WO2013107791A1 - Targeted mutant alpha-helical bundle cytokines - Google Patents

Targeted mutant alpha-helical bundle cytokines Download PDF

Info

Publication number
WO2013107791A1
WO2013107791A1 PCT/EP2013/050787 EP2013050787W WO2013107791A1 WO 2013107791 A1 WO2013107791 A1 WO 2013107791A1 EP 2013050787 W EP2013050787 W EP 2013050787W WO 2013107791 A1 WO2013107791 A1 WO 2013107791A1
Authority
WO
WIPO (PCT)
Prior art keywords
targeting
ifn
mutant
ifna2
cells
Prior art date
Application number
PCT/EP2013/050787
Other languages
French (fr)
Inventor
Jan Tavernier
Gilles UZÉ
Guillaume Cartron
Franciane PAUL
Jacob Piehler
Original Assignee
Vib Vzw
Universiteit Gent
Centre National De La Recherche Scientifique
Université Montpellier 2
Centre Hospitalier Regional Universitaire De Montpellier
Universität Osnabrück
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 KR1020147023041A priority Critical patent/KR102050119B1/en
Priority to CA2861927A priority patent/CA2861927C/en
Priority to EP13701014.6A priority patent/EP2804877B1/en
Priority to AU2013211059A priority patent/AU2013211059B8/en
Application filed by Vib Vzw, Universiteit Gent, Centre National De La Recherche Scientifique, Université Montpellier 2, Centre Hospitalier Regional Universitaire De Montpellier, Universität Osnabrück filed Critical Vib Vzw
Priority to BR112014017876-3A priority patent/BR112014017876B1/en
Priority to ES13701014.6T priority patent/ES2694180T3/en
Priority to JP2014552617A priority patent/JP6416628B2/en
Priority to US14/372,730 priority patent/US9492562B2/en
Priority to CN201380015170.0A priority patent/CN104245734B/en
Publication of WO2013107791A1 publication Critical patent/WO2013107791A1/en
Priority to HK15105433.7A priority patent/HK1205134A1/en
Priority to US15/278,854 priority patent/US9878014B2/en
Priority to US15/717,205 priority patent/US10034919B2/en
Priority to US16/020,447 priority patent/US10946070B2/en
Priority to US17/171,426 priority patent/US20210162010A1/en

