WO2015007520A1 - Targeting of cytokine antagonists - Google Patents

Targeting of cytokine antagonists Download PDF

Info

Publication number
WO2015007520A1
WO2015007520A1 PCT/EP2014/063976 EP2014063976W WO2015007520A1 WO 2015007520 A1 WO2015007520 A1 WO 2015007520A1 EP 2014063976 W EP2014063976 W EP 2014063976W WO 2015007520 A1 WO2015007520 A1 WO 2015007520A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
fusion protein
cytokine
antagonist
antibody
Prior art date
Application number
PCT/EP2014/063976
Other languages
French (fr)
Inventor
Jan Tavernier
Lennart Zabeau
Gilles UZÉ
Franciane PAUL
Yann BORDAT
Geneviève GARCIN
Original Assignee
Vib Vzw
Universiteit Gent
Centre National De La Recherche Scientifique
Université Montpellier 2
Centre Hospitalier Regional Universitaire De Montpellier
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US14/905,348 priority Critical patent/US9732135B2/en
Priority to KR1020167002677A priority patent/KR102305608B1/en
Priority to EP14734166.3A priority patent/EP3022230B1/en
Priority to CN201480040656.4A priority patent/CN105658669A/en
Application filed by Vib Vzw, Universiteit Gent, Centre National De La Recherche Scientifique, Université Montpellier 2, Centre Hospitalier Regional Universitaire De Montpellier filed Critical Vib Vzw
Priority to JP2016526507A priority patent/JP6475712B2/en
Priority to SG11201600165WA priority patent/SG11201600165WA/en
Priority to CA2918119A priority patent/CA2918119C/en
Priority to AU2014292355A priority patent/AU2014292355B2/en
Priority to MX2016000721A priority patent/MX2016000721A/en
Publication of WO2015007520A1 publication Critical patent/WO2015007520A1/en
Priority to IL24345916A priority patent/IL243459B/en
Priority to US15/642,989 priority patent/US10072059B2/en
Priority to US16/008,686 priority patent/US10947288B2/en
Priority to US16/008,695 priority patent/US20180334489A1/en
Priority to US17/171,433 priority patent/US20210238247A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/642Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2869Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against hormone receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor

Definitions

  • the present invention relates to a fusion protein, comprising a cytokine antagonist and a targeting moiety, preferably an antibody or antibody like molecule.
  • the cytokine antagonist is a modified cytokine which binds to the receptor, but doesn't induce the receptor signalling.
  • the invention relates further to a fusion protein according to the invention for use in treatment of cancer or for use in treatment of autoimmune diseases.
  • Cytokines are critical mediators of defence mechanisms against microbial invasion and tumorigenesis. However, their production and activities must be tightly regulated to prevent an excessive activity that can culminate in the uncontrolled inflammation and tissue injury, as characteristically observed with many autoimmune diseases.
  • Rheumatoid arthritis is the classic example of an autoimmune disease where TNFa, I L-1 , and IL-6 play a prominent role in the recruitment of lymphocytes and other types of leukocytes that mediate a progressive joint destruction. TNF inhibitors have been shown to decrease symptoms, slow disease progression, and improve the quality of life for many patients with rheumatoid arthritis (Moreland, 2009).
  • a mAb neutralizing IL-12 and IL-23 provides a potential therapy for psoriasis (Elliott et al., 2009) and a recombinant human IL-1 receptor antagonist, (anakinra, KineretTM), first approved by the FDA in 2001 for the treatment of rheumatoid arthritis, is a promising agent for the treatment of many IL-1 - mediated autoinflammatory diseases (Goldbach-Mansky, 2009).
  • IL17A is the best characterized member of the IL17 family of cytokines. This pleiotropic cytokine interacts with a receptor composed of IL17RA and IL17RC subunits.
  • the IL17RA chain is ubiquitously expressed, including haematopoietic, immune, epithelial, endothelial cell types, as well as fibroblasts.
  • IL17A is typically produced by Th17 cells upon activation by a subset of cytokines including IL-1 , IL-6, IL-21 and TGFp, and propagates early inflammatory signals that serve to bridge innate and adaptive immune responses.
  • IL17 is a potent activator of neutrophils and plays an important role in the immune defence against various extracellular pathogens.
  • IL17A promotes autoimmune pathologies (Gaffen, 2009; Shen & Gaffen, 2008).
  • Brodalumab, Secukinumab and Ixekizumab target the IL17A/IL17R axis for treatment of auto-immune diseases such as psoriasis and Crohn's disease. All may inflict adverse side effects including enhanced risk of infections (Hueber et al. 2012; Spuls & Hooft, 2012).
  • IL17A antagonists may therefore offer a significant advantage over completely antagonising IL17 function.
  • ILI a and ⁇ _ ⁇ are the founding members of the IL1 cytokine family. Both are pleiotropic and function through a ubiquitously expressed receptor complex composed of IL-1 receptor type-l (IL-1 RI) and IL-1 receptor accessory protein (IL-1 RAcP). Overactivation of this IL-1 axis is associated with many human pathologies including rheumatoid arthritis (RA), chronic obstructive pulmonary disease (COPD), asthma, inflammatory bowel diseases, multiple sclerosis, atherosclerosis and Alzheimer's disease.
  • RA rheumatoid arthritis
  • COPD chronic obstructive pulmonary disease
  • asthma inflammatory bowel diseases
  • multiple sclerosis multiple sclerosis
  • atherosclerosis atherosclerosis
  • Alzheimer's disease Alzheimer's disease.
  • IL-1 immune cells of different lineages are activated by IL-1 , including innate immune cells such as dendritic cells, macrophages and neutrophils, and also cells involved in the adaptive immune response including naive, Th17 and CD8+ T cells, and B cells (reviewed in Sims and Smith, 2010).
  • Recombinant human IL- 1 RA IL1 receptor antagonist, aka anakinra
  • autoinflammatory diseases Disperello, 201 1 .
  • One of the major side effects of prolonged treatment with anakinra is however the increased occurrence of infections.
  • IL-1 activity on only a subset of (immune) cells therefore may offer a safer alternative. It can be envisaged that targeted inhibition of IL-1 action on selected innate immune cells, leaving its activity on the T cell compartment intact, may still show efficacy for the treatment of inflammatory diseases, without affecting the host defence against pathogens.
  • TSLP thymic stromal lymphopoietin
  • IL-7Ra IL-7 receptor
  • TSLPRa TSLPRa
  • TSLP promotes Th2-type inflammation by acting on several distinct cell types, including dendritic cells, CD4 and CD8 T cells, B cells, NKT cells, mast cells, eosinophils and basophils.
  • TSLP can have a protective role in inflammatory diseases driven by exacerbated Th1 and Th17 responses, such as Inflammatory Bowel Disease (reviewed in He and Geha, 2010 and Roan et al., 2012).
  • the main problem with the therapeutic approaches aiming to neutralize cytokine actions is that the cytokine antagonists are not targeted towards cells or tissues that are specifically involved in the onset of the autoimmune or autoinflammatory diseases.
  • a long term systemic neutralization of type I IFN activity by a monoclonal antibody or an IFN receptor antagonist carry an important risk in term of viral infection susceptibility and tumor development since type I IFN is a family of proteins essential in the control of viral infections and for establishing immune responses, particularly those controlling cancer cell growth (Gajewski et al., 2012).
  • a systemic neutralization of IL-1 activity will impact the expansion, effector function, tissue localization, and memory response of antigen-cytotoxic T cells during immune responses (Ben-Sasson et al., 2013).
  • the invention is exemplified by targeting the action of a type I IFN antagonist to specific cell types expressing a given cell surface marker.
  • Such a method is applied to the design and construction of a targeted IFN antagonist that inhibits the action of endogenous IFN specifically on the cell subset culpably involved in the onset of autoimmune diseases, leaving the other cells and organs fully responsive.
  • Oncolytic viruses are advancing through clinical trials (Russell et al., 2012). Oncolytic viruses are often designed for having attenuated replication capacity in normal tissues by engineering their sensitivity to the normal cellular interferon-mediated antiviral responses.
  • An example is an oncolytic vesicular stomatitis virus coding for interferon ⁇ (Naik et al., 2012). The therapeutic effect of such viruses is expected to be a consequence of the defect of the IFN response exhibited by many tumor cells.
  • the genetic heterogeneity of tumors that impact the IFN response is highly variable and impairs the efficacy of virus-mediated tumor lysis (Naik and Russell, 2009).
  • a first aspect of the invention is a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody like molecule.
  • a cytokine antagonist as used here can be any cytokine antagonist known to the person skilled in the art, including but not limited to a soluble receptor, a cytokine binding antibody or a mutant cytokine.
  • said cytokine antagonist is a mutant cytokine, even more preferably a mutant which binds to the receptor, but is not or only weakly inducing the cytokine signalling.
  • the affinity of the mutant for the receptor is comparable to that of the wild type cytokine, even more preferable it has a higher affinity; preferably the signalling induced by the mutant is less than 20% of that of the wild type, even more preferably less than 10% of that of the wild type, even more preferably less than 5%, even more preferably less than 1 %. Most preferably, the binding of the mutant cytokine does not result in detectable signalling. Such mutant can act as a competitive inhibitor of cytokine signalling.
  • An antibody or antibody like molecule as used here is a protein specifically designed to bind another molecule, preferably a proteineous molecule, and comprising the specific binding domains.
  • said antibody or antibody like molecule can be a heavy chain antibody (hcAb), single domain antibody (sdAb), minibody (Tramontano et al., 1994), the variable domain of camelid heavy chain antibody (VHH), the variable domain of the new antigen receptor (VNAR), affibody (Nygren et al., 2008), alphabody (WO2010066740), designed ankyrin-repeat domain (DARPins) (Stumpp et al., 2008), anticalin (Skerra et al., 2008), knottin (Kolmar et al., 2008) and engineered CH2 domain (nanoantibodies; Dimitrov, 2009).
  • hcAb heavy chain antibody
  • sdAb single domain antibody
  • minibody Tramontano et al., 1994
  • VHH camelid heavy chain antibody
  • VNAR variable domain of the new antigen receptor
  • affibody Nygren et al., 2008
  • alphabody WO201006
  • said antibody or antibody like molecule consists of a single polypeptide chain, even more preferably, said antibody is not post-translationally modified.
  • Prost-translational modification indicates the modifications carried out by living cell during or after the protein synthesis, but excludes modifications, preferably chemical modifications, carried out on the isolated protein such as, but not limited to pegylation Even more preferably said antibody or antibody-like molecule comprises the complementary determining regions, derived from an antibody.
  • said targeting antibody or antibody-like molecule is a nanobody.
  • said cytokine antagonist and said targeting moiety are connected by a linker, preferably a GGS linker.
  • a linker preferably a GGS linker.
  • said GGS linker contains at least 5 GGS repeats, more preferably at least 10 GGS repeats, even more preferably at least 15 GGS repeats, most preferably at least 20 GGS repeats.
  • the cytokine antagonist according to the invention is an interferon antagonist; even more preferably, it is an IFNa2-R120E mutant.
  • the cytokine antagonist according to the invention is an antagonist of a cytokine of the IL17 family, preferably an IL17A antagonist.
  • the cytokine antagonist according to the invention is an antagonist of the IL1 cytokine family, preferably an ILI a or ILp antagonist.
  • the cytokine antagonist according to the invention is a TSLP antagonist.
  • the antibody or antibody-like molecule is directed against a cancer cell marker.
  • Cancer cell markers are known to the person skilled in the art, and include, but are not limited to CD19, CD20, CD22, CD30, CD33, CD37, CD56, CD70, CD74, CD138, AGS16, HER2, MUC1 , GPNMB and PMSA.
  • said cancer marker is CD20 or HER2.
  • the antibody or antibody-like molecule is directed against a marker on an immune cell, preferably an inflammatory cytokine producing immune cell.
  • An immune cell as used here, is a cell that belongs to the immune system, including but not limited to monocytes, dendritic cells and T-cells.
  • said immune cell is a proinflammatory cytokine producing cell.
  • Markers of inflammatory cytokine producing cells include but are not limited to CD4, CD1 1 b, CD26, sialoadhesin and flt3 receptor.
  • Another aspect of the invention is a fusion protein according to the invention for use in treatment of cancer. Still another aspect of the invention is a fusion protein according to the invention for use in treatment of autoimmune diseases.
  • Another aspect of the invention is a method to treat cancer, comprising (i) determination the type of cancer and the suitable targeting marker(s) for the cancer cells in a patient suffering from cancer (ii) providing to said patient in need of the treatment a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody-like molecule according to the invention, possibly with a suitable excipient. It is obvious for the person skilled in the art that the targeting moiety of step (ii) will be directed to the targeting marker identified in step (i).
  • Possible cancer cell markers are known to the person skilled in the art, and include, but are not limited to CD19, CD20, CD22, CD30, CD33, CD37, CD56, CD70, CD74, CD138, AGS16, HER2, MUC1 , GPNMB and PMSA.
  • Still another aspect of the invention is a method to treat an autoimmune disease, comprising (i) determination in a patient suffering from an autoimmune disease the suitable targeting marker(s) for the immune cells cells cells (ii) providing to said patient in need of the treatment a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody-like molecule according to the invention, possibly with a suitable excipient.
  • Immune cells include but are not limited to dendritic cells, CD4 and CD8 T cells, B cells, NKT cells, mast cells, eosinophils and basophils.
  • Figure 1 Representation of the structural elements of the nanobody-hlFNa2-R120E fusion protein.
  • Figure 2 Quantification of the luciferase activity induced by 10 pM hl FNa2 in the presence or absence (untreated) of the 4-1 1 -hl FNa2-R120E fusion protein on HL1 16 (A) and HL1 16- ml_R10 (B) cells.
  • Figure 3 Quantification of the luciferase activity induced by 1 pM IFN3 in the presence or absence (untreated) of the 4-1 1 -hl FNa2-R120E fusion protein on HL1 16 (A) and HL1 16- ml_R10 (B) cells.
  • Figure 4 FACS analysis of pY701 -STAT1 in CD19 positive and negative human PBMCs left untreated (left panel), treated with 50 pM of hl FNa2 (center) or with 50 pM of hl FNa2 in the presence of the CD20-targeted I FN antagonist.
  • Nanobody-I FN antagonist fusion construction Nanobody-I FN antagonist fusion construction.
  • Hek 293T cells were transfected with the protein fusion constructs using the standard lipofectamin method (Invitrogen). 48 hours after the transfection culture mediums were harvested and stored at -20°C. Cell lines
  • Hek 293T cells were grown in DMEM supplemented with 10% FCS.
  • the HL1 16 clone (Uze et al., 1994) is derived from the human HT1080 cell line. It contains the firefly luciferase gene controlled by the IFN-inducible 6-16 promoter.
  • the derived HL1 16-mLR10 clone which expresses the murine leptin receptor was described (PCT/EP2013/050787).
  • Antagonistic IFN activities were measured by quantifying the inhibition of the luciferase activity induced in HL1 16 cells and on the HL1 16-ml_R10 expressing the rmLR by IFNa2 or IFN3.
  • the IC50 values were calculated using nonlinear data regression with Prism software (GraphPad).
  • Luciferase activities were determined on a Berthold Centra LB960 luminometer using a luciferase substrate buffer (20 mM Tricine, 1.07 mM (MgC03)4Mg(OH)2 « 5H20, 2.67 mM MgS04 « 7H20, 0.1 mM EDTA, 33.3 mM dithiothreitol, 270 ⁇ coenzyme A, 470 ⁇ luciferin, 530 ⁇ ATP, final pH 7.8) after 6hr IFN stimulation.
  • a luciferase substrate buffer (20 mM Tricine, 1.07 mM (MgC03)4Mg(OH)2 « 5H20, 2.67 mM MgS04 « 7H20, 0.1 mM EDTA, 33.3 mM dithiothreitol, 270 ⁇ coenzyme A, 470 ⁇ luciferin, 530 ⁇ ATP, final pH 7.8) after 6hr IFN stimulation.
  • Example 1 The nanobody-IFNa2-R120E fusion protein.
  • the nanobody 4-1 1 directed against the murine leptin receptor was fused to the IFNa2 mutant R120E as described in the materials and methods
  • Figure 1 shows a schematic representation of the nanobody-IFN antagonist fusion protein constructed with the nanobody 4-1 1 against the murine leptin receptor and the human IFNa2- R120E (numbering as in Piehler et al., 2000).
  • Example 2 Targeted inhibition of IFNa activity on mLR-expressing cells
  • Example 3 Targeted inhibition of IFN activity on mLR-expressing cells
  • the IFN3 shows the highest affinity for the IFNa/ ⁇ receptor. We thus tested whether the 4-1 1 -IFNa2-R120E fusion protein exerts also an antagonistic activity against IFN3 action.
  • Parental HL1 16 cells and the derived HL1 16-mLR10 cells which express the mouse leptin receptor were treated for 6 hours with 1 pM IFN3 in the presence of several dilutions of culture medium conditioned by Hek 293T cells expressing the 4-1 1 -IFNa2-R120E fusion protein.
  • the 1 pM IFN3 dose was chosen because it corresponds to the IFN3 EC50 on both cell lines. Cells were then lysed and the IFN-induced luciferase activity was quantified.
  • the 4-1 1 -IFNa2-R120E fusion protein was unable to inhibit IFNa2 action on untargeted HL1 16 cells (Figure 3A).
  • its dose-dependent inhibition effect is clear on HL1 16-mLR10 cells which express the target of the 4-1 1 nanobody ( Figure 3B).
  • the type I IFN antagonist IFNa2-R120E was fused to the anti-human CD20 nanobody 2HCD25 through a linker sequence made with 20 repeats of GGS motif.
  • the fusion protein was produced in E. coli and purified by Immobilized Metal Affinity chromatography (IMAC).
  • IMAC Immobilized Metal Affinity chromatography
  • PBMCs peripheral blood mononuclear cells
  • FIG 4 shows that the IFN antagonist linked to the nanobody specific for CD20 inhibits the IFN action specifically in the major part of the B cell population, leaving intact the IFN response in the CD19 negative cell population.
  • Example 5 The CD20-targeted type I IFN antagonist inhibits the antiproliferative activity of type I IFN.
  • the fusion protein of the 2HCD25 nanobody and IFNa2-R120E inhibits IFN-induced STAT1 phosphorylation specifically in B-cells.
  • Daudi cells are a human lymphoblastoid B-cell line expressing CD20. Daudi cells were seeded at 2.0x105 cells/ml and were left untreated or cultured for 72 h in the presence of 2 pM IFNa2 alone or in combination with various CD20-targeted IFN antagonists. They were then counted to estimate the efficacy of the inhibition of proliferation induced by IFNa2.
  • Figure 5 shows that the CD20-targeted IFN antagonist fully inhibits the antiproliferative activity of IFNa2. It also shows that decreasing the IFN-IFNAR2 affinity decreases the antagonistic activity, proving that the inhibitory effect is indeed due to the binding of the targeted antagonist.
  • IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells. J Exp Med 210, 491 -502.
  • Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp Med 208, 469-478).
  • Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1 , Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood 1 15, 5312-5321 .

Abstract

The present invention relates to a fusion protein, comprising a cytokine antagonist and a targeting moiety, preferably an antibody or anti-body like molecule. In a preferred embodiment, the cytokine antagonist is a modified cytokine which binds to the receptor, but doesn't induce the receptor signalling. The invention relates further to a fusion protein according to the invention for use in treatment of cancer and immune- or inflammation- related disorders.

