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.
J Allergy Clin Immunol ; 151(4): 966-975, 2023 04.
Article in English | MEDLINE | ID: mdl-36592703

ABSTRACT

BACKGROUND: Type 2 endotype asthma is driven by IL-4 and IL-13 signaling via IL-4Ra, which is highly expressed on airway epithelium, airway smooth muscle, and immunocytes in the respiratory mucosa, suggesting potential advantages of an inhalable antagonist. Lipocalin 1 (Lcn1), a 16 kDa protein abundant in human periciliary fluid, has a robust drug-like structure well suited to protein engineering, but it has never been used to make an inhaled Anticalin protein therapeutic. OBJECTIVES: We sought to reengineer Lcn1 into an inhalable IL-4Ra antagonist and assess its pharmacodynamic/kinetic profile. METHODS: Lcn1 was systematically modified by directed protein mutagenesis yielding a high-affinity, slowly dissociating, long-acting full antagonist of IL-4Ra designated PRS-060 with properties analogous to dupilumab, competitively antagonizing IL-4Ra-dependent cell proliferation, mucus induction, and eotaxin expression in vitro. Because PRS-060 displayed exquisite specificity for human IL-4Ra, with no cross-reactivity to rodents or higher primates, we created a new triple-humanized mouse model substituting human IL-4Ra, IL-4, and IL-13 at their correct syntenic murine loci to model clinical dosing. RESULTS: Inhaled PRS-060 strongly suppressed acute allergic inflammation indexes in triple-humanized mice with a duration of action longer than its bulk clearance, suggesting that it may act locally in the lung. CONCLUSION: Lcn1 can be reengineered into the Anticalin antagonist PRS-060 (elarekibep), exemplifying a new class of inhaled topical, long-acting therapeutic drugs with the potential to treat type 2 endotype asthma.


Subject(s)
Asthma , Interleukin-13 , Animals , Humans , Mice , Asthma/drug therapy , Disease Models, Animal , Interleukin-4/genetics , Lung , Proteins , Nebulizers and Vaporizers , Receptors, Interleukin-4/immunology
2.
Clin Cancer Res ; 25(19): 5878-5889, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31138587

ABSTRACT

PURPOSE: 4-1BB (CD137) is a key costimulatory immunoreceptor and promising therapeutic target in cancer. To overcome limitations of current 4-1BB-targeting antibodies, we have developed PRS-343, a 4-1BB/HER2 bispecific molecule. PRS-343 is designed to facilitate T-cell costimulation by tumor-localized, HER2-dependent 4-1BB clustering and activation. EXPERIMENTAL DESIGN: PRS-343 was generated by the genetic fusion of 4-1BB-specific Anticalin proteins to a variant of trastuzumab with an engineered IgG4 isotype. Its activity was characterized using a panel of in vitro assays and humanized mouse models. The safety was assessed using ex vivo human cell assays and a toxicity study in cynomolgus monkeys. RESULTS: PRS-343 targets 4-1BB and HER2 with high affinity and binds both targets simultaneously. 4-1BB-expressing T cells are efficiently costimulated when incubated with PRS-343 in the presence of cancer cells expressing HER2, as evidenced by increased production of proinflammatory cytokines (IL2, GM-CSF, TNFα, and IFNγ). In a humanized mouse model engrafted with HER2-positive SK-OV-3 tumor cells and human peripheral blood mononuclear cells, PRS-343 leads to tumor growth inhibition and a dose-dependent increase of tumor-infiltrating lymphocytes. In IND-enabling studies, PRS-343 was found to be well tolerated, with no overt toxicity and no relevant drug-related toxicologic findings. CONCLUSIONS: PRS-343 facilitates tumor-localized targeting of T cells by bispecific engagement of HER2 and 4-1BB. This approach has the potential to provide a more localized activation of the immune system with higher efficacy and reduced peripheral toxicity compared with current monospecific approaches. The reported data led to initiation of a phase I clinical trial with this first-in-class molecule.See related commentary by Su et al., p. 5732.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating , Mice , T-Lymphocytes , Tumor Necrosis Factor Receptor Superfamily, Member 9
3.
PLoS One ; 13(11): e0207767, 2018.
Article in English | MEDLINE | ID: mdl-30500834