Links

Classifications

    • 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
    • A61K47/68Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6813Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin the drug being a peptidic cytokine, e.g. an interleukin or interferon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • 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
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • 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
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • 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 modified a-helical bundle cytokine, with reduced activity via a a-helical bundle cytokine receptor, wherein said a-helical bundle cytokine is specifically delivered to target cells.
  • said a-helical bundle cytokine is a mutant, more preferably it is a mutant interferon, with low affinity to the interferon receptor, wherein said mutant interferon is specifically delivered to target cells.
  • the targeting is realized by fusion of the modified a-helical bundle cytokine to a targeting moiety, preferably an antibody.
  • the invention relates further to the use of such targeted modified a-helical bundle cytokine to treat diseases.
  • a preferred embodiment is the use of a targeted mutant interferon, to treat diseases, preferably viral diseases and tumors.
  • Cytokines are small proteins that play an important role in intercellular communication. Cytokines can be classified based on their structure, the largest group being the four-a-helix bundle family. This family can, based on the use of receptors, further be divided in the interferon (IFN) and interleukin (IL)-2, -3, -10 and -12 subfamilies.
  • IFN interferon
  • IL-2 interleukin-2, -3, -10 and -12 subfamilies.
  • the a helical bundle cytokines are important as possible biopharmaceutical for treatment of human diseases; as a non-limiting example erythropoietin is used for treatment of anemia or red blood cell deficiency, somatotropin for treatment of growth hormone deficiency and interleukin-2 in the treatment of cancer.
  • type I IFNs belong to a cytokine family having important biological functions.
  • type I IFNs 13 ⁇ , ⁇ , ⁇ , ⁇ , ⁇
  • the assembling of the IFN-receptor complex initiates the activation of several signal transduction pathways that, depending of the cell type, modify cellular differentiation and/or functions.
  • type I I FN By acting on virtually every cell type, type I I FN is able to prevent productive viral infection. In addition, it exhibits marked antiangiogenic and proapoptotic effects. Type I IFNs are also deeply implicated in the regulation of several functions of the innate and adaptive immunity as well as on bone homeostasis. It acts particularly on the activation/differentiation of dendritic cells and osteoclasts. The type I IFN system is in fact critically important for the health of mammals.
  • mice Preclinical studies in mice have established a remarkable efficacy of type I IFN for the treatment of both viral or tumor diseases.
  • mice cured of an experimental tumor by IFN treatment have been found immunized against the initial tumor, suggesting that IFN acts not only to engage the processes of tumor rejection but also to break the immune tolerance against the tumor.
  • IFNa was approved in clinic for the treatment of both viral infection and cancer.
  • IFN3 was shown to be effective in relapsing- remitting multiple sclerosis and was also approved for this pathology.
  • the clinical efficacy of IFN was often found disappointing and today other therapeutic strategies such as specific antiviral compounds, chemotherapies and monoclonal antibodies have, when possible, largely supplanted IFN broad application.
  • IFN type I IFN
  • systemic toxicity and side effects including flu-like syndrome, depression, hepatotoxicity, autoimmune disease, thyroid dysfunction and weight loss. It would thus be highly worthwhile to target IFN activity toward only the cellular population which should be treated with IFN (e.g. infected organ or tumor mass) or activated by IFN (e.g. subsets of immune cells).
  • the patent application discloses the fusion of an antibody as targeting moiety with wild type IFN, but also with mutated IFN.
  • the IFN fragment should retain its endogeneous activity at a level of at least 80%, or even at a higher level than wild type IFN. Also in this case, the fusion is retaining the unwanted side effects of the wild type.
  • a modified a-helical bundle cytokine with a decreased affinity for the a-helical bundle cytokine receptor and a consequent decreased specific bioactivity can be fused to a targeting moiety, wherein the bioactivity is restored towards the targeted cells, but not towards cells that are not targeted by the construct.
  • Such construct has the advantage over the art of having less side effects, especially a lower systemic toxicity, while retaining the bioactivity against the target cells.
  • a first aspect of the invention is a targeting construct, comprising modified ⁇ -helical bundle cytokine, characterized by a reduced affinity for the a-helical bundle cytokine receptor, and a targeting moiety
  • ⁇ -helical bundle cytokines are known to the person skilled in the art, and include, but are not limited Cardiotrophin-like cytokine NNT-1 , Ciliary neurothrophic factor, Macrophage colony stimulating factor, Granulocyte-macrophage colony stimulating factor, Granulocyte colony stimulating factor, Cardiotrophin-1 , Erythropoietin, FLT3 ligand, Somatotropin, Interferon a-1 , 2, 4, 5, 6, 7, 8, 10, 13, 14, 16, 17, 21 ,, Interferon ⁇ , Interferon ⁇ , Interferon ⁇ , Interferon ⁇ -1 , Interferon co-1 , Interleukin 2, 3, 4, 5, 6, 7, 9, 10, 1 1 , 12 a chain, 13, 15, 19, 20, 21 , 22, 23,
  • a modified a-helical bundle cytokine means that the a-helical bundle cytokine has been changed to alter the affinity to the receptor, with as final result that the modified a-helical bundle cytokine has a reduced affinity for the receptor and a consequent reduced biological activity, as compared to the endogenous wild type cytokine that binds normally to the receptor.
  • Such a modification can be a modification that decreases the activity of the normal wild type cytokine, or it can be a modification that increases the affinity of a homologous, non-endogenous a-helical bundle cytokine (such as, but not limited to a mouse ⁇ -helical bundle cytokine, binding to a human ⁇ -helical bundle cytokine receptor).
  • Modifications can be any modification reducing or increasing the activity, known to the person skilled in the art, including but not limited to chemical and/or enzymatic modifications such as pegylation and glycosylation, fusion to other proteins and mutations.
  • said modification is a mutation, even more preferably it is a mutation decreasing the affinity of the- a-helical bundle cytokine.
  • a reduced affinity and a consequent reduced biological activity as used here means that the modified a-helical bundle cytokine has a biological activity of less than 70% of the biological activity of the a-helical bundle cytokine, even more preferably less than 60% of the biological activity of the a-helical bundle cytokine, more preferably less than 50% of the biological activity of the a-helical bundle cytokine, more preferably less than 40% of the biological activity of the a-helical bundle cytokine, more preferably less than 30% of the biological activity of the a-helical bundle cytokine, more preferably less than 20% of the biological activity of the a-helical bundle cytokine, most preferably less than 10% of the biological activity of the a-helical bundle cytokine as compared to the a-helical bundle cytokine that normally binds to the receptor.
  • the modified a-helical bundle cytokine is a mutant of the wild type ⁇ -helical bundle cytokine and the activity is compared with the wild type ⁇ -helical bundle cytokine.
  • the affinity and/or the activity can be measured by any method known to the person skilled in the art.
  • the activity is measured by measuring and quantifying STAT phosphorylation.
  • a preferred embodiment of the invention is a targeting construct, comprising a mutant IFN characterized by reduced affinity for the IFN receptor, and a targeting moiety.
  • IFN can be any IFN, including but not limited to IFNa, IFNp and ⁇ .
  • a mutant IFN as used here can be any mutant form that has a lower affinity for the receptor and as a consequence a lower antiproliferative activity and/or a lower antiviral activity. Indeed, as shown by Piehler et al. (2000), the relative affinity correlates directly with the relative antiproliferative activity and with the relative antiviral activity.
  • the affinity of the mutant IFN to the receptor in comparison to the affinity of the wild type IFN to the receptor can be measured by reflectometric interference spectroscopy under flow through conditions, as described by Brecht et al. (1993).
  • the mutant may be a point mutant, a deletion or an insertion mutant, or a combination thereof.
  • said mutant IFN is obtained by active mutagenesis, such as, but not limited to site directed mutagenesis by polymerase chain reaction amplification.
  • said mutant IFN has a biological activity of less than 70% of the biological activity of the wild type IFN, even more preferably less than 60% of the biological activity of the wild type IFN, more preferably less than 50% of the biological activity of the wild IFN, more preferably less than 40% of the biological activity of the wild IFN, more preferably less than 30% of the biological activity of the wild IFN, more preferably less than 20% of the biological activity of the wild IFN, most preferably less than 10% of the biological activity of the wild type of which it is deduced (i.e. the wild type IFN of which the coding sequence has been mutated to obtain the mutant IFN).
  • Mutant forms of IFN are known to the person skilled in the art.
  • IFNa2 mutants have been listed in Piehler et al. (2000).
  • said IFN is a type I IFN.
  • said mutant is an IFNa, even more preferably said mutant is an IFNa2.
  • said IFNa2 mutant is mutated in one or more amino acids of the region 144- 154, preferably at position 148, 149 and/or 153, even more preferably, said mutant IFNa2 is selected from the group consisting of IFNa2 L153A, IFNa2 R149A and IFNa2 M148A.
  • said mutant is selected from the group consisting of IFNa2 L153A and IFNa2 R149A.
  • said receptor is IFNAR2.
  • said targeting moiety is targeting to a marker expressed on an IFN receptor expressing cell, preferably a cell expressing IFNAR2.
  • said targeting moiety is directed to a tissue specific marker.
  • said tissue is a cancer tissue.
  • Said cancer can be any cancer, including but not limited to B cell lymphoma, lung cancer, breast cancer, colorectal cancer or prostate cancer.
  • said targeting moiety is directed to a marker selected from the group consisting of Her2 and CD20.
  • said targeting moiety is directed to a cell surface marker specific for viral infected cells such as but not limited to influenza M2 protein, LMP1 and EBV proteins).
  • said targeting moiety is directed towards an osteoclast marker such as DC-STAMP or RANK.
  • an osteoclast marker such as DC-STAMP or RANK.
  • IFN- ⁇ plays an important role in bone homeostasis, regulated by RANK and IFNAR coexpressing cells (Abraham et al., 2009).
  • said targeting moiety is directed towards a marker specifically expressed on the surface of an immune cell type on which IFN may regulates activity and/or differentiation.
  • the marker PDL2 specifically expressed on dendritic cells and some immune cells is an example.
  • a targeting moiety can be a protein as a part of a specifically binding protein complex, or any specifically binding protein or protein fragment, known to the person skilled in the art. It includes, but is not limited to carbohydrate binding domains (CBD) (Blake et al, 2006), lectin binding proteins, heavy chain antibodies (hcAb), single domain antibodies (sdAb), minibodies (Tramontano et al., 1994), the variable domain of camelid heavy chain antibodies (VHH), the variable domain of the new antigen receptors (VNAR), affibodies (Nygren et al., 2008), alphabodies (WO2010066740), designed ankyrin-repeat domains (DARPins) (Stumpp et al., 2008), anticalins (Skerra et al., 2008), knottins (Kolmar et al., 2008) and engineered CH2 domains (nanoantibodies; Dimitrov, 2009).
  • CBD carbohydrate binding domains
  • the targeting construct can be any targeting construct known to the person skilled in the art.
  • the targeting moiety may be chemically linked to the mutant interferon, or it may be a recombinant fusion protein.
  • said targeting construct is a recombinant fusion protein.
  • the targeting moiety may be fused directly to the mutant IFN, or it may be fused with the help of a linker fragment.
  • the targeting moiety may be fused at the aminoterminal or at the carboxyterminal end of the mutated IFN; preferably said targeting moiety is fused at the amino-terminal extremity of the mutated IFN molecule.
  • Another aspect of the invention is a targeting construct according to the invention for use as a medicament.
  • Still another aspect of the invention is the use of a targeting construct according to the invention for the manufacture of a medicament to treat cancer.
  • Still another aspect of the invention is the use of a targeting construct according to the invention for the manufacture of a medicament to treat a viral disease.
  • said viral disease may be HIV infection, HBV infection or HCV infection.
  • Another aspect of the invention is a targeting construct according to the invention for use in treatment of cancer.
  • Still another aspect of the invention is a targeting construct according to the invention for use in treatment of a viral disease.
  • said viral disease may be HIV infection, HBV infection or HCV infection.
  • Still another aspect of the invention is a targeting construct according to the invention for use in treatment of diseases involving bone degradation, such as, but not limited to osteoporosis.
  • Still another aspect of the invention is a pharmaceutical composition, comprising a targeting construct according to the invention and a suitable excipient. It is clear for the person skilled in the art that such a pharmaceutical composition can be used alone, or in a combination treatment, such as, but not limited to a combination with chemotherapy.
  • Figure 1 Representation of the structural elements of the nanobody-IFN fusion protein.
  • FIG. 2 Firefly luciferase activity induced by the indicated IFN preparation on HL1 16 cells (panels A and B) or HL1 16 cells expressing the murine leptin receptor (rmLR) (panels C and D).
  • Panels A and C on the one hand and panels B and D on the other hand were generated in two separate experiments. Consequently only vertical comparison (panel A versus panel C or panel B versus panel D) is possible.
  • Figure3 Renilla (light grey) and Firefly (dark grey) luciferase activity induced by the nanobody- IFNa2R149A or by the IFNa2 (7pM) in a 1 :1 coculture of cells expressing the leptin receptor and an IFN-inducible firefly luciferase or in cells expressing an IFN-inducible renilla luciferase but devoid of leptin receptor. Luciferase activities are expressed as a percentage of the luciferase activities induced by 3 nM IFN a2.
  • Figure 4 Activity of the purified constructs targeting the rmLR: A. Quantification of their specific activities on cells expressing the target (HL1 16-mLR) or on cells lacking the target (HL1 16). B. Calculation of the targeting efficiencies of the different constructs.
  • Figure 5 Activity of the construct 4-1 1 -IFNA2-R149A in presence and absence of the unconjugated leptin receptor binding nanobody.
  • HL1 16 cells expressing the rmLR were incubated for 6 hours with either the IFN-a2 (IFNA2) or the IFNA2-R149A fused to the nanobody 4-1 1 (Nanobody-IFNA2-R149A) at their respective EC50 concentration in the presence or absence (control) of a 100-fold molar excess of free 4-1 1 nanobody.
  • IFNA2 IFN-a2
  • Nanobody-IFNA2-R149A nanobody 4-1 1
  • Figure 6 Targeting the mutant IFN using the leptin binding nanobody 4-10.
  • Figure 7 Firefly luciferase activity induced in HL1 16 cells expressing the rmLR by the nanobody-IFNa2R149A in the presence of anti IFNAR1 monoclonal antibody 64G12 (Benoit et al. J. Immunol. 150, 707-716. 1993) or anti IFNAR2 monoclonal antibody MMHAR2 (PBL Interferon Source)
  • Figure 8 Specificity of the targeting to of 4-1 1 -IFNA2-R149A to cells expressing the rmLR.
  • A Cytopathic effect of the EMCV on HL1 16 cells (dark gray symbols) or on HL1 16-mLR (light grey symbols) of parental IFNA2 (upper left panel) or of the 4-1 1 -IFNA2-R149A (lower left panel).
  • B Upper panel: calculated EC50 for antiviral activity; lower panel: calculated targeting efficiencies
  • Figure 9 Specific activities (EC50) of IFNa2 (panel A) and the nanobody-IFNa2R149A (2R5A; panel B) on BXPC3 and BT474 cell lines which express different number of Her2 molecule at their surface (10.9 x 10 3 and 478 x 10 3 , respectively). The ordinate scale of panel A cannot be compared to the ordinate scale of panel B.
  • Figure 10 Targeting of the 1 R59B-IFNA2-Q124R to human Her2 expressing mouse cells. Quantification of the OASL2 mRNA expression in BTG9A cells with and without Her2 expression.
  • Figure 1 1 Targeting of mutant IFNA2 to human Her2 expressing mouse cells, using a single chain antibody. Quantification of the ISG15 mRNA expression in BTG9A cells with and without Her2 expression.