Description

TARGETING OF CYTOKINE ANTAGONISTS
The present invention relates to a fusion protein, comprising a cytokine antagonist and a targeting moiety, preferably an antibody or antibody like molecule. In a preferred embodiment, the cytokine antagonist is a modified cytokine which binds to the receptor, but doesn't induce the receptor signalling. The invention relates further to a fusion protein according to the invention for use in treatment of cancer or for use in treatment of autoimmune diseases.
Cytokines are critical mediators of defence mechanisms against microbial invasion and tumorigenesis. However, their production and activities must be tightly regulated to prevent an excessive activity that can culminate in the uncontrolled inflammation and tissue injury, as characteristically observed with many autoimmune diseases.
Rheumatoid arthritis is the classic example of an autoimmune disease where TNFa, I L-1 , and IL-6 play a prominent role in the recruitment of lymphocytes and other types of leukocytes that mediate a progressive joint destruction. TNF inhibitors have been shown to decrease symptoms, slow disease progression, and improve the quality of life for many patients with rheumatoid arthritis (Moreland, 2009). Similarly, a mAb neutralizing IL-12 and IL-23 (ustekinumab) provides a potential therapy for psoriasis (Elliott et al., 2009) and a recombinant human IL-1 receptor antagonist, (anakinra, Kineret™), first approved by the FDA in 2001 for the treatment of rheumatoid arthritis, is a promising agent for the treatment of many IL-1 - mediated autoinflammatory diseases (Goldbach-Mansky, 2009).
Several lines of evidence support the notion that overproduction of type I interferon by plasmacytoid dendritic cells is the primary pathogenesis of several autoimmune diseases, including systemic lupus erythematosus, a multi-system autoimmune disease that affects skin, kidney, musculoskeletal, and hematologic tissues, and Sjogren's syndrome, a disease characterized by the destruction of glands producing tears and saliva and which impacts 1 -3% of the human population. Indeed, if the natural IFN production is not regulated properly, the ensuing prolonged type I IFN exposure can drive autoantibody production which promotes the onset of systemic autoimmune disease (Kiefer et al., 2012). Accordingly, novel therapeutics targeting type I IFN have been developed. For instance, two monoclonal antibodies which neutralize IFNa (Sifalimumab and Rontalizumab) are currently in clinical trials (McBride et al., 2012; Merrill et al., 201 1 ) and a type I IFN antagonist has also been designed (Pan et al., 2008), (PCT/US2009/056366).
IL17A is the best characterized member of the IL17 family of cytokines. This pleiotropic cytokine interacts with a receptor composed of IL17RA and IL17RC subunits. The IL17RA chain is ubiquitously expressed, including haematopoietic, immune, epithelial, endothelial cell types, as well as fibroblasts. IL17A is typically produced by Th17 cells upon activation by a subset of cytokines including IL-1 , IL-6, IL-21 and TGFp, and propagates early inflammatory signals that serve to bridge innate and adaptive immune responses. IL17 is a potent activator of neutrophils and plays an important role in the immune defence against various extracellular pathogens. It is also well established that IL17A promotes autoimmune pathologies (Gaffen, 2009; Shen & Gaffen, 2008). Brodalumab, Secukinumab and Ixekizumab target the IL17A/IL17R axis for treatment of auto-immune diseases such as psoriasis and Crohn's disease. All may inflict adverse side effects including enhanced risk of infections (Hueber et al. 2012; Spuls & Hooft, 2012). Specific targeting of IL17A antagonists to selected cell types such as airway epithelium (asthma), astrocytes (multiple sclerosis), synoviocytes and monocytes/macrophages (rheumatoid arthritis) or keratinocytes (psoriasis) may therefore offer a significant advantage over completely antagonising IL17 function.
ILI a and ΙΙ_β are the founding members of the IL1 cytokine family. Both are pleiotropic and function through a ubiquitously expressed receptor complex composed of IL-1 receptor type-l (IL-1 RI) and IL-1 receptor accessory protein (IL-1 RAcP). Overactivation of this IL-1 axis is associated with many human pathologies including rheumatoid arthritis (RA), chronic obstructive pulmonary disease (COPD), asthma, inflammatory bowel diseases, multiple sclerosis, atherosclerosis and Alzheimer's disease. Many immune cells of different lineages are activated by IL-1 , including innate immune cells such as dendritic cells, macrophages and neutrophils, and also cells involved in the adaptive immune response including naive, Th17 and CD8+ T cells, and B cells (reviewed in Sims and Smith, 2010). Recombinant human IL- 1 RA (IL1 receptor antagonist, aka anakinra) can be used to treat rheumatoid arthritis and is being evaluated for use in a wide spectrum of autoinflammatory diseases (Dinarello, 201 1 ). One of the major side effects of prolonged treatment with anakinra is however the increased occurrence of infections. Selectively antagonising of IL-1 activity on only a subset of (immune) cells therefore may offer a safer alternative. It can be envisaged that targeted inhibition of IL-1 action on selected innate immune cells, leaving its activity on the T cell compartment intact, may still show efficacy for the treatment of inflammatory diseases, without affecting the host defence against pathogens.
Although the IL-7-related cytokine TSLP (thymic stromal lymphopoietin) is best studied in the context of promoting Th2 responses, it is now clear that it functions on various immune and non-immune cell types (reviewed in Roan et al., 2012). Its receptor is composed of the IL-7Ra, which is shared with IL-7, and the widely expressed TSLPRa, also known as CRLF2 (Pandey et al., 2000). TSLP promotes Th2-type inflammation by acting on several distinct cell types, including dendritic cells, CD4 and CD8 T cells, B cells, NKT cells, mast cells, eosinophils and basophils. It supports host defence against helminth parasites, but can contribute to allergic inflammation, and antagonising TSLP was suggested as a treatment for allergic diseases. Conversely, TSLP can have a protective role in inflammatory diseases driven by exacerbated Th1 and Th17 responses, such as Inflammatory Bowel Disease (reviewed in He and Geha, 2010 and Roan et al., 2012). It was recently also found that mutations in the TSLPRa are associated with cancer, including leukemias with poor prognosis (Harvey et al., 2010; Yoda et al., 2010; Ensor et al., 201 1 ), and TSLP levels are correlated with breast cancer progression (Olkhanud et al., 201 1 ) and reduced survival in pancreatic cancer (De Monte et al., 201 1 ). Selective targeting of TSLP antagonists to selected tumor cell types therefore may offer a selective antitumor strategy, and additional modulation by targeted antagonism of selected immune cells may be used to further optimise such strategy. Similar approaches could also be undertaken for non-malignant diseases. The main problem with the therapeutic approaches aiming to neutralize cytokine actions is that the cytokine antagonists are not targeted towards cells or tissues that are specifically involved in the onset of the autoimmune or autoinflammatory diseases. For example, It is easily foreseeable that a long term systemic neutralization of type I IFN activity by a monoclonal antibody or an IFN receptor antagonist carry an important risk in term of viral infection susceptibility and tumor development since type I IFN is a family of proteins essential in the control of viral infections and for establishing immune responses, particularly those controlling cancer cell growth (Gajewski et al., 2012). Similarly, it is expected that a systemic neutralization of IL-1 activity will impact the expansion, effector function, tissue localization, and memory response of antigen-cytotoxic T cells during immune responses (Ben-Sasson et al., 2013).
Surprisingly we found that specific targeting of the cytokine antagonist to a subset of target cells allows reaching the therapeutic effect, without having the negative side effects of systemic cytokine antagonist application. The invention is exemplified by targeting the action of a type I IFN antagonist to specific cell types expressing a given cell surface marker. Such a method is applied to the design and construction of a targeted IFN antagonist that inhibits the action of endogenous IFN specifically on the cell subset culpably involved in the onset of autoimmune diseases, leaving the other cells and organs fully responsive.