ABSTRACT

Subgroups of patients with severe asthma are insensitive to inhaled corticosteroids and require novel therapies on top of standard medical care. IL-13 is considered one of the key cytokines in the asthma pathogenesis, however, the effect of IL-13 was mostly studied in rodents. This study aimed to assess IL-13 effect in human lung tissue for the development of targeted therapy approaches such as inhibition of soluble IL-13 or its receptor IL-4Rα subunit. Precision-cut lung slices (PCLS) were prepared from lungs of rodents, non-human primates (NHP) and humans. Direct effect of IL-13 on human lung tissue was observed on inflammation, induction of mucin5AC, and airway constriction induced by methacholine and visualized by videomicroscopy. Anti-inflammatory treatment was evaluated by co-incubation of IL-13 with increasing concentrations of IL-13/IL-13 receptor inhibitors. IL-13 induced a two-fold increase in mucin5AC secretion in human bronchial tissue. Additionally, IL-13 induced release of proinflammatory cytokines eotaxin-3 and TARC in human PCLS. Anti-inflammatory treatment with four different inhibitors acting either on the IL-13 ligand itself (anti-IL-13 antibody, similar to Lebrikizumab) or the IL-4Rα chain of the IL-13/IL-4 receptor complex (anti-IL-4Rα #1, similar to AMG 317, and #2, similar to REGN668) and #3 PRS-060 (a novel anticalin directed against this receptor) could significantly attenuate IL-13 induced inflammation. Contrary to this, IL-13 did not induce airway hyperresponsiveness (AHR) in human and NHP PCLS, although it was effective in rodent PCLS. Overall, this study demonstrates that IL-13 stimulation induces production of mucus and biomarkers of allergic inflammation in human lung tissue ex-vivo but no airway hyperresponsiveness. The results of this study show a more distinct efficacy than known from animals models and a clear discrepancy in AHR induction. Moreover, it allows a translational approach in inhibitor profiling in human lung tissue.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Interleukin-13/pharmacology , Lung/drug effects , Bronchi/drug effects , Bronchi/metabolism , Chemokine CCL17/metabolism , Chemokine CCL26/metabolism , Humans , Lung/metabolism , Lung/pathology , Mucins/biosynthesis , Receptors, Interleukin-13/metabolism
4.
J Pharmacol Exp Ther ; 365(2): 368-378, 2018 05.
Article in English | MEDLINE | ID: mdl-29463608

ABSTRACT

Since it was recently reported that an antibody for proprotein convertase subtilisin/kexin type 9 (PCSK9) reduces the risk of cardiovascular events in a clinical context, PCSK9 inhibition is thought to be an attractive therapy for dyslipidemia. In the present study, we created a novel small biologic alternative to PCSK9 antibodies called DS-9001a, comprising an albumin binding domain fused to an artificial lipocalin mutein (ABD-fused Anticalin protein), which can be produced by a microbial production system. DS-9001a strongly interfered with PCSK9 binding to low-density-lipoprotein receptor (LDL-R) and PCSK9-mediated degradation of LDL-R. In cynomolgus monkeys, single DS-9001a administration significantly reduced the serum LDL-C level up to 21 days (62.4% reduction at the maximum). Moreover, DS-9001a reduced plasma non-high-density-lipoprotein cholesterol and oxidized LDL levels, and their further reductions were observed when atorvastatin and DS-9001a were administered in combination in human cholesteryl ester transfer protein/ApoB double transgenic mice. Additionally, their reductions on the combination of atorvastatin and DS-9001a were more pronounced than those on the combination of atorvastatin and anacetrapib. Besides its favorable pharmacologic profile, DS-9001a has a lower molecular weight (about 22 kDa), yielding a high stoichiometric drug concentration that might result in a smaller administration volume than that in existing antibody therapy. Since bacterial production systems are viewed as more suited to mass production at low cost, DS-9001a may provide a new therapeutic option to treat patients with dyslipidemia. In addition, considering the growing demand for antibody-like drugs, ABD-fused Anticalin proteins could represent a promising new class of small biologic molecules.


Subject(s)
Albumins/metabolism , Lipocalins/genetics , Proprotein Convertase 9/immunology , Recombinant Fusion Proteins/immunology , Animals , Atorvastatin/pharmacology , Cholesterol Ester Transfer Proteins , Drug Interactions , Hep G2 Cells , Humans , Lipocalins/chemistry , Lipoproteins, LDL/blood , Macaca fascicularis , Male , Mice , Oxazolidinones/pharmacology , Protein Domains , Rats , Rats, Sprague-Dawley , Receptors, LDL/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650228