  • Figure 12 Control of the activation of Her2 phosphorylation: Lane 1 3 to 76: no phosphorylated Her2 in extract of BTG9A cells expressing human Her2 treated with different concentration (200 pM for lane 3 to 5, 2 nM for lane 6) and time (lane 3: 5 min, lane 4 and 6: 10 min, lane 5: 30 min.) with the construct 1 R59B-IFNA2-Q124R.
  • Lane 7 and 8 control for the detection of phosphorylated Her2 in the human BT474 cell line. Lane 1 extract of BTG9A cells. Lane2: extract of BTG9A cells expressing human Her2.
  • Figure 13 Targeting the anti PD-L2 122-1 FNA2-Q124R to mouse primary cells endogenously expressing PD-L2.
  • the activation is measured as STAT phosphorylation.
  • the light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population.
  • FIG 14 In vivo targeting of 122-1 FNA2-Q124R to PD-L2 expressing cells. Mice were injected intraperitoneally (IP) or intravenous (IV) with either PBS, a control construct (nanobody against GFP fused to mutant IFNA2-Q124R, indicated as control) or a targeted mutant IFN (targeted to PD-L2, NM22-IFN2-Q124R, indicated as 122-Q124R. The light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population.
  • Figure 15 Dose response curve after IV injection of 122-IFN-Q124R in mice. The light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population.
  • Figure 16 Leptin-dependent growth induced by targeted mutant leptin: the loss in activity of a mutant leptin can be rescued in Ba/F3 cells expressing the human TNFR1 .
  • Upper panels experiment using the H6-leptin construct; lower panel, experiment using the mleptin construct. H6 indicated the his tag (6 xhis).
  • the nanobody 4-1 1 directed against the murine leptin receptor was described in Zabeau et al. (2012), and in the patent WO 2006/053883. Its coding sequence is cloned into the mammalian expression vector pMET7 (Takebe et al., 1988) in fusion with the Slgk leader peptide, the HA tag and albumin. Plasmid name: pMET7 SlgK-HA-4.1 1 -Albumin.
  • the nanobody 4-10 is also described in Zabeau et al. (2012).
  • the anti Her2 nanobodies 1 R59B and 2R5A are described in Vaneycken et al. (201 1 ). They were fused to the human IFNA2-Q124R and to the human IFNA2-R149A in the pMET7 vector. Fusion protein was produced by transfection of 293T cells.
  • the anti PD-L2 nanobody 122 was from Johan Grooten (VI B). It was fused to the human IFNA2-Q124R in the pMET7 vector. The fusion protein was produced by transfection of 293T cells and purified using the HisPur Ni-NTA purification kit (Pierce, Thermo Scientific).
  • the anti TNF nanobody was obtained from Claude Libert (VI B).
  • the anti Her2 ScFv was obtained from Andrea Pluckthun (Worn et al., 1998) It was fused to the human IFNA2-Q124R in the pMET7 vector. The fusion protein was produced by transfection of 293T cells. Control nanobody against GFP was obtained from Katrien Van Impe (University Ghent)
  • IFNa2 and the mutants L153A and R149A which show an IFNAR2 affinity reduced by a factor 10 and 100, respectively, have been described in Roisman et al., (2001 ).
  • IFN coding sequences are cloned in the pT3T7 vector (Stratagene) in fusion with the ybbR tag. Plasmid names: pT7T3ybbR-IFNa2, pT7T3ybbR-IFNa2-L153A, pT7T3ybbR-l FNa2-R149A.
  • the human IFNA2 Q124R has a high affinity for the murine IFNAR1 chain and a low affinity for the murine IFNAR2 chain. (Weber et al., 1987)
  • the coding sequence of the IFNa2, wild type, L153A and R149A were synthesized by PCR from the corresponding pT3T7ybbR IFNa2 plasmids using the Expand High Fidelity PCR system from Roche Diagnostics and the following primers: Forward: 5'GGGGGGTCCGGACCATCACCATCACCATCACCATCACCCTGCTTCTCCCGCC TCCCCAGCATCACCTGCCAGCCCAGCAAGTGATAGCCTGGAATTTATTGC3', Reverse: 5' CGTCTAG ATCATTC CTTACTTCTTAAAC3 ' .
  • This PCR introduces a His tag and a series of 5 Proline - Alanine - Serine (PAS) repeats at the amino terminal extremity of the IFNs.
  • the PCR products were digested with BspEI and Xbal and cloned into BspEI-Xbal digested pMET7 SlgK-HA-4.1 1 -Albumin vector to obtain pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2, pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2-L153A and pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2- R149A.
  • HEK293T cells were grown in DMEM supplemented with 10% FCS. They were transfected with pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2, pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2- L153A pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2-R149A, pMET7 SlgK-HA-2R5A-His-PAS- ybbr-l FNA2-R149A, pMET7 SlgK-HA-1 R59B-His-PAS-ybbr-IFNA2-Q124R, pMET7 SlgK-HA- 4D5-His-PAS-ybbr-IFNA2-Q124R or pMET7 SlgK-HA-122-His-PAS-ybbr-IFNA2-Q124R using lipo
  • sequences encoding the different nanobody-IFN fusions where subcloned into the baculovirus transfer plasmid pBAC-3 (Novagen). Proteins were produced by insect cells using the BacVector kit (Novagen) and purified to homogeneity using the HisPur Ni-NTA purification kit (Pierce, Thermo Scientific) and gel filtration. Protein concentrations were measured by absorbance at 280 nm. I FN reporter cell lines
  • 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 HL1 16 cells were co- transfected with an expression vector encoding the short isoform of the murine leptin receptor (pMET7 mLRsh-FLAG, Eyckerman et al., 1999) and pSV2neo (Southern and Berg 1982). Stable transfected clones were isolated in G418 containing medium.
  • the clone 10 was selected after analysis of the surface expression level of the murine leptin receptor by FACS, using the biotinylated anti-mouse leptin receptor antibody BAF497 from R&D and streptavidin-APC (BD bioscience)
  • HT1080 cells were cotransfected with p6-16-RL, a plasmid encoding the Renilla luciferase (from pRL-null, Promega) controlled by the IFN-inducible 6-16 promoter (from p1.8gpt-5, Pellegrini et al. 1989), pBB3 (Bourachot et al. 1982) and salmon sperm DNA (Sigma).
  • Stable transfected clones were isolated in HAT containing medium. The clone 4 was selected for a high level of renilla luciferase activity induction upon IFN induction.
  • the human pancreatic carcinoma BXPC3 (Tan et al., 1986; ATCC: CRL 1687) and breast cancer BT474 (Lasfargues et al., 1979; ATCC: HTB-20) cell lines were obtained from ATCC.
  • the mouse BTG9A cells were described in Uze et al. (1990).
  • IFN specific activities were measured by quantifying the luciferase activity induced in HL1 16 cells and on the HL1 16 clone 10 expressing the rmLR.
  • the EC50 were calculated using nonlinear data regression with GraphPad Prism software.
  • Luciferase activities were determined on a Berthold centra LB960 luminometer using either the Firefly Luciferase Assay System or the Dual-Luciferase Reporter Assay System from Promega after 6hr IFN stimulation.
  • the expression of the interferon inducible gene 6-16 was quantified by RT-PCR relatively to GAPDH or ⁇ -actin.
  • Cells were treated with targeted or control IFN for 4 hr.
  • Total RNA was purified with RNeasy columns (Qiagen).
  • Reverse transcriptions were primed with random primers and performed using Moloney murine leukemia virus reverse transcriptase (Invitrogen).
  • Quantitative real-time PCR qRT-PCR was performed using a Light Cycler as described (Coccia et al. 2004).
  • the transfection culture medium was assayed on murine BTG9A and BTG9A cells expressing human Her2 for expression of the OASL2 gene relatively to the expression of the ⁇ actin gene by quantitative RT-PCR using a Light Cycler (Roche) and the following primers: OASL2 forward: CAC-GAC-TGT-AGG-CCC-CAG-CGA; OASL2 reverse: AGC-AGC-TGT- CTC-TCC-CCT-CCG; 3actin forward: AGA-GGG-AAA-TCG-TGC-GTG-AC; 3actin reverse: CAA-TAG-TGA-TGA-CCT-GGC-CGT.
  • OASL2 forward CAC-GAC-TGT-AGG-CCC-CAG-CGA
  • OASL2 reverse AGC-AGC-TGT- CTC-TCC-CCT-CCG
  • 3actin forward AGA-GGG-AAA-TCG-TGC-GTG-AC
  • ISG15 forward GAG-CTA-GAG-CCT-GCA-GCA-AT
  • ISG15 reverse TTC-TGG-GCA-ATC- TGC-TTC-TT.
  • the antiviral assay was performed using the EMC virus and scoring the virus replication dependent cytopathic effect as described in Stewart (1979)
  • BTG9A cells expressing human Her2 were treated with 200 pM to 2 nM of 1 R59B-IFNA2- Q124R for 10 to 30 min. Cells were lysed in RIPA, and analysed by western blot on an Odyssey Fc (Licor Bioscience) after 7% SDS-PAGE (40 ⁇ g lane). Phopho-Her2 was detected with the anti Her2 Y-P 1248 (Upstate #06-229) and the Goat anti rabbit secondary antibody IRDye 680 (Licor Bioscience #926-32221 ).
  • STAT1 phosporylated on Y701 were detected by FACS using the STAT1 -PY701 (PE) (Beckton Dickinston #612564) and the manufacturer instruction for the PhosFlow technology.
  • the sequence of the targeted leptin constructs is given in Figure 17.
  • the L86 which is indicated is the amino acid that is mutated either to S or N.
  • Example 1 The nanobody-interferon fusion proteins
  • Figure 1 shows a schematic representation of the nanobody-IFN fusion proteins constructed with either IFNa2 wild type or the L153A and R149A mutants.
  • Example 2 IFN activity of the nanobody-IFN fusion proteins is targeted toward murine leptin receptor expressing cells
  • the three nanobody fusion proteins with IFNa2 WT, IFNa2 L153A or R149A were assayed on both HL1 16 and HL1 16-mLR-clone 10 cells which express the murine leptin receptor.
  • IFNa2 alone was also assayed in this assay system in order to check that the two cell clones do not differ in their IFN responsiveness. Indeed both HL1 16 and HL1 16-mLR-clone 10 cells are equally sensitive to this IFN (Fig 2A and 2C, black symbols).
  • the IFN activity of the three nanobody-IFN fusion proteins is however dramatically increased in cells expressing the leptin receptor compared to parental HL1 16 cells (compare Fig. 2A with Fig. 2C and Fig. 2B with Fig. 2D).
  • the nanobody- IFNa2R149A was assayed on a coculture of HL1 16-ml_R-clone10 and HT1080-6-16 renilla luciferase clone4. Both cell types will express luciferase activity in response to IFN stimulation but cells expressing the target of the nanobody will display a firefly luciferase activity whereas cells devoid of leptin receptor will display a renilla luciferase activity.
  • Fig. 3 shows clearly that the renilla luciferase activity is not induced upon stimulation of the co-culture with the nanobody- IFNa2R149A, indicating that the targeted IFN activity is delivered only on cells expressing the antigen recognized by the nanobody.
  • the efficacy of the targeting is further illustrated by comparing the activity of wild type and two types of mutant IFN (L153A and R149A) when added to HL1 16 expressing or not expressing the murine leptin receptor that is used for the targeting.
  • the results clearly show that the activity of the mutants is higher when the construct is targeted, and that the effect of targeting for the mutant is bigger than for wild type.
  • HL1 16 cells expressing the rmLR were incubated for 6 hours with either the IFN-a2 (indicated as IFNA2) or the IFNA2-R149A fused to the nanobody 4-1 1 (Nanobody-IFNA2-R149A) at their respective EC50 concentration in the presence or absence (control) of a 100-fold molar excess of free 4-1 1 nanobody.
  • Cells were lysed and the IFN-induced luciferase activities were measured.
  • the non-targeted IFN is not inhibited by the free nanobody, while the targeted construct is strongly inhibited, showing the specific effect of the targeting.
  • the targeting to the leptin receptor is independent of the epitope on the receptor: using the anti-leptin receptor nanobody 4-10 (Zabeau et al., 2012) which recognizes a different domain on the receptor than the nanobody 4-1 1 , a similar activation can be obtained using a targeted mutant IFN ( Figure 6).
  • Example 3 The IFN activity of the nanobody-IFN fusion proteins on cells expressing the leptin receptor is mediated by both IFN receptor chains
  • HL1 16 cells expressing the murine leptin receptor were pretreated with neutralizing antibodies against IFNAR1 or IFNAR2, and then stimulated with the nanobody-IFNA2-R149A fusion protein.
  • the activity of the IFN-induced luciferase was measured.
  • the figure 7 shows that both anti IFNAR1 and anti IFNAR2 neutralizing antibodies inhibit the IFN activity of the nanobody-IFNA2-R149A.
  • Example 4 Target-specific induction of antiviral activity by 4-11 -IFNA2-R149A in cells expressing the murine leptin receptor
  • Antiviral activity is an integrated part of the IFN response, implying the expression of several genes. Therefore, the antiviral activity on rmLR expressing cell was controlled, after targeting the mutant R149A IFN using the anti-leptin receptor antibody 4-1 1.
  • the results are summarized in Figure 8. The activity was measured as the cytopathic effect on HL1 16 cells, with or without leptin receptor expression.
  • the specific antiviral activity of the 4-1 1 -IFNA2- R149A nanobody-IFN fusion protein is 716-fold higher when assayed on leptin receptor expressing cells compared to HL1 16 cells.
  • Example 5 Targeting of IFN activity on Her2 expressing cells
  • Figure 9 shows the EC 50 determination of IFNA2 activity (Panel A) and 2R5A-IFNA2-R149A activity (Panel B) for the induction of the IFN-inducible gene 6-16 on BXPC3 and BT474 cell lines.
  • Panel A shows that BXPC3 and BT474 cell lines exhibit the same sensitivity to IFN-a2.
  • Panel B shows that the 2R5A-IFNA2-R149A nanobody-IFN fusion protein is much more potent on the BT474 cell line which expresses 40-fold more Her2 molecule than BXPC3.
  • the concept which consists to target type I IFN activity on cells expressing a specific cell surface antigen can be extended to untransfected human cells expressing another cell surface molecule from a different structural family, at a level naturally found in several types of breast carcinoma.
  • Example 6 Targeting of mutant IFNA2-Q149R to mouse cells expressing human Her2 Mutant human IFNA2 Q149R was targeted to murine cells, expressing the human Her2, using the nanobody 1 R59B in the 1 R59B-IFNA2-Q124R.
  • the IFNA2 Q124R has a high affinity for the murine IFNAR1 chain and a low activity for the murine IFNAR2 chain (Weber et al., 1987).
  • the induction by IFN was measured as expression of the OASL2 messenger RNA, by RT- QPCR. The results are shown in Figure 10. There is clearly a targeting-specific induction in the Her2 expressing cells, whereas there is no significant expression detected in untransfected BTG9A cells.
  • Example 7 The construct 1 R59B-IFNA2-Q124R does not activate the phosphorylation of Her2
  • Cells from a mouse peritoneal cavity were isolated and treated in vitro with Nb122-IFNA2- Q124R or natural mlFNa/ ⁇ for 30 min. Cells were, fixed, permeabilized, labelled with antibodies against PD-L2 (APC) (BD #560086) and STAT1 -PY701 (PE) (BD #612564) and analysed by FACS.
  • APC PD-L2
  • PE STAT1 -PY701
  • the PD-L2 positive cell population represents 20% of the total cell population present in the mouse peritoneal cavity.
  • Example 9 In vivo injection of 122-IFNA2-Q124R construct induces an IFN response only in PD-L2 -expressing cells
  • mice were injected (IP or IV) with either PBS, Nb122-IFNA2-Q124R or a control Nb (against GFP) fused to IFNA2-Q124R.
  • 30 min post injection mice were killed, cells from the peritoneal cavity were recovered by washing the peritoneal cavity with PBS, fixed (PhosLow Fix buffer I BD # 557870), permeabilized (PhosFlow Perm buffer III, BD #558050), labelled with Abs against PD-L2 (APC) (BD #560086) and STAT1 -PY701 (PE) (BD #612564) and analysed by FACS. The results are shown in Figure 14.
  • STAT1 -P coincides in PD-L2 positive and negative cells treated with either PBS or control nanobody. However, a clear induction in STAT1 -P (only in the PDL2 positive cell population) can be seen when the mice are injected with the targeted mutant IFN.
  • STAT1 -P was checked in mice, iv injected with different doses of natural mouse IFN (10 000, 100 000 or 1 000 000 units), and no difference in STAT1 -P could be detected between the PD-L2 positive and PD-L2 negative cells.
  • Figure 15 shows a similar dose response curve after iv injection of the Nb122-IFNA2-Q124R construct. A shift in STAT1 -P in the PD-L2 expressing cells can be noticed even at the lowest dose of 64ng.
  • Example 10 Targeting of mutant leptin to the leptin receptor, using a truncated TNFa receptor
  • Ba/F3 cells are growth-dependent on IL-3. After transfection with the rmLR, Ba/F3 cells also proliferate with leptin. Leptin mutants with reduced affinity for their receptor are less potent in inducing and sustaining proliferation of Ba/F3-ml_R cells. Leptin mutant L86S has a moderate and mutant L86N has a strong reduction in affinity and hence a moderate and strong reduced capacity to induce proliferation respectively.
  • Chimeric proteins consisting of leptin and a nanobody against human TNFR1 will bind to cells carrying the rmLR and to cells carrying the hTNFRI .
  • Chimeric proteins with leptin mutants L86S and L86N have reduced affinity for the LR but retain their affinity for the hTNFRL
  • Chimeric proteins were produced by transient transfection of Hek293T cells with expression plasmids. Supernatant was 0.45 ⁇ filtered and serially diluted in 96-well plates for the assay. A serial dilution of purified recombinant leptin was used as a reference. 3000 to 10000 cells were plated per well and proliferation was measured by staining with XTT four or five days later. OD was measured at 450 nm. The results are shown in Figure 16, for two experiments using a different leptin construct (see Figure 17).