Although not yet approved, oncolytic viruses are advancing through clinical trials (Russell et al., 2012). Oncolytic viruses are often designed for having attenuated replication capacity in normal tissues by engineering their sensitivity to the normal cellular interferon-mediated antiviral responses. An example is an oncolytic vesicular stomatitis virus coding for interferon β (Naik et al., 2012). The therapeutic effect of such viruses is expected to be a consequence of the defect of the IFN response exhibited by many tumor cells. However, the genetic heterogeneity of tumors that impact the IFN response is highly variable and impairs the efficacy of virus-mediated tumor lysis (Naik and Russell, 2009). Therefore, by inhibiting the IFN response specifically in tumor cells, a tumor-targeted IFN antagonist would permit the specific destruction of tumor cells by an oncolytic virus. A first aspect of the invention is a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody like molecule. A cytokine antagonist as used here can be any cytokine antagonist known to the person skilled in the art, including but not limited to a soluble receptor, a cytokine binding antibody or a mutant cytokine. Preferably said cytokine antagonist is a mutant cytokine, even more preferably a mutant which binds to the receptor, but is not or only weakly inducing the cytokine signalling. Preferably, the affinity of the mutant for the receptor is comparable to that of the wild type cytokine, even more preferable it has a higher affinity; preferably the signalling induced by the mutant is less than 20% of that of the wild type, even more preferably less than 10% of that of the wild type, even more preferably less than 5%, even more preferably less than 1 %. Most preferably, the binding of the mutant cytokine does not result in detectable signalling. Such mutant can act as a competitive inhibitor of cytokine signalling. An antibody or antibody like molecule as used here is a protein specifically designed to bind another molecule, preferably a proteineous molecule, and comprising the specific binding domains. As a non-limiting example, said antibody or antibody like molecule can be a heavy chain antibody (hcAb), single domain antibody (sdAb), minibody (Tramontano et al., 1994), the variable domain of camelid heavy chain antibody (VHH), the variable domain of the new antigen receptor (VNAR), affibody (Nygren et al., 2008), alphabody (WO2010066740), designed ankyrin-repeat domain (DARPins) (Stumpp et al., 2008), anticalin (Skerra et al., 2008), knottin (Kolmar et al., 2008) and engineered CH2 domain (nanoantibodies; Dimitrov, 2009). The definition, as used here, excludes the Fc tail (without the binding domains) of an antibody. Preferably, said antibody or antibody like molecule consists of a single polypeptide chain, even more preferably, said antibody is not post-translationally modified. Prost-translational modification, as used here, indicates the modifications carried out by living cell during or after the protein synthesis, but excludes modifications, preferably chemical modifications, carried out on the isolated protein such as, but not limited to pegylation Even more preferably said antibody or antibody-like molecule comprises the complementary determining regions, derived from an antibody. Most preferably, said targeting antibody or antibody-like molecule is a nanobody.
Preferably, said cytokine antagonist and said targeting moiety are connected by a linker, preferably a GGS linker. Preferably said GGS linker contains at least 5 GGS repeats, more preferably at least 10 GGS repeats, even more preferably at least 15 GGS repeats, most preferably at least 20 GGS repeats.
In a preferred embodiment, the cytokine antagonist according to the invention is an interferon antagonist; even more preferably, it is an IFNa2-R120E mutant. In another preferred embodiment, the cytokine antagonist according to the invention is an antagonist of a cytokine of the IL17 family, preferably an IL17A antagonist. In still another preferred embodiment, the cytokine antagonist according to the invention is an antagonist of the IL1 cytokine family, preferably an ILI a or ILp antagonist. In still another preferred embodiment, the cytokine antagonist according to the invention is a TSLP antagonist.
In one preferred embodiment, the antibody or antibody-like molecule is directed against a cancer cell marker. Cancer cell markers are known to the person skilled in the art, and include, but are not limited to CD19, CD20, CD22, CD30, CD33, CD37, CD56, CD70, CD74, CD138, AGS16, HER2, MUC1 , GPNMB and PMSA. Preferably, said cancer marker is CD20 or HER2. In another preferred embodiment, the antibody or antibody-like molecule is directed against a marker on an immune cell, preferably an inflammatory cytokine producing immune cell. An immune cell, as used here, is a cell that belongs to the immune system, including but not limited to monocytes, dendritic cells and T-cells. Preferably, said immune cell is a proinflammatory cytokine producing cell.
Markers of inflammatory cytokine producing cells are known to the person skilled in the art and include but are not limited to CD4, CD1 1 b, CD26, sialoadhesin and flt3 receptor.
Another aspect of the invention is a fusion protein according to the invention for use in treatment of cancer. Still another aspect of the invention is a fusion protein according to the invention for use in treatment of autoimmune diseases.
Another aspect of the invention is a method to treat cancer, comprising (i) determination the type of cancer and the suitable targeting marker(s) for the cancer cells in a patient suffering from cancer (ii) providing to said patient in need of the treatment a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody-like molecule according to the invention, possibly with a suitable excipient. It is obvious for the person skilled in the art that the targeting moiety of step (ii) will be directed to the targeting marker identified in step (i). Possible cancer cell markers are known to the person skilled in the art, and include, but are not limited to CD19, CD20, CD22, CD30, CD33, CD37, CD56, CD70, CD74, CD138, AGS16, HER2, MUC1 , GPNMB and PMSA.
Still another aspect of the invention is a method to treat an autoimmune disease, comprising (i) determination in a patient suffering from an autoimmune disease the suitable targeting marker(s) for the immune cells cells (ii) providing to said patient in need of the treatment a fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or an antibody-like molecule according to the invention, possibly with a suitable excipient. Immune cells, as used here, include but are not limited to dendritic cells, CD4 and CD8 T cells, B cells, NKT cells, mast cells, eosinophils and basophils. BRIEF DESCRIPTION OF THE FIGURES
Figure 1 : Representation of the structural elements of the nanobody-hlFNa2-R120E fusion protein. Figure 2: Quantification of the luciferase activity induced by 10 pM hl FNa2 in the presence or absence (untreated) of the 4-1 1 -hl FNa2-R120E fusion protein on HL1 16 (A) and HL1 16- ml_R10 (B) cells.
Figure 3: Quantification of the luciferase activity induced by 1 pM IFN3 in the presence or absence (untreated) of the 4-1 1 -hl FNa2-R120E fusion protein on HL1 16 (A) and HL1 16- ml_R10 (B) cells.
Figure 4: FACS analysis of pY701 -STAT1 in CD19 positive and negative human PBMCs left untreated (left panel), treated with 50 pM of hl FNa2 (center) or with 50 pM of hl FNa2 in the presence of the CD20-targeted I FN antagonist.
Figure 5: Density of the Daudi cell cultures treated by the following components:
A: Untreated
B: hl FNa2. 2 pM
C: hl FNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E.
Figure imgf000007_0001
D: hl FNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E. O.l g/ml
E: hlFNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E-R149A. 3^g/m\
F: hl FNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E-R149A.
Figure imgf000007_0002
G: hl FNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E-L153A. 3^g/m\
H: hl FNa2. 2 pM + 2HCD25-20xGGS-hl FNa2-R120E-L153A.
Figure imgf000007_0003
EXAMPLES
Materials & Methods to the examples
Nanobody-I FN antagonist fusion construction.
Using the QuikChange ll-E Site-Directed Mutagenesis Kit (Agilent), the mutation R120E which abrogates IFN-I FNAR1 binding and confers the antagonistic behaviour of human I FNa2 (Pan et al., 2008), (PCT/US2009/056366), was introduced into the pMET7 SlgK-HA-4.1 1 -His-PAS- ybbr-IFNa2 construct (PCT/EP2013/050787), which is a fusion between a nanobody against the murine leptin receptor and the human I FNa2. Production of the nanobody-I FN antagonist fusion protein
Hek 293T cells were transfected with the protein fusion constructs using the standard lipofectamin method (Invitrogen). 48 hours after the transfection culture mediums were harvested and stored at -20°C. Cell lines
Hek 293T cells were grown in DMEM supplemented with 10% FCS. The HL1 16 clone (Uze et al., 1994) is derived from the human HT1080 cell line. It contains the firefly luciferase gene controlled by the IFN-inducible 6-16 promoter. The derived HL1 16-mLR10 clone which expresses the murine leptin receptor was described (PCT/EP2013/050787).
Measurement of the luciferase activities
Antagonistic IFN activities were measured by quantifying the inhibition of the luciferase activity induced in HL1 16 cells and on the HL1 16-ml_R10 expressing the rmLR by IFNa2 or IFN3. The IC50 values were calculated using nonlinear data regression with Prism software (GraphPad). Luciferase activities were determined on a Berthold Centra LB960 luminometer using a luciferase substrate buffer (20 mM Tricine, 1.07 mM (MgC03)4Mg(OH)2«5H20, 2.67 mM MgS04«7H20, 0.1 mM EDTA, 33.3 mM dithiothreitol, 270 μΜ coenzyme A, 470 μΜ luciferin, 530 μΜ ATP, final pH 7.8) after 6hr IFN stimulation.
Example 1 : The nanobody-IFNa2-R120E fusion protein.
The nanobody 4-1 1 , directed against the murine leptin receptor was fused to the IFNa2 mutant R120E as described in the materials and methods
Figure 1 shows a schematic representation of the nanobody-IFN antagonist fusion protein constructed with the nanobody 4-1 1 against the murine leptin receptor and the human IFNa2- R120E (numbering as in Piehler et al., 2000). Example 2: Targeted inhibition of IFNa activity on mLR-expressing cells
Parental HL1 16 cells and the derived HL1 16-mLR10 cells which express the mouse leptin receptor were treated for 6 hours with 10 pM IFNa2 in the presence of several dilutions of culture medium conditioned by Hek 293T cells expressing the 4-1 1 -IFNa2-R120E fusion protein. The 10 pM IFNa2 dose was chosen because it corresponds to the IFNa2 EC50 on both cell lines. Cells were then lysed and the IFN-induced luciferase activity was quantified. At the higher concentration tested, the 4-1 1 -IFNa2-R120E fusion protein was unable to inhibit IFNa2 action on untargeted HL1 16 cells (Figure 2A). In contrast, its dose-dependent inhibition effect is clear on HL1 16-mLR10 cells which express the target of the 4-1 1 nanobody (Figure 2B).
Example 3: Targeted inhibition of IFN activity on mLR-expressing cells
Among the subtypes which constitute the human type I IFN, the IFN3 shows the highest affinity for the IFNa/β receptor. We thus tested whether the 4-1 1 -IFNa2-R120E fusion protein exerts also an antagonistic activity against IFN3 action.
