced pathologies in humans.


Subject(s)
Chromosome Mapping , Shock/chemically induced , Tumor Necrosis Factor-alpha/toxicity , Animals , Female , Interferon-gamma/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Quantitative Trait Loci , Species Specificity , Tumor Necrosis Factor-alpha/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...