Abstract

The present invention relates to a modified α-helical bundle cytokine, with reduced activity via a α-helical bundle cytokine receptor, wherein said α-helical bundle cytokine is specifically delivered to target cells. Preferably, said α-helical bundle cytokine is a mutant, more preferably it is a mutant interferon, with low affinity to the interferon receptor, wherein said mutant interferon is specifically delivered to target cells. The targeting is realized by fusion of the modified α-helical bundle cytokine to a targeting moiety, preferably an antibody. The invention relates further to the use of such targeted modified α-helical bundle cytokine to treat diseases. A preferred embodiment is the use of a targeted mutant interferon, to treat diseases, preferably viral diseases and tumors.

Description

TARGETED MUTANT ALPHA-HELICAL BUNDLE CYTOKINES
The present invention relates to a modified a-helical bundle cytokine, with reduced activity via a a-helical bundle cytokine receptor, wherein said a-helical bundle cytokine is specifically delivered to target cells. Preferably, said a-helical bundle cytokine is a mutant, more preferably it is a mutant interferon, with low affinity to the interferon receptor, wherein said mutant interferon is specifically delivered to target cells. The targeting is realized by fusion of the modified a-helical bundle cytokine to a targeting moiety, preferably an antibody. The invention relates further to the use of such targeted modified a-helical bundle cytokine to treat diseases. A preferred embodiment is the use of a targeted mutant interferon, to treat diseases, preferably viral diseases and tumors.
Cytokines are small proteins that play an important role in intercellular communication. Cytokines can be classified based on their structure, the largest group being the four-a-helix bundle family. This family can, based on the use of receptors, further be divided in the interferon (IFN) and interleukin (IL)-2, -3, -10 and -12 subfamilies. The a helical bundle cytokines are important as possible biopharmaceutical for treatment of human diseases; as a non-limiting example erythropoietin is used for treatment of anemia or red blood cell deficiency, somatotropin for treatment of growth hormone deficiency and interleukin-2 in the treatment of cancer.
Within the a helical bundle cytokines, type I IFNs belong to a cytokine family having important biological functions. In human, there are 17 different type I IFNs (13α, β, ε, κ, ω) which signal through an ubiquitously expressed cell surface receptor composed of two chains IFNAR1 and IFNAR2. The assembling of the IFN-receptor complex initiates the activation of several signal transduction pathways that, depending of the cell type, modify cellular differentiation and/or functions.
By acting on virtually every cell type, type I I FN is able to prevent productive viral infection. In addition, it exhibits marked antiangiogenic and proapoptotic effects. Type I IFNs are also deeply implicated in the regulation of several functions of the innate and adaptive immunity as well as on bone homeostasis. It acts particularly on the activation/differentiation of dendritic cells and osteoclasts. The type I IFN system is in fact critically important for the health of mammals.
Preclinical studies in mice have established a remarkable efficacy of type I IFN for the treatment of both viral or tumor diseases. Noteworthy, mice cured of an experimental tumor by IFN treatment have been found immunized against the initial tumor, suggesting that IFN acts not only to engage the processes of tumor rejection but also to break the immune tolerance against the tumor. Based on these studies, IFNa was approved in clinic for the treatment of both viral infection and cancer. More recently, IFN3 was shown to be effective in relapsing- remitting multiple sclerosis and was also approved for this pathology. Unfortunately, the clinical efficacy of IFN was often found disappointing and today other therapeutic strategies such as specific antiviral compounds, chemotherapies and monoclonal antibodies have, when possible, largely supplanted IFN broad application. Today, IFN is the first line therapeutic choice for only HBV and HCV chronic infections and for a limited number of tumors.
The efficacy of type I IFN in clinical practice is limited by ineffective dosing due to significant systemic toxicity and side effects, including flu-like syndrome, depression, hepatotoxicity, autoimmune disease, thyroid dysfunction and weight loss. It would thus be highly worthwhile to target IFN activity toward only the cellular population which should be treated with IFN (e.g. infected organ or tumor mass) or activated by IFN (e.g. subsets of immune cells).
In order to solve or limit the systemic toxicity of cytokines, specific targeting of cytokines by antibody-cytokine fusion proteins has been proposed (Ortiz-Sanchez et al., 2008). Rossi et al. (2009) specifically discloses CD20-targeted tetrameric IFNa, and its use in B-cell lymphoma therapy. However, the fusion maintains its biological activity, and is even more active than commercial pegylated IFN, which means that the unwanted side effects in human treatment would still be present, or would even be more severe. WO2009039409 discloses targeted IFN and its apoptotic and anti-tumor activities. The patent application discloses the fusion of an antibody as targeting moiety with wild type IFN, but also with mutated IFN. However, it is stated that the IFN fragment should retain its endogeneous activity at a level of at least 80%, or even at a higher level than wild type IFN. Also in this case, the fusion is retaining the unwanted side effects of the wild type.
Surprisingly we found that a modified a-helical bundle cytokine, with a decreased affinity for the a-helical bundle cytokine receptor and a consequent decreased specific bioactivity can be fused to a targeting moiety, wherein the bioactivity is restored towards the targeted cells, but not towards cells that are not targeted by the construct. Such construct has the advantage over the art of having less side effects, especially a lower systemic toxicity, while retaining the bioactivity against the target cells.
A first aspect of the invention is a targeting construct, comprising modified α-helical bundle cytokine, characterized by a reduced affinity for the a-helical bundle cytokine receptor, and a targeting moiety, α-helical bundle cytokines are known to the person skilled in the art, and include, but are not limited Cardiotrophin-like cytokine NNT-1 , Ciliary neurothrophic factor, Macrophage colony stimulating factor, Granulocyte-macrophage colony stimulating factor, Granulocyte colony stimulating factor, Cardiotrophin-1 , Erythropoietin, FLT3 ligand, Somatotropin, Interferon a-1 , 2, 4, 5, 6, 7, 8, 10, 13, 14, 16, 17, 21 ,, Interferon β, Interferon γ, Interferon κ, Interferon ε, Interferon τ-1 , Interferon co-1 , Interleukin 2, 3, 4, 5, 6, 7, 9, 10, 1 1 , 12 a chain, 13, 15, 19, 20, 21 , 22, 23, 24, 26, 27, 28A, 29, 31 , Stem cell factor, Leptin, Leukemia inhibitor factor, Oncostatin M, Prolactin, and Thrombopoietin. For a review on α-helical bundle cytokines, see Conklin (2004). A modified a-helical bundle cytokine means that the a-helical bundle cytokine has been changed to alter the affinity to the receptor, with as final result that the modified a-helical bundle cytokine has a reduced affinity for the receptor and a consequent reduced biological activity, as compared to the endogenous wild type cytokine that binds normally to the receptor. Such a modification can be a modification that decreases the activity of the normal wild type cytokine, or it can be a modification that increases the affinity of a homologous, non-endogenous a-helical bundle cytokine (such as, but not limited to a mouse α-helical bundle cytokine, binding to a human α-helical bundle cytokine receptor). Modifications can be any modification reducing or increasing the activity, known to the person skilled in the art, including but not limited to chemical and/or enzymatic modifications such as pegylation and glycosylation, fusion to other proteins and mutations. Preferably said modification is a mutation, even more preferably it is a mutation decreasing the affinity of the- a-helical bundle cytokine. A reduced affinity and a consequent reduced biological activity as used here means that the modified a-helical bundle cytokine has a biological activity of less than 70% of the biological activity of the a-helical bundle cytokine, even more preferably less than 60% of the biological activity of the a-helical bundle cytokine, more preferably less than 50% of the biological activity of the a-helical bundle cytokine, more preferably less than 40% of the biological activity of the a-helical bundle cytokine, more preferably less than 30% of the biological activity of the a-helical bundle cytokine, more preferably less than 20% of the biological activity of the a-helical bundle cytokine, most preferably less than 10% of the biological activity of the a-helical bundle cytokine as compared to the a-helical bundle cytokine that normally binds to the receptor. Preferably, the modified a-helical bundle cytokine is a mutant of the wild type α-helical bundle cytokine and the activity is compared with the wild type α-helical bundle cytokine. The affinity and/or the activity can be measured by any method known to the person skilled in the art. Preferably, the activity is measured by measuring and quantifying STAT phosphorylation.
A preferred embodiment of the invention is a targeting construct, comprising a mutant IFN characterized by reduced affinity for the IFN receptor, and a targeting moiety. IFN can be any IFN, including but not limited to IFNa, IFNp and ω. A mutant IFN as used here can be any mutant form that has a lower affinity for the receptor and as a consequence a lower antiproliferative activity and/or a lower antiviral activity. Indeed, as shown by Piehler et al. (2000), the relative affinity correlates directly with the relative antiproliferative activity and with the relative antiviral activity. The affinity of the mutant IFN to the receptor, in comparison to the affinity of the wild type IFN to the receptor can be measured by reflectometric interference spectroscopy under flow through conditions, as described by Brecht et al. (1993). The mutant may be a point mutant, a deletion or an insertion mutant, or a combination thereof. Preferably, said mutant IFN is obtained by active mutagenesis, such as, but not limited to site directed mutagenesis by polymerase chain reaction amplification. Preferably, said mutant IFN has a biological activity of less than 70% of the biological activity of the wild type IFN, even more preferably less than 60% of the biological activity of the wild type IFN, more preferably less than 50% of the biological activity of the wild IFN, more preferably less than 40% of the biological activity of the wild IFN, more preferably less than 30% of the biological activity of the wild IFN, more preferably less than 20% of the biological activity of the wild IFN, most preferably less than 10% of the biological activity of the wild type of which it is deduced (i.e. the wild type IFN of which the coding sequence has been mutated to obtain the mutant IFN). Mutant forms of IFN are known to the person skilled in the art. As a non-limiting example, IFNa2 mutants have been listed in Piehler et al. (2000). Preferably, said IFN is a type I IFN. Even more preferably, said mutant is an IFNa, even more preferably said mutant is an IFNa2. More preferably, said IFNa2 mutant is mutated in one or more amino acids of the region 144- 154, preferably at position 148, 149 and/or 153, even more preferably, said mutant IFNa2 is selected from the group consisting of IFNa2 L153A, IFNa2 R149A and IFNa2 M148A. Most preferably said mutant is selected from the group consisting of IFNa2 L153A and IFNa2 R149A.
Preferably, said receptor is IFNAR2.
Preferably, said targeting moiety is targeting to a marker expressed on an IFN receptor expressing cell, preferably a cell expressing IFNAR2. In one preferred embodiment, said targeting moiety is directed to a tissue specific marker. Preferably, said tissue is a cancer tissue. Said cancer can be any cancer, including but not limited to B cell lymphoma, lung cancer, breast cancer, colorectal cancer or prostate cancer. In another preferred embodiment, said targeting moiety is directed to a marker selected from the group consisting of Her2 and CD20. In still another preferred embodiment, said targeting moiety is directed to a cell surface marker specific for viral infected cells such as but not limited to influenza M2 protein, LMP1 and EBV proteins). In still another embodiment said targeting moiety is directed towards an osteoclast marker such as DC-STAMP or RANK. Indeed, it is know that IFN-β plays an important role in bone homeostasis, regulated by RANK and IFNAR coexpressing cells (Abraham et al., 2009). In still another embodiment said targeting moiety is directed towards a marker specifically expressed on the surface of an immune cell type on which IFN may regulates activity and/or differentiation. The marker PDL2 specifically expressed on dendritic cells and some immune cells is an example.
A targeting moiety, as used here, can be a protein as a part of a specifically binding protein complex, or any specifically binding protein or protein fragment, known to the person skilled in the art. It includes, but is not limited to carbohydrate binding domains (CBD) (Blake et al, 2006), lectin binding proteins, heavy chain antibodies (hcAb), single domain antibodies (sdAb), minibodies (Tramontano et al., 1994), the variable domain of camelid heavy chain antibodies (VHH), the variable domain of the new antigen receptors (VNAR), affibodies (Nygren et al., 2008), alphabodies (WO2010066740), designed ankyrin-repeat domains (DARPins) (Stumpp et al., 2008), anticalins (Skerra et al., 2008), knottins (Kolmar et al., 2008) and engineered CH2 domains (nanoantibodies; Dimitrov, 2009). Preferably, said targeting moiety consists of a single polypeptide chain and is not post-translationally modified. Even more preferably, said targeting moiety is a nanobody.
The targeting construct can be any targeting construct known to the person skilled in the art. As a non-limiting example, the targeting moiety may be chemically linked to the mutant interferon, or it may be a recombinant fusion protein. Preferably, said targeting construct is a recombinant fusion protein. The targeting moiety may be fused directly to the mutant IFN, or it may be fused with the help of a linker fragment. The targeting moiety may be fused at the aminoterminal or at the carboxyterminal end of the mutated IFN; preferably said targeting moiety is fused at the amino-terminal extremity of the mutated IFN molecule.