Parental HL1 16 cells and the derived HL1 16-mLR10 cells which express the mouse leptin receptor were treated for 6 hours with 1 pM IFN3 in the presence of several dilutions of culture medium conditioned by Hek 293T cells expressing the 4-1 1 -IFNa2-R120E fusion protein. The 1 pM IFN3 dose was chosen because it corresponds to the IFN3 EC50 on both cell lines. Cells were then lysed and the IFN-induced luciferase activity was quantified. At the higher concentration tested, the 4-1 1 -IFNa2-R120E fusion protein was unable to inhibit IFNa2 action on untargeted HL1 16 cells (Figure 3A). In contrast, its dose-dependent inhibition effect is clear on HL1 16-mLR10 cells which express the target of the 4-1 1 nanobody (Figure 3B).
Example 4. Specific inhibition of IFNa2-induced STAT1 phosphorylation in B-cells within human whole PBMCs
The type I IFN antagonist IFNa2-R120E was fused to the anti-human CD20 nanobody 2HCD25 through a linker sequence made with 20 repeats of GGS motif. The fusion protein was produced in E. coli and purified by Immobilized Metal Affinity chromatography (IMAC). Human peripheral blood mononuclear cells (PBMCs) are expected to contain =4% of B-cells which can be characterized by the cell surface expression of CD19. The large majority of circulating B-cells are also positive for the expression of CD20.
PBMCs were isolated over ficoll gradient (histopaque-1077, Sigma-Aldrich) from blood samples of healthy donors. Cells were left untreated or were incubated for 15 minutes with 50 pM of human IFNa2 in the absence or presence of 10μg ml of the 2HCD25 nanobody - IFNa2-R120E fusion protein.
Cells were then fixed (BD Fix Buffer I), permeabilized (BD Perm Buffer III) and labelled with PE-labelled anti pSTATI (BD#612564) and APC-labelled anti human CD19 (BD #555415). FACS data were acquired using a BD FACS Canto and analyzed using Diva (BD Biosciences) software for the fluorescence associated with pSTATI in CD19 positive and negative cell populations.
Figure 4 shows that the IFN antagonist linked to the nanobody specific for CD20 inhibits the IFN action specifically in the major part of the B cell population, leaving intact the IFN response in the CD19 negative cell population.
Example 5. The CD20-targeted type I IFN antagonist inhibits the antiproliferative activity of type I IFN.
Having established that the fusion protein of the 2HCD25 nanobody and IFNa2-R120E inhibits IFN-induced STAT1 phosphorylation specifically in B-cells, we tested if it can inhibit the antiproliferative activity of type I IFN. In addition, we evaluated the effect of the IFN mutations L153A and R149A that decrease the affinity of IFNa2 for IFNAR2 by a factor of 10 and 100, respectively, in combination with the inhibiting mutation R120E.
Daudi cells are a human lymphoblastoid B-cell line expressing CD20. Daudi cells were seeded at 2.0x105 cells/ml and were left untreated or cultured for 72 h in the presence of 2 pM IFNa2 alone or in combination with various CD20-targeted IFN antagonists. They were then counted to estimate the efficacy of the inhibition of proliferation induced by IFNa2. Figure 5 shows that the CD20-targeted IFN antagonist fully inhibits the antiproliferative activity of IFNa2. It also shows that decreasing the IFN-IFNAR2 affinity decreases the antagonistic activity, proving that the inhibitory effect is indeed due to the binding of the targeted antagonist.
REFERENCES
Ben-Sasson, S.Z., Hogg, A., Hu-Li, J., Wingfield, P., Chen, X., Crank, M., Caucheteux, S., Ratner-Hurevich, M., Berzofsky, J.A., Nir-Paz, R et al. (2013). IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells. J Exp Med 210, 491 -502.
De Monte, L, Reni, M., Tassi, E., Clavenna, D., Papa, I., Recalde, H., Braga, M., Di Carlo, V., Doglioni, C. and Protti, M.P. (201 1 ). Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp Med 208, 469-478).
Dimitrov, D.S. (2009) Engineered CH2 domains (nanoantibodies). mAbs 1 , 26-28. Dinarello, C.A. (201 1 ). Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 1 17, 3720-3732.
Ensor, H.M., Schwab, C, Russell, L.J., Richards, S.M., Morrison, H., Masic, D., Jones, L, Kinsey, S.E., Vora, A.J., Mitchell, CD., Harrison, C.J. and Moorman, A.V. (201 1 ). Demographic, clinical, and outcome features of children with acute lymphoblastic leukemia and CRLF2 deregulation: results from the MRC ALL97 clinical trial. Blood 1 17, 2129-2136
Elliott, M., Benson, J., Blank, M., Brodmerkel, C, Baker, D., Sharpies, K.R., and Szapary, P. (2009). Ustekinumab: lessons learned from targeting interleukin-12/23p40 in immune- mediated diseases. Ann N Y Acad Sci 1182, 97-1 10.
Gaffen, S.L. (2009). Structure and signalling in the IL-17 receptor family. Nat Rev Immunol 9, 556-567. Gajewski, T.F., Fuertes, M.B., and Woo, S.R. (2012). Innate immune sensing of cancer: clues from an identified role for type I IFNs. Cancer Immunol Immunother 61, 1343-1347.
Goldbach-Mansky, R. (2009). Blocking interleukin-1 in rheumatic diseases. Ann N Y Acad Sci 1182, 1 1 1 -123.
Harvey, R.C., Mullighan, C.G., Chen, I.M., Wharton, W., Mikhail, F.M., Carroll, A.J., Kang, H., Liu, W., Dobbin, K.K., Smith, M.A., Carroll, W.L., Devidas, M., Bowman, W.P., Camitta, B.M., Reaman, G.H., Hunger, S.P., Downing, J.R. and Willman, C.L. (2010). Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1 , Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood 1 15, 5312-5321 .
He, R. and Geha, R.S. (2010). Thymic stromal lymphopoietin. Ann N Y Acad Sci (2010) 1 183, 13-24.
Hueber, W., Sands, B.E., Lewitzky, S., Vandemeulebroecke, M., Reinisch, W., Higgins, P.D., Wehkamp, J., Feagan, B.G., Yao, M.D., Karczewski, M., Karczewski, J., Pezous, N., Bek, S., Bruin, G., Mellgard, B., Berger, C, Londei, M., Bertolino, A.P., Tougas, G. and Travis S.P. (2012). Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 61 , 1693-1700.
Kiefer, K., Oropallo, M.A., Cancro, M.P., and Marshak-Rothstein, A. (2012). Role of type I interferons in the activation of autoreactive B cells. Immunol Cell Biol 90, 498-504.
Kolmar, H. (2008) Alternative binding proteins: biological activity and therapeutic potential of cysteine-knot miniproteins. FEBS J. 275, 2684-2690.
McBride, J.M., Jiang, J., Abbas, A.R., Morimoto, A., Li, J., Maciuca, R., Townsend, M., Wallace, D.J., Kennedy, W.P., and Drappa, J. (2012). Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase I, placebo- controlled, double-blind, dose-escalation study. Arthritis Rheum 64, 3666-3676.
Merrill, J.T., Wallace, D.J., Petri, M., Kirou, K.A., Yao, Y., White, W.I., Robbie, G., Levin, R., Berney, S.M., Chindalore, V., et al. (201 1 ). Safety profile and clinical activity of sifalimumab, a fully human anti-interferon alpha monoclonal antibody, in systemic lupus erythematosus: a phase I, multicentre, double-blind randomised study. Ann Rheum Dis 70, 1905-1913.
Moreland, L.W. (2009). Cytokines as targets for anti-inflammatory agents. Ann N Y Acad Sci 1182, 88-96. Naik, S., Nace, R., Barber, G.N., and Russell, S.J. (2012). Potent systemic therapy of multiple myeloma utilizing oncolytic vesicular stomatitis virus coding for interferon-beta. Cancer Gene Ther 19, 443-450. Naik, S., and Russell, S.J. (2009). Engineering oncolytic viruses to exploit tumor specific defects in innate immune signaling pathways. Expert Opin Biol Ther 9, 1 163-1 176.
Russell, S.J., Peng, K.W., and Bell, J.C. (2012). Oncolytic virotherapy. Nat Biotechnol 30, 658-670.
Nygren, P-A. (2008) Alternative binding proteins: affibody binding proteins developed from a small three-helix bundle scaffold. FEBS J. 275, 2668-2676. Olkhanud, P.B., Rochman, Y., Bodogai, M., Malchinkhuu, E., Wejksza, K., Xu, M., Gress, R.E., Hesdorffer, C, Leonard, W.J. and Biragyn, A. (201 1 ). Thymic stromal lymphopoietin is a key mediator of breast cancer progression. J. Immunol. 186, 5656-5662.
Pan, M., Kalie, E., Scaglione, B.J., Raveche, E.S., Schreiber, G., and Langer, J.A. (2008). Mutation of the IFNAR-1 receptor binding site of human IFN-alpha2 generates type I IFN competitive antagonists. Biochemistry 47, 12018-12027.
Pandey, A., Ozaki, K., Baumann, H., Levin, S.D., Puel, A., Farr, A.G., Ziegler, S.F., Leonard, W.J. and Lodish, H.F. (2000). Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin. Nat Immunol. 1 , 59-64.
Piehler, J., Roisman, L.C. and Schreiber, G. (2000). New structural and functional aspects of the type I interferon-receptor interaction revealed by comprehensive mutational analysis of the binding interface. J. Biol. Chem 275, 40425-40433.
Roan, F., Bell, B.D., Stoklasek, T.A., Kitajima, M., Han, H. and Ziegler, S.F. (2012). The multiple facets of thymic stromal lymphopoietin (TSLP) during allergic inflammation and beyond. J Leuk Biol, 91 , 877-886. Shen, F. and Gaffen, S.L. (2008). Structure-function relationships in the IL-17 receptor: implications for signal transduction and therapy. Cytokine 41 , 92-104.
Sims, J.E. and Smith, D.E. (2010). The IL-1 family: regulators of immunity. Nat Rev Immunol 10, 89-102.
Skerra, A. (2008) Alternative binding proteins: anticalins - harnessing the structural plasticity of the lipocalin ligand pocket to engineer novel binding activities. FEBS J. 275, 2677-2683. Spuls, P.I. and Hooft, L. (2012). Brodalumab and ixekizumab, anti-interleukin-17-receptor antibodies for psoriasis: a critical appraisal. Br J Dermatol 167, 710-713. Stump, M.T., Binz, H.K., Amstutz, P. (2008) DARPins: a new generation of protein therapeutics. Drug iscov. Today 13, 695-701.
Tramontano, A., Bianchi, E., Venturini, S., Martin, F., Pessi, A and Sollazzo, M. (1994) The making of the minibody: an engineered beta-protein for the display of conformationally constrained peptides. J. Mol. Recognition 7, 9-24.
Uze, G., Di Marco, S., Mouchel-Vielh, E., Monneron, D., Bandu, M.T., Horisberger, M.A., Dorques, A., Lutfalla, G., and Mogensen, K.E. (1994). Domains of interaction between alpha interferon and its receptor components. J Mol Biol 243, 245-257. Yoda, A., Yoda, Y., Chiaretti, S., Bar-Natan, M., Mani, K., Rodig, S.J., West, N., Xiao, Y., Brown, J.R., Mitsiades, C, Sattler, M., Kutok, J.L., DeAngelo, D.J., Wadleigh, M., Piciocchi, A., Dal Cin, P., Bradner, J.E., Griffin, J.D., Anderson, K.C., Stone, R.M., Ritz, J., Foa, R., Aster, J.C., Frank, D.A., Weinstock, D.M. (2010). Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA 107, 252-257