ABSTRACT

Human tear lipocalin (Tlc) was utilized as a protein scaffold to engineer an Anticalin that specifically binds and functionally blocks vascular endothelial growth factor A (VEGF-A), a pivotal inducer of physiological angiogenesis that also plays a crucial role in several neovascular diseases. Starting from a naive combinatorial library where residues that form the natural ligand-binding site of Tlc were randomized, followed by affinity maturation, the final Anticalin PRS-050 was selected to bind all major splice forms of VEGF-A with picomolar affinity. Moreover, this Anticalin cross-reacts with the murine ortholog. PRS-050 efficiently antagonizes the interaction between VEGF-A and its cellular receptors, and it inhibits VEGF-induced mitogenic signaling as well as proliferation of primary human endothelial cells with subnanomolar IC50 values. Intravitreal administration of the Anticalin suppressed VEGF-induced blood-retinal barrier breakdown in a rabbit model. To allow lasting systemic neutralization of VEGF-A in vivo, the plasma half-life of the Anticalin was extended by site-directed PEGylation. The modified Anticalin efficiently blocked VEGF-mediated vascular permeability as well as growth of tumor xenografts in nude mice, concomitantly with reduction in microvessel density. In contrast to bevacizumab, the Anticalin did not trigger platelet aggregation and thrombosis in human FcγRIIa transgenic mice, thus suggesting an improved safety profile. Since neutralization of VEGF-A activity is well known to exert beneficial effects in cancer and other neovascular diseases, including wet age-related macular degeneration, this Anticalin offers a novel potent small protein antagonist for differentiated therapeutic intervention in oncology and ophthalmology.


Subject(s)
Lipocalins/pharmacology , Molecular Targeted Therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antigen-Antibody Complex/metabolism , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Blood-Retinal Barrier/pathology , Capillary Permeability , Cell Proliferation/drug effects , Female , Half-Life , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipocalins/pharmacokinetics , Lipocalins/therapeutic use , Mice, Nude , Mice, Transgenic , Mitogens/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Polyethylene Glycols/chemistry , Protein Engineering , Rabbits , Receptors, IgG/metabolism , Signal Transduction , Surface Plasmon Resonance , Thrombosis/pathology , Vascular Endothelial Growth Factor A/metabolism
6.
Mol Cancer Ther ; 12(11): 2459-71, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002935

ABSTRACT

Activation of the MET oncogenic pathway has been implicated in the development of aggressive cancers that are difficult to treat with current chemotherapies. This has led to an increased interest in developing novel therapies that target the MET pathway. However, most existing drug modalities are confounded by their inability to specifically target and/or antagonize this pathway. Anticalins, a novel class of monovalent small biologics, are hypothesized to be "fit for purpose" for developing highly specific and potent antagonists of cancer pathways. Here, we describe a monovalent full MET antagonist, PRS-110, displaying efficacy in both ligand-dependent and ligand-independent cancer models. PRS-110 specifically binds to MET with high affinity and blocks hepatocyte growth factor (HGF) interaction. Phosphorylation assays show that PRS-110 efficiently inhibits HGF-mediated signaling of MET receptor and has no agonistic activity. Confocal microscopy shows that PRS-110 results in the trafficking of MET to late endosomal/lysosomal compartments in the absence of HGF. In vivo administration of PRS-110 resulted in significant, dose-dependent tumor growth inhibition in ligand-dependent (U87-MG) and ligand-independent (Caki-1) xenograft models. Analysis of MET protein levels on xenograft biopsy samples show a significant reduction in total MET following therapy with PRS-110 supporting its ligand-independent mechanism of action. Taken together, these data indicate that the MET inhibitor PRS-110 has potentially broad anticancer activity that warrants evaluation in patients.


Subject(s)
Lipocalins/pharmacology , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Proteins/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites/drug effects , CHO Cells , Cell Line, Tumor , Cricetulus , Dose-Response Relationship, Drug , Epitope Mapping , Female , HT29 Cells , Hepatocyte Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Lipocalins/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/therapeutic use , Proteins/therapeutic use , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
J Mol Biol ; 425(4): 780-802, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23238252