Another aspect of the invention is a targeting construct according to the invention for use as a medicament.
Still another aspect of the invention is the use of a targeting construct according to the invention for the manufacture of a medicament to treat cancer.
Still another aspect of the invention is the use of a targeting construct according to the invention for the manufacture of a medicament to treat a viral disease. As a non-limiting example said viral disease may be HIV infection, HBV infection or HCV infection.
Another aspect of the invention is a targeting construct according to the invention for use in treatment of cancer.
Still another aspect of the invention is a targeting construct according to the invention for use in treatment of a viral disease. As a non-limiting example said viral disease may be HIV infection, HBV infection or HCV infection.
Still another aspect of the invention is a targeting construct according to the invention for use in treatment of diseases involving bone degradation, such as, but not limited to osteoporosis.
Still another aspect of the invention is a pharmaceutical composition, comprising a targeting construct according to the invention and a suitable excipient. It is clear for the person skilled in the art that such a pharmaceutical composition can be used alone, or in a combination treatment, such as, but not limited to a combination with chemotherapy.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 : Representation of the structural elements of the nanobody-IFN fusion protein.
Figure 2: Firefly luciferase activity induced by the indicated IFN preparation on HL1 16 cells (panels A and B) or HL1 16 cells expressing the murine leptin receptor (rmLR) (panels C and D). Panels A and C on the one hand and panels B and D on the other hand were generated in two separate experiments. Consequently only vertical comparison (panel A versus panel C or panel B versus panel D) is possible.
Figure3: Renilla (light grey) and Firefly (dark grey) luciferase activity induced by the nanobody- IFNa2R149A or by the IFNa2 (7pM) in a 1 :1 coculture of cells expressing the leptin receptor and an IFN-inducible firefly luciferase or in cells expressing an IFN-inducible renilla luciferase but devoid of leptin receptor. Luciferase activities are expressed as a percentage of the luciferase activities induced by 3 nM IFN a2.
Figure 4: Activity of the purified constructs targeting the rmLR: A. Quantification of their specific activities on cells expressing the target (HL1 16-mLR) or on cells lacking the target (HL1 16). B. Calculation of the targeting efficiencies of the different constructs.
Figure 5: Activity of the construct 4-1 1 -IFNA2-R149A in presence and absence of the unconjugated leptin receptor binding nanobody. HL1 16 cells expressing the rmLR were incubated for 6 hours with either the IFN-a2 (IFNA2) or the IFNA2-R149A fused to the nanobody 4-1 1 (Nanobody-IFNA2-R149A) at their respective EC50 concentration in the presence or absence (control) of a 100-fold molar excess of free 4-1 1 nanobody.
Figure 6: Targeting the mutant IFN using the leptin binding nanobody 4-10.
Figure 7: Firefly luciferase activity induced in HL1 16 cells expressing the rmLR by the nanobody-IFNa2R149A in the presence of anti IFNAR1 monoclonal antibody 64G12 (Benoit et al. J. Immunol. 150, 707-716. 1993) or anti IFNAR2 monoclonal antibody MMHAR2 (PBL Interferon Source)
Figure 8: Specificity of the targeting to of 4-1 1 -IFNA2-R149A to cells expressing the rmLR.
A: Cytopathic effect of the EMCV on HL1 16 cells (dark gray symbols) or on HL1 16-mLR (light grey symbols) of parental IFNA2 (upper left panel) or of the 4-1 1 -IFNA2-R149A (lower left panel). B: Upper panel: calculated EC50 for antiviral activity; lower panel: calculated targeting efficiencies Figure 9: Specific activities (EC50) of IFNa2 (panel A) and the nanobody-IFNa2R149A (2R5A; panel B) on BXPC3 and BT474 cell lines which express different number of Her2 molecule at their surface (10.9 x 103 and 478 x 103, respectively). The ordinate scale of panel A cannot be compared to the ordinate scale of panel B. Figure 10: Targeting of the 1 R59B-IFNA2-Q124R to human Her2 expressing mouse cells. Quantification of the OASL2 mRNA expression in BTG9A cells with and without Her2 expression.
Figure 1 1 : Targeting of mutant IFNA2 to human Her2 expressing mouse cells, using a single chain antibody. Quantification of the ISG15 mRNA expression in BTG9A cells with and without Her2 expression.
Figure 12: Control of the activation of Her2 phosphorylation: Lane 1 3 to 76: no phosphorylated Her2 in extract of BTG9A cells expressing human Her2 treated with different concentration (200 pM for lane 3 to 5, 2 nM for lane 6) and time (lane 3: 5 min, lane 4 and 6: 10 min, lane 5: 30 min.) with the construct 1 R59B-IFNA2-Q124R.
Lane 7 and 8: control for the detection of phosphorylated Her2 in the human BT474 cell line. Lane 1 extract of BTG9A cells. Lane2: extract of BTG9A cells expressing human Her2.
Figure 13: Targeting the anti PD-L2 122-1 FNA2-Q124R to mouse primary cells endogenously expressing PD-L2. The activation is measured as STAT phosphorylation. The light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population.
Figure 14: In vivo targeting of 122-1 FNA2-Q124R to PD-L2 expressing cells. Mice were injected intraperitoneally (IP) or intravenous (IV) with either PBS, a control construct (nanobody against GFP fused to mutant IFNA2-Q124R, indicated as control) or a targeted mutant IFN (targeted to PD-L2, NM22-IFN2-Q124R, indicated as 122-Q124R. The light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population. Figure 15: Dose response curve after IV injection of 122-IFN-Q124R in mice. The light gray area represents the PD-L2 negative cell population; the dark gray area represents the PD-L2 positive population.
Figure 16: Leptin-dependent growth induced by targeted mutant leptin: the loss in activity of a mutant leptin can be rescued in Ba/F3 cells expressing the human TNFR1 . Upper panels: experiment using the H6-leptin construct; lower panel, experiment using the mleptin construct. H6 indicated the his tag (6 xhis).
Figure 17: construction of the targeted leptin constructs
EXAMPLES Materials & Methods to the examples
Nanobodies and ScFv
The nanobody 4-1 1 directed against the murine leptin receptor was described in Zabeau et al. (2012), and in the patent WO 2006/053883. Its coding sequence is cloned into the mammalian expression vector pMET7 (Takebe et al., 1988) in fusion with the Slgk leader peptide, the HA tag and albumin. Plasmid name: pMET7 SlgK-HA-4.1 1 -Albumin.
The nanobody 4-10 is also described in Zabeau et al. (2012).
The anti Her2 nanobodies 1 R59B and 2R5A are described in Vaneycken et al. (201 1 ). They were fused to the human IFNA2-Q124R and to the human IFNA2-R149A in the pMET7 vector. Fusion protein was produced by transfection of 293T cells.
The anti PD-L2 nanobody 122 was from Johan Grooten (VI B). It was fused to the human IFNA2-Q124R in the pMET7 vector. The fusion protein was produced by transfection of 293T cells and purified using the HisPur Ni-NTA purification kit (Pierce, Thermo Scientific).
The anti TNF nanobody was obtained from Claude Libert (VI B).
The anti Her2 ScFv was obtained from Andrea Pluckthun (Worn et al., 1998) It was fused to the human IFNA2-Q124R in the pMET7 vector. The fusion protein was produced by transfection of 293T cells. Control nanobody against GFP was obtained from Katrien Van Impe (University Ghent)
Interferons The IFNa2 and the mutants L153A and R149A which show an IFNAR2 affinity reduced by a factor 10 and 100, respectively, have been described in Roisman et al., (2001 ). IFN coding sequences are cloned in the pT3T7 vector (Stratagene) in fusion with the ybbR tag. Plasmid names: pT7T3ybbR-IFNa2, pT7T3ybbR-IFNa2-L153A, pT7T3ybbR-l FNa2-R149A.
The human IFNA2 Q124R has a high affinity for the murine IFNAR1 chain and a low affinity for the murine IFNAR2 chain. (Weber et al., 1987)
Nanobody-IFN fusion construction
The coding sequence of the IFNa2, wild type, L153A and R149A were synthesized by PCR from the corresponding pT3T7ybbR IFNa2 plasmids using the Expand High Fidelity PCR system from Roche Diagnostics and the following primers: Forward: 5'GGGGGGTCCGGACCATCACCATCACCATCACCATCACCATCACCCTGCTTCTCCCGCC TCCCCAGCATCACCTGCCAGCCCAGCAAGTGATAGCCTGGAATTTATTGC3', Reverse: 5' CGTCTAG ATCATTC CTTACTTCTTAAAC3 ' . This PCR introduces a His tag and a series of 5 Proline - Alanine - Serine (PAS) repeats at the amino terminal extremity of the IFNs. The PCR products were digested with BspEI and Xbal and cloned into BspEI-Xbal digested pMET7 SlgK-HA-4.1 1 -Albumin vector to obtain pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2, pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2-L153A and pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2- R149A.
In a similar way the human mutant Q124R was fused to the 1 R59B nanobody and to the anti- PD-L2 nanobody.
Production of the nanobody-IFN fusion protein
HEK293T cells were grown in DMEM supplemented with 10% FCS. They were transfected with pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2, pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2- L153A pMET7 SlgK-HA-4.1 1 -His-PAS-ybbr-IFNA2-R149A, pMET7 SlgK-HA-2R5A-His-PAS- ybbr-l FNA2-R149A, pMET7 SlgK-HA-1 R59B-His-PAS-ybbr-IFNA2-Q124R, pMET7 SlgK-HA- 4D5-His-PAS-ybbr-IFNA2-Q124R or pMET7 SlgK-HA-122-His-PAS-ybbr-IFNA2-Q124R using lipofectamin (Invitrogen). 48 hours after the transfection culture mediums were harvested and stored at -20°C.
Alternatively, sequences encoding the different nanobody-IFN fusions where subcloned into the baculovirus transfer plasmid pBAC-3 (Novagen). Proteins were produced by insect cells using the BacVector kit (Novagen) and purified to homogeneity using the HisPur Ni-NTA purification kit (Pierce, Thermo Scientific) and gel filtration. Protein concentrations were measured by absorbance at 280 nm. I FN reporter cell lines
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 HL1 16 cells were co- transfected with an expression vector encoding the short isoform of the murine leptin receptor (pMET7 mLRsh-FLAG, Eyckerman et al., 1999) and pSV2neo (Southern and Berg 1982). Stable transfected clones were isolated in G418 containing medium. The clone 10 was selected after analysis of the surface expression level of the murine leptin receptor by FACS, using the the biotinylated anti-mouse leptin receptor antibody BAF497 from R&D and streptavidin-APC (BD bioscience)
HT1080 cells were cotransfected with p6-16-RL, a plasmid encoding the Renilla luciferase (from pRL-null, Promega) controlled by the IFN-inducible 6-16 promoter (from p1.8gpt-5, Pellegrini et al. 1989), pBB3 (Bourachot et al. 1982) and salmon sperm DNA (Sigma). Stable transfected clones were isolated in HAT containing medium. The clone 4 was selected for a high level of renilla luciferase activity induction upon IFN induction.
The human pancreatic carcinoma BXPC3 (Tan et al., 1986; ATCC: CRL 1687) and breast cancer BT474 (Lasfargues et al., 1979; ATCC: HTB-20) cell lines were obtained from ATCC. The mouse BTG9A cells were described in Uze et al. (1990).
Measurement of the luciferase activities
IFN specific activities were measured by quantifying the luciferase activity induced in HL1 16 cells and on the HL1 16 clone 10 expressing the rmLR. The EC50 were calculated using nonlinear data regression with GraphPad Prism software.
Luciferase activities were determined on a Berthold centra LB960 luminometer using either the Firefly Luciferase Assay System or the Dual-Luciferase Reporter Assay System from Promega after 6hr IFN stimulation.
Quantitative RT-PCR
The expression of the interferon inducible gene 6-16 was quantified by RT-PCR relatively to GAPDH or β-actin. Cells were treated with targeted or control IFN for 4 hr. Total RNA was purified with RNeasy columns (Qiagen). Reverse transcriptions were primed with random primers and performed using Moloney murine leukemia virus reverse transcriptase (Invitrogen). Quantitative real-time PCR (qRT-PCR) was performed using a Light Cycler as described (Coccia et al. 2004).
For Her2, the transfection culture medium was assayed on murine BTG9A and BTG9A cells expressing human Her2 for expression of the OASL2 gene relatively to the expression of the β actin gene by quantitative RT-PCR using a Light Cycler (Roche) and the following primers: OASL2 forward: CAC-GAC-TGT-AGG-CCC-CAG-CGA; OASL2 reverse: AGC-AGC-TGT- CTC-TCC-CCT-CCG; 3actin forward: AGA-GGG-AAA-TCG-TGC-GTG-AC; 3actin reverse: CAA-TAG-TGA-TGA-CCT-GGC-CGT. In a similar way the ISG expression in Her2 targeted cells was measured using the same 3actin primers and the following primer ISG15 primers: ISG15 forward: GAG-CTA-GAG-CCT-GCA-GCA-AT; ISG15 reverse: TTC-TGG-GCA-ATC- TGC-TTC-TT.
Antiviral Assay
The antiviral assay was performed using the EMC virus and scoring the virus replication dependent cytopathic effect as described in Stewart (1979)
Measurement of Her2 phosphorylation
BTG9A cells expressing human Her2 were treated with 200 pM to 2 nM of 1 R59B-IFNA2- Q124R for 10 to 30 min. Cells were lysed in RIPA, and analysed by western blot on an Odyssey Fc (Licor Bioscience) after 7% SDS-PAGE (40 μg lane). Phopho-Her2 was detected with the anti Her2 Y-P 1248 (Upstate #06-229) and the Goat anti rabbit secondary antibody IRDye 680 (Licor Bioscience #926-32221 ).
Measurement of STAT1 phosphorylation
STAT1 phosporylated on Y701 were detected by FACS using the STAT1 -PY701 (PE) (Beckton Dickinston #612564) and the manufacturer instruction for the PhosFlow technology.
Targeted leptin constructs
The sequence of the targeted leptin constructs is given in Figure 17. The L86 which is indicated is the amino acid that is mutated either to S or N.
Example 1 : The nanobody-interferon fusion proteins
Figure 1 shows a schematic representation of the nanobody-IFN fusion proteins constructed with either IFNa2 wild type or the L153A and R149A mutants. Example 2: IFN activity of the nanobody-IFN fusion proteins is targeted toward murine leptin receptor expressing cells
The three nanobody fusion proteins with IFNa2 WT, IFNa2 L153A or R149A were assayed on both HL1 16 and HL1 16-mLR-clone 10 cells which express the murine leptin receptor. The
IFNa2 alone was also assayed in this assay system in order to check that the two cell clones do not differ in their IFN responsiveness. Indeed both HL1 16 and HL1 16-mLR-clone 10 cells are equally sensitive to this IFN (Fig 2A and 2C, black symbols). The IFN activity of the three nanobody-IFN fusion proteins is however dramatically increased in cells expressing the leptin receptor compared to parental HL1 16 cells (compare Fig. 2A with Fig. 2C and Fig. 2B with Fig. 2D).