Claims

I . A fusion protein comprising a cytokine antagonist and a targeting moiety consisting of an antibody or antibody-like molecule.
2. The fusion protein according to claim 1 , wherein said cytokine antagonist is a mutant cytokine.
3. The fusion protein according to claim 1 or 2, wherein said targeting moiety is a nanobody.
4. The fusion protein according to any of the preceding claims, wherein said cytokine antagonist is an interferon antagonist.
5. The fusion protein according to claim 4, wherein said interferon antagonist is an IFNa2- R120E mutant.
6. The fusion protein according to any of the preceding claims, wherein said targeting moiety is specifically targeted to a marker of a cancer cell.
7. The fusion protein according to any of the claims 1 -5, wherein said targeting moiety is specifically targeted to a marker of an immune cell.
8. A fusion protein according to any of the claims 1 -6, for use in treatment of cancer.
9. A fusion protein according to any of the claims 1 -5 and 7, for use in treatment of an autoimmune disease.
10. The fusion protein for use in treatment of cancer according to claim 8 wherein the targeting moiety is specifically targeted to a marker of a cancer cell.
I I . The fusion protein for use in treatment of cancer according to claim 10, wherein the cytokine antagonist is an IFNa2-R120E mutant.
12. The fusion protein for use in treatment of an autoimmune disease according to claim 9, wherein the targeting moiety is specifically targeted to a marker of an immune cell.
13. The fusion protein for use in treatment of an autoimmune disease according to claim 12, wherein the cytokine antagonist is an IFNa2-R120E mutant.
PCT/EP2014/063976 2013-07-19 2014-07-01 Targeting of cytokine antagonists WO2015007520A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
SG11201600165WA SG11201600165WA (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists
EP14734166.3A EP3022230B1 (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists
CN201480040656.4A CN105658669A (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists
AU2014292355A AU2014292355B2 (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists
JP2016526507A JP6475712B2 (en) 2013-07-19 2014-07-01 Targeting cytokine antagonists
KR1020167002677A KR102305608B1 (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists
CA2918119A CA2918119C (en) 2013-07-19 2014-07-01 Targeting of ifn.alpha.2 antagonists to treat autoimmune diseases
US14/905,348 US9732135B2 (en) 2013-07-19 2014-07-01 Targeting of human interferon antagonists
MX2016000721A MX2016000721A (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists.
IL24345916A IL243459B (en) 2013-07-19 2016-01-04 Targeting of cytokine antagonists
US15/642,989 US10072059B2 (en) 2013-07-19 2017-07-06 Targeting of human interferon antagonists
US16/008,686 US10947288B2 (en) 2013-07-19 2018-06-14 Targeting of human interferon antagonists
US16/008,695 US20180334489A1 (en) 2013-07-19 2018-06-14 Targeting of cytokine antagonists
US17/171,433 US20210238247A1 (en) 2013-07-19 2021-02-09 Targeting of human interferon antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13306045.9 2013-07-19
EP13306045 2013-07-19

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/905,348 A-371-Of-International US9732135B2 (en) 2013-07-19 2014-07-01 Targeting of human interferon antagonists
US15/642,989 Continuation US10072059B2 (en) 2013-07-19 2017-07-06 Targeting of human interferon antagonists

Publications (1)

Publication Number Publication Date
WO2015007520A1 true WO2015007520A1 (en) 2015-01-22

Family

ID=48874233

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/063976 WO2015007520A1 (en) 2013-07-19 2014-07-01 Targeting of cytokine antagonists

Country Status (11)

Country Link
US (5) US9732135B2 (en)
EP (1) EP3022230B1 (en)
JP (1) JP6475712B2 (en)
KR (1) KR102305608B1 (en)
CN (1) CN105658669A (en)
AU (1) AU2014292355B2 (en)
CA (1) CA2918119C (en)
IL (1) IL243459B (en)
MX (1) MX2016000721A (en)
SG (2) SG11201600165WA (en)
WO (1) WO2015007520A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017077382A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
WO2017134305A1 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Bispecific signaling agents and uses thereof
WO2017153345A1 (en) * 2016-03-07 2017-09-14 Vib Vzw Cd20 binding agents and uses thereof
WO2017153402A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
WO2017194782A2 (en) 2016-05-13 2017-11-16 Orionis Biosciences Nv Therapeutic targeting of non-cellular structures
WO2017194783A1 (en) 2016-05-13 2017-11-16 Orionis Biosciences Nv Targeted mutant interferon-beta and uses thereof
WO2018077893A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
WO2018141964A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof
WO2018144999A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
WO2018146074A1 (en) 2017-02-07 2018-08-16 Vib Vzw Immune-cell targeted bispecific chimeric proteins and uses thereof
WO2021009332A1 (en) 2019-07-18 2021-01-21 Enyo Pharma Method for decreasing adverse-effects of interferon
US11440943B2 (en) 2019-03-28 2022-09-13 Orionis Biosciences, Inc. Therapeutic interferon alpha 1 proteins
WO2022200525A1 (en) 2021-03-26 2022-09-29 Innate Pharma Multispecific proteins comprising an nkp46-binding site, a cancer antgienge binding site fused to a cytokine for nk cell engaging
US11498966B2 (en) 2017-08-09 2022-11-15 Orionis Biosciences Inc. PD-1 and PD-L1 binding agents
WO2022258662A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp46, a cytokine receptor, a tumour antigen and cd16a
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
US11566072B2 (en) 2017-08-09 2023-01-31 Orionis Biosciences, Inc. CD8 binding agents
US11661455B2 (en) 2016-02-05 2023-05-30 Orionis Biosciences BV Chimeric protein comprising an interferon alpha 2mutant and a PD-L1 binding moiety
US11896643B2 (en) 2018-02-05 2024-02-13 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
WO2024040249A1 (en) 2022-08-18 2024-02-22 Regeneron Pharmaceuticals, Inc. Interferon receptor agonists and uses thereof
WO2024040247A1 (en) 2022-08-18 2024-02-22 Regeneron Pharmaceuticals, Inc. Interferon proproteins and uses thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2804877B1 (en) * 2012-01-20 2018-08-22 VIB vzw Targeted mutant alpha-helical bundle cytokines
KR102305608B1 (en) * 2013-07-19 2021-09-28 브이아이비 브이지더블유 Targeting of cytokine antagonists
CN111499718B (en) * 2019-01-30 2022-06-14 复旦大学 Human alpha interferon receptor binding related site mutant and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010066740A1 (en) 2008-12-08 2010-06-17 Complix Nv Single-chain antiparallel coiled coil proteins
US20130183298A1 (en) * 1991-03-18 2013-07-18 New York University Methods of Treating Hepatitis With Anti-TNF alpha Antibodies

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0489116B1 (en) 1989-08-22 1994-04-06 Immunex Corporation Fusion proteins comprising gm-csf and il-3
US5914254A (en) 1993-08-02 1999-06-22 Celtrix Pharmaceuticals, Inc. Expression of fusion polypeptides transported out of the cytoplasm without leader sequences
ES2243995T3 (en) * 1996-04-26 2005-12-01 Beth Israel Deaconess Medical Center, Inc. INTERLEUCINE ANTAGONISTS-15.
US6617135B1 (en) * 1999-08-09 2003-09-09 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
US20020193569A1 (en) * 2001-06-04 2002-12-19 Idec Pharmaceuticals Corporation Bispecific fusion protein and method of use for enhancing effector cell killing of target cells
CA2585549A1 (en) 2004-11-18 2006-05-26 Vib Vzw Novel type leptin receptor antagonist
WO2006115800A2 (en) 2005-04-15 2006-11-02 The Regents Of The University Of California Enhanced wound healing utilizing an anti-her2 antibody coupled to a tnf alpha
EP2476428B1 (en) * 2005-06-29 2013-08-21 Yeda Research and Development Co. Ltd. Recombinant interferon alpha 2 (IFN alpha 2) mutants
CA2693326A1 (en) 2006-08-02 2008-02-07 Mcgill University Gm-csf and il-15 fusokines and methods for modulation of the immune response
WO2008124086A2 (en) 2007-04-05 2008-10-16 President And Fellows Of Harvard College Chimeric activators: quantitatively designed protein therapeutics and uses thereof
CN104109200B (en) * 2007-05-11 2018-03-20 阿尔托生物科学有限公司 Fusion molecule and the variants of IL 15
WO2009003145A1 (en) 2007-06-26 2008-12-31 University Of Miami Antibody-endostatin fusion protein and its variants
EP2853267B1 (en) 2007-09-21 2016-12-07 The Regents of the University of California Targeted interferon demonstrates potent apoptotic and anti-tumor activities
US20110224407A1 (en) * 2008-09-09 2011-09-15 University Of Medicine And Dentistry Of New Jersey Type I Interferon Antagonists
WO2010036918A2 (en) 2008-09-26 2010-04-01 University Of Massachusetts Intracellular dna receptor
ES2534085T3 (en) 2009-08-17 2015-04-17 Roche Glycart Ag Targeted Immunoconjugates
EP2475397A1 (en) 2009-09-10 2012-07-18 Cytos Biotechnology AG Use of interleukin-1 beta mutein conjugates in the treatment of diabetes
US9534056B2 (en) 2011-06-06 2017-01-03 Immungene Inc Engineered TAA antibody-TNFSF member ligand fusion molecules
JP6184965B2 (en) * 2011-10-28 2017-08-23 テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド Polypeptide constructs and uses thereof
EP2804877B1 (en) * 2012-01-20 2018-08-22 VIB vzw Targeted mutant alpha-helical bundle cytokines
EP2822575B1 (en) 2012-03-03 2020-05-06 ImmunGene, Inc. Engineered antibody-interferon mutant fusion molecules
KR102305608B1 (en) * 2013-07-19 2021-09-28 브이아이비 브이지더블유 Targeting of cytokine antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130183298A1 (en) * 1991-03-18 2013-07-18 New York University Methods of Treating Hepatitis With Anti-TNF alpha Antibodies
WO2010066740A1 (en) 2008-12-08 2010-06-17 Complix Nv Single-chain antiparallel coiled coil proteins