ABSTRACT

The oncofetal isoform of the extracellular matrix protein fibronectin (Fn), which carries the extra-domain B (ED-B) and is exclusively expressed in neovasculature, has gained interest for tumor diagnosis and therapy using engineered antibody fragments. We have employed the human lipocalin 2 (Lcn2) as a small and robust non-immunoglobulin scaffold to select ED-B-specific Anticalins from a new advanced random library using bacterial phage display and ELISA screening against appropriately engineered Fn fragments. As a result, we have isolated and biochemically characterized four different Anticalins that all show low nanomolar affinities for ED-B, right in the range between the monomeric and dimeric forms of the single-chain variable antibody fragment L19 that has been widely applied in this area before. All Anticalins can be readily expressed in Escherichia coli as soluble and strictly monomeric proteins, and they show specific staining of ED-B-positive tumor cells in immunofluorescence microscopy while BIAcore affinity analyses indicate recognition of distinct ED-B epitopes. The crystal structure for one Anticalin, N7A, in complex with the Fn7B8 fragment, was solved at 2.6Å resolution and reveals binding to the gfcc' sheet and cc' loop on ED-B. This is the second example of a protein-specific Lcn2-based Anticalin, which illustrates the remarkable plasticity of the calyx-like ligand pocket of lipocalins with their four structurally hypervariable loops supported by a highly conserved ß-barrel. The ED-B-specific Anticalins resulting from this study should provide useful reagents in research and biomedical drug development, both for in vivo imaging and for directed cancer therapy.


Subject(s)
Antibodies/metabolism , Fibronectins/metabolism , Lipocalins/immunology , Peptide Library , Amino Acid Sequence , Antibodies/chemistry , Antibodies/genetics , Binding Sites/genetics , Binding, Competitive , Caco-2 Cells , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Fibronectins/chemistry , Fibronectins/genetics , Humans , Lipocalins/genetics , Lipocalins/metabolism , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Surface Plasmon Resonance
8.
J Mol Biol ; 400(4): 783-802, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20630471

ABSTRACT

We demonstrate that small engineered single-chain binding proteins based on the lipocalin scaffold, so-called Anticalins, can be functionally displayed on the Gram-negative bacterial cell envelope. To this end, the beta-domains of five different bacterial autotransporters (the IgA protease from Neisseria gonorrhoeae, the esterase EstA from Pseudomonas aeruginosa, the YpjA autotransporter from E. coli K12, the AIDA-I adhesin from enteropathogenic E. coli O127:H27 strain 2787 and the protease EspP from enterohemorrhagic E. coli O157:H7 strain EDL933) were compared with respect to display level, functional variance, and bacterial cell viability. Use of the EspP autotransporter led to a system with high genetic stability for the display of fully functional Anticalins in high density on the cell surface of E. coli as shown by quantitative flow cytofluorimetry. This system was applied to engineer an immunostimulatory Anticalin that binds and blocks the extracellular region of human CTLA-4 to achieve a slower dissociation rate. A combinatorial library of the original Anticalin was generated by error-prone PCR, subjected to E. coli cell surface display, and applied to repeated cycles of cell sorting after incubation with the fluorescently labelled target protein under competition with the unlabelled extracellular domain of CTLA-4. The resulting Anticalin variants, which were expressed and purified as soluble proteins, showed more than eightfold decelerated target dissociation, as revealed by real time surface plasmon resonance analysis. Hence, the EspP autotransporter-mediated E. coli surface display in combination with high-throughput fluorescence-activated cell sorting (FACS) provides an efficient strategy to select for Anticalins, and possibly other small protein scaffolds, with improved binding properties, which is particularly useful for in vitro affinity maturation but may also serve for the selection of novel target specificity from naive libraries.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lipocalins/metabolism , Serine Endopeptidases/metabolism , Antigens, CD/metabolism , CTLA-4 Antigen , Escherichia coli/genetics , Escherichia coli K12/enzymology , Escherichia coli K12/genetics , Escherichia coli O157/enzymology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Flow Cytometry , Lipocalins/genetics , Microbial Viability , Models, Biological , Neisseria gonorrhoeae/enzymology , Neisseria gonorrhoeae/genetics , Protein Binding , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics , Surface Plasmon Resonance
9.
Bioorg Med Chem Lett ; 12(6): 985-8, 2002 Mar 25.
Article in English | MEDLINE | ID: mdl-11959009

ABSTRACT

Based on bifunctional diketopiperazines as templates and m-aminomethyl-phenylalanine as arginine mimetic, we have synthesized a new class of structurally related dibasic tryptase inhibitors with systematically increasing spacer length. These compounds were used to scan the distance between the active sites of two neighboring subunits of the beta-tryptase tetramer. The K(i)-values obtained are a function of the distance between the terminal amino groups and indicate optimal binding of inhibitors with 29-31 bonds between the amino groups. These experimental data are in full agreement with predictions derived from a novel modeling program that allows the docking of bivalent ligands.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Models, Molecular , Serine Endopeptidases/chemistry , Algorithms , Humans , Ligands , Serine Endopeptidases/drug effects , Tryptases
SELECTION OF CITATIONS
SEARCH DETAIL
...