We estimated that cells expressing the leptin receptor are 10, 100 and 1000-fold more sensitive than parental HL1 16 cells to the nanobody-IFN WT, L153A and R149A, respectively. Since the affinities for I FNAR2 of the I FN mutant L153A and R149A are 0.1 and 0.01 relatively to the WT, there is a correlation between the loss of activity caused by mutations in the IFNAR2 binding site and the targeting efficiency by the nanobody.
In order to determine whether the IFN activity of the nanobody-IFN fusion proteins is delivered only on cells expressing the nanobody target or also on neighboring cells, the nanobody- IFNa2R149A was assayed on a coculture of HL1 16-ml_R-clone10 and HT1080-6-16 renilla luciferase clone4. Both cell types will express luciferase activity in response to IFN stimulation but cells expressing the target of the nanobody will display a firefly luciferase activity whereas cells devoid of leptin receptor will display a renilla luciferase activity. The dilution of the nanobody-IFNa2R149A protein was chosen at 1/30, a dilution which induces a maximal response in cells carrying the leptin receptor and a minimal response on cells devoid of the nanobody target (see Fig.2B and D, black curves). Fig. 3 shows clearly that the renilla luciferase activity is not induced upon stimulation of the co-culture with the nanobody- IFNa2R149A, indicating that the targeted IFN activity is delivered only on cells expressing the antigen recognized by the nanobody.
The efficacy of the targeting is further illustrated by comparing the activity of wild type and two types of mutant IFN (L153A and R149A) when added to HL1 16 expressing or not expressing the murine leptin receptor that is used for the targeting. The results clearly show that the activity of the mutants is higher when the construct is targeted, and that the effect of targeting for the mutant is bigger than for wild type. (Figure 4A and B)
In order to prove that the targeting was nanobody specific, HL1 16 cells expressing the rmLR were incubated for 6 hours with either the IFN-a2 (indicated as IFNA2) or the IFNA2-R149A fused to the nanobody 4-1 1 (Nanobody-IFNA2-R149A) at their respective EC50 concentration in the presence or absence (control) of a 100-fold molar excess of free 4-1 1 nanobody. Cells were lysed and the IFN-induced luciferase activities were measured. As shown in Figure 5, the non-targeted IFN is not inhibited by the free nanobody, while the targeted construct is strongly inhibited, showing the specific effect of the targeting.
The targeting to the leptin receptor is independent of the epitope on the receptor: using the anti-leptin receptor nanobody 4-10 (Zabeau et al., 2012) which recognizes a different domain on the receptor than the nanobody 4-1 1 , a similar activation can be obtained using a targeted mutant IFN (Figure 6). Example 3: The IFN activity of the nanobody-IFN fusion proteins on cells expressing the leptin receptor is mediated by both IFN receptor chains
In order to determine whether the IFN activity of the nanobody-IFN fusion proteins needs the activation of the IFN receptor, HL1 16 cells expressing the murine leptin receptor were pretreated with neutralizing antibodies against IFNAR1 or IFNAR2, and then stimulated with the nanobody-IFNA2-R149A fusion protein. The activity of the IFN-induced luciferase was measured. The figure 7 shows that both anti IFNAR1 and anti IFNAR2 neutralizing antibodies inhibit the IFN activity of the nanobody-IFNA2-R149A. Example 4: Target-specific induction of antiviral activity by 4-11 -IFNA2-R149A in cells expressing the murine leptin receptor
Antiviral activity is an integrated part of the IFN response, implying the expression of several genes. Therefore, the antiviral activity on rmLR expressing cell was controlled, after targeting the mutant R149A IFN using the anti-leptin receptor antibody 4-1 1. The results are summarized in Figure 8. The activity was measured as the cytopathic effect on HL1 16 cells, with or without leptin receptor expression. The specific antiviral activity of the 4-1 1 -IFNA2- R149A nanobody-IFN fusion protein is 716-fold higher when assayed on leptin receptor expressing cells compared to HL1 16 cells. Example 5: Targeting of IFN activity on Her2 expressing cells
In order to demonstrate that the concept is not restricted to cytokine receptor targeting, we generated similar fusion protein using the nanobody 2R5A against Her2 (Vaneycken et al., 201 1 ) and the mutant IFN alpha2 R149A (2R5A-IFNA2-R149A). This molecule was assayed on BXPC3 (Pancreatic cancer, from ATCC) and BT474 (Breast cancer, from ATCC) cell lines and compared with the activity of IFN-a2 (IFNA2) for the induction of the 6-16 IFN-inducible gene as determined relatively to GAPDH by quantitative RT-PCR. The BXPC3 and BT474 cells lines differ by their number of Her2 molecules expressed at their surface (10.9 x 103 and 478 x 103, respectively as reported by Gaborit et al. (201 1 )).
Figure 9 shows the EC50 determination of IFNA2 activity (Panel A) and 2R5A-IFNA2-R149A activity (Panel B) for the induction of the IFN-inducible gene 6-16 on BXPC3 and BT474 cell lines. Panel A shows that BXPC3 and BT474 cell lines exhibit the same sensitivity to IFN-a2.
Panel B shows that the 2R5A-IFNA2-R149A nanobody-IFN fusion protein is much more potent on the BT474 cell line which expresses 40-fold more Her2 molecule than BXPC3.
In conclusion, the concept which consists to target type I IFN activity on cells expressing a specific cell surface antigen, as shown on human cells expressing the mouse leptin receptor, can be extended to untransfected human cells expressing another cell surface molecule from a different structural family, at a level naturally found in several types of breast carcinoma.
Example 6: Targeting of mutant IFNA2-Q149R to mouse cells expressing human Her2 Mutant human IFNA2 Q149R was targeted to murine cells, expressing the human Her2, using the nanobody 1 R59B in the 1 R59B-IFNA2-Q124R. The IFNA2 Q124R has a high affinity for the murine IFNAR1 chain and a low activity for the murine IFNAR2 chain (Weber et al., 1987). The induction by IFN was measured as expression of the OASL2 messenger RNA, by RT- QPCR. The results are shown in Figure 10. There is clearly a targeting-specific induction in the Her2 expressing cells, whereas there is no significant expression detected in untransfected BTG9A cells.
Similar results were obtained when the Her2 specific ScFv against Her2 was used to target the mutant IFN Q124R. In this case, the IFN induction was measured using the ISG15 messenger RNA expression. The results are shown in Figure 1 1. Again, a specific induction of ISG15 is seen in the cells expressing Her2, while there is little effect of the mutant IFN on the cells that do not express Her2.
Example 7: The construct 1 R59B-IFNA2-Q124R does not activate the phosphorylation of Her2
To check whether targeting of Her2 is resulting in a Her2 activation, Her2 phosphorylation was controlled in targeted cells. The results are shown in Figure 12, clearly demonstrating that no phosphorylated Her2 could be detected in 1 R59B-IFNA2-Q124R targeted cells, irrespective of the concentration or time of treatment. Example 8: The anti PD-L2 Nb122-IFNA2-Q124R construct activity is targeted on mouse primary cells expressing PD-L2
Cells from a mouse peritoneal cavity were isolated and treated in vitro with Nb122-IFNA2- Q124R or natural mlFNa/β for 30 min. Cells were, fixed, permeabilized, labelled with antibodies against PD-L2 (APC) (BD #560086) and STAT1 -PY701 (PE) (BD #612564) and analysed by FACS.
The PD-L2 positive cell population represents 20% of the total cell population present in the mouse peritoneal cavity.
The results are shown in Figure 13. It is clear from this figure that in untreated cells, or in non- targeted, murine IFN treated cells the peaks of STAT1 -P for PD-L2 expressing and non- expressing cells coincide. Moreover, a clear induction in STAT1 -P can be seen by murine IFN treatment. Treatment with the targeted mutant IFN however results in a specific shift in the STAT1 -P only for the PD-L2 expressing cells. A same result is obtained if the IFN response of splenocytes is analysed in a similar experiment. The PD-L2 positive cell population represents 1 % of the total cell population present in mouse spleen, indicating that also a minor cell population can be targeted in an efficient way.
Example 9: In vivo injection of 122-IFNA2-Q124R construct induces an IFN response only in PD-L2 -expressing cells
Mice were injected (IP or IV) with either PBS, Nb122-IFNA2-Q124R or a control Nb (against GFP) fused to IFNA2-Q124R. 30 min post injection, mice were killed, cells from the peritoneal cavity were recovered by washing the peritoneal cavity with PBS, fixed (PhosLow Fix buffer I BD # 557870), permeabilized (PhosFlow Perm buffer III, BD #558050), labelled with Abs against PD-L2 (APC) (BD #560086) and STAT1 -PY701 (PE) (BD #612564) and analysed by FACS. The results are shown in Figure 14. STAT1 -P coincides in PD-L2 positive and negative cells treated with either PBS or control nanobody. However, a clear induction in STAT1 -P (only in the PDL2 positive cell population) can be seen when the mice are injected with the targeted mutant IFN.
As a control, STAT1 -P was checked in mice, iv injected with different doses of natural mouse IFN (10 000, 100 000 or 1 000 000 units), and no difference in STAT1 -P could be detected between the PD-L2 positive and PD-L2 negative cells.
Figure 15 shows a similar dose response curve after iv injection of the Nb122-IFNA2-Q124R construct. A shift in STAT1 -P in the PD-L2 expressing cells can be noticed even at the lowest dose of 64ng.
Example 10: Targeting of mutant leptin to the leptin receptor, using a truncated TNFa receptor
Ba/F3 cells are growth-dependent on IL-3. After transfection with the rmLR, Ba/F3 cells also proliferate with leptin. Leptin mutants with reduced affinity for their receptor are less potent in inducing and sustaining proliferation of Ba/F3-ml_R cells. Leptin mutant L86S has a moderate and mutant L86N has a strong reduction in affinity and hence a moderate and strong reduced capacity to induce proliferation respectively.
Additional transfection of Ba/F3-mLR cells with the human TNFa Receptor 1 (hTNFRI ) lacking its intracellular domain introduces a non-functional receptor which can function as a membrane bound extracellular marker.
Chimeric proteins consisting of leptin and a nanobody against human TNFR1 (here nb96) will bind to cells carrying the rmLR and to cells carrying the hTNFRI . Chimeric proteins with leptin mutants L86S and L86N have reduced affinity for the LR but retain their affinity for the hTNFRL
Chimeric proteins were produced by transient transfection of Hek293T cells with expression plasmids. Supernatant was 0.45 μηη filtered and serially diluted in 96-well plates for the assay. A serial dilution of purified recombinant leptin was used as a reference. 3000 to 10000 cells were plated per well and proliferation was measured by staining with XTT four or five days later. OD was measured at 450 nm. The results are shown in Figure 16, for two experiments using a different leptin construct (see Figure 17). For both constructs, a hTNFR depending growth stimulation can be seen for the mutant constructs, whereas the hTNFR expression does not affect the growth of the cells treated with wt (non-targeted) leptin. It is clear from these results that the targeting can compensate for the negative effect of the mutation.
REFERENCES
- Abraham, A.K., Ramanathan, M. Weinstock-Guttman, B. and Mager, D.E. (2009).
Biochemical pharmacology 77, 1757-1762.
- Benoit, P., Maguire, D., Plavec, I., Kocher, H., Tovey, M. and Meyer, F. (1993). J.
Immunol., 150,707-716
- Blake, A.W., McCartney, L, Flint,J., Bolam, D.N., Boraston, A.B., Gilbert, H.J., and Knox, J. P. (2006). J. Biol. Chem., 281 , 29321 -29329.
Bourachot, B., Jouanneau, J., Giri, I., Katinka, M., Cereghini, S., and Yaniv, M. (1982). EMBO J., 1 , 895-900.
- Brecht, A., Gauglitz, G. And Polster, J. (1993). Biosens. Bioelectron.. 8, 387-392.
Coccia, E.M., Severa, M, Giacomini, E., Monneron, D., Remoli, M-E., Julkunen, I., Cella, M., Lande, R. and Uze, G. (2004). Eur. J. Immunol. 34. 796-805.
- Conklin, D. (2004). J. Computiational Biol. 1 1 , 1 189-1200.
- Dimitrov, D.S. (2009). mAbs 1 , 26-28.
Eyckerman, S., Waelput, W., Verhee, A., Broekaert, D., Vendekerckhove, J., and Tavernier, J. (1999). Eur. Cytok. Netw. 10, 549-559.
Gaborit, N., Labouret, C, Vallaghe, J., Peyrusson, F., Bascoul-Mollevi, C. ? Crapez, E., Azria, D., Chardes, T., Poul, M.A., Mathis, G., Bazin, H., Pelegrin, A. (201 1 ). J. Biol. Chem. 286, 1 1337-1 1345.
- Ortiz-Sanchez, E., Helguera, G., Daniels, T.R. and Penichet, M.L. (2008). Expert Opin.
Biol. Ther., 8, 609-632.
- Pellegrini, S., John, J., Shearer, M., Kerr, I.M. and Stark, G.R. (1989). Mol. Cell. Biol. 9, 4605-4012.
- Piehler, J., Roisman, L.C. and Schreiber, G. (2000). J. Biol. Chem. 275, 40425-40433.
- Lasfargues, E.Y., Coutino, W.G. and Dion, A.S. (1979). In Vitro, 15, 723-729.
- Nygren, P.A. (2008). FEBS J., 275, 2668-2676.
Roisman, L.C, Piehler, J., Trosset, J.Y., Scheraga, H.A. and Schreiber, G. (2001 ). Proc. Natl. Acad. Sci. YSA, 98, 13231 -13236.
- Rossi, E.A., Goldenberg, D.M., Cardillo, T.M., Stein, R. And Chang, C.H. (2009) Blood, 1 14, 3964-3871 .
- Southern, P.J. and Berg, P. (1982) J. Mol. Appl. Genet. 1 , 327-341. Skerra, A. (2008).
FEBS J., 275, 2677-2683.
Stewart II W.E. The interferon system. Springer-Verlag, Wien, New York. 1979.
- Stumpp, M.T., Binz, H.K. and Amstutz, P (2008). Drug Discov. Today 13, 695-701. - Takabe, Y., Seiki, M., Fujisawa, J., Hoy, P., Yokata, K. and Arai, N. (1988). Mol. Cell. Biol. 8, 466-472.
- Tan, M.H., Nowak, N.J., Loor, R., Ochi, H., Sandberg, A.A., Lopez, C, Pickren, J.W., Berjian, R., Douglass, H.O. jr. and Chu, T.M. (1986) Cancer Invest. 4, 15-23.
Tramontano, A., Bianchi, E., Venturini, S., Martin, F., Pessi, A and Sollazzo, M. (1994). J. Mol. Recognition 7, 9-24.
- Uze et al. Cell 60, 225-234 (1990).
- Uze, G., Di Marco, S., Mouchel-Veilh, E., Monneron, D., Bandu, M.T., Horisberger, M.A., Dorques, A., Lutfalla, G., and Mogensen, K.E. (1994). J. Mol. Biol. 243, 245-257.
Vaneycken, I., Devoogdt, N., Van Gassen, N., Vincke, C, Xavier, C, Wernery, U., Muyldermans, S., Lahoutte, T. And Caveliers, V. (201 1 ). FASEB J. 25, 2433-2446.
- Weber et al. (1987) EMBO J. 6, 591 -598.
- Worn et al., FEBS Lett. 427, 357-361 (1998)
- Zabeau, L., Verhee, A., Catteeuw, D., Faes, L., Seeuws, S., Decruy, T., Elewaut, D., Peelman, F. And Tavernier, J. (2012). Biochem. J. . 441 , 425-434.