Non-Patent Citations (36)

* Cited by examiner, † Cited by third party
Title
BEN-SASSON, S.Z.; HOGG, A.; HU-LI, J.; WINGFIELD, P.; CHEN, X.; CRANK, M.; CAUCHETEUX, S.; RATNER-HUREVICH, M.; BERZOFSKY, J.A.; N: "IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells", J EXP MED, vol. 210, 2013, pages 491 - 502
DE MONTE, L.; RENI, M.; TASSI, E.; CLAVENNA, D.; PAPA, I.; RECALDE, H.; BRAGA, M.; DI CARLO, V.; DOGLIONI, C.; PROTTI, M.P.: "Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer", J. EXP MED, vol. 208, 2011, pages 469 - 478
DIMITROV DIMITER S: "Engineered CH2 domains (nanoantibodies)", MABS, LANDES BIOSCIENCE, US, vol. 1, no. 1, 1 January 2009 (2009-01-01), pages 26 - 28, XP002596414, ISSN: 1942-0870 *
DIMITROV, D.S.: "Engineered CH2 domains (nanoantibodies", MABS, vol. 1, 2009, pages 26 - 28, XP002596414
DINARELLO, C.A.: "Interleukin-1 in the pathogenesis and treatment of inflammatory diseases", BLOOD, vol. 117, 2011, pages 3720 - 3732, XP055146700, DOI: doi:10.1182/blood-2010-07-273417
ELLIOTT, M.; BENSON, J.; BLANK, M.; BRODMERKEL, C.; BAKER, D.; SHARPLES, K.R.; SZAPARY, P.: "Ustekinumab: lessons learned from targeting interleukin-12/23p40 in immune-mediated diseases", ANN N Y ACAD SCI, vol. 1182, 2009, pages 97 - 110
ENSOR, H.M.; SCHWAB, C.; RUSSELL, L.J.; RICHARDS, S.M.; MORRISON, H.; MASIC, D.; JONES, L.; KINSEY, S.E.; VORA, A.J.; MITCHELL, C.: "Demographic, clinical, and outcome features of children with acute lymphoblastic leukemia and CRLF2 deregulation: results from the MRC ALL97 clinical trial", BLOOD, vol. 117, 2011, pages 2129 - 2136
GAFFEN, S.L.: "Structure and signalling in the IL- 7 receptor family", NAT REV IMMUNOL, vol. 9, 2009, pages 556 - 567, XP055052444, DOI: doi:10.1038/nri2586
GAJEWSKI, T.F.; FUERTES, M.B.; WOO, S.R.: "Innate immune sensing of cancer: clues from an identified role for type I IFNs", CANCER IMMUNOL IMMUNOTHER, vol. 61, 2012, pages 1343 - 1347, XP035088690, DOI: doi:10.1007/s00262-012-1305-6
GOLDBACH-MANSKY, R.: "Blocking interleukin-1 in rheumatic diseases", ANN N Y ACAD SCI, vol. 1182, 2009, pages 111 - 123
HARVEY, R.C.; MULLIGHAN, C.G.; CHEN, I.M.; WHARTON, W.; MIKHAIL, F.M.; CARROLL, A.J.; KANG, H.; LIU, W.; DOBBIN, K.K.; SMITH, M.A.: "Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia", BLOOD, vol. 115, 2010, pages 5312 - 5321
HE, R.; GEHA, R.S.: "Thymic stromal lymphopoietin", ANN N Y ACAD SCI, vol. 1183, 2010, pages 13 - 24, XP008152987, DOI: doi:DOI: 10.1111/j.1749-6632.2009.05128.x
HUEBER, W.; SANDS, B.E.; LEWITZKY, S.; VANDEMEULEBROECKE, M.; REINISCH, W.; HIGGINS, P.D.; WEHKAMP, J.; FEAGAN, B.G.; YAO, M.D.; K: "Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial", GUT, vol. 61, 2012, pages 1693 - 1700
KIEFER, K.; OROPALLO, M.A.; CANCRO, M.P.; MARSHAK-ROTHSTEIN, A.: "Role of type I interferons in the activation of autoreactive B cells", IMMUNOL CELL BIOL, vol. 90, 2012, pages 498 - 504
KOLMAR, H.: "Alternative binding proteins: biological activity and therapeutic potential of cysteine-knot miniproteins", FEBS J., vol. 275, 2008, pages 2684 - 2690, XP055417456, DOI: doi:10.1111/j.1742-4658.2008.06440.x
MANJING PAN ET AL: "Mutation of the IFNAR-1 Receptor Binding Site of Human IFN-[alpha]2 Generates Type I IFN Competitive Antagonists +", BIOCHEMISTRY, vol. 47, no. 46, 18 November 2008 (2008-11-18), pages 12018 - 12027, XP055145910, ISSN: 0006-2960, DOI: 10.1021/bi801588g *
MCBRIDE, J.M.; JIANG, J.; ABBAS, A.R.; MORIMOTO, A.; LI, J.; MACIUCA, R.; TOWNSEND, M.; WALLACE, D.J.; KENNEDY, W.P.; DRAPPA, J.: "Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase , placebo-controlled, double-blind, dose-escalation study", ARTHRITIS RHEUM, vol. 64, 2012, pages 3666 - 3676, XP002697719, DOI: doi:10.1002/ART.34632
MERRILL, J.T.; WALLACE, D.J.; PETRI, M.; KIROU, K.A.; YAO, Y.; WHITE, W.I.; ROBBIE, G.; LEVIN, R.; BERNEY, S.M.; CHINDALORE, V. ET: "Safety profile and clinical activity of sifalimumab, a fully human anti-interferon alpha monoclonal antibody, in systemic lupus erythematosus: a phase , multicentre, double-blind randomised study", ANN RHEUM DIS, vol. 70, 2011, pages 1905 - 1913, XP009169832, DOI: doi:10.1136/ard.2010.144485
MORELAND, L.W.: "Cytokines as targets for anti-inflammatory agents", ANN N Y ACAD SCI, vol. 1182, 2009, pages 88 - 96
NAIK, S.; NACE, R.; BARBER, G.N.; RUSSELL, S.J.: "Potent systemic therapy of multiple myeloma utilizing oncolytic vesicular stomatitis virus coding for interferon-beta", CANCER GENE THER, vol. 19, 2012, pages 443 - 450
NAIK, S.; RUSSELL, S.J.: "Engineering oncolytic viruses to exploit tumor specific defects in innate immune signaling pathways", EXPERT OPIN BIOL THER, vol. 9, 2009, pages 1163 - 1176
NYGREN, P-A.: "Alternative binding proteins: affibody binding proteins developed from a small three-helix bundle scaffold", FEBS J., vol. 275, 2008, pages 2668 - 2676, XP002601623, DOI: doi:10.1111/J.1742-4658.2008.06438.X
OLKHANUD, P.B.; ROCHMAN, Y.; BODOGAI, M.; MALCHINKHUU, E.; WEJKSZA, K.; XU, M.; GRESS, R.E.; HESDORFFER, C.; LEONARD, W.J.; BIRAGY: "Thymic stromal lymphopoietin is a key mediator of breast cancer progression", J. IMMUNOL., vol. 186, 2011, pages 5656 - 5662
PAN, M.; KALIE, E.; SCAGLIONE, B.J.; RAVECHE, E.S.; SCHREIBER, G.; LANGER, J.A.: "Mutation of the IFNAR-1 receptor binding site of human IFN-alpha2 generates type I IFN competitive antagonists", BIOCHEMISTRY, vol. 47, 2008, pages 12018 - 12027, XP055145910, DOI: doi:10.1021/bi801588g
PANDEY, A.; OZAKI, K.; BAUMANN, H.; LEVIN, S.D.; PUEL, A.; FARR, A.G.; ZIEGLER, S.F.; LEONARD, W.J.; LODISH, H.F.: "Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin", NAT IMMUNOL., vol. 1, 2000, pages 59 - 64, XP008014551, DOI: doi:10.1038/76923
PIEHLER, J.; ROISMAN, L.C.; SCHREIBER, G.: "New structural and functional aspects of the type I interferon-receptor interaction revealed by comprehensive mutational analysis of the binding interface", J. BIOL. CHEM, vol. 275, 2000, pages 40425 - 40433
ROAN, F.; BELL, B.D.; STOKLASEK, T.A.; KITAJIMA, M.; HAN, H.; ZIEGLER, S.F.: "The multiple facets of thymic stromal lymphopoietin (TSLP) during allergic inflammation and beyond", J LEUK BIOL, vol. 91, 2012, pages 877 - 886, XP008152989, DOI: doi:10.1189/jlb.1211622
RUSSELL, S.J.; PENG, K.W.; BELL, J.C.: "Oncolytic virotherapy", NAT BIOTECHNOL, vol. 30, 2012, pages 658 - 670, XP055164204, DOI: doi:10.1038/nbt.2287
SHEN, F.; GAFFEN, S.L.: "Structure-function relationships in the IL-17 receptor: implications for signal transduction and therapy", CYTOKINE, vol. 41, 2008, pages 92 - 104, XP022458527, DOI: doi:10.1016/j.cyto.2007.11.013
SIMS, J.E.; SMITH, D.E.: "The IL-1 family: regulators of immunity", NAT REV IMMUNOL, vol. 10, 2010, pages 89 - 102
SKERRA, A.: "Alternative binding proteins: anticalins - harnessing the structural plasticity of the lipocalin ligand pocket to engineer novel binding activities", FEBS J., vol. 275, 2008, pages 2677 - 2683, XP055099855, DOI: doi:10.1111/j.1742-4658.2008.06439.x
SPULS, P.I.; HOOFT, L.: "Brodalumab and ixekizumab, anti-interleukin-17-receptor antibodies for psoriasis: a critical appraisal", BR J DERMATOL, vol. 167, 2012, pages 710 - 713
STUMP, M.T.; BINZ, H.K.; AMSTUTZ, P.: "DARPins: a new generation of protein therapeutics", DRUG ISCOV. TODAY, vol. 13, 2008, pages 695 - 701, XP023440383, DOI: doi:10.1016/j.drudis.2008.04.013
TRAMONTANO, A.; BIANCHI, E.; VENTURINI, S.; MARTIN, F.; PESSI, A; SOLLAZZO, M.: "The making of the minibody: an engineered beta-protein for the display of conformationally constrained peptides", J. MOL. RECOGNITION, vol. 7, 1994, pages 9 - 24
UZE, G.; DI MARCO, S.; MOUCHEL-VIELH, E.; MONNERON, D.; BANDU, M.T.; HORISBERGER, M.A.; DORQUES, A.; LUTFALLA, G.; MOGENSEN, K.E.: "Domains of interaction between alpha interferon and its receptor components", J MOL BIOL, vol. 243, 1994, pages 245 - 257, XP024008163, DOI: doi:10.1006/jmbi.1994.1651
YODA, A.; YODA, Y.; CHIARETTI, S.; BAR-NATAN, M.; MANI, K.; RODIG, S.J.; WEST, N.; XIAO, Y.; BROWN, J.R.; MITSIADES, C.: "Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia", PROC NATL ACAD SCI USA, vol. 107, 2010, pages 252 - 257, XP055169038, DOI: doi:10.1073/pnas.0911726107