Claims

1 . A targeting construct, comprising a modified a-helical bundle cytokine characterized by a reduced affinity for the a-helical bundle cytokine receptor, and a targeting moiety.
2. The targeting construct, according to claim 1 , wherein said modified α-helical bundle
cytokine is a mutant a-helical bundle cytokine.
3. The targeting construct according to claim 1 or 2, wherein said targeting moiety is
targeting to a marker expressed on a a-helical bundle cytokine receptor expressing cell.
4. The targeting construct, according to claim 2, wherein said mutant α-helical bundle
cytokine is a mutant interferon.
5. The targeting construct, according to claim 4, wherein said targeting moiety is targeting to a marker expressed on an interferon receptor expressing cell.
6. The targeting construct according to claim 5, wherein said interferon receptor is IFNAR2.
7. The targeting construct according to any of the preceding claims, wherein said targeting moiety is directed to a tissue specific marker
8. The targeting construct according to any of the claims 1 -6, wherein said targeting moiety is directed to a marker selected from the group consisting of Her2, DC-STAMP and CD20.
9. The targeting construct according to any of the claims 1 -6, wherein said targeting moiety is directed to a cell surface marker specific for viral infected cells
10. The targeting construct according to any of the previous claims, wherein said targeting moiety is an antibody.
1 1 . The targeting construct according to claim 10, wherein said antibody is a nanobody.
12. The targeting construct according to claim 4, wherein the mutant interferon is a mutant interferon alpha 2.
13. The targeting construct according to claim 12, wherein said mutant interferon alpha 2 is mutated in one or more amino acids of the region 144-154.
14. The targeting construct according to claim 12 , wherein said mutant interferon alpha 2 is selected from the group consisting of IFNa2 L153A, IFNa2 R149A and IFNa2 M148A.
15. A targeting construct according to any of the claims 1 -14 for use as a medicament.
16. The targeting construct according to any of the claims 1 -14 for use in treatment of cancer.
17. The targeting construct according to any of the claims 1 -14 for use in treatment of a viral disease.
18. The targeting construct according to any of the claims 1 -14 for use in treatment of
diseases involving bone degradation.
19. A pharmaceutical composition, comprising a targeting construct according to any of the claims 1 -14, and a suitable excipient.
PCT/EP2013/050787 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines WO2013107791A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
ES13701014.6T ES2694180T3 (en) 2012-01-20 2013-01-17 Alpha-helical directed cytokines directed mutants
EP13701014.6A EP2804877B1 (en) 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines
AU2013211059A AU2013211059B8 (en) 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines
US14/372,730 US9492562B2 (en) 2012-01-20 2013-01-17 Targeted human-interferon fusion proteins
BR112014017876-3A BR112014017876B1 (en) 2012-01-20 2013-01-17 DIRECTORY CONSTRUCT COMPRISING MUTANT HUMAN INTERFERON ALPHA 2 AND PHARMACEUTICAL COMPOSITION CONTAINING THE SAME
CA2861927A CA2861927C (en) 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines
JP2014552617A JP6416628B2 (en) 2012-01-20 2013-01-17 Targeted variant alpha helix bundle cytokine
KR1020147023041A KR102050119B1 (en) 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines
CN201380015170.0A CN104245734B (en) 2012-01-20 2013-01-17 Target mutant alpha helical bundle cell factor
HK15105433.7A HK1205134A1 (en) 2012-01-20 2015-06-08 Targeted mutant alpha helical bundle cytokines
US15/278,854 US9878014B2 (en) 2012-01-20 2016-09-28 Targeted human-interferon fusion proteins
US15/717,205 US10034919B2 (en) 2012-01-20 2017-09-27 Targeted human-interferon fusion proteins
US16/020,447 US10946070B2 (en) 2012-01-20 2018-06-27 Targeted and mutated human-interferon fusion proteins
US17/171,426 US20210162010A1 (en) 2012-01-20 2021-02-09 Targeted human-interferon fusion proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12305075.9 2012-01-20
EP12305075 2012-01-20

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/372,730 A-371-Of-International US9492562B2 (en) 2012-01-20 2013-01-17 Targeted human-interferon fusion proteins
US15/278,854 Continuation US9878014B2 (en) 2012-01-20 2016-09-28 Targeted human-interferon fusion proteins

Publications (1)

Publication Number Publication Date
WO2013107791A1 true WO2013107791A1 (en) 2013-07-25

Family

ID=47603651

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/050787 WO2013107791A1 (en) 2012-01-20 2013-01-17 Targeted mutant alpha-helical bundle cytokines

Country Status (11)

Country Link
US (5) US9492562B2 (en)
EP (1) EP2804877B1 (en)
JP (1) JP6416628B2 (en)
KR (1) KR102050119B1 (en)
CN (1) CN104245734B (en)
AU (1) AU2013211059B8 (en)
BR (1) BR112014017876B1 (en)
CA (1) CA2861927C (en)
ES (1) ES2694180T3 (en)
HK (1) HK1205134A1 (en)
WO (1) WO2013107791A1 (en)

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8980267B2 (en) 2012-03-03 2015-03-17 Immungene Inc Engineered antibody-interferon mutant fusion molecules
WO2015007536A3 (en) * 2013-07-18 2015-03-19 Vib Vzw Fusokines involving cytokines with strongly reduced receptor binding affinities
US9492562B2 (en) 2012-01-20 2016-11-15 Vib Vzw Targeted human-interferon fusion proteins
US9611322B2 (en) 2011-10-28 2017-04-04 Teva Pharmaceuticals Australia Pty Ltd Fusions of antibodies to CD38 and attenuated interferon alpha
WO2017077382A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
WO2017134302A2 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Targeted therapeutic agents and uses thereof
US9732135B2 (en) 2013-07-19 2017-08-15 Vib Vzw Targeting of human interferon antagonists
WO2017153402A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
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
US9914759B2 (en) 2013-07-19 2018-03-13 Vib Vzw Targeted modified TNF family members
US9932409B2 (en) 2013-07-19 2018-04-03 Vib Vzw Targeted modified IL-1 family members
WO2018077893A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
US9963515B2 (en) 2013-04-29 2018-05-08 Teva Pharmaceuticals Australia Pty Ltd. Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
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
US10232041B2 (en) 2014-05-01 2019-03-19 Teva Pharmaceuticals Pty Ltd Combination of lenalidomide and polypeptide construct, and uses thereof
US10544199B2 (en) 2014-10-29 2020-01-28 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants
CN112074267A (en) * 2018-02-05 2020-12-11 奥里尼斯生物科学公司股份有限公司 Fibroblast binding agents and uses thereof
WO2021009332A1 (en) 2019-07-18 2021-01-21 Enyo Pharma Method for decreasing adverse-effects of interferon
US11059876B2 (en) 2018-02-28 2021-07-13 Pfizer Inc. IL-15 variants and uses thereof
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
EP3783035A4 (en) * 2018-04-16 2022-07-20 Institute of Biophysics Chinese Academy of Sciences Fusion protein of ifn and anti-pd-l1 antibody and application thereof
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
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, 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
WO2022258662A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp46, a cytokine receptor, a tumour antigen and cd16a
US11566072B2 (en) 2017-08-09 2023-01-31 Orionis Biosciences, Inc. CD8 binding agents
US11584794B2 (en) 2016-10-14 2023-02-21 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Ralpha Fc-fusion proteins and immune checkpoint antibody fragments
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
US11661455B2 (en) 2016-02-05 2023-05-30 Orionis Biosciences BV Chimeric protein comprising an interferon alpha 2mutant and a PD-L1 binding moiety
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
US11932675B2 (en) 2019-10-11 2024-03-19 Genentech, Inc. PD-1 targeted IL-15/IL-15Rα Fc fusion proteins with improved properties

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201504859D0 (en) 2015-03-23 2015-05-06 Vib Vzw Viral particle based small molecule-protein interaction trap
EP3946409A4 (en) * 2019-03-28 2023-06-21 Orionis Biosciences, Inc. Fms-like tyrosine kinase 3 ligand (flt3l)-based chimeric proteins

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006053883A1 (en) 2004-11-18 2006-05-26 Vib Vzw Fibronectin iii domain as leptin receptor antagonists
WO2008124086A2 (en) 2007-04-05 2008-10-16 President And Fellows Of Harvard College Chimeric activators: quantitatively designed protein therapeutics and uses thereof
WO2009039409A1 (en) 2007-09-21 2009-03-26 The Regents Of The University Of Californina Targeted interferon demonstrates potent apoptotic and anti-tumor activities
WO2010066740A1 (en) 2008-12-08 2010-06-17 Complix Nv Single-chain antiparallel coiled coil proteins
WO2013059885A2 (en) 2011-10-28 2013-05-02 Cephalon Australia Pty Ltd Polypeptide constructs and uses thereof

Family Cites Families (19)

* 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
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
IL108584A (en) * 1994-02-07 2008-03-20 Yeda Res & Dev Cloning of interferon -alpha/beta binding protein
US5914254A (en) 1993-08-02 1999-06-22 Celtrix Pharmaceuticals, Inc. Expression of fusion polypeptides transported out of the cytoplasm without leader sequences
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
DE10161767T1 (en) * 2002-07-03 2018-06-07 Honjo Tasuku Immunopotentiating compositions containing an anti-PD-L1 antibody
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
CA2693326A1 (en) 2006-08-02 2008-02-07 Mcgill University Gm-csf and il-15 fusokines and methods for modulation of the immune response
WO2008071447A2 (en) * 2006-12-15 2008-06-19 Ablynx N.V. Amino acid sequences that modulate the interaction between cells of the immune system
WO2009003145A1 (en) 2007-06-26 2008-12-31 University Of Miami Antibody-endostatin fusion protein and its variants
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
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
US11661455B2 (en) * 2016-02-05 2023-05-30 Orionis Biosciences BV Chimeric protein comprising an interferon alpha 2mutant and a PD-L1 binding moiety
WO2018144999A1 (en) * 2017-02-06 2018-08-09 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006053883A1 (en) 2004-11-18 2006-05-26 Vib Vzw Fibronectin iii domain as leptin receptor antagonists
WO2008124086A2 (en) 2007-04-05 2008-10-16 President And Fellows Of Harvard College Chimeric activators: quantitatively designed protein therapeutics and uses thereof
WO2009039409A1 (en) 2007-09-21 2009-03-26 The Regents Of The University Of Californina Targeted interferon demonstrates potent apoptotic and anti-tumor activities
WO2010066740A1 (en) 2008-12-08 2010-06-17 Complix Nv Single-chain antiparallel coiled coil proteins
WO2013059885A2 (en) 2011-10-28 2013-05-02 Cephalon Australia Pty Ltd Polypeptide constructs and uses thereof

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
ABRAHAM, A.K.; RAMANATHAN, M.; WEINSTOCK-GUTTMAN, B.; MAGER, D.E., BIOCHEMICAL PHARMACOLOGY, vol. 77, 2009, pages 1757 - 1762
BENOIT ET AL., J. IMMUNOL., vol. 150, 1993, pages 707 - 716
BENOIT, P.; MAGUIRE, D.; PLAVEC, I.; KOCHER, H.; TOVEY, M.; MEYER, F., J. IMMUNOL., vol. 150, 1993, pages 707 - 716
BLAKE, A.W.; MCCARTNEY, L.; FLINT,J.; BOLAM, D.N.; BORASTON, A.B.; GILBERT, H.J.; KNOX, J.P., J. BIOL. CHEM., vol. 281, 2006, pages 29321 - 29329
BOURACHOT, B.; JOUANNEAU, J.; GIRI, I.; KATINKA, M.; CEREGHINI, S.; YANIV, M., EMBO J., vol. 1, 1982, pages 895 - 900
BRECHT, A.; GAUGLITZ, G.; POLSTER, J., BIOSENS. BIOELECTRON., vol. 8, 1993, pages 387 - 392
COCCIA, E.M.; SEVERA, M; GIACOMINI, E.; MONNERON, D.; REMOLI, M-E.; JULKUNEN, I.; CELLA, M.; LANDE, R.; UZE, G., EUR. J. IMMUNOL., vol. 34, 2004, pages 796 - 805
CONKLIN, D., J. COMPUTIATIONAL BIOL., vol. 11, 2004, pages 1189 - 1200
COULSTOCK EDWARD ET AL: "Liver-targeting of interferon-alpha with tissue-specific domain antibodies.", PLOS ONE, vol. 8, no. 2, E57263, February 2013 (2013-02-01), pages 1 - 11, XP002697904, ISSN: 1932-6203 *
DIMITROV, D.S., MABS, vol. 1, 2009, pages 26 - 28
EYCKERMAN, S.; WAELPUT, W.; VERHEE, A.; BROEKAERT, D.; VENDEKERCKHOVE, J.; TAVERNIER, J., EUR. CYTOK. NETW., vol. 10, 1999, pages 549 - 559
GABORIT, N.; LABOURET, C.; VALLAGHE, J.; PEYRUSSON, F.; BASCOUL-MOLLEVI, C. ?; CRAPEZ, E.; AZRIA, D.; CHARDÈS, T.; POUL, M.A.; MA, J. BIOL. CHEM., vol. 286, 2011, pages 11337 - 11345
LASFARGUES, E.Y.; COUTINO, W.G.; DION, A.S., IN VITRO, vol. 15, 1979, pages 723 - 729
MASCI P ET AL: "NEW AND MOFIFIED INTERFERON ALFAS: PRECLINICAL AND CLINICAL DATA", CURRENT ONCOLOGY REPORTS, vol. 5, no. 2, 1 March 2003 (2003-03-01), CURRENT SCIENCE, GB, pages 108 - 113, XP008037765, ISSN: 1523-3790 *
NYGREN, P.A., FEBS J., vol. 275, 2008, pages 2668 - 2676
ORTIZ-SANCHEZ, E.; HELGUERA, G.; DANIELS, T.R.; PENICHET, M.L., EXPERT OPIN. BIOL. THER., vol. 8, 2008, pages 609 - 632
PELLEGRINI, S.; JOHN, J.; SHEARER, M.; KERR, I.M.; STARK, G.R., MOL. CELL. BIOL., vol. 9, 1989, pages 4605 - 4012
PIEHLER, J.; ROISMAN, L.C.; SCHREIBER, G., J. BIOL. CHEM., vol. 275, 2000, pages 40425 - 40433
ROISMAN L C ET AL: "Structure of the interferon-receptor complex determined by distance constraints from double-mutant cycles and flexible docking", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 98, no. 23, 6 November 2001 (2001-11-06), US, pages 13231 - 13236, XP002242615, ISSN: 0027-8424, DOI: 10.1073/PNAS.221290398 *
ROISMAN, L.C.; PIEHLER, J.; TROSSET, J.Y.; SCHERAGA, H.A.; SCHREIBER, G., PROC. NATL. ACAD. SCI. YSA, vol. 98, 2001, pages 13231 - 13236
ROSSI, E.A.; GOLDENBERG, D.M.; CARDILLO, T.M.; STEIN, R.; CHANG, C.H., BLOOD, vol. 114, 2009, pages 3964 - 3871
SKERRA, A., FEBS J., vol. 275, 2008, pages 2677 - 2683
SOUTHERN, P.J.; BERG, P., J. MOL. APPL. GENET., vol. 1, 1982, pages 327 - 341
STEWART II W.E.: "The interferon system", 1979, SPRINGER-VERLAG
STUMPP, M.T.; BINZ, H.K.; AMSTUTZ, P, DRUG DISCOV. TODAY, vol. 13, 2008, pages 695 - 701
TAKABE, Y.; SEIKI, M.; FUJISAWA, J.; HOY, P.; YOKATA, K.; ARAI, N., MOL. CELL. BIOL., vol. 8, 1988, pages 466 - 472
TAN, M.H.; NOWAK, N.J.; LOOR, R.; OCHI, H.; SANDBERG, A.A.; LOPEZ, C.; PICKREN, J.W.; BERJIAN, R.; DOUGLASS, H.O. JR.; CHU, T.M., CANCER INVEST., vol. 4, 1986, pages 15 - 23
TRAMONTANO, A.; BIANCHI, E.; VENTURINI, S.; MARTIN, F.; PESSI, A; SOLLAZZO, M., J. MOL. RECOGNITION, vol. 7, 1994, pages 9 - 24
UZE ET AL., CELL, vol. 60, 1990, pages 225 - 234
UZE, G.; DI MARCO, S.; MOUCHEL-VEILH, E.; MONNERON, D.; BANDU, M.T.; HORISBERGER, M.A.; DORQUES, A.; LUTFALLA, G.; MOGENSEN, K.E., J. MOL. BIOL., vol. 243, 1994, pages 245 - 257
VANEYCKEN, I.; DEVOOGDT, N.; VAN GASSEN, N.; VINCKE, C.; XAVIER, C.; WERNERY, U.; MUYLDERMANS, S.; LAHOUTTE, T.; CAVELIERS, V., FASEB J, vol. 25, 2011, pages 2433 - 2446
WEBER ET AL., EMBO J., vol. 6, 1987, pages 591 - 598
WORN ET AL., FEBS LETT., vol. 427, 1998, pages 357 - 361
ZABEAU, L.; VERHEE, A.; CATTEEUW, D.; FAES, L.; SEEUWS, S.; DECRUY, T.; ELEWAUT, D.; PEELMAN, F.; TAVERNIER, J., BIOCHEM. J., vol. 441, 2012, pages 425 - 434

Cited By (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9611322B2 (en) 2011-10-28 2017-04-04 Teva Pharmaceuticals Australia Pty Ltd Fusions of antibodies to CD38 and attenuated interferon alpha
US10981986B2 (en) 2011-10-28 2021-04-20 Teva Pharmaceuticals Australia Pty Ltd Fusions of antibodies to CD38 and attenuated interferon alpha
US10946070B2 (en) 2012-01-20 2021-03-16 Vib Vzw Targeted and mutated human-interferon fusion proteins
US9878014B2 (en) 2012-01-20 2018-01-30 Vib Vzw Targeted human-interferon fusion proteins
US9492562B2 (en) 2012-01-20 2016-11-15 Vib Vzw Targeted human-interferon fusion proteins
US10034919B2 (en) 2012-01-20 2018-07-31 Vib Vzw Targeted human-interferon fusion proteins
US10259854B2 (en) 2012-03-03 2019-04-16 Immungene Inc Engineered antibody-interferon mutant fusion molecules
US9534057B2 (en) 2012-03-03 2017-01-03 Immungene Inc Methods of treating tumors with engineered antibody-interferon mutant fusion molecules
US8980267B2 (en) 2012-03-03 2015-03-17 Immungene Inc Engineered antibody-interferon mutant fusion molecules
US9963515B2 (en) 2013-04-29 2018-05-08 Teva Pharmaceuticals Australia Pty Ltd. Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
EP3299466A1 (en) * 2013-07-18 2018-03-28 VIB vzw Fusokines involving cytokines with strongly reduced receptor binding affinities
US10640542B2 (en) 2013-07-18 2020-05-05 Vib Vzw Fusokines involving cytokines with strongly reduced receptor binding affinities
US11358997B2 (en) 2013-07-18 2022-06-14 Vib Vzw Fusokines involving cytokines with strongly reduced receptor binding affinities
WO2015007536A3 (en) * 2013-07-18 2015-03-19 Vib Vzw Fusokines involving cytokines with strongly reduced receptor binding affinities
US11396532B2 (en) 2013-07-19 2022-07-26 Vib Vzw Targeted modified TNF family members
US9932409B2 (en) 2013-07-19 2018-04-03 Vib Vzw Targeted modified IL-1 family members
US9914759B2 (en) 2013-07-19 2018-03-13 Vib Vzw Targeted modified TNF family members
US10407480B2 (en) 2013-07-19 2019-09-10 Vib Vzw Targeted modified TNF family members
AU2014292355B2 (en) * 2013-07-19 2019-08-01 Centre Hospitalier Regional Universitaire De Montpellier Targeting of cytokine antagonists
US10035835B2 (en) 2013-07-19 2018-07-31 Vib Vzw Targeted modified TNF family members
US9732135B2 (en) 2013-07-19 2017-08-15 Vib Vzw Targeting of human interferon antagonists
US10787493B2 (en) 2013-07-19 2020-09-29 Vib Vzw Targeted modified TNF family members
US10947288B2 (en) 2013-07-19 2021-03-16 Vib Vzw Targeting of human interferon antagonists
US10072059B2 (en) 2013-07-19 2018-09-11 Vib Vzw Targeting of human interferon antagonists
US10232041B2 (en) 2014-05-01 2019-03-19 Teva Pharmaceuticals Pty Ltd Combination of lenalidomide and polypeptide construct, and uses thereof
US11253591B2 (en) 2014-05-01 2022-02-22 Teva Pharmaceuticals Australia Pty Ltd. Combination of lenalidomide and polypeptide construct, and uses thereof
US11319356B2 (en) 2014-10-29 2022-05-03 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants
US10544199B2 (en) 2014-10-29 2020-01-28 Teva Pharmaceuticals Australia Pty Ltd Interferon alpha 2B variants
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
US10988538B2 (en) 2016-02-05 2021-04-27 Orionis Biosciences BV Bispecific signaling agents and uses thereof
EP3909978A1 (en) 2016-02-05 2021-11-17 Orionis Biosciences BV Clec9a binding agents and use 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
EP4059957A1 (en) 2016-02-05 2022-09-21 Orionis Biosciences BV Bispecific signaling agents and uses thereof
WO2017134302A2 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Targeted therapeutic 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
US11236166B2 (en) 2016-02-05 2022-02-01 Orionis Biosciences BV CD8 binding agents
US11001631B2 (en) 2016-02-05 2021-05-11 Orionis Biosciences BV Clec9A binding agents
US11248057B2 (en) 2016-03-07 2022-02-15 Vib Vzw CD20 binding single domain antibodies
WO2017153402A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
EP4276114A2 (en) 2016-03-07 2023-11-15 Vib Vzw Cd20 binding single domain antibodies
EP4276114A3 (en) * 2016-03-07 2024-02-21 Vib Vzw Cd20 binding single domain antibodies
WO2017194782A3 (en) * 2016-05-13 2017-12-21 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
US11753463B2 (en) 2016-05-13 2023-09-12 Orionis Biosciences BV Therapeutic targeting of non-cellular structures
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
US11584794B2 (en) 2016-10-14 2023-02-21 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Ralpha Fc-fusion proteins and immune checkpoint antibody fragments
US11084859B2 (en) 2016-10-24 2021-08-10 Orionis Biosciences BV Targeted mutant interferon-gamma and uses thereof
WO2018077893A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
US11384154B2 (en) 2017-02-06 2022-07-12 Orionis Biosciences BV Targeted chimeric proteins and uses thereof
WO2018141964A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv 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
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
US11498966B2 (en) 2017-08-09 2022-11-15 Orionis Biosciences Inc. PD-1 and PD-L1 binding agents
US11566072B2 (en) 2017-08-09 2023-01-31 Orionis Biosciences, Inc. CD8 binding agents
US11896643B2 (en) 2018-02-05 2024-02-13 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
CN112074267A (en) * 2018-02-05 2020-12-11 奥里尼斯生物科学公司股份有限公司 Fibroblast binding agents and uses thereof
EP3749295A4 (en) * 2018-02-05 2022-04-27 Orionis Biosciences, Inc. Fibroblast binding agents and use thereof
US11059876B2 (en) 2018-02-28 2021-07-13 Pfizer Inc. IL-15 variants and uses thereof
EP3783035A4 (en) * 2018-04-16 2022-07-20 Institute of Biophysics Chinese Academy of Sciences Fusion protein of ifn and anti-pd-l1 antibody and application thereof
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
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
US11932675B2 (en) 2019-10-11 2024-03-19 Genentech, Inc. PD-1 targeted IL-15/IL-15Rα Fc fusion proteins with improved properties
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
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
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
CN104245734A (en) 2014-12-24
CA2861927A1 (en) 2013-07-25
KR102050119B1 (en) 2019-11-28
AU2013211059A8 (en) 2017-12-21
AU2013211059B8 (en) 2017-12-21
BR112014017876A2 (en) 2017-06-27
AU2013211059A1 (en) 2014-08-21
CA2861927C (en) 2021-01-26
US20170072020A1 (en) 2017-03-16
EP2804877A1 (en) 2014-11-26
HK1205134A1 (en) 2015-12-11
US20180333465A1 (en) 2018-11-22
AU2013211059B2 (en) 2017-11-02
US9878014B2 (en) 2018-01-30
BR112014017876B1 (en) 2023-04-11
US20180028616A1 (en) 2018-02-01
EP2804877B1 (en) 2018-08-22
ES2694180T3 (en) 2018-12-18
US20140348789A1 (en) 2014-11-27
CN104245734B (en) 2017-09-05
KR20140138129A (en) 2014-12-03
US10034919B2 (en) 2018-07-31
US10946070B2 (en) 2021-03-16
JP2015511219A (en) 2015-04-16
JP6416628B2 (en) 2018-10-31
US20210162010A1 (en) 2021-06-03
US9492562B2 (en) 2016-11-15

Similar Documents

Publication Publication Date Title
US20210162010A1 (en) Targeted human-interferon fusion proteins
US20220106392A1 (en) Albumin binding domain fusion proteins
CN112996805A (en) Multi-chain chimeric polypeptides and uses thereof
WO2009039409A1 (en) Targeted interferon demonstrates potent apoptotic and anti-tumor activities
EP2941439B1 (en) Human serum albumin binding compounds and fusion proteins thereof
US20220195051A1 (en) Fc-MODIFIED BIOLOGICALS FOR LOCAL DELIVERY TO COMPARTMENT, IN PARTICULAR TO THE CNS
WO2022057909A1 (en) Bispecific recombinant protein and use thereof
Class et al. Patent application title: TARGETED MUTANT ALPHA-HELICAL BUNDLE CYTOKINES Inventors: Jan Tavernier (Balegem, BE) Jan Tavernier (Balegem, BE) Gilles Uze (Montpellier, FR) Guillaume Cartron (Combaillaux, FR) Franciane Paul (Montpellier, FR) Jacob Piehler (Osnabruck, DE)
TW202248218A (en) Cytokine conjugates

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: 13701014

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14372730

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2861927

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2014552617

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20147023041

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2013701014

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013211059

Country of ref document: AU

Date of ref document: 20130117

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014017876

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014017876

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140721