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11702477B2 (en) 2015-11-06 2023-07-18 Orionis Biosciences BV Bi-functional chimeric proteins and uses thereof
WO2017077382A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
EP3998281A1 (en) 2016-02-05 2022-05-18 Orionis Biosciences BV Cd8 binding agents
WO2017134305A1 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Bispecific signaling agents and uses thereof
WO2017134302A2 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Targeted therapeutic agents and uses thereof
US11236166B2 (en) 2016-02-05 2022-02-01 Orionis Biosciences BV CD8 binding agents
EP3909978A1 (en) 2016-02-05 2021-11-17 Orionis Biosciences BV Clec9a binding agents and use thereof
US11001631B2 (en) 2016-02-05 2021-05-11 Orionis Biosciences BV Clec9A binding agents
US10988538B2 (en) 2016-02-05 2021-04-27 Orionis Biosciences BV Bispecific signaling agents and uses thereof
US11661455B2 (en) 2016-02-05 2023-05-30 Orionis Biosciences BV Chimeric protein comprising an interferon alpha 2mutant and a PD-L1 binding moiety
EP4059957A1 (en) 2016-02-05 2022-09-21 Orionis Biosciences BV Bispecific signaling agents and uses thereof
JP2019507762A (en) * 2016-03-07 2019-03-22 ブイアイビー ブイゼットダブリュー CD20 binding single domain antibody
CN109069573A (en) * 2016-03-07 2018-12-21 弗拉芒区生物技术研究所 In conjunction with the single domain antibody of CD20
EP4276114A3 (en) * 2016-03-07 2024-02-21 Vib Vzw Cd20 binding single domain antibodies
EP4276114A2 (en) 2016-03-07 2023-11-15 Vib Vzw Cd20 binding single domain antibodies
WO2017153402A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
WO2017153345A1 (en) * 2016-03-07 2017-09-14 Vib Vzw Cd20 binding agents and uses thereof
US11248057B2 (en) 2016-03-07 2022-02-15 Vib Vzw CD20 binding single domain antibodies
EP3426278B1 (en) * 2016-03-07 2024-01-03 Vib Vzw Cd20 binding single domain antibodies
CN109069573B (en) * 2016-03-07 2022-04-05 弗拉芒区生物技术研究所 Single domain antibodies that bind CD20
US11753463B2 (en) 2016-05-13 2023-09-12 Orionis Biosciences BV Therapeutic targeting of non-cellular structures
WO2017194783A1 (en) 2016-05-13 2017-11-16 Orionis Biosciences Nv Targeted mutant interferon-beta and uses thereof
WO2017194782A2 (en) 2016-05-13 2017-11-16 Orionis Biosciences Nv Therapeutic targeting of non-cellular structures
US11236141B2 (en) 2016-05-13 2022-02-01 Orionis Biosciences BV Targeted mutant interferon-beta and uses thereof
WO2018077893A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
US11084859B2 (en) 2016-10-24 2021-08-10 Orionis Biosciences BV Targeted mutant interferon-gamma and uses thereof
US11384154B2 (en) 2017-02-06 2022-07-12 Orionis Biosciences BV Targeted chimeric proteins and uses thereof
US10906985B2 (en) 2017-02-06 2021-02-02 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
WO2018144999A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
WO2018141964A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof
WO2018146074A1 (en) 2017-02-07 2018-08-16 Vib Vzw Immune-cell targeted bispecific chimeric proteins and uses thereof
US11246911B2 (en) 2017-02-07 2022-02-15 Vib Vzw Immune-cell targeted bispecific chimeric proteins and uses thereof
US11566072B2 (en) 2017-08-09 2023-01-31 Orionis Biosciences, Inc. CD8 binding agents
US11498966B2 (en) 2017-08-09 2022-11-15 Orionis Biosciences Inc. PD-1 and PD-L1 binding agents
US11896643B2 (en) 2018-02-05 2024-02-13 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
US11440943B2 (en) 2019-03-28 2022-09-13 Orionis Biosciences, Inc. Therapeutic interferon alpha 1 proteins
WO2021009332A1 (en) 2019-07-18 2021-01-21 Enyo Pharma Method for decreasing adverse-effects of interferon
WO2022200525A1 (en) 2021-03-26 2022-09-29 Innate Pharma Multispecific proteins comprising an nkp46-binding site, a cancer antgienge binding site fused to a cytokine for nk cell engaging
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
WO2022258662A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp46, a cytokine receptor, a tumour antigen and cd16a
WO2024040249A1 (en) 2022-08-18 2024-02-22 Regeneron Pharmaceuticals, Inc. Interferon receptor agonists and uses thereof
WO2024040247A1 (en) 2022-08-18 2024-02-22 Regeneron Pharmaceuticals, Inc. Interferon proproteins and uses thereof

Also Published As

Publication number Publication date
US20160159875A1 (en) 2016-06-09
SG11201600165WA (en) 2016-02-26
KR102305608B1 (en) 2021-09-28
JP2016529232A (en) 2016-09-23
AU2014292355A1 (en) 2016-02-04
US9732135B2 (en) 2017-08-15
US20180334489A1 (en) 2018-11-22
US10947288B2 (en) 2021-03-16
US20180334488A1 (en) 2018-11-22
EP3022230A1 (en) 2016-05-25
US20210238247A1 (en) 2021-08-05
IL243459A0 (en) 2016-03-31
IL243459B (en) 2019-10-31
KR20160108292A (en) 2016-09-19
MX2016000721A (en) 2016-09-07
EP3022230B1 (en) 2019-11-06
US20180057555A1 (en) 2018-03-01
CA2918119C (en) 2022-11-29
JP6475712B2 (en) 2019-02-27
SG10202010429YA (en) 2020-11-27
CA2918119A1 (en) 2015-01-22
AU2014292355B2 (en) 2019-08-01
CN105658669A (en) 2016-06-08
US10072059B2 (en) 2018-09-11

Similar Documents

Publication Publication Date Title
US20210238247A1 (en) Targeting of human interferon antagonists
Larochette et al. IL-26, a cytokine with roles in extracellular DNA-induced inflammation and microbial defense
Hamilton et al. Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases
Whibley et al. Antibody blockade of IL-17 family cytokines in immunity to acute murine oral mucosal candidiasis
Campanella et al. CXCL10 can inhibit endothelial cell proliferation independently of CXCR3
Wolk et al. Biology of interleukin-22
Hoge et al. IL-6 controls the innate immune response against Listeria monocytogenes via classical IL-6 signaling
RU2419450C2 (en) Method of treating and preventing fibrosis by il-21/il-21r antagonists
Doodes et al. IFN-γ regulates the requirement for IL-17 in proteoglycan-induced arthritis
US20050214296A1 (en) Methods of modulating cytokine activity; related reagents
Lokau et al. Biological functions and therapeutic opportunities of soluble cytokine receptors
Lalani et al. Biology of IL-5 in health and disease
Chiricozzi et al. Emerging treatment options for the treatment of moderate to severe plaque psoriasis and psoriatic arthritis: evaluating bimekizumab and its therapeutic potential
JP2009543579A (en) WSX-1 / p28 as a target for anti-inflammatory response
Ruzek et al. Dual blockade of interleukin-1β and interleukin-17A reduces murine arthritis pathogenesis but also leads to spontaneous skin infections in nonhuman primates
JP2023538919A (en) CD25-biased anti-IL-2 antibody
TAVERNIER TARGETING OF HUMAN INTERFERON ANTAGONISTS-Patent Information
BR112016001122B1 (en) FUSION PROTEIN COMPRISING AN INTERFERON ANTAGONIST AND A TARGETING FRACTION
TAVERNIER et al. Patent 2918119 Summary
Zhou The function of IL-17F in infection and inflammation
CN116783213A (en) CD25 biased anti-IL-2 antibodies
Moncada Vélez et al. Human TYK2 deficiency: Mycobacterial and viral infections without hyper-IgE syndrome
Ono et al. Accessory Protein-Like Is Essential for

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14734166

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 243459

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2918119

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016526507

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14905348

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/000721

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20167002677

Country of ref document: KR

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016001122

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2014292355

Country of ref document: AU

Date of ref document: 20140701

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014734166

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112016001122

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160118